Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 24
Filtrar
Mais filtros








Base de dados
Intervalo de ano de publicação
1.
Front Cell Dev Biol ; 10: 1033684, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-36393867

RESUMO

The human DEAD-box protein DDX3X is an RNA remodelling enzyme that has been implicated in various aspects of RNA metabolism. In addition, like many DEAD-box proteins, it has non-conventional functions that are independent of its enzymatic activity, e.g., DDX3X acts as an adaptor molecule in innate immune signalling pathways. DDX3X has been linked to several human diseases. For example, somatic mutations in DDX3X were identified in various human cancers, and de novo germline mutations cause a neurodevelopmental condition now termed 'DDX3X syndrome'. DDX3X is also an important host factor in many different viral infections, where it can have pro-or anti-viral effects depending on the specific virus. The regulation of translation initiation for specific mRNA transcripts is likely a central cellular function of DDX3X, yet many questions regarding its exact targets and mechanisms of action remain unanswered. In this review, we explore the current knowledge about DDX3X's physiological RNA targets and summarise its interactions with the translation machinery. A role for DDX3X in translational reprogramming during cellular stress is emerging, where it may be involved in the regulation of stress granule formation and in mediating non-canonical translation initiation. Finally, we also discuss the role of DDX3X-mediated translation regulation during viral infections. Dysregulation of DDX3X's function in mRNA translation likely contributes to its involvement in disease pathophysiology. Thus, a better understanding of its exact mechanisms for regulating translation of specific mRNA targets is important, so that we can potentially develop therapeutic strategies for overcoming the negative effects of its dysregulation.

2.
Biochim Biophys Acta Gene Regul Mech ; 1865(2): 194787, 2022 02.
Artigo em Inglês | MEDLINE | ID: mdl-35121200

RESUMO

DEAD-box protein 3X (DDX3X) is a human DEAD-box protein with conventional roles in RNA metabolism and unconventional functions in signalling pathways that do not require its enzymatic activity. For example, DDX3X acts as a multifunctional adaptor molecule in anti-viral innate immune signalling pathways, where it interacts with and regulates the kinase IKB-kinase-epsilon (IIKKε). Interestingly, both DDX3X and IKKɛ have also independently been shown to act as breast cancer oncogenes. IKKɛ's oncogenic functions are likely multifactorial, but it was suggested to phosphorylate the transcription factor Estrogen receptor alpha (ERα) at Serine 167, which drives expression of Erα target genes in an estrogen-independent manner. In this study, we identified a novel physical interaction between DDX3X and ERα that positively regulates ERα activation. DDX3X knockdown in ER+ breast cancer cell lines resulted in reduced ERα phosphorylation, reduced Estrogen Response Element (ERE)-controlled reporter gene expression, decreased expression of ERα target genes, and decreased cell proliferation. Vice versa, overexpression of DDX3X resulted in enhanced ERα phosphorylation and activity. Furthermore, we provide evidence that DDX3X physically binds to ERα from co-immunoprecipitation and pulldown experiments. Based on our data, we propose that DDX3X acts as an adaptor to facilitate IKKε-mediated ERα activation, akin to the mechanism we previously elucidated for IKKε-mediated Interferon Regulatory factor 3 (IRF3) activation in innate immune signalling. In conclusion, our research provides a novel molecular mechanism that might contribute to the oncogenic effect of DDX3X in breast cancer, potentially linking it to the development of resistance against endocrine therapy.


Assuntos
Neoplasias da Mama , RNA Helicases DEAD-box , Receptor alfa de Estrogênio , Neoplasias da Mama/genética , Neoplasias da Mama/metabolismo , Proliferação de Células , RNA Helicases DEAD-box/genética , RNA Helicases DEAD-box/metabolismo , Receptor alfa de Estrogênio/genética , Receptor alfa de Estrogênio/metabolismo , Feminino , Humanos , Receptores de Estrogênio , Transdução de Sinais
3.
Front Immunol ; 12: 653883, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34054816

RESUMO

Tight regulation of inflammatory cytokine and interferon (IFN) production in innate immunity is pivotal for optimal control of pathogens and avoidance of immunopathology. The human Nod-like receptor (NLR) NLRP11 has been shown to regulate type I IFN and pro-inflammatory cytokine responses. Here, we identified the ATP-dependent RNA helicase DDX3X as a novel binding partner of NLRP11, using co-immunoprecipitation and LC-MS/MS. DDX3X is known to enhance type I IFN responses and NLRP3 inflammasome activation. We demonstrate that NLRP11 can abolish IKKϵ-mediated phosphorylation of DDX3X, resulting in lower type I IFN induction upon viral infection. These effects were dependent on the LRR domain of NLRP11 that we mapped as the interaction domain for DDX3X. In addition, NLRP11 also suppressed NLRP3-mediated caspase-1 activation in an LRR domain-dependent manner, suggesting that NLRP11 might sequester DDX3X and prevent it from promoting NLRP3-induced inflammasome activation. Taken together, our data revealed DDX3X as a central target of NLRP11, which can mediate the effects of NLRP11 on type I IFN induction as well as NLRP3 inflammasome activation. This expands our knowledge of the molecular mechanisms underlying NLRP11 function in innate immunity and suggests that both NLRP11 and DDX3X might be promising targets for modulation of innate immune responses.


Assuntos
RNA Helicases DEAD-box/metabolismo , Inflamassomos/metabolismo , Interferon Tipo I/metabolismo , Proteína 3 que Contém Domínio de Pirina da Família NLR/metabolismo , Proteínas NLR/metabolismo , Cromatografia Líquida , Citocinas/metabolismo , Humanos , Imunidade Inata , Mediadores da Inflamação , Ligação Proteica , Espectrometria de Massas em Tandem
4.
FASEB J ; 33(8): 8732-8744, 2019 08.
Artigo em Inglês | MEDLINE | ID: mdl-31163989

RESUMO

Viruses use a spectrum of immune evasion strategies that enable infection and replication. The acute phase of hepatitis C virus (HCV) infection is characterized by nonspecific and often mild clinical symptoms, suggesting an immunosuppressive mechanism that, unless symptomatic liver disease presents, allows the virus to remain largely undetected. We previously reported that HCV induced the regulatory protein suppressor of cytokine signaling (SOCS)3, which inhibited TNF-α-mediated inflammatory responses. However, the mechanism by which HCV up-regulates SOCS3 remains unknown. Here we show that the HCV protein, p7, enhances both SOCS3 mRNA and protein expression. A p7 inhibitor reduced SOCS3 induction, indicating that p7's ion channel activity was required for optimal up-regulation of SOCS3. Short hairpin RNA and chemical inhibition revealed that both the Janus kinase-signal transducer and activator of transcription (JAK-STAT) and MAPK pathways were required for p7-mediated induction of SOCS3. HCV-p7 expression suppressed TNF-α-mediated IκB-α degradation and subsequent NF-κB promoter activity, revealing a new and functional, anti-inflammatory effect of p7. Together, these findings identify a molecular mechanism by which HCV-p7 induces SOCS3 through STAT3 and ERK activation and demonstrate that p7 suppresses proinflammatory responses to TNF-α, possibly explaining the lack of inflammatory symptoms observed during early HCV infection.-Convery, O., Gargan, S., Kickham, M., Schroder, M., O'Farrelly, C., Stevenson, N. J. The hepatitis C virus (HCV) protein, p7, suppresses inflammatory responses to tumor necrosis factor (TNF)-α via signal transducer and activator of transcription (STAT)3 and extracellular signal-regulated kinase (ERK)-mediated induction of suppressor of cytokine signaling (SOCS)3.


Assuntos
Hepatite C/metabolismo , Sistema de Sinalização das MAP Quinases , Fator de Transcrição STAT3/metabolismo , Proteína 3 Supressora da Sinalização de Citocinas/genética , Fator de Necrose Tumoral alfa/metabolismo , Proteínas Virais/metabolismo , Linhagem Celular Tumoral , Células HEK293 , Humanos , Proteína Quinase 1 Ativada por Mitógeno/metabolismo , Proteína Quinase 3 Ativada por Mitógeno/metabolismo , NF-kappa B/metabolismo , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Proteína 3 Supressora da Sinalização de Citocinas/metabolismo , Regulação para Cima
5.
Biochem J ; 475(22): 3595-3607, 2018 11 20.
Artigo em Inglês | MEDLINE | ID: mdl-30341167

RESUMO

DDX3 is a DEAD-box RNA helicase that we and others have previously implicated in antiviral immune signalling pathways leading to type I interferon (IFN) induction. We previously demonstrated that it directly interacts with the kinase IKKε (IκB kinase ε), enhances it activation, and then facilitates phosphorylation of the transcription factor IRF3 by IKKε. However, the TLR7/9 (Toll-like receptor 7/9)-mediated pathway, one of the most physiologically relevant IFN induction pathways, proceeds independently of IKKε or the related kinase TBK1 (TANK-binding kinase 1). This pathway induces type I IFN production via the kinases NIK (NF-κB-inducing kinase) and IKKα and is activated when plasmacytoid dendritic cells sense viral nucleic acids. In the present study, we demonstrate that DDX3 also directly interacts with IKKα and enhances its autophosphorylation and -activation. Modulation of DDX3 expression consequently affected NIK/IKKα-mediated IRF7 phosphorylation and induction of type I interferons. In addition, alternative NF-κB (nuclear factor-κB) activation, another pathway regulated by NIK and IKKα, was also down-regulated in DDX3 knockdown cells. This substantially broadens the effects of DDX3 in innate immune signalling to pathways beyond TBK1/IKKε and IFN induction. Dysregulation of these pathways is involved in disease states, and thus, our research might implicate DDX3 as a potential target for their therapeutic manipulation.


Assuntos
RNA Helicases DEAD-box/metabolismo , Quinase I-kappa B/metabolismo , Interferon Tipo I/metabolismo , Transdução de Sinais , RNA Helicases DEAD-box/genética , Ativação Enzimática , Células HEK293 , Humanos , Fator Regulador 7 de Interferon/metabolismo , NF-kappa B/metabolismo , Fosforilação , Ligação Proteica , Proteínas Serina-Treonina Quinases/metabolismo , Interferência de RNA , Células THP-1 , Quinase Induzida por NF-kappaB
6.
Eur J Cell Biol ; 97(7): 501-511, 2018 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-30131165

RESUMO

The human DEAD-box helicase DDX3 is a multi-functional protein involved in the regulation of gene expression and additional non-conventional roles as signalling adaptor molecule that are independent of its enzymatic RNA remodeling activity. It is a nucleo-cytoplasmic shuttling protein and it has previously been suggested that dysregulation of its subcellular localization could contribute to tumourigenesis. Indeed, both tumour suppressor and oncogenic functions have been attributed to DDX3. In this study, we investigated the regulation of DDX3's nucleocytoplasmic shuttling. We confirmed that an N-terminal conserved Nuclear Export Signal (NES) is required for export of human DDX3 from the nucleus, and identified three regions within DDX3 that can independently facilitate its nuclear import. We also aimed to identify conditions that alter DDX3's subcellular localisation. Viral infection, cytokine treatment and DNA damage only induced minor changes in DDX3's subcellular distribution as determined by High Content Analysis. However, DDX3's nuclear localization increased in early mitotic cells (during prophase) concomitant with an increase in DDX3 expression levels. Our results are likely to have implications for the proposed use of (nuclear) DDX3 as a prognostic biomarker in cancer.


Assuntos
Núcleo Celular/metabolismo , RNA Helicases DEAD-box/metabolismo , Transporte Ativo do Núcleo Celular , Sequência de Aminoácidos , Ciclo Celular , Sequência Conservada , RNA Helicases DEAD-box/química , Células HEK293 , Células HeLa , Humanos , Carioferinas/metabolismo , Mutação/genética , Sinais de Exportação Nuclear , Sinais de Localização Nuclear/metabolismo , Fosforilação , Fosfotreonina/metabolismo , Ligação Proteica , Domínios Proteicos , Receptores Citoplasmáticos e Nucleares/metabolismo , Frações Subcelulares/metabolismo , Regulação para Cima/genética , Proteína Exportina 1
7.
Biochem J ; 474(4): 571-587, 2017 02 15.
Artigo em Inglês | MEDLINE | ID: mdl-27980081

RESUMO

The human DEAD-box helicase 3 (DDX3) has been shown to contribute to type I interferon (IFN) induction downstream from antiviral pattern recognition receptors. It binds to TANK-binding kinase 1 and IκB-kinase-ε (IKKε), the two key kinases mediating activation of IFN regulatory factor (IRF) 3 and IRF7. We previously demonstrated that DDX3 facilitates IKKε activation downstream from RIG-I and then links the activated kinase to IRF3. In the present study, we probed the interactions between DDX3 and other key signalling molecules in the RIG-I pathway and identified a novel direct interaction between DDX3 and TNF receptor-associated factor 3 (TRAF3) mediated by a TRAF-interaction motif in the N-terminus of DDX3, which was required for TRAF3 ubiquitination. Interestingly, we observed two waves of K63-linked TRAF3 ubiquitination following RIG-I activation by Sendai virus (SeV) infection, both of which were suppressed by DDX3 knockdown. We also investigated the spatiotemporal formation of endogenous downstream signalling complexes containing the mitochondrial antiviral signalling (MAVS) adaptor, DDX3, IκB-kinase-ε (IKKε), TRAF3 and IRF3. DDX3 was recruited to MAVS early after SeV infection, suggesting that it might mediate subsequent recruitment of other molecules. Indeed, knockdown of DDX3 prevented the formation of TRAF3-MAVS and TRAF3-IKKε complexes. Based on our data, we propose that early TRAF3 ubiquitination is required for the formation of a stable MAVS-TRAF3 complex, while the second wave of TRAF3 ubiquitination mediates IRF3 recruitment and activation. Our study characterises DDX3 as a multifunctional adaptor molecule that co-ordinates assembly of different TRAF3, IKKε and IRF3-containing signalling complexes downstream from MAVS. Additionally, it provides novel insights into the role of TRAF3 in RIG-I signalling.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/metabolismo , RNA Helicases DEAD-box/metabolismo , Interações Hospedeiro-Patógeno , Vírus Sendai/metabolismo , Fator 3 Associado a Receptor de TNF/metabolismo , Células A549 , Proteínas Adaptadoras de Transdução de Sinal/genética , Proteína DEAD-box 58/genética , Proteína DEAD-box 58/metabolismo , RNA Helicases DEAD-box/antagonistas & inibidores , RNA Helicases DEAD-box/genética , Regulação da Expressão Gênica , Células HEK293 , Humanos , Quinase I-kappa B/genética , Quinase I-kappa B/metabolismo , Interferons/biossíntese , Interferons/genética , RNA Interferente Pequeno/genética , RNA Interferente Pequeno/metabolismo , Receptores Imunológicos , Vírus Sendai/genética , Vírus Sendai/crescimento & desenvolvimento , Transdução de Sinais , Fator 3 Associado a Receptor de TNF/genética , Ubiquitinação
8.
J Immunol ; 194(7): 3136-46, 2015 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-25750433

RESUMO

We previously showed that the T cell activation inhibitor, mitochondrial (Tcaim) is highly expressed in grafts of tolerance-developing transplant recipients and that the encoded protein is localized within mitochondria. In this study, we show that CD11c(+) dendritic cells (DCs), as main producers of TCAIM, downregulate Tcaim expression after LPS stimulation or in vivo alloantigen challenge. LPS-stimulated TCAIM-overexpressing bone marrow-derived DC (BMDCs) have a reduced capacity to induce proliferation of and cytokine expression by cocultured allogeneic T cells; this is not due to diminished upregulation of MHC or costimulatory molecules. Transcriptional profiling also revealed normal LPS-mediated upregulation of the majority of genes involved in TLR signaling. However, TCAIM BMDCs did not induce Il2 mRNA expression upon LPS stimulation in comparison with Control-BMDCs. In addition, TCAIM overexpression abolished LPS-mediated Ca(2+) influx and mitochondrial reactive oxygen species formation. Addition of IL-2 to BMDC-T cell cocultures restored the priming capacity of TCAIM BMDCs for cocultured allogeneic CD8(+) T cells. Furthermore, BMDCs of IL-2-deficient mice showed similarly abolished LPS-induced T cell priming as TCAIM-overexpressing wild type BMDCs. Thus, TCAIM interferes with TLR4 signaling in BMDCs and subsequently impairs their T cell priming capacity, which supports its role for tolerance induction.


Assuntos
Cálcio/metabolismo , Células Dendríticas/imunologia , Interleucina-2/biossíntese , Proteínas Mitocondriais/genética , Linfócitos T/imunologia , Linfócitos T/metabolismo , Receptores Toll-Like/metabolismo , Animais , Antígeno B7-2/metabolismo , Proteína beta Intensificadora de Ligação a CCAAT/metabolismo , Análise por Conglomerados , Expressão Gênica , Perfilação da Expressão Gênica , Regulação da Expressão Gênica , Antígenos de Histocompatibilidade Classe II/metabolismo , Interleucina-2/genética , Interleucina-2/farmacologia , Lipopolissacarídeos/imunologia , Ativação Linfocitária/efeitos dos fármacos , Ativação Linfocitária/genética , Ativação Linfocitária/imunologia , Masculino , Camundongos , Proteínas Mitocondriais/metabolismo , NF-kappa B/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Transplante de Pele , Linfócitos T/efeitos dos fármacos , Transplante Homólogo
9.
Biochim Biophys Acta ; 1829(8): 854-65, 2013 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-23567047

RESUMO

Traditional functions of DExD/H-box helicases are concerned with RNA metabolism; they have been shown to play a part in nearly every cellular process that involves RNA. On the other hand, it is accepted that DexD/H-box helicases also engage in activities that do not require helicase activity. A number of DExD/H-box helicases have been shown to be involved in anti-viral immunity. The RIG-like helicases, RIG-I, mda5 and lgp2, act as important cytosolic pattern recognition receptors for viral RNA. Detection of viral nucleic acids by the RIG-like helicases or other anti-viral pattern recognition receptors leads to the induction of type I interferons and pro-inflammatory cytokines. More recently, additional DExD/H-box helicases have also been implicated to act as cytosolic sensors of viral nucleic acids, including DDX3, DDX41, DHX9, DDX60, DDX1 and DHX36. However, there is evidence that at least some of these helicases might have more downstream functions in pattern recognition receptor signalling pathways, as signalling adaptors or transcriptional regulators. In an interesting twist, a lot of DExD/H-box helicases have also been identified as essential host factors for the replication of different viruses, suggesting that viruses 'hijack' their RNA helicase activities for their benefit. Interestingly, DDX3, DDX1 and DHX9 are among the helicases that are required for the replication of a diverse range of viruses. This might suggest that these helicases are highly contested targets in the ongoing 'arms race' between viruses and the host immune system. This article is part of a Special Issue entitled: The Biology of RNA helicases - Modulation for life.


Assuntos
RNA Helicases DEAD-box/imunologia , Imunidade Inata/imunologia , Replicação Viral/fisiologia , Animais , RNA Helicases DEAD-box/genética , RNA Helicases DEAD-box/metabolismo , Humanos , Imunidade Inata/genética , RNA Viral/genética , RNA Viral/imunologia , RNA Viral/metabolismo , Replicação Viral/genética , Replicação Viral/imunologia
10.
Mol Cell Biol ; 33(10): 2004-15, 2013 May.
Artigo em Inglês | MEDLINE | ID: mdl-23478265

RESUMO

The human DEAD box protein 3 (DDX3) has been implicated in different processes contributing to gene expression. Interestingly, DDX3 is required as an essential host factor for the replication of HIV and hepatitis C virus (HCV) and is therefore considered a potential drug target. On the other hand, DDX3 interacts with IκB kinase ε (IKKε) and TANK-binding kinase 1 (TBK1) and contributes to the induction of antiviral type I interferons (IFNs). However, the molecular mechanism by which DDX3 contributes to IFN induction remains unclear. Here we show that DDX3 mediates phosphorylation of interferon regulatory factor 3 (IRF3) by the kinase IKKε. DDX3 directly interacts with IKKε and enhances its autophosphorylation and activation. IKKε then phosphorylates several serine residues in the N terminus of DDX3. Phosphorylation of DDX3 at serine 102 (S102) is required for recruitment of IRF3 to DDX3, facilitating its phosphorylation by IKKε. Mutation of S102 to alanine disrupted the interaction between DDX3 and IRF3 but not that between DDX3 and IKKε. The S102A mutation failed to enhance ifnb promoter activation, suggesting that the DDX3-IRF3 interaction is crucial for this effect. Our data implicates DDX3 as a scaffolding adaptor that directly facilitates phosphorylation of IRF3 by IKKε. DDX3 might thus be involved in pathway-specific activation of IRF3.


Assuntos
RNA Helicases DEAD-box/metabolismo , Quinase I-kappa B/metabolismo , Fator Regulador 3 de Interferon/metabolismo , Sequência de Aminoácidos , Substituição de Aminoácidos , RNA Helicases DEAD-box/química , RNA Helicases DEAD-box/genética , Ativação Enzimática , Expressão Gênica , Genes Reporter , Células HEK293 , Humanos , Quinase I-kappa B/química , Interferon beta/genética , Dados de Sequência Molecular , Mutagênese Sítio-Dirigida , Fragmentos de Peptídeos/química , Mapeamento de Peptídeos , Fosforilação , Regiões Promotoras Genéticas , Ligação Proteica , Domínios e Motivos de Interação entre Proteínas , Processamento de Proteína Pós-Traducional , Ativação Transcricional
11.
J Immunol ; 190(6): 2844-56, 2013 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-23396947

RESUMO

IRAK1 is involved in the regulation of type I IFN production downstream of TLR3. Previous work indicated that IRAK1 negatively regulates TRIF-mediated activation of IRF3 and IRF7. We report that IRAK1 limits the activation of the TLR3-NF-κB pathway. Following TLR3 stimulation, IRAK1-deficient macrophages produced increased levels of IL-6 and IFN-ß compared with wild type macrophages. Pharmacological inhibition of TAK1 reduced this increase in IFN-ß, together with the heightened activation of IRF3 and p65 found in TLR3-ligand stimulated IRAK1-deficient macrophages. Recently, IKKε and TANK-binding kinase 1 (TBK1) were reported to limit activation of the NF-κB pathway downstream of IL-1R, TNFR1, and TLRs. We show that TBK1 has a positive role in the TLR3-NF-κB pathway, because we detected reduced levels of IL-6 and reduced activation of p65 in TBK1-deficient macrophages. In contrast, we show that IKKε limits the activation of the TLR3-NF-κB pathway. Furthermore, we show that IRAK1 is required for the activation of IKKε downstream of TLR3. We report impaired activation of ERK1/2 in IRAK1- and IKKε-deficient macrophages, a novel finding for both kinases. Importantly, this work provides novel mechanistic insight into the regulation of the TLR3-signaling pathway, providing strong evidence that an IRAK1-IKKε-signaling axis acts to limit the production of both type I IFNs and proinflammatory cytokines by regulating TAK1 activity.


Assuntos
Proteínas de Transporte/metabolismo , Regulação para Baixo/imunologia , Quinase I-kappa B/fisiologia , Quinases Associadas a Receptores de Interleucina-1/fisiologia , MAP Quinase Quinase Quinases/metabolismo , Sistema de Sinalização das MAP Quinases/imunologia , Receptor 3 Toll-Like/metabolismo , Animais , Proteínas de Transporte/antagonistas & inibidores , Linhagem Celular , Regulação para Baixo/genética , Células HEK293 , Humanos , Quinase I-kappa B/genética , Inflamação/enzimologia , Inflamação/genética , Inflamação/imunologia , Quinases Associadas a Receptores de Interleucina-1/genética , Peptídeos e Proteínas de Sinalização Intracelular , MAP Quinase Quinase Quinases/antagonistas & inibidores , MAP Quinase Quinase Quinases/genética , Sistema de Sinalização das MAP Quinases/genética , Camundongos , Microglia/enzimologia , Microglia/imunologia , Microglia/patologia , Mapeamento de Interação de Proteínas , Receptor 3 Toll-Like/genética , Fatores de Elongação da Transcrição
12.
Biochem Soc Trans ; 39(2): 679-83, 2011 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-21428961

RESUMO

Human DDX3 is a DEAD (Asp-Glu-Ala-Asp)-box RNA helicase that appears to be a prime target for viral manipulation. While two viruses that manifest major global health threats, HIV and HCV (hepatitis C virus), utilize DDX3 for their replication, other viruses inhibit DDX3's newly identified function in innate antiviral signalling. This review discusses the role of DDX3 in antiviral immunity and its inhibition or exploitation by different viruses.


Assuntos
RNA Helicases DEAD-box/fisiologia , Fenômenos Fisiológicos Virais , Animais , RNA Helicases DEAD-box/metabolismo , Replicação do DNA/genética , Replicação do DNA/fisiologia , Regulação para Baixo/fisiologia , Humanos , Imunidade Inata/genética , Imunidade Inata/fisiologia , Modelos Biológicos , Fenômenos Fisiológicos Virais/genética , Replicação Viral/genética , Replicação Viral/fisiologia , Vírus/genética , Vírus/crescimento & desenvolvimento , Vírus/imunologia , Vírus/metabolismo
13.
Biochem Pharmacol ; 79(3): 297-306, 2010 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-19782656

RESUMO

The human DEAD-box RNA helicase DDX3 has been implicated to play a role in the whole repertoire of processes regulating gene expression, including transcription, splicing, mRNA export and translation. It has also been suggested to be involved in cell cycle control and the regulation of apoptosis. In addition, DDX3 was recently shown to be part of innate immune signalling pathways and to contribute to the induction of anti-viral mediators, such as type I interferon. Interestingly, DDX3 appears to be a prime target for viral manipulation: at least four different viruses, namely Hepatitis C virus (HCV), Hepatitis B virus (HBV), Human Immunodeficiency Virus (HIV) and poxviruses, encode proteins that interact with DDX3 and modulate its function. HIV and HCV seem to co-opt DDX3 and require it for their replication. It has therefore been suggested that DDX3 could be a novel target for the development of drugs against these two viruses, both of which still pose major global health threats. However, in the light of the apparent multifunctionality of DDX3 in the cell, drug development strategies targeting DDX3 will have to be carefully evaluated. This review summarises the available data on the cellular functions of DDX3 and discusses their manipulation by the different viruses known to target DDX3. Understanding the viral strategies for manipulating or co-opting DDX3 in functional and molecular detail can provide valuable insights for the development of strategies to therapeutically target DDX3.


Assuntos
Antivirais/farmacologia , Ciclo Celular/fisiologia , RNA Helicases DEAD-box/fisiologia , Sistemas de Liberação de Medicamentos , Regulação da Expressão Gênica/fisiologia , Animais , Antivirais/metabolismo , Ciclo Celular/efeitos dos fármacos , RNA Helicases DEAD-box/antagonistas & inibidores , Sistemas de Liberação de Medicamentos/métodos , Regulação da Expressão Gênica/efeitos dos fármacos , Vírus de Hepatite/efeitos dos fármacos , Vírus de Hepatite/fisiologia , Humanos
14.
Structure ; 17(11): 1528-37, 2009 Nov 11.
Artigo em Inglês | MEDLINE | ID: mdl-19913487

RESUMO

Poxviruses are DNA viruses that express numerous proteins to subvert the host immune response. Vaccinia virus protein K7 adopts a Bcl-2 fold and displays structural and functional similarities to Toll-like receptor antagonist A52. Both proteins interact with IRAK2 and TRAF6 and suppress TLR-dependent NF-kappaB activation. However, unlike A52, K7 also forms a complex with RNA helicase DDX3 and antagonizes interferon-beta promoter induction. We have narrowed the K7 binding site to an N-terminal peptide motif of DDX3 ahead of its core RNA-helicase domains. The crystal structure of full-length K7 in complex with the DDX3 peptide reveals a thumblike projection of tandem phenalyalanine residues of DDX3 into a deep hydrophobic cleft. Mutagenesis of these phenylalanines abolishes the effects of DDX3 on interferon-beta promoter induction. The structure of K7-DDX3 reveals a novel binding mode by a viral Bcl-2 protein that antagonizes a key pathway in innate immunity.


Assuntos
RNA Helicases DEAD-box/química , Imunidade Inata/imunologia , Modelos Moleculares , Vaccinia virus/química , Proteínas Virais/metabolismo , Sequência de Aminoácidos , Sítios de Ligação/genética , Cristalização , RNA Helicases DEAD-box/metabolismo , Humanos , Interferon beta/antagonistas & inibidores , Quinases Associadas a Receptores de Interleucina-1/metabolismo , Dados de Sequência Molecular , Mutagênese , Alinhamento de Sequência , Fator 6 Associado a Receptor de TNF/metabolismo , Proteínas Virais/genética
15.
Eur J Immunol ; 39(4): 1066-77, 2009 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-19266486

RESUMO

IL-10 is a potent immunoregulatory and anti-inflammatory cytokine. However, therapeutic trials in chronic inflammation have been largely disappointing. It is well established that IL-10 can inhibit Th1 and Th2 cytokine production via indirect effects on APC. Less data are available about the influence of IL-10 on IL-17 production, a cytokine which has been recently linked to chronic inflammation. Furthermore, there are only few reports about a direct effect of IL-10 on T cells. We demonstrate here that IL-10 can directly interfere with TCR-induced IFN-gamma production in freshly isolated memory T cells in the absence of APC. This effect was independent of the previously described effects of IL-10 on T cells, namely inhibition of IL-2 production and inhibition of CD28 signaling. In contrast, IL-10 did not affect anti-CD3/anti-CD28-induced IL-17 production from memory T cells even in the presence of APC. This might have implications for the interpretation of therapeutic trials in patients with chronic inflammation where Th17 cells contribute to pathogenesis.


Assuntos
Interferon gama/antagonistas & inibidores , Interleucina-10/farmacologia , Interleucina-17/biossíntese , Receptores de Antígenos de Linfócitos T/metabolismo , Subpopulações de Linfócitos T/efeitos dos fármacos , Células Apresentadoras de Antígenos/imunologia , Antígenos de Fungos/imunologia , Candida albicans/imunologia , Células Cultivadas , Humanos , Memória Imunológica/efeitos dos fármacos , Memória Imunológica/imunologia , Interferon gama/metabolismo , Interleucina-17/antagonistas & inibidores , Interleucina-2/imunologia , Interleucina-2/metabolismo , Neutrófilos/efeitos dos fármacos , Neutrófilos/imunologia , Proteína Tirosina Fosfatase não Receptora Tipo 6/imunologia , Proteína Tirosina Fosfatase não Receptora Tipo 6/metabolismo , Receptores de Antígenos de Linfócitos T/agonistas , Fator de Transcrição STAT3/metabolismo , Proteína 3 Supressora da Sinalização de Citocinas , Proteínas Supressoras da Sinalização de Citocina/metabolismo , Subpopulações de Linfócitos T/imunologia
16.
J Mol Biol ; 385(3): 843-53, 2009 Jan 23.
Artigo em Inglês | MEDLINE | ID: mdl-18845156

RESUMO

Poxviruses have evolved numerous strategies to evade host innate immunity. Vaccinia virus K7 is a 149-residue protein with previously unknown structure that is highly conserved in the orthopoxvirus family. K7 bears sequence and functional similarities to A52, which interacts with interleukin receptor-associated kinase 2 and tumor necrosis factor receptor-associated factor 6 to suppress nuclear factor kappaB activation and to stimulate the secretion of the anti-inflammatory cytokine interleukin-10. In contrast to A52, K7 forms a complex with DEAD box RNA helicase DDX3, thereby suppressing DDX3-mediated ifnb promoter induction. We determined the NMR solution structure of K7 to provide insight into the structural basis for poxvirus antagonism of innate immune signaling. The structure reveals an alpha-helical fold belonging to the Bcl-2 family despite an unrelated primary sequence. NMR chemical-shift mapping studies have localized the binding surface for DDX3 on a negatively charged face of K7. Furthermore, thermodynamic studies have mapped the K7-binding region to a 30-residue N-terminal fragment of DDX3, ahead of the core RNA helicase domains.


Assuntos
RNA Helicases DEAD-box/metabolismo , Poxviridae/metabolismo , Proteínas Virais/metabolismo , Cromatografia em Gel , Humanos , Espectroscopia de Ressonância Magnética , Ligação Proteica
17.
EMBO J ; 27(15): 2147-57, 2008 Aug 06.
Artigo em Inglês | MEDLINE | ID: mdl-18636090

RESUMO

Viruses are detected by different classes of pattern recognition receptors (PRRs), such as Toll-like receptors and RIG-like helicases. Engagement of PRRs leads to activation of interferon (IFN)-regulatory factor 3 (IRF3) and IRF7 through IKKepsilon and TBK1 and consequently IFN-beta induction. Vaccinia virus (VACV) encodes proteins that manipulate host signalling, sometimes by targeting uncharacterised proteins. Here, we describe a novel VACV protein, K7, which can inhibit PRR-induced IFN-beta induction by preventing TBK1/IKKepsilon-mediated IRF activation. We identified DEAD box protein 3 (DDX3) as a host target of K7. Expression of DDX3 enhanced Ifnb promoter induction by TBK1/IKKepsilon, whereas knockdown of DDX3 inhibited this, and virus- or dsRNA-induced IRF3 activation. Further, dominant-negative DDX3 inhibited virus-, dsRNA- and cytosolic DNA-stimulated Ccl5 promoter induction, which is also TBK1/IKKepsilon dependent. Both K7 binding and enhancement of Ifnb induction mapped to the N-terminus of DDX3. Furthermore, virus infection induced an association between DDX3 and IKKepsilon. Therefore, this study shows for the first time the involvement of a DEAD box helicase in TBK1/IKKepsilon-mediated IRF activation and Ifnb promoter induction.


Assuntos
RNA Helicases DEAD-box/metabolismo , Quinase I-kappa B/metabolismo , Fator Regulador 3 de Interferon/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , Proteínas Virais/fisiologia , Sequência de Aminoácidos , Linhagem Celular , Núcleo Celular/metabolismo , Quimiocina CCL5/genética , Citoplasma/metabolismo , Humanos , Fator Regulador 7 de Interferon/metabolismo , Interferon beta/biossíntese , Dados de Sequência Molecular , Regiões Promotoras Genéticas , Receptores de Reconhecimento de Padrão/metabolismo , Vaccinia virus/metabolismo
18.
Nat Immunol ; 7(10): 1074-81, 2006 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-16964262

RESUMO

Toll-like receptors discriminate between different pathogen-associated molecules and activate signaling cascades that lead to immune responses. The specificity of Toll-like receptor signaling occurs by means of adaptor proteins containing Toll-interleukin 1 receptor (TIR) domains. Activating functions have been assigned to four TIR adaptors: MyD88, Mal, TRIF and TRAM. Here we characterize a fifth TIR adaptor, SARM, as a negative regulator of TRIF-dependent Toll-like receptor signaling. Expression of SARM blocked gene induction 'downstream' of TRIF but not of MyD88. SARM associated with TRIF, and 'knockdown' of endogenous SARM expression by interfering RNA led to enhanced TRIF-dependent cytokine and chemokine induction. Thus, the fifth mammalian TIR adaptor SARM is a negative regulator of Toll-like receptor signaling.


Assuntos
Proteínas Adaptadoras de Transporte Vesicular/antagonistas & inibidores , Proteínas do Domínio Armadillo/metabolismo , Citocinas/genética , Proteínas do Citoesqueleto/metabolismo , Regulação da Expressão Gênica , Receptores Toll-Like/metabolismo , Proteínas Adaptadoras de Transporte Vesicular/genética , Proteínas Adaptadoras de Transporte Vesicular/metabolismo , Proteínas do Domínio Armadillo/antagonistas & inibidores , Proteínas do Domínio Armadillo/genética , Células Cultivadas , Quimiocinas/genética , Proteínas do Citoesqueleto/antagonistas & inibidores , Proteínas do Citoesqueleto/genética , Humanos , Fator Regulador 7 de Interferon/antagonistas & inibidores , Fator 88 de Diferenciação Mieloide/metabolismo , NF-kappa B/metabolismo , Estrutura Terciária de Proteína/genética , Interferência de RNA , RNA Interferente Pequeno/farmacologia , Transdução de Sinais/genética , Receptor 3 Toll-Like/metabolismo , Receptor 4 Toll-Like/metabolismo , Ativação Transcricional
19.
J Biol Chem ; 280(35): 30838-44, 2005 Sep 02.
Artigo em Inglês | MEDLINE | ID: mdl-15998638

RESUMO

Vaccinia virus (VV) has many mechanisms to suppress and modulate the host immune response. The VV protein A52R was previously shown to act as an intracellular inhibitor of nuclear factor kappaB (NFkappaB) signaling by Toll-like receptors (TLRs). Co-immunoprecipitation studies revealed that A52R interacted with both tumor necrosis factor receptor-associated factor 6 (TRAF6) and interleukin-1 receptor-associated kinase 2 (IRAK2). The effect of A52R on signals other than NFkappaB was not determined. Here, we show that A52R does not inhibit TLR-induced p38 or c-Jun amino N-terminal kinase (JNK) mitogen activating protein (MAP) kinase activation. Rather, A52R could drive activation of these kinases. Two lines of evidence suggested that the A52R/TRAF6 interaction was critical for these effects. First, A52R-induced p38 MAP kinase activation was inhibited by overexpression of the TRAF domain of TRAF6, which sequestered A52R and inhibited its interaction with endogenous TRAF6. Second, a truncated version of A52R, which interacted with IRAK2 and not TRAF6, was unable to activate p38. Because interleukin 10 (IL-10) production is strongly p38-dependent, we examined the effect of A52R on IL-10 gene induction. A52R was found to be capable of inducing the IL-10 promoter through a TRAF6-dependent mechanism. Furthermore, A52R enhanced lipopolysaccharide/TLR4-induced IL-10 production, while inhibiting the TLR-induced NFkappaB-dependent genes IL-8 and RANTES. These results show that although A52R inhibits NFkappaB activation by multiple TLRs it can simultaneously activate MAP kinases. A52R-mediated enhancement of TLR-induced IL-10 may be important to virulence, given the role of IL-10 in immunoregulation.


Assuntos
Interleucina-10/metabolismo , Lipopolissacarídeos/metabolismo , Sistema de Sinalização das MAP Quinases/fisiologia , Proteínas Virais/metabolismo , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo , Animais , Linhagem Celular , Ativação Enzimática , Regulação da Expressão Gênica , Genes Reporter , Humanos , Quinases Associadas a Receptores de Interleucina-1 , Interleucina-10/genética , Proteínas Quinases JNK Ativadas por Mitógeno/metabolismo , Camundongos , NF-kappa B/metabolismo , Regiões Promotoras Genéticas , Proteínas Quinases/genética , Proteínas Quinases/metabolismo , Estrutura Terciária de Proteína , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/metabolismo , Fator 6 Associado a Receptor de TNF/genética , Fator 6 Associado a Receptor de TNF/metabolismo , Ativação Transcricional , Proteínas Virais/genética , Proteínas Quinases p38 Ativadas por Mitógeno/genética
20.
Trends Immunol ; 26(9): 462-8, 2005 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-16027039

RESUMO

Toll-like receptor 3 (TLR3), which recognizes double-stranded (ds)RNA, was the first identified antiviral TLR and, because dsRNA is a universal viral molecular pattern, TLR3 has been assumed to have a central role in the host response to viruses. However, this role has recently been questioned by in vivo studies and the discovery of several other antiviral pattern-recognition receptors. In this review, the function of TLR3 in the context of these other receptors, namely TLR7, 8 and 9 and the newly identified dsRNA-receptor retinoic-acid inducible gene-I (RIG-I) is discussed. Also, recent research concerning the expression profile of TLR3, its evasion by viruses and a potential role in crosspriming is addressed, which reveals a clearer appreciation of the contribution of TLR3 to antiviral immunity.


Assuntos
Viroses/imunologia , Animais , Antígenos Virais/imunologia , Proteína DEAD-box 58 , RNA Helicases DEAD-box , Humanos , Células Matadoras Naturais/imunologia , Modelos Imunológicos , RNA Helicases/imunologia , RNA de Cadeia Dupla/imunologia , Receptores Imunológicos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA