Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 33
Filtrar
Mais filtros








Base de dados
Intervalo de ano de publicação
1.
J Virol ; 97(3): e0197722, 2023 03 30.
Artigo em Inglês | MEDLINE | ID: mdl-36815839

RESUMO

African swine fever (ASF) is an acute and severe infectious disease caused by the ASF virus (ASFV). The mortality rate of ASF in pigs can reach 100%, causing huge economic losses to the pig industry. Here, we found that ASFV protein MGF505-7R inhibited the beta interferon (IFN-ß)-mediated Janus-activated kinase-signal transducer and activation of transcription (JAK-STAT) signaling. Our results demonstrate that MGF505-7R inhibited interferon-stimulated gene factor 3 (ISGF3)-mediated IFN-stimulated response element (ISRE) promoter activity. Importantly, we observed that MGF505-7R inhibits ISGF3 heterotrimer formation by interacting with interferon regulatory factor 9 (IRF9) and inhibits the nuclear translocation of ISGF3. Moreover, to demonstrate the role of MGF505-7R in IFN-I signal transduction during ASFV infection, we constructed and evaluated ASFV-ΔMGF505-7R recombinant viruses. ASFV-ΔMGF505-7R restored STAT2 and STAT1 phosphorylation, alleviated the inhibition of ISGF3 nuclear translocation, and showed increased susceptibility to IFN-ß, unlike the parental GZ201801 strain. In conclusion, our study shows that ASFV protein MGF505-7R plays a key role in evading IFN-I-mediated innate immunity, revealing a new mode of evasion for ASFV. IMPORTANCE ASF, caused by ASFV, is currently prevalent in Eurasia, with mortality rates reaching 100% in pigs. At present, there are no safe or effective vaccines against ASFV. In this study, we found that the ASFV protein MGF505-7R hinders IFN-ß signaling by interacting with IRF9 and inhibiting the formation of ISGF3 heterotrimers. Of note, we demonstrated that MGF505-7R plays a role in the immune evasion of ASFV in infected hosts and that recombinant viruses alleviated the effect on type I IFN (IFN-I) signaling and exhibited increased susceptibility to IFN-ß. This study provides a theoretical basis for developing vaccines against ASFV using strains with MGF505-7R gene deletions.


Assuntos
Vírus da Febre Suína Africana , Febre Suína Africana , Interferon Tipo I , Fator Gênico 3 Estimulado por Interferon, Subunidade gama , Replicação Viral , Animais , Febre Suína Africana/imunologia , Febre Suína Africana/virologia , Vírus da Febre Suína Africana/genética , Vírus da Febre Suína Africana/imunologia , Imunidade Inata , Interferon Tipo I/imunologia , Fator Gênico 3 Estimulado por Interferon, Subunidade gama/imunologia , Transdução de Sinais , Suínos , Proteínas Virais/genética , Proteínas Virais/imunologia , Replicação Viral/fisiologia , Transporte Ativo do Núcleo Celular/genética , Evasão da Resposta Imune/genética
2.
J Immunol ; 207(2): 613-625, 2021 07 15.
Artigo em Inglês | MEDLINE | ID: mdl-34272232

RESUMO

Alphaherpesviruses are large dsDNA viruses with an ability to establish persistent infection in hosts, which rely partly on their ability to evade host innate immune responses, notably the type I IFN response. However, the relevant molecular mechanisms are not well understood. In this study, we report the UL42 proteins of alphaherpesvirus pseudorabies virus (PRV) and HSV type 1 (HSV1) as a potent antagonist of the IFN-I-induced JAK-STAT signaling pathway. We found that ectopic expression of UL42 in porcine macrophage CRL and human HeLa cells significantly suppresses IFN-α-mediated activation of the IFN-stimulated response element (ISRE), leading to a decreased transcription and expression of IFN-stimulated genes (ISGs). Mechanistically, UL42 directly interacts with ISRE and interferes with ISG factor 3 (ISGF3) from binding to ISRE for efficient gene transcription, and four conserved DNA-binding sites of UL42 are required for this interaction. The substitution of these DNA-binding sites with alanines results in reduced ISRE-binding ability of UL42 and impairs for PRV to evade the IFN response. Knockdown of UL42 in PRV remarkably attenuates the antagonism of virus to IFN in porcine kidney PK15 cells. Our results indicate that the UL42 protein of alphaherpesviruses possesses the ability to suppress IFN-I signaling by preventing the association of ISGF3 and ISRE, thereby contributing to immune evasion. This finding reveals UL42 as a potential antiviral target.


Assuntos
DNA Polimerase Dirigida por DNA/imunologia , Exodesoxirribonucleases/imunologia , Herpesvirus Suídeo 1/imunologia , Interferon Tipo I/imunologia , Fator Gênico 3 Estimulado por Interferon, Subunidade gama/imunologia , Proteínas Virais/imunologia , Animais , Linhagem Celular , Linhagem Celular Tumoral , Células HEK293 , Células HeLa , Herpesvirus Humano 1/imunologia , Humanos , Evasão da Resposta Imune/imunologia , Imunidade Inata/imunologia , Pseudorraiva/imunologia , Elementos de Resposta/imunologia , Transdução de Sinais/imunologia , Suínos , Transcrição Gênica/imunologia
3.
J Virol ; 94(21)2020 10 14.
Artigo em Inglês | MEDLINE | ID: mdl-32847859

RESUMO

Interferon (IFN) family cytokines stimulate genes (interferon-stimulated genes [ISGs]) that are integral to antiviral host defense. Type I IFNs act systemically, whereas type III IFNs act preferentially at epithelial barriers. Among barrier cells, intestinal epithelial cells (IECs) are particularly dependent on type III IFN for the control and clearance of virus infection, but the physiological basis of this selective IFN response is not well understood. Here, we confirm that type III IFN treatment elicits robust and uniform ISG expression in neonatal mouse IECs and inhibits the replication of IEC-tropic rotavirus. In contrast, type I IFN elicits a marginal ISG response in neonatal mouse IECs and does not inhibit rotavirus replication. In vitro treatment of IEC organoids with type III IFN results in ISG expression that mirrors the in vivo type III IFN response. However, IEC organoids have increased expression of the type I IFN receptor relative to neonate IECs, and the response of IEC organoids to type I IFN is strikingly increased in magnitude and scope relative to type III IFN. The expanded type I IFN-specific response includes proapoptotic genes and potentiates toxicity triggered by tumor necrosis factor alpha (TNF-α). The ISGs stimulated in common by type I and III IFNs have strong interferon-stimulated response element (ISRE) promoter motifs, whereas the expanded set of type I IFN-specific ISGs, including proapoptotic genes, have weak ISRE motifs. Thus, the preferential responsiveness of IECs to type III IFN in vivo enables selective ISG expression during infection that confers antiviral protection but minimizes disruption of intestinal homeostasis.IMPORTANCE Enteric viral infections are a major cause of gastroenteritis worldwide and have the potential to trigger or exacerbate intestinal inflammatory diseases. Prior studies have identified specialized innate immune responses that are active in the intestinal epithelium following viral infection, but our understanding of the benefits of such an epithelium-specific response is incomplete. Here, we show that the intestinal epithelial antiviral response is programmed to enable protection while minimizing epithelial cytotoxicity that can often accompany an inflammatory response. Our findings offer new insight into the benefits of a tailored innate immune response at the intestinal barrier and suggest how dysregulation of this response could promote inflammatory disease.


Assuntos
Citocinas/imunologia , Mucosa Intestinal/imunologia , Infecções por Rotavirus/imunologia , Fator de Transcrição STAT1/imunologia , Fator de Transcrição STAT2/imunologia , Fator de Necrose Tumoral alfa/toxicidade , Animais , Animais Recém-Nascidos , Citocinas/genética , Células Epiteliais/efeitos dos fármacos , Células Epiteliais/imunologia , Células Epiteliais/virologia , Regulação da Expressão Gênica , Humanos , Fator Gênico 3 Estimulado por Interferon, Subunidade gama/genética , Fator Gênico 3 Estimulado por Interferon, Subunidade gama/imunologia , Mucosa Intestinal/efeitos dos fármacos , Mucosa Intestinal/virologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Knockout , Organoides/efeitos dos fármacos , Organoides/imunologia , Organoides/virologia , Elementos de Resposta , Rotavirus/efeitos dos fármacos , Rotavirus/crescimento & desenvolvimento , Rotavirus/patogenicidade , Infecções por Rotavirus/genética , Infecções por Rotavirus/patologia , Infecções por Rotavirus/virologia , Fator de Transcrição STAT1/genética , Fator de Transcrição STAT2/genética , Transdução de Sinais , Replicação Viral
4.
Fish Shellfish Immunol ; 103: 220-228, 2020 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-32439513

RESUMO

As a NAD+-dependent deacetylase, SIRT1 is widely involved in apoptosis and cellular inflammation via multiple pathways such as p53, NF-кB and STAT. More and more studies have shown that p53 is the first non-histone deacetylation target of SIRT1. SIRT1-p53 axis thus plays an important role in mammalian cells. IRF9 is an important member of interferon regulator factor family and performs an important role in innate immunity against foreign virus invasion. More importantly, human IRF9 can suppress the SIRT1-p53 axis. However, the functions and relationship between IRF9 and SIRT1-p53 axis are rarely studied in fish. To this end, we made a preliminary research on the functions of grass carp (Ctenopharyngodon idella) IRF9, SIRT1 and p53 in apoptosis and innate immunity. Firstly, we cloned and identified the ORF of SIRT1 (named CiSIRT1, MN125614) from C. idella and demonstrated that CiIRF9 promoted apoptosis, while CiSIRT1 inhibited apoptosis by flow cytometry and TUNEL experiments. Next, we found the interaction between CiSIRT1 and Cip53 in vivo by co-immunoprecipitation experiments. Moreover, the colocalization analysis also showed CiSIRT1 and Cip53 were mainly distributed in nucleus. Thirdly, we got a conclusion that CiIRF9 can repress the expression of CiSIRT1, implying that CiIRF9 regulates CiSIRT1-p53 axis. Finally, CiSIRT1 mRNA level was significantly up-regulated and the expression reached the highest level at 24 h post poly (I:C) stimulation in CIK cells. So, CiSIRT1 may exert an important function in innate immunity. Furthermore, we found CiSIRT1 down-regulated the expression of CiIFN1. In summary, CiIRF9 promotes apoptosis and innate immunity by inhibiting SIRT1-p53 axis. These findings will provide a new theoretical basis for the research on teleost innate immunity.


