Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 34
Filtrar
1.
Stem Cells ; 39(2): 183-195, 2021 02.
Artigo em Inglês | MEDLINE | ID: mdl-33252829

RESUMO

Hematopoietic stem cells (HSCs) maintain quiescence under steady state; however, they are compelled to proliferate and expand to replenish the blood system under stress. The molecular basis underlying stress hematopoiesis remains to be fully understood. In this study, we reported that IRF7 represents an important regulator of stress hematopoiesis. Interferon regulatory factor 7 (IRF7) was dispensable for normal hematopoiesis, whereas its deficiency significantly enhanced hematopoietic stem and progenitor cells (HSPCs) regeneration and improved long-term repopulation of HSCs under stress. Mechanistic studies showed that CXCR4 was identified as a downstream target of IRF7. Overexpression of CXCR4 abrogated the enhanced proliferation and regeneration of IRF7-deficient HSPCs under stress. Similar results were obtained in HSCs from human umbilical cord blood. These observations demonstrated that IRF7 plays an important role in hematopoietic regeneration under stress.


Assuntos
Transplante de Células-Tronco de Sangue do Cordão Umbilical/métodos , Hematopoese/fisiologia , Fator Regulador 7 de Interferon/metabolismo , Estresse Oxidativo/fisiologia , Receptores CXCR4/metabolismo , Animais , Células Cultivadas , Sangue Fetal/metabolismo , Sangue Fetal/transplante , Humanos , Fator Regulador 7 de Interferon/antagonistas & inibidores , Fator Regulador 7 de Interferon/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Receptores CXCR4/genética
2.
Genes (Basel) ; 11(4)2020 04 02.
Artigo em Inglês | MEDLINE | ID: mdl-32252379

RESUMO

Interferon regulatory factor 7 (IRF7) is known as the master transcription factor of the type I interferon response in mammalian species along with IRF3. Yet birds only have IRF7, while they are missing IRF3, with a smaller repertoire of immune-related genes, which leads to a distinctive immune response in chickens compared to in mammals. In order to understand the functional role of IRF7 in the regulation of the antiviral response against avian influenza virus in chickens, we generated IRF7-/- chicken embryonic fibroblast (DF-1) cell lines and respective controls (IRF7wt) by utilizing the CRISPR/Cas9 (clustered regularly interspaced short palindromic repeats/CRISPR-associated protein 9) system. IRF7 knockout resulted in increased viral titers of low pathogenic avian influenza viruses. Further RNA-sequencing performed on H6N2-infected IRF7-/- and IRF7wt cell lines revealed that the deletion of IRF7 resulted in the significant down-regulation of antiviral effectors and the differential expression of genes in the MAPK (mitogen-activated protein kinase) and mTOR (mechanistic target of rapamycin) signaling pathways. Dynamic gene expression profiling of the host response between the wildtype and IRF7 knockout revealed potential signaling pathways involving AP1 (activator protein 1), NF-κB (nuclear factor kappa B) and inflammatory cytokines that may complement chicken IRF7. Our findings in this study provide novel insights that have not been reported previously, and lay a solid foundation for enhancing our understanding of the host antiviral response against the avian influenza virus in chickens.


Assuntos
Sistemas CRISPR-Cas , Vírus da Influenza A/imunologia , Influenza Aviária/imunologia , Fator Regulador 7 de Interferon/antagonistas & inibidores , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Serina-Treonina Quinases TOR/metabolismo , Animais , Embrião de Galinha , Galinhas , Perfilação da Expressão Gênica , Influenza Aviária/metabolismo , Influenza Aviária/virologia , Fator Regulador 7 de Interferon/genética , Fator Regulador 7 de Interferon/metabolismo , Proteínas Quinases Ativadas por Mitógeno/genética , Serina-Treonina Quinases TOR/genética
3.
J Neuroimmune Pharmacol ; 14(4): 551-564, 2019 12.
Artigo em Inglês | MEDLINE | ID: mdl-31154625

RESUMO

Nicotine, the active ingredient in tobacco smoke, suppresses antiviral responses. Interferon regulatory factors (IRFs) regulate transcription of type I interferons (IFNs) and IFN-stimulated genes (ISGs) in this response. IRF7 is a key member of the IRF family. Expression of Irf7 is elevated in the brains of virus-infected animals, including human immunodeficiency virus-1 transgenic (HIV-1Tg) rats. We hypothesized that IRF7 affects nicotine's modulation of antiviral responses. Using CRISPR/Cas9 system, IRF7-mutant cell lines were created from human embryonic kidney 293FT cells in which 16 nicotinic acetylcholine receptors (nAChRs) were detected. Decreased expression of IRF7 was confirmed at both the mRNA and protein levels, as was IRF7-regulated cell growth in two IRF7-mutant cell lines, designated IRF7-Δ7 and IRF7-Δ11. In IRF7-Δ7 cells, expression of two nAChR genes, CHRNA3 and CHRNA9, changed modestly. After stimulation with polyinosinic-polycytidylic acid (poly I:C) (0.25 µg/ml) for 4 h to mimic viral infection, 293FT wild-type (WT) and IRF7-Δ7 cells were treated with 0, 1, or 100 µM nicotine for 24 h, which increased IFN-ß expression in both types of cells but elevation was higher in WT cells (p < 0.001). Expression was significantly suppressed in WT cells (p < 0.001) but not in IRF7-Δ7 cells by 24-h nicotine exposure. Poly I:C stimulation increased mRNA expression of retinoic-acid-inducible protein I (RIG-I), melanoma-differentiation-associated gene 5 (MDA5), IFN-stimulated gene factor 3 (ISG3) complex, and IFN-stimulated genes (IRF7, ISG15, IFIT1, OAS1); nicotine attenuated mRNA expression only in WT cells. Overall, IRF7 is critical to nicotine's effect on the antiviral immune response. Graphical Abstract Involvement of IRF7 in nicotine's suppression of poly I:C-induced antiviral immune responses. PAMPs, such as a synthetic viral analogue of dsRNA poly I:C attack cells, will be recognized by PRRs, and the host innate immunity against viral infection will be activated. PRRs signaling trigger phosphorylation of IRF7 and IRF3 to induce their translocation to the nucleus and result in the production of type I IFNs. Then IFNs bind to IFNAR to activate the transcription factor ISGF3, a complex consisting of STAT1, STAT2, and IRF9. Further, it induces the expression of ISGs, including IFIT1, OAS1, IRF7, ISG15, etc. Nicotine suppresses the immune responses stimulated by poly I:C. In the IRF7-mutant cells, nicotine's suppressive effects on poly I:C-stimulated immune responses were restrained.


Assuntos
Antivirais/farmacologia , Imunidade Celular/fisiologia , Imunidade Inata/fisiologia , Fator Regulador 7 de Interferon/imunologia , Nicotina/toxicidade , Poli I-C/farmacologia , Sequência de Bases , Células HEK293 , Humanos , Imunidade Celular/efeitos dos fármacos , Imunidade Inata/efeitos dos fármacos , Fator Regulador 7 de Interferon/antagonistas & inibidores , Fator Regulador 7 de Interferon/biossíntese , Viroses/imunologia , Viroses/metabolismo
4.
AIDS Res Hum Retroviruses ; 35(1): 40-48, 2019 01.
Artigo em Inglês | MEDLINE | ID: mdl-30073840

RESUMO

Interferon-α (IFN-α) plays a vital role in combating viral infections especially in the early control after infection. However, the HIV infection has shown substantial level of suppression of IFN-α secretion during initial phase of infection. The reasons behind this impairment are still obscure. As plasmacytoid dendritic cells (pDCs) are the major producers of this cytokine, the mechanisms of HIV-1-mediated suppression of IFN-α production by pDCs using the primary pDCs were explored. The nuclear translocation of the interferon regulatory factor (IRF)-7, a transcription factor for IFN-α genes, is essential for the initiation of IFN-α production in pDCs. The HIV-1-exposed pDCs did not show the translocation of IRF-7 into the nucleus in our experiments. Furthermore, it was also observed that HIV-1 inhibited AKT phosphorylation of PI3K/akt pathway in pDCs, an important step for IRF-7 translocation to nucleus. HIV-1-induced inhibition of AKT phosphorylation and IRF-7 translocation was evident even in the presence of Toll-like receptor-7 agonist stimulation and correlated with IFN-α suppression. The findings suggest that HIV-1 may alter AKT phosphorylation to inhibit the translocation of IRF-7 into pDC nucleus, leading to IFN-α suppression, and this may be the reason for IFN-α abrogation observed in recently infected HIV patients. Understanding of interactions between HIV-1 and signaling pathways leading to IFN-α secretion may provide targets for immune intervention.


Assuntos
Células Dendríticas/imunologia , Infecções por HIV/imunologia , Interações Hospedeiro-Patógeno , Fator Regulador 7 de Interferon/antagonistas & inibidores , Interferon-alfa/antagonistas & inibidores , Fosfatidilinositol 3-Quinases/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , HIV-1/imunologia , Humanos , Evasão da Resposta Imune , Fosforilação , Processamento de Proteína Pós-Traducional , Transdução de Sinais
5.
Proc Natl Acad Sci U S A ; 115(16): E3798-E3807, 2018 04 17.
Artigo em Inglês | MEDLINE | ID: mdl-29610295

RESUMO

Sterile alpha motif and HD-domain-containing protein 1 (SAMHD1) blocks replication of retroviruses and certain DNA viruses by reducing the intracellular dNTP pool. SAMHD1 has been suggested to down-regulate IFN and inflammatory responses to viral infections, although the functions and mechanisms of SAMHD1 in modulating innate immunity remain unclear. Here, we show that SAMHD1 suppresses the innate immune responses to viral infections and inflammatory stimuli by inhibiting nuclear factor-κB (NF-κB) activation and type I interferon (IFN-I) induction. Compared with control cells, infection of SAMHD1-silenced human monocytic cells or primary macrophages with Sendai virus (SeV) or HIV-1, or treatment with inflammatory stimuli, induces significantly higher levels of NF-κB activation and IFN-I induction. Exogenous SAMHD1 expression in cells or SAMHD1 reconstitution in knockout cells suppresses NF-κB activation and IFN-I induction by SeV infection or inflammatory stimuli. Mechanistically, SAMHD1 inhibits NF-κB activation by interacting with NF-κB1/2 and reducing phosphorylation of the NF-κB inhibitory protein IκBα. SAMHD1 also interacts with the inhibitor-κB kinase ε (IKKε) and IFN regulatory factor 7 (IRF7), leading to the suppression of the IFN-I induction pathway by reducing IKKε-mediated IRF7 phosphorylation. Interactions of endogenous SAMHD1 with NF-κB and IFN-I pathway proteins were validated in human monocytic cells and primary macrophages. Comparing splenocytes from SAMHD1 knockout and heterozygous mice, we further confirmed SAMHD1-mediated suppression of NF-κB activation, suggesting an evolutionarily conserved property of SAMHD1. Our findings reveal functions of SAMHD1 in down-regulating innate immune responses to viral infections and inflammatory stimuli, highlighting the importance of SAMHD1 in modulating antiviral immunity.


Assuntos
Imunidade Inata , Inflamação/imunologia , Interferon-alfa/biossíntese , NF-kappa B/metabolismo , Proteína 1 com Domínio SAM e Domínio HD/fisiologia , Viroses/imunologia , Animais , Células Cultivadas , Regulação para Baixo , Regulação da Expressão Gênica/efeitos dos fármacos , Inativação Gênica , Células HEK293 , HIV/fisiologia , Humanos , Quinase I-kappa B/antagonistas & inibidores , Fator Regulador 7 de Interferon/antagonistas & inibidores , Interferon-alfa/genética , Macrófagos/imunologia , Macrófagos/virologia , Masculino , Camundongos , Inibidor de NF-kappaB alfa/metabolismo , Fosforilação , Processamento de Proteína Pós-Traducional , Proteínas Recombinantes/imunologia , Vírus Sendai/fisiologia , Transdução de Sinais/imunologia , Células THP-1
6.
PLoS One ; 11(7): e0159329, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27434537

RESUMO

Despite recent advances in understanding macrophage activation, little is known regarding how human alveolar macrophages in health calibrate its transcriptional response to canonical TLR4 activation. In this study, we examined the full spectrum of LPS activation and determined whether the transcriptomic profile of human alveolar macrophages is distinguished by a TIR-domain-containing adapter-inducing interferon-ß (TRIF)-dominant type I interferon signature. Bronchoalveolar lavage macrophages were obtained from healthy volunteers, stimulated in the presence or absence of ultrapure LPS in vitro, and whole transcriptomic profiling was performed by RNA sequencing (RNA-Seq). LPS induced a robust type I interferon transcriptional response and Ingenuity Pathway Analysis predicted interferon regulatory factor (IRF)7 as the top upstream regulator of 89 known gene targets. Ubiquitin-specific peptidase (USP)-18, a negative regulator of interferon α/ß responses, was among the top up-regulated genes in addition to IL10 and USP41, a novel gene with no known biological function but with high sequence homology to USP18. We determined whether IRF-7 and USP-18 can influence downstream macrophage effector cytokine production such as IL-10. We show that IRF-7 siRNA knockdown enhanced LPS-induced IL-10 production in human monocyte-derived macrophages, and USP-18 overexpression attenuated LPS-induced production of IL-10 in RAW264.7 cells. Quantitative PCR confirmed upregulation of USP18, USP41, IL10, and IRF7. An independent cohort confirmed LPS induction of USP41 and IL10 genes. These results suggest that IRF-7 and predicted downstream target USP18, both elements of a type I interferon gene signature identified by RNA-Seq, may serve to fine-tune early cytokine response by calibrating IL-10 production in human alveolar macrophages.


Assuntos
Endopeptidases/genética , Fator Regulador 7 de Interferon/genética , Interleucina-10/genética , Proteases Específicas de Ubiquitina/genética , Adolescente , Adulto , Idoso , Animais , Líquido da Lavagem Broncoalveolar/citologia , Feminino , Regulação da Expressão Gênica/efeitos dos fármacos , Técnicas de Silenciamento de Genes , Voluntários Saudáveis , Sequenciamento de Nucleotídeos em Larga Escala , Humanos , Fator Regulador 7 de Interferon/antagonistas & inibidores , Lipopolissacarídeos/administração & dosagem , Lipopolissacarídeos/metabolismo , Ativação de Macrófagos/efeitos dos fármacos , Ativação de Macrófagos/genética , Macrófagos Alveolares/efeitos dos fármacos , Macrófagos Alveolares/metabolismo , Masculino , Camundongos , Pessoa de Meia-Idade , Células RAW 264.7 , Transcriptoma/genética , Ubiquitina Tiolesterase
7.
Sci Rep ; 6: 29891, 2016 07 14.
Artigo em Inglês | MEDLINE | ID: mdl-27412723

RESUMO

Plasmacytoid dendritic cells (pDC) are specialized immune cells that produce massive levels of type I interferon in response to pathogens. Unfortunately, pDC are fragile and extremely rare, rendering their functional study a tough challenge. However, because of their central role in numerous pathologies, there is a considerable need for an efficient and reproducible protocol for gene silencing in these cells. In this report, we tested six different methods for siRNA delivery into primary human pDC including viral-based, lipid-based, electroporation, and poly-ethylenimine (PEI) technologies. We show that lipid-based reagent DOTAP was extremely efficient for siRNA delivery into pDC, and did not induce cell death or pDC activation. We successfully silenced Toll-Like Receptor 7 (TLR7), CXCR4 and IFN regulatory factor 7 (IRF-7) gene expression in pDC as assessed by RT-qPCR or cytometry. Finally, we showed that TLR7 or IRF-7 silencing in pDC specifically suppressed IFN-α production upon stimulation, providing a functional validation of our transfection protocol.


Assuntos
Células Dendríticas/metabolismo , Ácidos Graxos Monoinsaturados/química , Técnicas de Transferência de Genes , Fator Regulador 7 de Interferon/antagonistas & inibidores , Compostos de Amônio Quaternário/química , Receptores CXCR4/antagonistas & inibidores , Receptor 7 Toll-Like/antagonistas & inibidores , Células Dendríticas/citologia , Eletroporação/métodos , Inativação Gênica , Vetores Genéticos/química , Vetores Genéticos/metabolismo , Humanos , Fator Regulador 7 de Interferon/genética , Fator Regulador 7 de Interferon/metabolismo , Interferon-alfa/antagonistas & inibidores , Interferon-alfa/metabolismo , Lentivirus/genética , Lentivirus/metabolismo , Polietilenoimina/química , Cultura Primária de Células , RNA Interferente Pequeno/genética , RNA Interferente Pequeno/metabolismo , Receptores CXCR4/genética , Receptores CXCR4/metabolismo , Receptor 7 Toll-Like/genética , Receptor 7 Toll-Like/metabolismo
8.
FEBS Lett ; 589(24 Pt B): 4112-8, 2015 Dec 21.
Artigo em Inglês | MEDLINE | ID: mdl-26602079

RESUMO

Little is known about the role of microRNA during influenza A virus (IAV) infection. We observed that NIK 3'UTR luciferase activity was elevated during IAV infection. Further studies demonstrated that miR-302c reduced NIK expression, resulting in the reduction of IFNß mRNA expression. We found that miR-302c prevented the translocation of NF-κB from the cytosol to the nucleus. Furthermore, IAV infection downregulated miR-302c expression, leading to the activation of IFNß expression and the inhibition of viral replication. Compared to miR-302c, miR-520e cannot promote viral replication and production, although the two microRNAs target the same site of the NIK 3'UTR. Collectively, our work defines a novel signaling pathway implicated in the control of IFNß mRNA expression during IAV infection.


Assuntos
Imunidade Inata , Imunidade nas Mucosas , Vírus da Influenza A Subtipo H3N2/imunologia , Interferon beta/antagonistas & inibidores , MicroRNAs/metabolismo , Proteínas Serina-Treonina Quinases/antagonistas & inibidores , Mucosa Respiratória/imunologia , Regiões 3' não Traduzidas , Transporte Ativo do Núcleo Celular , Linhagem Celular Tumoral , Repressão Enzimática , Regulação da Expressão Gênica , Interações Hospedeiro-Patógeno , Humanos , Vírus da Influenza A Subtipo H3N2/fisiologia , Fator Regulador 3 de Interferon/antagonistas & inibidores , Fator Regulador 3 de Interferon/genética , Fator Regulador 3 de Interferon/metabolismo , Fator Regulador 7 de Interferon/antagonistas & inibidores , Fator Regulador 7 de Interferon/genética , Fator Regulador 7 de Interferon/metabolismo , Interferon beta/agonistas , Interferon beta/genética , Interferon beta/metabolismo , NF-kappa B/agonistas , NF-kappa B/antagonistas & inibidores , NF-kappa B/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , RNA Mensageiro/metabolismo , Mucosa Respiratória/enzimologia , Mucosa Respiratória/metabolismo , Mucosa Respiratória/virologia , Elementos de Resposta , Transdução de Sinais , Replicação Viral , Quinase Induzida por NF-kappaB
9.
Cell Death Dis ; 5: e1557, 2014 Dec 04.
Artigo em Inglês | MEDLINE | ID: mdl-25476905

RESUMO

Mutations in tuberous sclerosis (TSC) genes cause the genetic disorder TSC, as well as other neoplasms, including lymphangioleiomyomatosis (LAM) and angiomyolipomas (AMLs). AMLs are benign renal tumors occur both in sporadic LAM and in TSC. As they carry the same mutations, AML cell lines serve as a model for TSC and LAM. Rheb/mammalian target of rapamycin complex 1 (mTORC1) pathway is chronically activated in TSC-deficient cells, and this activation can be diminished using the appropriate inhibitors. Rapamycin (sirolimus) is a known specific inhibitor of mTORC1, whereas S-trans,trans-farnesylthiosalicylic acid (FTS; salirasib) has been shown to inhibit Rheb. To examine the effect of the Rheb/mTOR inhibition pathway, we used human TSC2-deficient AML cells, derived from a LAM patient. FTS indeed inhibited Rheb in these cells and attenuated their proliferation. After comparative treatments with FTS or rapamycin or by re-expression of TSC2, we carried out a gene array analysis. This yielded a substantial number of commonly altered genes, many of which we identified as downstream targets of the interferon (IFN) regulatory factor 7 (IRF7) transcription factor, a central activator of the IFN type 1 immune response. Furthermore, nuclear localization of IRF7 was impaired by each of the three treatments. Interestingly, the phenomena seen on FTS or rapamycin treatment were selective for TSC2-deficient cells. Moreover, knockdown of IRF7 by siRNA mimicked the decrease in number of the abovementioned genes and also inhibited AML cell proliferation. Altogether, these findings support FTS as a potential treatment for TSC and its related pathologies and IRF7 as a novel target for treatment.


Assuntos
Angiomiolipoma/genética , Regulação Neoplásica da Expressão Gênica , Fator Regulador 7 de Interferon/genética , Linfangioleiomiomatose/genética , Proteínas Monoméricas de Ligação ao GTP/genética , Neuropeptídeos/genética , Serina-Treonina Quinases TOR/genética , Proteínas Supressoras de Tumor/genética , Angiomiolipoma/metabolismo , Angiomiolipoma/patologia , Proliferação de Células/efeitos dos fármacos , Farneseno Álcool/análogos & derivados , Farneseno Álcool/farmacologia , Perfilação da Expressão Gênica , Humanos , Fator Regulador 7 de Interferon/antagonistas & inibidores , Fator Regulador 7 de Interferon/metabolismo , Rim/metabolismo , Rim/patologia , Linfangioleiomiomatose/metabolismo , Linfangioleiomiomatose/patologia , Análise em Microsséries , Proteínas Monoméricas de Ligação ao GTP/antagonistas & inibidores , Proteínas Monoméricas de Ligação ao GTP/metabolismo , Neuropeptídeos/antagonistas & inibidores , Neuropeptídeos/metabolismo , RNA Interferente Pequeno/genética , RNA Interferente Pequeno/metabolismo , Proteína Enriquecida em Homólogo de Ras do Encéfalo , Salicilatos/farmacologia , Transdução de Sinais , Sirolimo/farmacologia , Serina-Treonina Quinases TOR/antagonistas & inibidores , Serina-Treonina Quinases TOR/metabolismo , Proteína 2 do Complexo Esclerose Tuberosa , Células Tumorais Cultivadas , Proteínas Supressoras de Tumor/deficiência
10.
J Virol ; 88(19): 11140-53, 2014 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-25031341

RESUMO

UNLABELLED: Several studies have demonstrated that the delivery of type I, II, or III interferons (IFNs) by inoculation of a replication-defective human adenovirus 5 (Ad5) vector expressing IFNs can effectively control foot-and-mouth disease (FMD) in cattle and swine during experimental infections. However, relatively high doses are required to achieve protection. In this study, we identified the functional properties of a porcine fusion protein, poIRF7/3(5D), as a biotherapeutic and enhancer of IFN activity against FMD virus (FMDV). We showed that poIRF7/3(5D) is a potent inducer of type I IFNs, including alpha IFN (IFN-α), IFN-ß, and IFN-ω but not type III IFN (interleukin-28B), without inducing cytotoxicity. Expression of poIRF7/3(5D) significantly and steadily reduced FMDV titers by up to 6 log10 units in swine and bovine cell lines. Treatment with an IFN receptor inhibitor (B18R) combined with an anti-IFN-α antibody neutralized the antiviral activity in the supernatants of cells transduced with an Ad5 vector expressing poIRF7/3(5D) [Ad5-poIRF7/3(5D)]. However, several transcripts with known antiviral function, including type I IFNs, were still highly upregulated (range of increase, 8-fold to over 500-fold) by poIRF7/3(5D) in the presence of B18R. Furthermore, the sera of mice treated with Ad5-poIRF7/3(5D) showed antiviral activity that was associated with the induction of high levels of IFN-α and resulted in complete protection against FMDV challenge at 6, 24, or 48 h posttreatment. This study highlights for the first time the antiviral potential of Ad5-poIRF7/3(5D) in vitro and in vivo against FMDV. IMPORTANCE: FMD remains one of the most devastating diseases that affect livestock worldwide. Effective vaccine formulations are available but are serotype specific and require approximately 7 days before they are able to elicit protective immunity. We have shown that vector-delivered IFN is an option to protect animals against many FMDV serotypes as soon as 24 h and for about 4 days postadministration. Here we demonstrate that delivery of a constitutively active transcription factor that induces the production of endogenous IFNs and potentially other antiviral genes is a viable strategy to protect against FMD.


Assuntos
Adenoviridae/imunologia , Vírus da Febre Aftosa/imunologia , Febre Aftosa/prevenção & controle , Fator Regulador 7 de Interferon/imunologia , Proteínas Recombinantes de Fusão/imunologia , Vacinas Virais/imunologia , Adenoviridae/genética , Animais , Bovinos , Linhagem Celular , Febre Aftosa/imunologia , Febre Aftosa/virologia , Vírus da Febre Aftosa/genética , Expressão Gênica/imunologia , Vetores Genéticos , Humanos , Indutores de Interferon/antagonistas & inibidores , Indutores de Interferon/imunologia , Fator Regulador 7 de Interferon/antagonistas & inibidores , Fator Regulador 7 de Interferon/genética , Interferon Tipo I/antagonistas & inibidores , Interferon Tipo I/biossíntese , Interferon Tipo I/imunologia , Camundongos , Proteínas Recombinantes de Fusão/genética , Suínos , Vacinação , Vacinas Sintéticas , Proteínas Virais/farmacologia , Vacinas Virais/administração & dosagem , Replicação Viral/imunologia
11.
J Interferon Cytokine Res ; 34(1): 16-21, 2014 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-23962002

RESUMO

Hepatitis C virus (HCV) establishes chronic infection in a large number of infected individuals. We have previously shown that HCV infection in hepatocytes blocks poly (I-C) or interferon (IFN)-α-mediated IRF-7 nuclear translocation (Raychoudhuri and others 2010). However, the mechanism of IRF-7 regulation by HCV remained unknown. In this study, we have observed that HCV NS5A physically associates with IRF-7. A subsequent study suggested that the HCV NS5A protein blocks IRF-7-mediated IFN-α14 promoter activation. Further analyses demonstrated that site-specific mutagenesis of the 2 basic arginine residues (amino acids Arg(216) and Arg(217)) in the NS5A is critical for IRF-7-mediated IFN-α14 promoter regulation. Together, our results suggested that the HCV NS5A protein limits the IFN-α-signaling pathway in association with IRF-7, and may, in part, be responsible for the establishment of chronic infection.


Assuntos
Fator Regulador 7 de Interferon/metabolismo , Interferon-alfa/metabolismo , Transdução de Sinais , Proteínas não Estruturais Virais/metabolismo , Células Cultivadas , Humanos , Fator Regulador 7 de Interferon/antagonistas & inibidores , Proteínas não Estruturais Virais/genética
12.
Cell Res ; 23(12): 1356-68, 2013 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-24165894

RESUMO

Previous studies on developmental hematopoiesis have mainly focused on signaling and transcription factors, while the appreciation of epigenetic regulation including that of microRNAs is recent. Here, we show that in zebrafish and mouse, miR-142-3p is specifically expressed in hematopoietic stem cells (HSCs). Knockdown of miR-142a-3p in zebrafish led to a reduced population of HSCs in the aorta-gonad-mesonephros (AGM) region as well as T-cell defects in the thymus. Mechanistically, miR-142a-3p regulates HSC formation and differentiation through the repression of interferon regulatory factor 7 (irf7)-mediated inflammation signaling. Finally, we show that miR-142-3p is also involved in the development of HSCs in mouse AGM, suggesting that it has a highly conserved role in vertebrates. Together, these findings unveil the pivotal roles that miR-142a-3p plays in the formation and differentiation of HSCs by repressing irf7 signaling.


Assuntos
Diferenciação Celular , Regulação da Expressão Gênica no Desenvolvimento , Células-Tronco Hematopoéticas/citologia , Células-Tronco Hematopoéticas/metabolismo , MicroRNAs/metabolismo , Regiões 3' não Traduzidas , Animais , Animais Geneticamente Modificados , Sequência de Bases , Subunidade alfa 2 de Fator de Ligação ao Core/antagonistas & inibidores , Subunidade alfa 2 de Fator de Ligação ao Core/genética , Subunidade alfa 2 de Fator de Ligação ao Core/metabolismo , Embrião de Mamíferos/metabolismo , Embrião não Mamífero/metabolismo , Células HEK293 , Humanos , Fator Regulador 7 de Interferon/antagonistas & inibidores , Fator Regulador 7 de Interferon/genética , Fator Regulador 7 de Interferon/metabolismo , Camundongos , MicroRNAs/antagonistas & inibidores , MicroRNAs/genética , Óxido Nítrico/metabolismo , Oligonucleotídeos Antissenso/metabolismo , Interferência de RNA , Linfócitos T/citologia , Linfócitos T/imunologia , Linfócitos T/metabolismo , Peixe-Zebra/genética , Peixe-Zebra/crescimento & desenvolvimento , Peixe-Zebra/metabolismo
13.
J Virol ; 87(3): 1690-8, 2013 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-23175366

RESUMO

Enterovirus 71 (EV71) is a positive-stranded RNA virus which is capable of inhibiting innate immunity. Among virus-encoded proteins, the 3C protein compromises the type I interferon (IFN-I) response mediated by retinoid acid-inducible gene-I (RIG-I) or Toll-like receptor 3 that activates interferon regulatory 3 (IRF3) and IRF7. In the present study, we report that enterovirus 71 downregulates IRF7 through the 3C protein, which inhibits the function of IRF7. When expressed in mammalian cells, the 3C protein mediates cleavage of IRF7 rather than that of IRF3. This process is insensitive to inhibitors of caspase, proteasome, lysosome, and autophagy. H40D substitution in the 3C active site abolishes its activity, whereas R84Q or V154S substitution in the RNA binding motif has no effect. Furthermore, 3C-mediated cleavage occurs at the Q189-S190 junction within the constitutive activation domain of IRF7, resulting in two cleaved IRF7 fragments that are incapable of activating IFN expression. Ectopic expression of wild-type IRF7 limits EV71 replication. On the other hand, expression of the amino-terminal domain of IRF7 enhances EV71 infection, which correlates with its ability to interact with and inhibit IRF3. These results suggest that control of IRF7 by the 3C protein may represent a viral mechanism to escape cellular responses.


Assuntos
Cisteína Endopeptidases/metabolismo , Enterovirus Humano A/patogenicidade , Evasão da Resposta Imune , Fator Regulador 7 de Interferon/antagonistas & inibidores , Fator Regulador 7 de Interferon/metabolismo , Proteínas Virais/metabolismo , Proteases Virais 3C , Substituição de Aminoácidos , Domínio Catalítico , Linhagem Celular , Cisteína Endopeptidases/genética , Enterovirus Humano A/enzimologia , Humanos , Hidrólise , Proteínas Mutantes/genética , Proteínas Mutantes/metabolismo , Mutação de Sentido Incorreto , Proteínas Virais/genética
14.
J Immunol ; 189(6): 2860-8, 2012 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-22896628

RESUMO

Type I IFNs are induced by pathogens to protect the host from infection and boost the immune response. We have recently demonstrated that this IFN response is not restricted to pathogens, as the Gram-positive bacterium Lactobacillus acidophilus, a natural inhabitant of the intestine, induces high levels of IFN-ß in dendritic cells. In the current study, we investigate the intracellular pathways involved in IFN-ß upon stimulation of dendritic cells with L. acidophilus and reveal that this IFN-ß induction requires phagosomal uptake and processing but bypasses the endosomal receptors TLR7 and TLR9. The IFN-ß production is fully dependent on the TIR adapter molecule MyD88, partly dependent on IFN regulatory factor (IRF)1, but independent of the TIR domain-containing adapter inducing IFN-ß MyD88 adapter-like, IRF and IRF7. However, our results suggest that IRF3 and IRF7 have complementary roles in IFN-ß signaling. The IFN-ß production is strongly impaired by inhibitors of spleen tyrosine kinase (Syk) and PI3K. Our results indicate that L. acidophilus induces IFN-ß independently of the receptors typically used by bacteria, as it requires MyD88, Syk, and PI3K signaling and phagosomal processing to activate IRF1 and IRF3/IRF7 and thereby the release of IFN-ß.


Assuntos
Células Dendríticas/imunologia , Células Dendríticas/microbiologia , Fator Regulador 1 de Interferon/fisiologia , Fator Regulador 3 de Interferon/fisiologia , Fator Regulador 7 de Interferon/fisiologia , Interferon beta/metabolismo , Lactobacillus acidophilus/imunologia , Fator 88 de Diferenciação Mieloide/fisiologia , Animais , Células Cultivadas , Células Dendríticas/metabolismo , Endossomos/imunologia , Endossomos/metabolismo , Endossomos/microbiologia , Fator Regulador 1 de Interferon/deficiência , Fator Regulador 3 de Interferon/antagonistas & inibidores , Fator Regulador 3 de Interferon/deficiência , Fator Regulador 7 de Interferon/antagonistas & inibidores , Fator Regulador 7 de Interferon/deficiência , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Fator 88 de Diferenciação Mieloide/deficiência , Fagossomos/imunologia , Fagossomos/metabolismo , Processamento de Proteína Pós-Traducional/imunologia , Transdução de Sinais/genética , Transdução de Sinais/imunologia
15.
Nat Med ; 18(8): 1224-31, 2012 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-22820642

RESUMO

Breast cancer metastasis is a key determinant of long-term patient survival. By comparing the transcriptomes of primary and metastatic tumor cells in a mouse model of spontaneous bone metastasis, we found that a substantial number of genes suppressed in bone metastases are targets of the interferon regulatory factor Irf7. Restoration of Irf7 in tumor cells or administration of interferon led to reduced bone metastases and prolonged survival time. In mice deficient in the interferon (IFN) receptor or in natural killer (NK) and CD8(+) T cell responses, metastasis was accelerated, indicating that Irf7-driven suppression of metastasis was reliant on IFN signaling to host immune cells. We confirmed the clinical relevance of these findings in over 800 patients in which high expression of Irf7-regulated genes in primary tumors was associated with prolonged bone metastasis-free survival. This gene signature may identify patients that could benefit from IFN-based therapies. Thus, we have identified an innate immune pathway intrinsic to breast cancer cells, the suppression of which restricts immunosurveillance to enable metastasis.


Assuntos
Neoplasias da Mama/genética , Regulação Neoplásica da Expressão Gênica , Inativação Gênica , Fator Regulador 7 de Interferon/fisiologia , Neoplasias Mamárias Experimentais/imunologia , Proteínas de Neoplasias/fisiologia , Evasão Tumoral/fisiologia , Animais , Neoplasias da Mama/imunologia , Linfócitos T CD8-Positivos/imunologia , Feminino , Perfilação da Expressão Gênica , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Humanos , Vigilância Imunológica , Fator Regulador 7 de Interferon/antagonistas & inibidores , Fator Regulador 7 de Interferon/biossíntese , Fator Regulador 7 de Interferon/genética , Fator Gênico 3 Estimulado por Interferon, Subunidade gama/antagonistas & inibidores , Fator Gênico 3 Estimulado por Interferon, Subunidade gama/genética , Fator Gênico 3 Estimulado por Interferon, Subunidade gama/fisiologia , Interferon-alfa/farmacologia , Células Matadoras Naturais/imunologia , Neoplasias Mamárias Experimentais/genética , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos NOD , Camundongos SCID , Metástase Neoplásica/fisiopatologia , Proteínas de Neoplasias/antagonistas & inibidores , Proteínas de Neoplasias/genética , Receptores de Interferon/deficiência , Receptores de Interferon/fisiologia , Proteínas Recombinantes/metabolismo , Imunodeficiência Combinada Severa/genética , Imunodeficiência Combinada Severa/imunologia , Subpopulações de Linfócitos T/imunologia , Evasão Tumoral/genética
16.
Biol Cell ; 104(8): 462-75, 2012 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-22509910

RESUMO

BACKGROUND INFORMATION: The interferon (IFN)-inducible protein TRIM22 (Staf50) is a member of the tripartite motif protein family and has been suggested a role in the regulation of viral replication as well as of protein ubiquitylation. In addition, we have previously shown that TRIM22 is a direct target gene for the tumour suppressor p53. Consistently, over-expression of TRIM22 inhibits the clonogenic growth of monoblastic U937 cells, suggesting anti-proliferative or cell death-inducing effects. RESULTS: Here, we demonstrate that TRIM22 directly or indirectly interacts with the eukaryotic translation initiation factor (eIF)4E, and inhibits the binding of eIF4E to eIF4G, thus disturbing the assembly of the eIF4F complex, which is necessary for cap-dependent translation. Furthermore, TRIM22 exerts a repressive effect on luciferase reporter protein levels and to some extent on radiolabelled methionine incorporation. Even though all nuclear mRNAs are capped, some are more dependent on eIF4F than others for translation. The translation of one of these mRNAs, IRF-7C, was indeed found to be repressed in the presence of TRIM22. CONCLUSIONS: Our data suggest TRIM22 to repress protein translation preferably of some specific mRNAs. Taken together, we show that TRIM22 represses translation by inhibiting the binding of eIF4E to eIF4G, suggesting a mechanism for regulation of protein translation, which may be of importance in response to p53 and/or IFN signalling.


Assuntos
Fator de Iniciação 4E em Eucariotos , Fator de Iniciação Eucariótico 4G , Biossíntese de Proteínas , Proteínas Repressoras , Fator de Iniciação 4E em Eucariotos/genética , Fator de Iniciação 4E em Eucariotos/metabolismo , Fator de Iniciação Eucariótico 4G/genética , Fator de Iniciação Eucariótico 4G/metabolismo , Regulação da Expressão Gênica , Células HEK293 , Humanos , Fator Regulador 7 de Interferon/antagonistas & inibidores , Fator Regulador 7 de Interferon/genética , Fator Regulador 7 de Interferon/metabolismo , Interferons/metabolismo , Antígenos de Histocompatibilidade Menor , Ligação Proteica , Proteínas Repressoras/genética , Proteínas Repressoras/metabolismo , Proteínas com Motivo Tripartido , Proteína Supressora de Tumor p53/metabolismo
17.
J Immunol ; 187(12): 6456-62, 2011 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-22095711

RESUMO

IRF7 is known as the master regulator in virus-triggered induction of type I IFNs (IFN-I). In this study, we identify GBP4 virus-induced protein interacting with IRF7 as a negative regulator for IFN-I response. Overexpression of GBP4 inhibits virus-triggered activation of IRF7-dependent signaling, but has no effect on NF-κB signaling, whereas the knockdown of GBP4 has opposite effects. Furthermore, the supernatant from Sendai virus-infected cells in which GBP4 have been silenced inhibits the replication of vesicular stomatitis virus more efficiently. Competitive coimmunoprecipitation experiments indicate that overexpression of GBP4 disrupts the interactions between TRAF6 and IRF7, resulting in impaired TRAF6-mediated IRF7 ubiquitination. Our results suggest that GBP4 is a negative regulator of virus-triggered IFN-I production, and it is identified as a novel protein targeting IRF7 and inhibiting its function.


Assuntos
Regulação para Baixo/imunologia , Proteínas de Ligação ao GTP/fisiologia , Marcação de Genes , Fator Regulador 7 de Interferon/antagonistas & inibidores , Interferon-alfa/antagonistas & inibidores , Interferon-alfa/biossíntese , Vírus Sendai/imunologia , Animais , Linhagem Celular , Linhagem Celular Tumoral , Regulação para Baixo/genética , Proteínas de Ligação ao GTP/biossíntese , Proteínas de Ligação ao GTP/genética , Técnicas de Silenciamento de Genes , Células HEK293 , Humanos , Fator Regulador 7 de Interferon/metabolismo , Camundongos , Proteínas Recombinantes/antagonistas & inibidores , Proteínas Recombinantes/genética , Fator 6 Associado a Receptor de TNF/antagonistas & inibidores , Fator 6 Associado a Receptor de TNF/metabolismo , Ativação Transcricional/genética , Ativação Transcricional/imunologia , Regulação para Cima/genética , Regulação para Cima/imunologia
18.
J Immunol ; 187(9): 4754-63, 2011 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-21940674

RESUMO

IFN regulatory factor 7 (IRF7) is a potent transcription factor of type I IFNs and IFN-stimulated genes and is known as the master regulator of type I IFN-dependent immune responses. Because excessive responses could harm the host, IRF7 itself is delicately regulated at the transcriptional, translational, and posttranslational levels. Modification of IRF7 by small ubiquitin-related modifiers (SUMOs) has been shown to regulate IFN expression and antiviral responses negatively, but the specific E3 ligase needed for IRF7 SUMOylation has remained unknown. As reported in this article, we have identified the tripartite motif-containing protein 28 (TRIM28) as a binding partner of IRF7. We have demonstrated that TRIM28 also interacts with the SUMO E2 enzyme and increases SUMOylation of IRF7 both in vivo and in vitro, suggesting it acts as a SUMO E3 ligase of IRF7. Unlike the common SUMO E3 ligase, protein inhibitor of activated STAT1, the E3 activity of TRIM28 is specific to IRF7, because it has little effect on IRF7's close relative IRF3. TRIM28 is therefore, so far as we know, the first IRF7-specific SUMO E3 reported. TRIM28-mediated SUMOylation of IRF7 is increased during viral infection, and SUMOylation of transcription factors usually results in transcriptional repression. Overexpression of TRIM28 therefore inhibits IRF7 transactivation activity, whereas knockdown of TRIM28 has the opposite effect and potentiates IFN production and antiviral responses. Collectively, our results suggest that TRIM28 is a specific SUMO E3 ligase and negative regulator of IRF7.


Assuntos
Regulação para Baixo/imunologia , Fator Regulador 7 de Interferon/antagonistas & inibidores , Proteínas Repressoras/fisiologia , Proteínas Modificadoras Pequenas Relacionadas à Ubiquitina/fisiologia , Ubiquitina-Proteína Ligases/fisiologia , Motivos de Aminoácidos/imunologia , Linhagem Celular Tumoral , Células HEK293 , Humanos , Fator Regulador 7 de Interferon/metabolismo , Proteínas Repressoras/química , Proteínas Modificadoras Pequenas Relacionadas à Ubiquitina/química , Especificidade por Substrato/imunologia , Proteína 28 com Motivo Tripartido , Ubiquitina-Proteína Ligases/química
19.
Cell Immunol ; 271(2): 342-9, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-21872224

RESUMO

The type I interferon (IFN) response plays a critical role in autoimmunity and is induced by innate receptor ligation and activation of IFN-regulatory factors (IRF). The present study investigated the roles and functional hierarchy of IRF3, IRF5, and IRF7 in expression of cytokines, chemokines, and matrix metalloproteinases in human THP1 monocytic cells. Targeted IRF knockdown was followed by evaluation of gene expression, promoter activation, and mRNA stability to determine the role of IRF as potential targets for modulating IFN responses in patients with autoimmune diseases. IRF played a distinct role in regulation of type I IFN gene expression in human monocytic cells and specifically regulated gene expression through the IFN-stimulated response element, with no contribution to transcription of traditionally AP-1 or NF-kB regulated genes. IRF7 regulated IL-6 gene expression by increasing IL-6 mRNA stability. IRF regulation of inflammation and induction of the IFN signature might contribute to the pathogenesis of autoimmune diseases and therefore represent novel therapeutic targets.


Assuntos
Doenças Autoimunes/imunologia , Doenças Autoimunes/terapia , Fatores Reguladores de Interferon/antagonistas & inibidores , Interferon Tipo I/biossíntese , Doenças Autoimunes/etiologia , Linhagem Celular , Regulação da Expressão Gênica , Técnicas de Silenciamento de Genes , Humanos , Fator Regulador 3 de Interferon/antagonistas & inibidores , Fator Regulador 3 de Interferon/genética , Fator Regulador 3 de Interferon/imunologia , Fator Regulador 7 de Interferon/antagonistas & inibidores , Fator Regulador 7 de Interferon/genética , Fator Regulador 7 de Interferon/imunologia , Fatores Reguladores de Interferon/deficiência , Fatores Reguladores de Interferon/genética , Fatores Reguladores de Interferon/imunologia , Interleucina-6/genética , Monócitos/imunologia , Monócitos/metabolismo , Regiões Promotoras Genéticas , Estabilidade de RNA , RNA Interferente Pequeno/genética
20.
J Immunol ; 186(5): 3006-14, 2011 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-21270393

RESUMO

Bone marrow-derived plasmacytoid dendritic cells (pDCs) from IL-1R-associated kinase (IRAK)2-deficient mice produced more IFNs than did wild-type pDCs upon stimulation with the TLR9 ligand CpG. Furthermore, in CpG-stimulated IRAK2-deficient pDCs there was increased nuclear translocation of IFN regulatory factor 7, the key transcription factor for IFN gene transcription in these cells. In IRAK2-deficient macrophages, enhanced NF-κB activation and increased expression of CpG-induced genes were detected within 2 h after treatment. However, at later times, NF-κB activation was decreased and, in contrast to the results with IFN, there was less secretion of other proinflammatory cytokines (such as TNF-α) and chemokines in CpG-stimulated IRAK2-deficient pDCs and macrophages. Therefore, although IRAK2 is a negative regulator of TLR9-mediated IFN production through its modulation of the transcriptional activity of IFN regulatory factor 7, it is also a positive regulator of TLR9-mediated proinflammatory cytokine and chemokine production at some level subsequent to transcription.


Assuntos
Citocinas/biossíntese , Mediadores da Inflamação/metabolismo , Interferon Tipo I/biossíntese , Interferon beta/biossíntese , Quinases Associadas a Receptores de Interleucina-1/fisiologia , Receptor Toll-Like 9/fisiologia , Animais , Células Cultivadas , Ilhas de CpG/imunologia , Citocinas/antagonistas & inibidores , Citocinas/fisiologia , Células Dendríticas/imunologia , Células Dendríticas/metabolismo , Regulação para Baixo/imunologia , Infecções por Herpesviridae/enzimologia , Infecções por Herpesviridae/imunologia , Infecções por Herpesviridae/patologia , Mediadores da Inflamação/antagonistas & inibidores , Mediadores da Inflamação/fisiologia , Fator Regulador 7 de Interferon/antagonistas & inibidores , Fator Regulador 7 de Interferon/fisiologia , Interferon Tipo I/antagonistas & inibidores , Interferon-alfa , Interferon beta/antagonistas & inibidores , Quinases Associadas a Receptores de Interleucina-1/deficiência , Ligantes , Camundongos , Camundongos Knockout , Proteínas Recombinantes , Rhadinovirus/imunologia , Receptor Toll-Like 9/metabolismo , Infecções Tumorais por Vírus/enzimologia , Infecções Tumorais por Vírus/imunologia , Infecções Tumorais por Vírus/patologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA