Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 24
Filter
1.
FASEB J ; 33(8): 8732-8744, 2019 08.
Article in English | MEDLINE | ID: mdl-31163989

ABSTRACT

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.


Subject(s)
Hepatitis C/metabolism , MAP Kinase Signaling System , STAT3 Transcription Factor/metabolism , Suppressor of Cytokine Signaling 3 Protein/genetics , Tumor Necrosis Factor-alpha/metabolism , Viral Proteins/metabolism , Cell Line, Tumor , HEK293 Cells , Humans , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , NF-kappa B/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Suppressor of Cytokine Signaling 3 Protein/metabolism , Up-Regulation
2.
Biochem J ; 475(22): 3595-3607, 2018 11 20.
Article in English | MEDLINE | ID: mdl-30341167

ABSTRACT

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.


Subject(s)
DEAD-box RNA Helicases/metabolism , I-kappa B Kinase/metabolism , Interferon Type I/metabolism , Signal Transduction , DEAD-box RNA Helicases/genetics , Enzyme Activation , HEK293 Cells , Humans , Interferon Regulatory Factor-7/metabolism , NF-kappa B/metabolism , Phosphorylation , Protein Binding , Protein Serine-Threonine Kinases/metabolism , RNA Interference , THP-1 Cells , NF-kappaB-Inducing Kinase
3.
Biochem J ; 474(4): 571-587, 2017 02 15.
Article in English | MEDLINE | ID: mdl-27980081

ABSTRACT

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.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , DEAD-box RNA Helicases/metabolism , Host-Pathogen Interactions , Sendai virus/metabolism , TNF Receptor-Associated Factor 3/metabolism , A549 Cells , Adaptor Proteins, Signal Transducing/genetics , DEAD Box Protein 58/genetics , DEAD Box Protein 58/metabolism , DEAD-box RNA Helicases/antagonists & inhibitors , DEAD-box RNA Helicases/genetics , Gene Expression Regulation , HEK293 Cells , Humans , I-kappa B Kinase/genetics , I-kappa B Kinase/metabolism , Interferons/biosynthesis , Interferons/genetics , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Receptors, Immunologic , Sendai virus/genetics , Sendai virus/growth & development , Signal Transduction , TNF Receptor-Associated Factor 3/genetics , Ubiquitination
4.
J Immunol ; 194(7): 3136-46, 2015 Apr 01.
Article in English | MEDLINE | ID: mdl-25750433

ABSTRACT

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.


Subject(s)
Calcium/metabolism , Dendritic Cells/immunology , Interleukin-2/biosynthesis , Mitochondrial Proteins/genetics , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Toll-Like Receptors/metabolism , Animals , B7-2 Antigen/metabolism , CCAAT-Enhancer-Binding Protein-beta/metabolism , Cluster Analysis , Gene Expression , Gene Expression Profiling , Gene Expression Regulation , Histocompatibility Antigens Class II/metabolism , Interleukin-2/genetics , Interleukin-2/pharmacology , Lipopolysaccharides/immunology , Lymphocyte Activation/drug effects , Lymphocyte Activation/genetics , Lymphocyte Activation/immunology , Male , Mice , Mitochondrial Proteins/metabolism , NF-kappa B/metabolism , Reactive Oxygen Species/metabolism , Skin Transplantation , T-Lymphocytes/drug effects , Transplantation, Homologous
5.
J Immunol ; 190(6): 2844-56, 2013 Mar 15.
Article in English | MEDLINE | ID: mdl-23396947

ABSTRACT

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.


Subject(s)
Carrier Proteins/metabolism , Down-Regulation/immunology , I-kappa B Kinase/physiology , Interleukin-1 Receptor-Associated Kinases/physiology , MAP Kinase Kinase Kinases/metabolism , MAP Kinase Signaling System/immunology , Toll-Like Receptor 3/metabolism , Animals , Carrier Proteins/antagonists & inhibitors , Cell Line , Down-Regulation/genetics , HEK293 Cells , Humans , I-kappa B Kinase/genetics , Inflammation/enzymology , Inflammation/genetics , Inflammation/immunology , Interleukin-1 Receptor-Associated Kinases/genetics , Intracellular Signaling Peptides and Proteins , MAP Kinase Kinase Kinases/antagonists & inhibitors , MAP Kinase Kinase Kinases/genetics , MAP Kinase Signaling System/genetics , Mice , Microglia/enzymology , Microglia/immunology , Microglia/pathology , Protein Interaction Mapping , Toll-Like Receptor 3/genetics , Transcriptional Elongation Factors
6.
Biochim Biophys Acta ; 1829(8): 854-65, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23567047

ABSTRACT

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.


Subject(s)
DEAD-box RNA Helicases/immunology , Immunity, Innate/immunology , Virus Replication/physiology , Animals , DEAD-box RNA Helicases/genetics , DEAD-box RNA Helicases/metabolism , Humans , Immunity, Innate/genetics , RNA, Viral/genetics , RNA, Viral/immunology , RNA, Viral/metabolism , Virus Replication/genetics , Virus Replication/immunology
7.
EMBO J ; 27(15): 2147-57, 2008 Aug 06.
Article in English | MEDLINE | ID: mdl-18636090

ABSTRACT

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.


Subject(s)
DEAD-box RNA Helicases/metabolism , I-kappa B Kinase/metabolism , Interferon Regulatory Factor-3/metabolism , Protein Serine-Threonine Kinases/metabolism , Viral Proteins/physiology , Amino Acid Sequence , Cell Line , Cell Nucleus/metabolism , Chemokine CCL5/genetics , Cytoplasm/metabolism , Humans , Interferon Regulatory Factor-7/metabolism , Interferon-beta/biosynthesis , Molecular Sequence Data , Promoter Regions, Genetic , Receptors, Pattern Recognition/metabolism , Vaccinia virus/metabolism
8.
Front Cell Dev Biol ; 10: 1033684, 2022.
Article in English | MEDLINE | ID: mdl-36393867

ABSTRACT

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.

9.
Biochim Biophys Acta Gene Regul Mech ; 1865(2): 194787, 2022 02.
Article in English | MEDLINE | ID: mdl-35121200

ABSTRACT

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.


Subject(s)
Breast Neoplasms , DEAD-box RNA Helicases , Estrogen Receptor alpha , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Cell Proliferation , DEAD-box RNA Helicases/genetics , DEAD-box RNA Helicases/metabolism , Estrogen Receptor alpha/genetics , Estrogen Receptor alpha/metabolism , Female , Humans , Receptors, Estrogen , Signal Transduction
10.
J Exp Med ; 201(6): 1007-18, 2005 Mar 21.
Article in English | MEDLINE | ID: mdl-15767367

ABSTRACT

Viral immune evasion strategies target key aspects of the host antiviral response. Recently, it has been recognized that Toll-like receptors (TLRs) have a role in innate defense against viruses. Here, we define the function of the vaccinia virus (VV) protein A46R and show it inhibits intracellular signalling by a range of TLRs. TLR signalling is triggered by homotypic interactions between the Toll-like-interleukin-1 resistance (TIR) domains of the receptors and adaptor molecules. A46R contains a TIR domain and is the only viral TIR domain-containing protein identified to date. We demonstrate that A46R targets the host TIR adaptors myeloid differentiation factor 88 (MyD88), MyD88 adaptor-like, TIR domain-containing adaptor inducing IFN-beta (TRIF), and the TRIF-related adaptor molecule and thereby interferes with downstream activation of mitogen-activated protein kinases and nuclear factor kappaB. TRIF mediates activation of interferon (IFN) regulatory factor 3 (IRF3) and induction of IFN-beta by TLR3 and TLR4 and suppresses VV replication in macrophages. Here, A46R disrupted TRIF-induced IRF3 activation and induction of the TRIF-dependent gene regulated on activation, normal T cell expressed and secreted. Furthermore, we show that A46R is functionally distinct from another described VV TLR inhibitor, A52R. Importantly, VV lacking the A46R gene was attenuated in a murine intranasal model, demonstrating the importance of A46R for VV virulence.


Subject(s)
Adaptor Proteins, Vesicular Transport/metabolism , Antigens, Differentiation/metabolism , MAP Kinase Signaling System/physiology , Membrane Glycoproteins/metabolism , Receptors, Cell Surface/metabolism , Receptors, Immunologic/metabolism , Vaccinia virus/metabolism , Viral Proteins/metabolism , ATP-Binding Cassette Transporters , Adaptor Proteins, Signal Transducing , Amino Acid Sequence , Animals , Cell Line , DNA-Binding Proteins/metabolism , Disease Models, Animal , Gene Expression Regulation, Viral/genetics , Gene Expression Regulation, Viral/physiology , Humans , Interferon Regulatory Factor-3 , Interferon-beta/biosynthesis , Lymphocyte Activation/genetics , Lymphocyte Activation/physiology , MAP Kinase Signaling System/genetics , Mice , Molecular Sequence Data , Myeloid Differentiation Factor 88 , Periplasmic Binding Proteins , Protein Structure, Tertiary/genetics , Protein Structure, Tertiary/physiology , T-Lymphocytes/physiology , Toll-Like Receptor 3 , Toll-Like Receptor 4 , Toll-Like Receptors , Transcription Factors/metabolism , Vaccinia virus/genetics , Vaccinia virus/pathogenicity , Viral Proteins/genetics , Virus Diseases/genetics , Virus Diseases/physiopathology , Virus Replication/genetics , Virus Replication/physiology
11.
Biochem Soc Trans ; 39(2): 679-83, 2011 Apr.
Article in English | MEDLINE | ID: mdl-21428961

ABSTRACT

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.


Subject(s)
DEAD-box RNA Helicases/physiology , Virus Physiological Phenomena , Animals , DEAD-box RNA Helicases/metabolism , DNA Replication/genetics , DNA Replication/physiology , Down-Regulation/physiology , Humans , Immunity, Innate/genetics , Immunity, Innate/physiology , Models, Biological , Virus Physiological Phenomena/genetics , Virus Replication/genetics , Virus Replication/physiology , Viruses/genetics , Viruses/growth & development , Viruses/immunology , Viruses/metabolism
12.
Front Immunol ; 12: 653883, 2021.
Article in English | MEDLINE | ID: mdl-34054816

ABSTRACT

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.


Subject(s)
DEAD-box RNA Helicases/metabolism , Inflammasomes/metabolism , Interferon Type I/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , NLR Proteins/metabolism , Chromatography, Liquid , Cytokines/metabolism , Humans , Immunity, Innate , Inflammation Mediators , Protein Binding , Tandem Mass Spectrometry
13.
Eur J Immunol ; 39(4): 1066-77, 2009 Apr.
Article in English | MEDLINE | ID: mdl-19266486

ABSTRACT

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.


Subject(s)
Interferon-gamma/antagonists & inhibitors , Interleukin-10/pharmacology , Interleukin-17/biosynthesis , Receptors, Antigen, T-Cell/metabolism , T-Lymphocyte Subsets/drug effects , Antigen-Presenting Cells/immunology , Antigens, Fungal/immunology , Candida albicans/immunology , Cells, Cultured , Humans , Immunologic Memory/drug effects , Immunologic Memory/immunology , Interferon-gamma/metabolism , Interleukin-17/antagonists & inhibitors , Interleukin-2/immunology , Interleukin-2/metabolism , Neutrophils/drug effects , Neutrophils/immunology , Protein Tyrosine Phosphatase, Non-Receptor Type 6/immunology , Protein Tyrosine Phosphatase, Non-Receptor Type 6/metabolism , Receptors, Antigen, T-Cell/agonists , STAT3 Transcription Factor/metabolism , Suppressor of Cytokine Signaling 3 Protein , Suppressor of Cytokine Signaling Proteins/metabolism , T-Lymphocyte Subsets/immunology
14.
Eur J Cell Biol ; 97(7): 501-511, 2018 Sep.
Article in English | MEDLINE | ID: mdl-30131165

ABSTRACT

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.


Subject(s)
Cell Nucleus/metabolism , DEAD-box RNA Helicases/metabolism , Active Transport, Cell Nucleus , Amino Acid Sequence , Cell Cycle , Conserved Sequence , DEAD-box RNA Helicases/chemistry , HEK293 Cells , HeLa Cells , Humans , Karyopherins/metabolism , Mutation/genetics , Nuclear Export Signals , Nuclear Localization Signals/metabolism , Phosphorylation , Phosphothreonine/metabolism , Protein Binding , Protein Domains , Receptors, Cytoplasmic and Nuclear/metabolism , Subcellular Fractions/metabolism , Up-Regulation/genetics , Exportin 1 Protein
15.
J Leukoc Biol ; 75(5): 792-7, 2004 May.
Article in English | MEDLINE | ID: mdl-14742639

ABSTRACT

Signal transducers and activators of transcription (STATs) are crucial molecules in cytokine signaling. In the conventional model of STAT activation, STAT molecules are recruited from a latent pool of cytoplasmic monomers to the activated cytokine receptor. After binding to the receptor, they get tyrosine-phosphorylated, dissociate from the receptor, and translocate to the nucleus as activation-induced dimers. Recently, several publications questioned this model of STAT activation and showed the existence of preassociated STAT molecules before activation. We were able to demonstrate the existence of these preassociated STAT3 molecules in living mammalian cells using bioluminescence resonance energy transfer. Our results support the new hypothesis that STAT molecules exist in the cytoplasm as dimers or multimers and point to an activation-induced change in STAT3 conformation. Therefore, we propose a new model of STAT activation and discuss a hypothetical structure of "cytoplasmic" STAT dimers as opposed to the known "activation-induced" dimer.


Subject(s)
DNA-Binding Proteins/metabolism , Trans-Activators/metabolism , Animals , COS Cells , Chlorocebus aethiops , DNA-Binding Proteins/genetics , Dimerization , Energy Transfer , Luciferases , Luminescent Measurements , Protein Binding , Recombinant Fusion Proteins , STAT3 Transcription Factor , Trans-Activators/genetics , Transfection
16.
Virchows Arch ; 444(2): 149-52, 2004 Feb.
Article in English | MEDLINE | ID: mdl-14691721

ABSTRACT

The human trophoblast has the capacity to invade maternal tissue in a controlled fashion and to produce a wide range of hormones. The transcription factors belonging to the CCAAT/enhancer-binding protein (C/EBP) family are regulators of intracellular processes and mediators of hormone action. C/EBP binding sites have been described in the promoters of several placenta-expressed target genes. In the present study, we used immunohistochemistry and Western-blot analysis to investigate the expression pattern of the three most important members of this family, C/EBP-alpha, -beta, and -delta, in the normal human placenta as well as in isolated trophoblast cell populations. We found C/EBP-alpha and C/EBP-beta expression in the villous syncytiotrophophoblast (ST) and the extravillous (intermediate) trophoblast (EVT), but it was absent from the villous cytotrophoblast (CT). Interestingly, expression of C/EBP-beta continued to be very strong up to the third trimester of pregnancy, especially in the ST. C/EBP-delta showed overall lower expression levels, stronger only in the EVT, while CT/ST showed very low/negative expression. These data show for the first time the expression pattern and tissue localization of C/EBP factors in the human placenta, indicating that these factors (especially C/EBP-beta) may play important roles in the regulation of placenta-specific genes and processes.


Subject(s)
CCAAT-Enhancer-Binding Proteins/biosynthesis , Placenta/metabolism , Blotting, Western , Cells, Cultured , Female , Gene Expression Regulation , Humans , Immunohistochemistry , Pregnancy , Pregnancy Trimesters , Trophoblasts/metabolism
17.
Mol Cell Biol ; 33(10): 2004-15, 2013 May.
Article in English | MEDLINE | ID: mdl-23478265

ABSTRACT

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.


Subject(s)
DEAD-box RNA Helicases/metabolism , I-kappa B Kinase/metabolism , Interferon Regulatory Factor-3/metabolism , Amino Acid Sequence , Amino Acid Substitution , DEAD-box RNA Helicases/chemistry , DEAD-box RNA Helicases/genetics , Enzyme Activation , Gene Expression , Genes, Reporter , HEK293 Cells , Humans , I-kappa B Kinase/chemistry , Interferon-beta/genetics , Molecular Sequence Data , Mutagenesis, Site-Directed , Peptide Fragments/chemistry , Peptide Mapping , Phosphorylation , Promoter Regions, Genetic , Protein Binding , Protein Interaction Domains and Motifs , Protein Processing, Post-Translational , Transcriptional Activation
18.
Biochem Pharmacol ; 79(3): 297-306, 2010 Feb 01.
Article in English | MEDLINE | ID: mdl-19782656

ABSTRACT

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.


Subject(s)
Antiviral Agents/pharmacology , Cell Cycle/physiology , DEAD-box RNA Helicases/physiology , Drug Delivery Systems , Gene Expression Regulation/physiology , Animals , Antiviral Agents/metabolism , Cell Cycle/drug effects , DEAD-box RNA Helicases/antagonists & inhibitors , Drug Delivery Systems/methods , Gene Expression Regulation/drug effects , Hepatitis Viruses/drug effects , Hepatitis Viruses/physiology , Humans
19.
Structure ; 17(11): 1528-37, 2009 Nov 11.
Article in English | MEDLINE | ID: mdl-19913487

ABSTRACT

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.


Subject(s)
DEAD-box RNA Helicases/chemistry , Immunity, Innate/immunology , Models, Molecular , Vaccinia virus/chemistry , Viral Proteins/metabolism , Amino Acid Sequence , Binding Sites/genetics , Crystallization , DEAD-box RNA Helicases/metabolism , Humans , Interferon-beta/antagonists & inhibitors , Interleukin-1 Receptor-Associated Kinases/metabolism , Molecular Sequence Data , Mutagenesis , Sequence Alignment , TNF Receptor-Associated Factor 6/metabolism , Viral Proteins/genetics
20.
J Mol Biol ; 385(3): 843-53, 2009 Jan 23.
Article in English | MEDLINE | ID: mdl-18845156

ABSTRACT

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.


Subject(s)
DEAD-box RNA Helicases/metabolism , Poxviridae/metabolism , Viral Proteins/metabolism , Chromatography, Gel , Humans , Magnetic Resonance Spectroscopy , Protein Binding
SELECTION OF CITATIONS
SEARCH DETAIL