Assuntos
Apoptose/genética , Carpas/imunologia , Proteínas de Peixes/imunologia , Imunidade Inata/genética , Fator Gênico 3 Estimulado por Interferon, Subunidade gama/imunologia , Sirtuína 1/imunologia , Proteína Supressora de Tumor p53/imunologia , Animais , Carpas/genética , Proteínas de Peixes/genética , Regulação da Expressão Gênica/imunologia , Fator Gênico 3 Estimulado por Interferon, Subunidade gama/genética , Sirtuína 1/genética , Proteína Supressora de Tumor p53/genética
5.
Dev Comp Immunol ; 103: 103528, 2020 02.
Artigo em Inglês | MEDLINE | ID: mdl-31654647

RESUMO

Interferon regulatory factor 9 (IRF9) plays a crucial role in JAK-STAT signaling in human and mammal. However, the relationship between IRF9 and STAT1 in teleost fish remains largely unknown. The previous study has elucidated that two STAT1 isoforms (bcSTAT1a and bcSTAT1b) of black carp (Mylopharyngodon piceus) play an important role during the innate immune activation initiated by grass carp reovirus (GCRV). In this paper, black carp IRF9 (bcIRF9) has been identified and characterized. bcIRF9 was distributed majorly in the nucleus and the linker domain (LD) of bcIRF9 was vital for its nuclear localization. bcIRF9 showed ISRE-inducing activity in reporter assay and presented antiviral activity against GCRV in plaque assay, in which both DNA binding domain (DBD) and LD of bcIRF9 were essential for its antiviral signaling. bcIRF9 was identified to interact with both bcSTAT1a and bcSTAT1b in the co-immunoprecipitation assay. It was interesting that bcIRF9-mediated antiviral signaling was up-regulated by bcSTAT1a; however, down-regulated by bcSTAT1b. Thus, our data support the conclusion that bcIRF9 plays an important role in the innate immune defense against GCRV, in which two STAT1 proteins function differently.


Assuntos
Carpas/imunologia , Proteínas de Peixes/imunologia , Fator Gênico 3 Estimulado por Interferon, Subunidade gama/imunologia , Fator de Transcrição STAT1/imunologia , Animais , Doenças dos Peixes/imunologia , Doenças dos Peixes/virologia , Reoviridae/imunologia , Infecções por Reoviridae/imunologia , Infecções por Reoviridae/veterinária
6.
Sci Rep ; 9(1): 7981, 2019 05 28.
Artigo em Inglês | MEDLINE | ID: mdl-31138840

RESUMO

The association between gene polymorphisms and plasma virus load at the set point (SP-PVL) was investigated in Mauritian macaques inoculated with SIV. Among 44 macaques inoculated with 50 AID50, six individuals were selected: three with SP-PVL among the highest and three with SP-PVL among the lowest. The exons of 390 candidate genes of these six animals were sequenced. Twelve non-synonymous single nucleotide polymorphisms (NS-SNPs) lying in nine genes potentially associated with PVL were genotyped in 23 animals. Three NS-SNPs with probabilities of association with PVL less than 0.05 were genotyped in a total of 44 animals. One NS-SNP lying in exon 1 of the IL37 gene displayed a significant association (p = 3.33 × 10-4) and a strong odds ratio (19.52). Multiple linear regression modeling revealed three significant predictors of SP-PVL, including the IL37 exon 1 NS-SNP (p = 0.0004) and the MHC Class IB haplotypes M2 (p = 0.0007) and M6 (p = 0.0013). These three factors in conjunction explained 48% of the PVL variance (p = 4.8 × 10-6). The potential role of IL37 in the control of SIV infection is discussed.


Assuntos
Interações Hospedeiro-Patógeno/imunologia , Interleucina-1/genética , Polimorfismo de Nucleotídeo Único , Síndrome de Imunodeficiência Adquirida dos Símios/imunologia , Vírus da Imunodeficiência Símia/imunologia , Carga Viral/imunologia , Animais , Sequência de Bases , Éxons , Expressão Gênica , Loci Gênicos , Haplótipos , Sequenciamento de Nucleotídeos em Larga Escala , Antígenos de Histocompatibilidade Classe I/genética , Antígenos de Histocompatibilidade Classe I/imunologia , Interações Hospedeiro-Patógeno/genética , Fator Gênico 3 Estimulado por Interferon, Subunidade gama/genética , Fator Gênico 3 Estimulado por Interferon, Subunidade gama/imunologia , Interleucina-1/imunologia , Modelos Lineares , Macaca fascicularis , Masculino , Razão de Chances , Alinhamento de Sequência , Síndrome de Imunodeficiência Adquirida dos Símios/genética , Síndrome de Imunodeficiência Adquirida dos Símios/patologia , Vírus da Imunodeficiência Símia/crescimento & desenvolvimento , Vírus da Imunodeficiência Símia/patogenicidade
7.
J Virol ; 92(21)2018 11 01.
Artigo em Inglês | MEDLINE | ID: mdl-30089701

RESUMO

Both type I and type II interferons (IFNs) have been implicated in the host defense against varicella-zoster virus (VZV), a common human herpesvirus that causes varicella and zoster. The purpose of this study was to compare their contributions to the control of VZV replication, to identify the signaling pathways that are critical for mediating their antiviral activity, and to define the mechanisms by which the virus counteracts their effects. Gamma interferon (IFN-γ) was much more potent than IFN-α in blocking VZV infection, which was associated with a differential induction of the interferon regulatory factor (IRF) proteins IRF1 and IRF9, respectively. These observations account for the clinical experience that while the formation of VZV skin lesions is initially controlled by local immunity, adaptive virus-specific T cell responses are required to prevent life-threatening VZV infections.IMPORTANCE While both type I and type II IFNs are involved in the control of herpesvirus infections in the human host, to our knowledge, their relative contributions to the restriction of viral replication and spread have not been assessed. We report that IFN-γ has more potent activity than IFN-α against VZV. Findings from this comparative analysis show that the IFN-α-IRF9 axis functions as a first line of defense to delay the onset of viral replication and spread, whereas the IFN-γ-IRF1 axis has the capacity to block the infectious process. Our findings underscore the importance of IRFs in IFN regulation of herpesvirus infection and account for the clinical experience of the initial control of VZV skin infection attributable to IFN-α production, together with the requirement for induction of adaptive IFN-γ-producing VZV-specific T cells to resolve the infection.


Assuntos
Herpesvirus Humano 3/imunologia , Fator Regulador 1 de Interferon/imunologia , Fator Gênico 3 Estimulado por Interferon, Subunidade gama/imunologia , Interferon-alfa/imunologia , Interferon gama/imunologia , Infecção pelo Vírus da Varicela-Zoster/imunologia , Linhagem Celular Tumoral , Células HEK293 , Humanos , Fator de Transcrição STAT1/imunologia , Fator de Transcrição STAT2/imunologia , Transdução de Sinais/imunologia , Linfócitos T/imunologia , Infecção pelo Vírus da Varicela-Zoster/virologia , Replicação Viral/imunologia
8.
J Exp Med ; 215(10): 2567-2585, 2018 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-30143481

RESUMO

Life-threatening pulmonary influenza can be caused by inborn errors of type I and III IFN immunity. We report a 5-yr-old child with severe pulmonary influenza at 2 yr. She is homozygous for a loss-of-function IRF9 allele. Her cells activate gamma-activated factor (GAF) STAT1 homodimers but not IFN-stimulated gene factor 3 (ISGF3) trimers (STAT1/STAT2/IRF9) in response to IFN-α2b. The transcriptome induced by IFN-α2b in the patient's cells is much narrower than that of control cells; however, induction of a subset of IFN-stimulated gene transcripts remains detectable. In vitro, the patient's cells do not control three respiratory viruses, influenza A virus (IAV), parainfluenza virus (PIV), and respiratory syncytial virus (RSV). These phenotypes are rescued by wild-type IRF9, whereas silencing IRF9 expression in control cells increases viral replication. However, the child has controlled various common viruses in vivo, including respiratory viruses other than IAV. Our findings show that human IRF9- and ISGF3-dependent type I and III IFN responsive pathways are essential for controlling IAV.


Assuntos
Alelos , Homozigoto , Influenza Humana , Fator Gênico 3 Estimulado por Interferon, Subunidade gama/deficiência , Orthomyxoviridae/imunologia , Pneumonia Viral , Feminino , Humanos , Lactente , Influenza Humana/genética , Influenza Humana/imunologia , Influenza Humana/patologia , Interferon alfa-2/genética , Interferon alfa-2/imunologia , Fator Gênico 3 Estimulado por Interferon, Subunidade gama/imunologia , Pneumonia Viral/genética , Pneumonia Viral/imunologia , Pneumonia Viral/patologia
9.
J Virol ; 92(19)2018 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-29997210

RESUMO

Type I interferons (IFNs), as major components of the innate immune system, play a vital role in host resistance to a variety of pathogens. Canonical signaling mediated by type I IFNs activates the Janus kinase-signal transducer and activator of transcription (JAK-STAT) pathway through binding to the IFN-α/ß receptor (IFNAR), resulting in transcription of IFN-stimulated genes (ISGs). However, viruses have evolved multiple strategies to evade this process. Here, we report that herpes simplex virus 1 (HSV-1) ubiquitin-specific protease (UL36USP) abrogates the type I IFN-mediated signaling pathway independent of its deubiquitinase (DUB) activity. In this study, ectopically expressed UL36USP inhibited IFN-ß-induced activation of ISRE promoter and transcription of ISGs, and overexpression of UL36USP lacking DUB activity did not influence this effect. Furthermore, UL36USP was demonstrated to antagonize IFN-ß-induced activation of JAKs and STATs via specifically binding to the IFNAR2 subunit and blocking the interaction between JAK1 and IFNAR2. More importantly, knockdown of HSV-1 UL36USP restored the formation of JAK1-IFNAR2 complex. These findings underline the roles of UL36USP-IFNAR2 interaction in counteracting the type I IFN-mediated signaling pathway and reveal a novel evasion mechanism of antiviral innate immunity by HSV-1.IMPORTANCE Type I IFNs mediate transcription of numerous antiviral genes, creating a remarkable antiviral state in the host. Viruses have evolved various mechanisms to evade this response. Our results indicated that HSV-1 encodes a ubiquitin-specific protease (UL36USP) as an antagonist to subvert type I IFN-mediated signaling. UL36USP was identified to significantly inhibit IFN-ß-induced signaling independent of its deubiquitinase (DUB) activity. The underlying mechanism of UL36USP antagonizing type I IFN-mediated signaling was to specifically bind with IFNAR2 and disassociate JAK1 from IFNAR2. For the first time, we identify UL36USP as a crucial suppressor for HSV-1 to evade type I IFN-mediated signaling. Our findings also provide new insights into the innate immune evasion by HSV-1 and will facilitate our understanding of host-virus interplay.


Assuntos
Herpesvirus Humano 1/genética , Evasão da Resposta Imune , Imunidade Inata , Interferon-alfa/genética , Interferon beta/genética , Receptor de Interferon alfa e beta/genética , Proteínas Virais/genética , Animais , Chlorocebus aethiops , Regulação da Expressão Gênica , Células HEK293 , Herpesvirus Humano 1/imunologia , Humanos , Fator Gênico 3 Estimulado por Interferon, Subunidade gama/genética , Fator Gênico 3 Estimulado por Interferon, Subunidade gama/imunologia , Interferon-alfa/imunologia , Interferon beta/imunologia , Janus Quinase 1/genética , Janus Quinase 1/imunologia , RNA Interferente Pequeno/genética , RNA Interferente Pequeno/imunologia , Receptor de Interferon alfa e beta/imunologia , Fatores de Transcrição STAT/genética , Fatores de Transcrição STAT/imunologia , Transdução de Sinais , Células Vero , Proteínas Virais/imunologia
10.
World J Gastroenterol ; 23(32): 5895-5903, 2017 Aug 28.
Artigo em Inglês | MEDLINE | ID: mdl-28932081

RESUMO

AIM: To investigate the effect of (-)-epigallocatechin-3-gallate (EGCG) on polyinosinic-polycytidylic acid (poly I:C)-triggered intracellular innate immunity against hepatitis C virus (HCV) in hepatocytes. METHODS: A cell culture model of HCV infection was generated by infecting a hepatoma cell line, Huh7, with HCV JFH-1 strain (JFH-1-Huh7). Poly I:C with a high molecular weight and EGCG were used to stimulate the JFH-1-Huh7 cells. Real-time reverse transcription-polymerase chain reaction was used to detect the expression levels of intracellular mRNAs and of intracellular and extracellular HCV RNA. Enzyme-linked immunosorbent assay was used to evaluate the interferon (IFN)-λ1 protein level in the cell culture supernatant. Immunostaining was used to examine HCV core protein expression in Huh7 cells. RESULTS: Our recent study showed that HCV replication could impair poly I:C-triggered intracellular innate immune responses in hepatocytes. In the current study, we showed that EGCG treatment significantly increased the poly I:C-induced expression of Toll-like receptor 3 (TLR3), retinoic acid-inducible gene I, and IFN-λ1 in JFH-1-Huh7 cells. In addition, supplementation with EGCG increased the poly I:C-mediated antiviral activity in JFH-1-Huh7 cells at the intracellular and extracellular HCV RNA and protein levels. Further investigation of the mechanisms showed that EGCG treatment significantly enhanced the poly I:C-induced expression of IFN-regulatory factor 9 and several antiviral IFN-stimulated genes, including ISG15, ISG56, myxovirus resistance A, and 2'-5'-oligoadenylate synthetase 1, which encode the key antiviral elements in the IFN signaling pathway. CONCLUSION: Our observations provide experimental evidence that EGCG has the ability to enhance poly I:C-induced intracellular antiviral innate immunity against HCV replication in hepatocytes.


Assuntos
Antivirais/farmacologia , Catequina/análogos & derivados , Hepacivirus/fisiologia , Hepatite C/imunologia , Imunidade Inata/efeitos dos fármacos , Interferon gama/imunologia , Poli I-C/imunologia , Antivirais/uso terapêutico , Catequina/farmacologia , Catequina/uso terapêutico , Linhagem Celular Tumoral , Proteína DEAD-box 58/imunologia , Proteína DEAD-box 58/metabolismo , Ensaio de Imunoadsorção Enzimática , Hepatite C/tratamento farmacológico , Hepatite C/virologia , Hepatócitos , Humanos , Fator Gênico 3 Estimulado por Interferon, Subunidade gama/imunologia , Fator Gênico 3 Estimulado por Interferon, Subunidade gama/metabolismo , Interferon gama/metabolismo , RNA Viral/isolamento & purificação , Receptores Imunológicos , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transdução de Sinais/imunologia , Receptor 3 Toll-Like/metabolismo , Proteínas do Core Viral/metabolismo , Replicação Viral/efeitos dos fármacos , Replicação Viral/imunologia
11.
J Virol ; 91(22)2017 11 15.
Artigo em Inglês | MEDLINE | ID: mdl-28878077

RESUMO

Effective CD8+ T cell responses play an important role in determining the course of a viral infection. Overwhelming antigen exposure can result in suboptimal CD8+ T cell responses, leading to chronic infection. This altered CD8+ T cell differentiation state, termed exhaustion, is characterized by reduced effector function, upregulation of inhibitory receptors, and altered expression of transcription factors. Prevention of overwhelming antigen exposure to limit CD8+ T cell exhaustion is of significant interest for the control of chronic infection. The transcription factor interferon regulatory factor 9 (IRF9) is a component of type I interferon (IFN-I) signaling downstream of the IFN-I receptor (IFNAR). Using acute infection of mice with lymphocytic choriomeningitis virus (LCMV) strain Armstrong, we show here that IRF9 limited early LCMV replication by regulating expression of interferon-stimulated genes and IFN-I and by controlling levels of IRF7, a transcription factor essential for IFN-I production. Infection of IRF9- or IFNAR-deficient mice led to a loss of early restriction of viral replication and impaired antiviral responses in dendritic cells, resulting in CD8+ T cell exhaustion and chronic infection. Differences in the antiviral activities of IRF9- and IFNAR-deficient mice and dendritic cells provided further evidence of IRF9-independent IFN-I signaling. Thus, our findings illustrate a CD8+ T cell-extrinsic function for IRF9, as a signaling factor downstream of IFNAR, in preventing overwhelming antigen exposure resulting in CD8+ T cell exhaustion and, ultimately, chronic infection.IMPORTANCE During early viral infection, overwhelming antigen exposure can cause functional exhaustion of CD8+ T cells and lead to chronic infection. Here we show that the transcription factor interferon regulatory factor 9 (IRF9) plays a decisive role in preventing CD8+ T cell exhaustion. Using acute infection of mice with LCMV strain Armstrong, we found that IRF9 limited early LCMV replication by regulating expression of interferon-stimulated genes and Irf7, encoding a transcription factor crucial for type I interferon (IFN-I) production, as well as by controlling the levels of IFN-I. Infection of IRF9-deficient mice led to a chronic infection that was accompanied by CD8+ T cell exhaustion due to defects extrinsic to T cells. Our findings illustrate an essential role for IRF9, as a mediator downstream of IFNAR, in preventing overwhelming antigen exposure causing CD8+ T cell exhaustion and leading to chronic viral infection.


Assuntos
Linfócitos T CD8-Positivos/imunologia , Fator Gênico 3 Estimulado por Interferon, Subunidade gama/imunologia , Coriomeningite Linfocítica/imunologia , Vírus da Coriomeningite Linfocítica/imunologia , Transdução de Sinais/imunologia , Doença Aguda , Animais , Linfócitos T CD8-Positivos/patologia , Doença Crônica , Fator Regulador 7 de Interferon , Interferon Tipo I/genética , Interferon Tipo I/imunologia , Fator Gênico 3 Estimulado por Interferon, Subunidade gama/genética , Coriomeningite Linfocítica/genética , Coriomeningite Linfocítica/patologia , Vírus da Coriomeningite Linfocítica/genética , Camundongos , Camundongos Knockout , Receptor de Interferon alfa e beta/genética , Receptor de Interferon alfa e beta/imunologia , Transdução de Sinais/genética
12.
J Cell Mol Med ; 21(11): 3087-3099, 2017 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-28560754

RESUMO

Interleukin (IL)-6-type cytokines have no direct antiviral activity; nevertheless, they display immune-modulatory functions. Oncostatin M (OSM), a member of the IL-6 family, has recently been shown to induce a distinct number of classical interferon stimulated genes (ISG). Most of them are involved in antigen processing and presentation. However, induction of retinoic acid-inducible gene (RIG)-I-like receptors (RLR) has not been investigated. Here we report that OSM has the capability to induce the expression of the DExD/H-Box RNA helicases RIG-I and melanoma differentiation antigen 5 (MDA5) as well as of the transcription factors interferon regulatory factor (IRF)1, IRF7 and IRF9 in primary fibroblasts. Induction of the helicases depends on tyrosine as well as serine phosphorylation of STAT1. Moreover, we could show that the OSM-induced STAT1 phosphorylation is predominantly counter-regulated by a strong STAT3-dependent SOCS3 induction, as Stat3 as well as Socs3 knock-down results in an enhanced and prolonged helicase and IRF expression. Other factors involved in regulation of STAT1 or IRF1 activity, like protein tyrosine phosphatase, non-receptor type 2 (PTPN2), promyelocytic leukaemia protein (PML) or small ubiquitin-related modifier 1 (SUMO1), play a minor role in OSM-mediated induction of RLR. Remarkably, OSM and interferon-γ (IFN-γ) synergize to mediate transcription of RLR and pre-treatment of fibroblasts with OSM fosters the type I interferon production in response to a subsequent encounter with double-stranded RNA. Together, these findings suggest that the OSM-induced JAK/STAT1 signalling is implicated in virus protection of non-professional immune cells and may cooperate with interferons to enhance RLR expression in these cells.


Assuntos
Proteína DEAD-box 58/genética , Fibroblastos/efeitos dos fármacos , Imunidade Inata , Helicase IFIH1 Induzida por Interferon/genética , Oncostatina M/farmacologia , Fator de Transcrição STAT1/genética , Linhagem Celular Tumoral , Proteína DEAD-box 58/antagonistas & inibidores , Proteína DEAD-box 58/imunologia , Fibroblastos/citologia , Fibroblastos/metabolismo , Regulação da Expressão Gênica , Humanos , Fator Regulador 1 de Interferon/genética , Fator Regulador 1 de Interferon/imunologia , Fator Regulador 7 de Interferon/genética , Fator Regulador 7 de Interferon/imunologia , Helicase IFIH1 Induzida por Interferon/antagonistas & inibidores , Helicase IFIH1 Induzida por Interferon/imunologia , Fator Gênico 3 Estimulado por Interferon, Subunidade gama/genética , Fator Gênico 3 Estimulado por Interferon, Subunidade gama/imunologia , Interferon gama/farmacologia , Interleucina-6/farmacologia , Fator Inibidor de Leucemia/farmacologia , Subunidade alfa de Receptor de Fator Inibidor de Leucemia/genética , Subunidade alfa de Receptor de Fator Inibidor de Leucemia/imunologia , Lipopolissacarídeos/farmacologia , Pulmão/citologia , Pulmão/efeitos dos fármacos , Pulmão/metabolismo , Osteoblastos/citologia , Osteoblastos/efeitos dos fármacos , Osteoblastos/metabolismo , Cultura Primária de Células , RNA Interferente Pequeno/genética , RNA Interferente Pequeno/metabolismo , Receptores Imunológicos , Fator de Transcrição STAT1/imunologia , Fator de Transcrição STAT3/genética , Fator de Transcrição STAT3/imunologia , Transdução de Sinais , Pele/citologia , Pele/efeitos dos fármacos , Pele/metabolismo , Proteína 3 Supressora da Sinalização de Citocinas/genética , Proteína 3 Supressora da Sinalização de Citocinas/imunologia
13.
Fish Shellfish Immunol ; 66: 524-530, 2017 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-28546020

RESUMO

Interferon regulatory factors (IRFs) are transcription factors which play important roles in regulating the expression of type I interferons (IFNs) and IFN-stimulated genes. IRF9 is one of the IRF family gene members which belongs to the IRF4 subfamily. Mammalian IRF9 has been known to be involved in antiviral responses as the DNA sequence recognition subunit of IFN-stimulated gene factor 3 (ISGF3) complex. In fish, only a few studies investigated the characteristics of IRF9 and the role in IFN signaling. In this study, we identified the IRF9 gene from miiuy croaker (mmiIRF9) and studied its feature and function. Sequence analysis showed the similarity of mmiIRF9 and other fish IRF9 genes. Structural and syntenic analysis showed the conservatism in fish IRF9 genes. The result of expression analysis in normal tissues and infected tissues and macrophages showed that mmiIRF9 expressed in all tested normal tissues and up-regulated expression in liver, kidney and macrophages after stimulated with poly(I:C). Luciferase reporter assays demonstrated the mmiIRF9 can induced IFNα and IFNß luciferase reporters and the cellular localization of mmiIRF9 was mainly distributed in the cytoplasm in Hela cells. Furthermore, the evolutionary analysis of IRF4 subfamily showed the IRF4 and IRF8 may be the most ancient and conservative genes in the evolution of this subfamily.


Assuntos
Evolução Molecular , Proteínas de Peixes/genética , Imunidade Inata , Fator Gênico 3 Estimulado por Interferon, Subunidade gama/genética , Perciformes/genética , Sequência de Aminoácidos , Animais , DNA Complementar/genética , DNA Complementar/metabolismo , Proteínas de Peixes/química , Proteínas de Peixes/imunologia , Regulação da Expressão Gênica , Fator Gênico 3 Estimulado por Interferon, Subunidade gama/química , Fator Gênico 3 Estimulado por Interferon, Subunidade gama/imunologia , Perciformes/classificação , Perciformes/imunologia , Perciformes/metabolismo , Filogenia , Poli I-C/farmacologia , Alinhamento de Sequência/veterinária , Distribuição Tecidual
14.
Virology ; 507: 161-169, 2017 07.
Artigo em Inglês | MEDLINE | ID: mdl-28441586

RESUMO

Emerged porcine kobuvirus (PKV) has adversely affected the global swine industry since 2008, but the etiological biology of PKV is unclear. Screening PKV-encoded structural and non-structural proteins with a type I IFN-responsive luciferase reporter showed that PKV VP3 protein inhibited the IFN-ß-triggered signaling pathway, resulting in the decrease of VSV-GFP replication. QPCR data showed that IFN-ß downstream cytokine genes were suppressed without cell-type specificity as well. The results from biochemical experiments indicated that PKV VP3 associated with STAT2 and IRF9, and interfered with the formation of the STAT2-IRF9 and STAT2-STAT2 complex, impairing nuclear translocation of STAT2 and IRF9. Taken together, these data reveal a new mechanism for immune evasion of PKV.


Assuntos
Fator Gênico 3 Estimulado por Interferon, Subunidade gama/imunologia , Interferon beta/imunologia , Kobuvirus/imunologia , Infecções por Picornaviridae/imunologia , Fator de Transcrição STAT2/imunologia , Proteínas Virais/imunologia , Animais , Linhagem Celular , Dimerização , Humanos , Evasão da Resposta Imune , Fator Gênico 3 Estimulado por Interferon, Subunidade gama/genética , Interferon beta/genética , Kobuvirus/genética , Camundongos , Infecções por Picornaviridae/genética , Infecções por Picornaviridae/virologia , Fator de Transcrição STAT2/química , Fator de Transcrição STAT2/genética , Transdução de Sinais , Proteínas Virais/genética
16.
Mol Immunol ; 85: 123-129, 2017 05.
Artigo em Inglês | MEDLINE | ID: mdl-28236773

RESUMO

Proinflammatory cytokines and type I IFNs were produced by TLR signaling and these responses are crucial for host defensive responses against pathogens. In order to avoid harmful and inappropriate inflammatory responses, there are multiple mechanisms to negatively regulate TLR signaling. In this paper, we have firstly studied IRF9 functions as a negative regulator involved in TRIF-mediated NF-κB pathway. Moreover, we found inhibitory effect of IRF9 primary depends on DBD domain. Interestingly, we also demonstrated that else mutants of IRF9, except for IRF9-ΔDBD, have different inhibitory effects upon TRIF-mediated NF-κB pathway. This study provides a novel evidence about the negatively regulation of innate immune signaling pathway in teleost fish. In addition, this finding provides new insights into the regulatory mechanism in mammals.


Assuntos
Proteínas Adaptadoras de Transporte Vesicular/imunologia , Imunidade Inata/imunologia , Fator Gênico 3 Estimulado por Interferon, Subunidade gama/imunologia , NF-kappa B/imunologia , Perciformes/imunologia , Proteínas Adaptadoras de Transporte Vesicular/metabolismo , Animais , Western Blotting , Imuno-Histoquímica , Fator Gênico 3 Estimulado por Interferon, Subunidade gama/metabolismo , NF-kappa B/metabolismo , Perciformes/metabolismo , Reação em Cadeia da Polimerase , Transdução de Sinais/imunologia
17.
Cytokine Growth Factor Rev ; 29: 71-81, 2016 06.
Artigo em Inglês | MEDLINE | ID: mdl-27053489

RESUMO

In the canonical pathway of IFN-I-mediated signaling, phosphorylation of STAT1 and STAT2 leads to heterodimerization and interaction with IRF9. This complex, also known as IFN-stimulated gene factor 3 (ISGF3), then translocates into the nucleus and binds the IFN-I-stimulated response element (ISRE) leading to the activation of transcription of over 300 interferon stimulated genes (ISGs). In addition, STAT1 homodimers [known as γ-activated factor (GAF)] are formed and translocate to the nucleus, where they target genes containing the γ-activated sequence (GAS). The primary function of ISGF3 is to mediate a rapid and robust IFN-I activated response by regulating transient transcription of antiviral ISGs. This requires the quick assembly of ISGF3 from its pre-existing components STAT1, STAT2 and IRF9 and transport to the nucleus to bind ISRE-containing ISGs. The exact events that take place in formation, nuclear translocation and DNA-binding of active ISGF3 are still not clear. Over the years many studies have provided evidence for the existence of a multitude of alternative STAT2-containing (ISRE or GAS-binding) complexes involved in IFN-I signaling, emphasizing the importance of STAT2 in the regulation of specific IFN-I-induced transcriptional programs, independent of its involvement in the classical ISGF3 complex. This review describes the unique role of STAT2 in differential complex formation of unphosphorylated and phosphorylated ISGF3 components that direct constitutive and IFN-I-stimulated transcriptional responses. In addition, we highlight the existence of a STAT1-independent IFN-I signaling pathway, where STAT2/IRF9 can potentially substitute for the role of ISGF3 and offer a back-up response against viral infection.


Assuntos
Interferon Tipo I/imunologia , Fator de Transcrição STAT2/imunologia , Transdução de Sinais/imunologia , Transcrição Gênica/imunologia , Viroses/imunologia , Animais , Humanos , Fator Gênico 3 Estimulado por Interferon, Subunidade gama/imunologia , Elementos de Resposta/imunologia
18.
Cytokine Growth Factor Rev ; 29: 35-43, 2016 06.
Artigo em Inglês | MEDLINE | ID: mdl-26987614

RESUMO

The host response to viral infections relies on tightly regulated and intricate signaling pathways involving type I interferons (IFN-Is). The IFN-Is mediate their antiviral effects predominantly through a signaling factor complex that comprises the transcription factors, interferon regulatory factor 9 (IRF9) and the signal transducers and activators of transcription (STAT) 1 and STAT2. While STAT1 and STAT2 have been studied extensively, the biological significance of IRF9 is only beginning to emerge. Recent studies have revealed a unique role for IRF9 as a conductor of the cellular responses to IFN-Is. Intriguingly, novel roles for IRF9 outside of the antiviral response are also being identified. Thus IRF9 may have a more extensive influence on cellular processes than previously recognized, ranging from antiviral immune responses to oncogenesis and gut homeostasis. In this review, we will focus on the distinct and emerging roles of IRF9 in the antiviral host response and beyond.


Assuntos
Fator Gênico 3 Estimulado por Interferon, Subunidade gama/imunologia , Transdução de Sinais/imunologia , Viroses/imunologia , Animais , Humanos , Fator de Transcrição STAT1/imunologia , Fator de Transcrição STAT2/imunologia , Viroses/patologia
19.
Arthritis Rheumatol ; 68(5): 1233-44, 2016 05.
Artigo em Inglês | MEDLINE | ID: mdl-26636548

RESUMO

OBJECTIVE: Type I interferon (IFN) signaling is a central pathogenic pathway in systemic lupus erythematosus (SLE), and therapeutics targeting type I IFN signaling are in development. Multiple proteins with overlapping functions play a role in IFN signaling, but the signaling events downstream of receptor engagement are unclear. This study was undertaken to investigate the roles of the type I and type II IFN signaling components IFN-α/ß/ω receptor 2 (IFNAR-2), IFN regulatory factor 9 (IRF-9), and STAT-1 in a mouse model of SLE. METHODS: We used immunohistochemical staining and highly multiplexed assays to characterize pathologic changes in histology, autoantibody production, cytokine/chemokine profiles, and STAT phosphorylation in order to investigate the individual roles of IFNAR-2, IRF-9, and STAT-1 in MRL/lpr mice. RESULTS: We found that STAT-1(-/-) mice, but not IRF-9(-/-) or IFNAR-2(-/-) mice, developed interstitial nephritis characterized by infiltration with retinoic acid receptor-related orphan nuclear receptor γt-positive lymphocytes, macrophages, and eosinophils. Despite pronounced interstitial kidney disease and abnormal kidney function, STAT-1(-/-) mice had decreased proteinuria, glomerulonephritis, and autoantibody production. Phosphospecific flow cytometry revealed shunting of STAT phosphorylation from STAT-1 to STAT-3/4. CONCLUSION: We describe unique contributions of STAT-1 to pathology in different kidney compartments in a mouse model, and provide potentially novel insight into tubulointerstitial nephritis, a poorly understood complication that predicts end-stage kidney disease in SLE patients.


Assuntos
Fator Gênico 3 Estimulado por Interferon, Subunidade gama/genética , Lúpus Eritematoso Sistêmico/genética , Nefrite Intersticial/genética , Receptor de Interferon alfa e beta/genética , Fator de Transcrição STAT1/genética , Células Th17/imunologia , Animais , Formação de Anticorpos , Autoanticorpos/imunologia , Imunofluorescência , Glomerulonefrite/genética , Glomerulonefrite/imunologia , Interferon Tipo I , Fator Gênico 3 Estimulado por Interferon, Subunidade gama/imunologia , Interferon gama , Rim/patologia , Lúpus Eritematoso Sistêmico/imunologia , Camundongos , Camundongos Endogâmicos MRL lpr , Camundongos Knockout , Nefrite Intersticial/imunologia , Nefrite Intersticial/patologia , Proteinúria/genética , Proteinúria/imunologia , Receptor de Interferon alfa e beta/imunologia , Fator de Transcrição STAT1/imunologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA