Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 23
Filter
Add more filters










Publication year range
1.
Front Immunol ; 15: 1279920, 2024.
Article in English | MEDLINE | ID: mdl-38495890

ABSTRACT

Type I interferons play a fundamental role in innate host defense against viral infections by eliciting the induction of an antiviral gene program that serves to inhibit viral replication. Activation of type I interferon is regulated by the IRF3 transcription factor, which undergoes phosphorylation-dependent activation by the upstream kinase, TBK1, during viral infection. However, the mechanisms by which TBK1 achieves activation to support signaling to IRF3 remain incompletely understood. Here we identified the E3 ubiquitin ligase, tripartite motif containing 28 (TRIM28), as a positive regulator of type I interferon activation by facilitating TBK1 signaling. Genetic deletion of TRIM28 via CRISPR-Cas9 editing resulted in impaired type I interferon activation upon both RNA and DNA virus challenge, corresponding with increased susceptibility to virus infections in TRIM28 knockout cells. Mechanistically, TRIM28 interacted with TBK1 and mediated the assembly of K63-linked ubiquitin chains onto TBK1, a post-translational modification shown to augment TBK1 signal transmission events. TRIM28 knockout cells further displayed defective TBK1 phosphorylation and complex assembly with IRF3, resulting in impaired IRF3 phosphorylation. Altogether, our data demonstrate TBK1 to be a novel substrate for TRIM28 and identify TRIM28 as an essential regulatory factor in controlling innate antiviral immune responses.


Subject(s)
Interferon Type I , Protein Serine-Threonine Kinases , Protein Serine-Threonine Kinases/metabolism , Signal Transduction , Phosphorylation , Interferon-beta
2.
Front Physiol ; 13: 887702, 2022.
Article in English | MEDLINE | ID: mdl-36479348

ABSTRACT

The rising prevalence of obesity presents a world-wide challenge as it is associated with numerous comorbidities including cardiovascular disease, insulin resistance and hypertension. Obesity-associated illnesses are estimated to cause nearly 4 million deaths globally per year, therefore there is a critical need to better understand associated pathogenesis, identify new therapeutic targets, and develop new interventions. Emerging data identify a key role for chronic inflammation in mediating obesity related disease states and reveal higher incidence of autoimmune disease development. Of the multiple potential mechanisms linking obesity and autoimmunity, the strongest link has been shown for leptin, a hormone secreted at high levels from obese white adipose tissue. Numerous studies have demonstrated that leptin enhances activation of both arms of the immune system, while its absence protects against development of autoimmunity. Other potential newly discovered mechanisms that contribute to autoimmune pathogenesis are not directly connected but also associated with obesity including sustained platelet activation, gut dysbiosis, and aging. Here we review how obesity instigates autoimmunity, particularly in the context of immune cell activations and adipokine secretion.

3.
Acta Pharm Sin B ; 12(4): 1624-1635, 2022 Apr.
Article in English | MEDLINE | ID: mdl-35251918

ABSTRACT

SARS-CoV-2 is an emerging viral pathogen and a major global public health challenge since December of 2019, with limited effective treatments throughout the pandemic. As part of the innate immune response to viral infection, type I interferons (IFN-I) trigger a signaling cascade that culminates in the activation of hundreds of genes, known as interferon stimulated genes (ISGs), that collectively foster an antiviral state. We report here the identification of a group of type I interferon suppressed genes, including fatty acid synthase (FASN), which are involved in lipid metabolism. Overexpression of FASN or the addition of its downstream product, palmitate, increased viral infection while knockout or knockdown of FASN reduced infection. More importantly, pharmacological inhibitors of FASN effectively blocked infections with a broad range of viruses, including SARS-CoV-2 and its variants of concern. Thus, our studies not only suggest that downregulation of metabolic genes may present an antiviral strategy by type I interferon, but they also introduce the potential for FASN inhibitors to have a therapeutic application in combating emerging infectious diseases such as COVID-19.

4.
Front Cell Infect Microbiol ; 11: 789462, 2021.
Article in English | MEDLINE | ID: mdl-35083167

ABSTRACT

Corona virus disease 2019 (COVID-19) pathogenesis is intimately linked to the severe acute respiratory syndrome corona virus 2 (SARS-CoV-2) and disease severity has been associated with compromised induction of type I interferon (IFN-I) cytokines which coordinate the innate immune response to virus infections. Here we identified the SARS-CoV-2 encoded protein, Spike, as an inhibitor of IFN-I that antagonizes viral RNA pattern recognition receptor RIG-I signaling. Ectopic expression of SARS-CoV-2 Spike blocked RIG-I mediated activation of IFNß and downstream induction of interferon stimulated genes. Consequently, SARS-CoV-2 Spike expressing cells harbored increased RNA viral burden compared to control cells. Co-immunoprecipitation experiments revealed SARS-CoV-2 Spike associated with interferon regulatory factor 3 (IRF3), a key transcription factor that governs IFN-I activation. Co-expression analysis via immunoassays further indicated Spike specifically suppressed IRF3 expression as NF-κB and STAT1 transcription factor levels remained intact. Further biochemical experiments uncovered SARS-CoV-2 Spike potentiated proteasomal degradation of IRF3, implicating a novel mechanism by which SARS-CoV-2 evades the host innate antiviral immune response to facilitate COVID-19 pathogenesis.


Subject(s)
Antiviral Restriction Factors/immunology , COVID-19 , Immunity, Innate , Interferon Regulatory Factor-3 , Spike Glycoprotein, Coronavirus/immunology , COVID-19/immunology , HEK293 Cells , Humans , Interferon Regulatory Factor-3/metabolism , Interferon Type I/metabolism , SARS-CoV-2
5.
Antimicrob Agents Chemother ; 63(12)2019 09 09.
Article in English | MEDLINE | ID: mdl-31527024

ABSTRACT

Azithromycin (AZM) is a widely used antibiotic, with additional antiviral and anti-inflammatory properties that remain poorly understood. Although Zika virus (ZIKV) poses a significant threat to global health, there are currently no vaccines or effective therapeutics against it. Herein, we report that AZM effectively suppresses ZIKV infection in vitro by targeting a late stage in the viral life cycle. Besides that, AZM upregulates the expression of host type I and III interferons and several of their downstream interferon-stimulated genes (ISGs) in response to ZIKV infection. In particular, we found that AZM upregulates the expression of MDA5 and RIG-I, pathogen recognition receptors (PRRs) induced by ZIKV infection, and increases the levels of phosphorylated TBK1 and IRF3. Interestingly, AZM treatment upregulates phosphorylation of TBK1, without inducing phosphorylation of IRF3 by itself. These findings highlight the potential use of AZM as a broad antiviral agent to combat viral infection and prevent ZIKV associated devastating clinical outcomes, such as congenital microcephaly.

6.
J Virol ; 93(23)2019 12 01.
Article in English | MEDLINE | ID: mdl-31511375

ABSTRACT

In spite of several decades of research focused on understanding the biology of human herpes simplex virus 1 (HSV-1), no tool has been developed to study its genome in a high-throughput fashion. Here, we describe the creation of a transposon insertion mutant library of the HSV-1 genome. Using this tool, we aimed to identify novel viral regulators of type I interferon (IFN-I). HSV-1 evades the host immune system by encoding viral proteins that inhibit the type I interferon response. Applying differential selective pressure, we identified the three strongest viral IFN-I regulators in HSV-1. We report that the viral polymerase processivity factor UL42 interacts with the host transcription factor IFN regulatory factor 3 (IRF-3), inhibiting its phosphorylation and downstream beta interferon (IFN-ß) gene transcription. This study represents a proof of concept for the use of high-throughput screening of the HSV-1 genome in investigating viral biology and offers new targets both for antiviral therapy and for oncolytic vector design.IMPORTANCE This work is the first to report the use of a high-throughput mutagenesis method to study the genome of HSV-1. We report three novel viral proteins potentially involved in regulating the host type I interferon response. We describe a novel mechanism by which the viral protein UL42 is able to suppress the production of beta interferon. The tool we introduce in this study can be used to study the HSV-1 genome in great detail to better understand viral gene functions.


Subject(s)
DNA-Directed DNA Polymerase/metabolism , Exodeoxyribonucleases/metabolism , Herpesvirus 1, Human/genetics , Interferon Type I/metabolism , Mutagenesis , Viral Proteins/metabolism , A549 Cells , Antiviral Agents/pharmacology , DNA-Directed DNA Polymerase/genetics , Exodeoxyribonucleases/genetics , HEK293 Cells , Herpesvirus 1, Human/physiology , Humans , Interferon Regulatory Factor-3/metabolism , Interferon-beta/genetics , Receptor, Interferon alpha-beta/genetics , Viral Proteins/genetics
7.
J Clin Invest ; 129(5): 1926-1939, 2019 04 02.
Article in English | MEDLINE | ID: mdl-30939123

ABSTRACT

IL-26 is an antimicrobial protein secreted by Th17 cells that has the ability to directly kill extracellular bacteria. To ascertain whether IL-26 contributes to host defense against intracellular bacteria, we studied leprosy, caused by the obligate intracellular pathogen Mycobacterium leprae, as a model. Analysis of leprosy skin lesions by gene expression profiling and immunohistology revealed that IL-26 was more strongly expressed in lesions from the self-limited tuberculoid compared with expression in progressive lepromatous patients. IL-26 directly bound to M. leprae in axenic culture and reduced bacteria viability. Furthermore, IL-26, when added to human monocyte-derived macrophages infected with M. leprae, entered the infected cell, colocalized with the bacterium, and reduced bacteria viability. In addition, IL-26 induced autophagy via the cytoplasmic DNA receptor stimulator of IFN genes (STING), as well as fusion of phagosomes containing bacilli with lysosomal compartments. Altogether, our data suggest that the Th17 cytokine IL-26 contributes to host defense against intracellular bacteria.


Subject(s)
Interleukins/immunology , Leprosy, Lepromatous/microbiology , Leprosy, Tuberculoid/microbiology , Th17 Cells/immunology , Autophagy , Cytokines/immunology , Gene Expression Profiling , Humans , Lysosomes/immunology , Lysosomes/microbiology , Macrophages/immunology , Monocytes/cytology , Mycobacterium leprae , Mycobacterium tuberculosis , Phagosomes/immunology , Recombinant Proteins/immunology , Signal Transduction
8.
JCI Insight ; 4(8)2019 04 18.
Article in English | MEDLINE | ID: mdl-30996142

ABSTRACT

DC, through the uptake, processing, and presentation of antigen, are responsible for activation of T cell responses to defend the host against infection, yet it is not known if they can directly kill invading bacteria. Here, we studied in human leprosy, how Langerhans cells (LC), specialized DC, contribute to host defense against bacterial infection. IFN-γ treatment of LC isolated from human epidermis and infected with Mycobacterium leprae (M. leprae) activated an antimicrobial activity, which was dependent on the upregulation of the antimicrobial peptide cathelicidin and induction of autophagy. IFN-γ induction of autophagy promoted fusion of phagosomes containing M. leprae with lysosomes and the delivery of cathelicidin to the intracellular compartment containing the pathogen. Autophagy enhanced the ability of M. leprae-infected LC to present antigen to CD1a-restricted T cells. The frequency of IFN-γ labeling and LC containing both cathelicidin and autophagic vesicles was greater in the self-healing lesions vs. progressive lesions, thus correlating with the effectiveness of host defense against the pathogen. These data indicate that autophagy links the ability of DC to kill and degrade an invading pathogen, ensuring cell survival from the infection while facilitating presentation of microbial antigens to resident T cells.


Subject(s)
Antigen Presentation , Autophagy , Langerhans Cells/immunology , Leprosy/immunology , Mycobacterium leprae/immunology , Antigens, Bacterial/immunology , Antimicrobial Cationic Peptides/immunology , Antimicrobial Cationic Peptides/metabolism , Autophagosomes/immunology , Autophagosomes/metabolism , Autophagosomes/microbiology , Biopsy , Cells, Cultured , Epidermis/immunology , Epidermis/microbiology , Epidermis/pathology , Humans , Interferon-gamma/immunology , Langerhans Cells/microbiology , Langerhans Cells/ultrastructure , Leprosy/microbiology , Leprosy/pathology , Lysosomes/immunology , Lysosomes/metabolism , Lysosomes/microbiology , Microscopy, Electron, Transmission , Mycobacterium leprae/isolation & purification , Primary Cell Culture , Recombinant Proteins/immunology , T-Lymphocytes/immunology , Up-Regulation/immunology , Cathelicidins
9.
Nat Commun ; 9(1): 2770, 2018 07 17.
Article in English | MEDLINE | ID: mdl-30018345

ABSTRACT

Detection of viral genomes by the innate immune system elicits an antiviral gene program mediated by type I interferons (IFNs). While viral RNA and DNA species induce IFN via separate pathways, the mechanisms by which these pathways are differentially modulated are unknown. Here we show that the positive regulator of IFN in the RNA pathway, TRAF3, has an inhibitory function in the DNA pathway. Loss of TRAF3 coincides with increased expression of the alternative NF-κB-inducing molecule, NIK, which interacts with the DNA pathway adaptor, STING, to enhance IFN induction. Cells lacking NIK display defective IFN activation in the DNA pathway due to impaired STING signaling, and NIK-deficient mice are more susceptible to DNA virus infection. Mechanistically, NIK operates independently from alternative NF-κB signaling components and instead requires autophosphorylation and oligomerization to activate STING. Thus a previously undescribed pathway for NIK exists in activating IFN in the DNA pathway.


Subject(s)
DNA, Viral/genetics , Herpesvirus 1, Human/genetics , Host-Pathogen Interactions , Protein Serine-Threonine Kinases/genetics , RNA, Viral/genetics , TNF Receptor-Associated Factor 3/genetics , Vesicular stomatitis Indiana virus/genetics , A549 Cells , Animals , DNA, Viral/immunology , Female , Gene Expression Regulation , Genes, Reporter , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/immunology , HEK293 Cells , Herpesvirus 1, Human/immunology , Humans , Immunity, Innate , Interferon-alpha/genetics , Interferon-alpha/immunology , Interferon-beta/genetics , Interferon-beta/immunology , Membrane Proteins/genetics , Membrane Proteins/immunology , Mice , Mice, Knockout , NF-kappa B/genetics , NF-kappa B/immunology , Protein Serine-Threonine Kinases/immunology , RNA, Viral/immunology , Signal Transduction , THP-1 Cells , TNF Receptor-Associated Factor 3/immunology , Vesicular stomatitis Indiana virus/immunology , NF-kappaB-Inducing Kinase
10.
Mol Cell Biol ; 37(1)2017 01 01.
Article in English | MEDLINE | ID: mdl-27736772

ABSTRACT

The host response to RNA virus infection consists of an intrinsic innate immune response and the induction of apoptosis as mechanisms to restrict viral replication. The mitochondrial adaptor molecule MAVS plays critical roles in coordinating both virus-induced type I interferon production and apoptosis; however, the regulation of MAVS-mediated apoptosis is poorly understood. Here, we show that the adaptor protein TAX1BP1 functions as a negative regulator of virus-induced apoptosis. TAX1BP1-deficient cells are highly sensitive to apoptosis in response to infection with the RNA viruses vesicular stomatitis virus and Sendai virus and to transfection with poly(I·C). TAX1BP1 undergoes degradation during RNA virus infection, and loss of TAX1BP1 is associated with apoptotic cell death. TAX1BP1 deficiency augments virus-induced activation of proapoptotic c-Jun N-terminal kinase (JNK) signaling. Virus infection promotes the mitochondrial localization of TAX1BP1 and concomitant interaction with the mitochondrial adaptor MAVS. TAX1BP1 recruits the E3 ligase Itch to MAVS to trigger its ubiquitination and degradation, and loss of TAX1BP1 or Itch results in increased MAVS protein expression. Together, these results indicate that TAX1BP1 functions as an adaptor molecule for Itch to target MAVS during RNA virus infection and thus restrict virus-induced apoptosis.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Mitochondria/metabolism , Neoplasm Proteins/metabolism , RNA Viruses/pathogenicity , Repressor Proteins/metabolism , Ubiquitin-Protein Ligases/metabolism , Animals , Apoptosis , HEK293 Cells , HeLa Cells , Humans , Mice , Sendai virus/pathogenicity , Ubiquitination , Vesiculovirus/pathogenicity
11.
Mol Cell Biol ; 36(12): 1776-92, 2016 06 15.
Article in English | MEDLINE | ID: mdl-27090639

ABSTRACT

We and others have shown that the cystatin E/M gene is inactivated in primary human tumors, pointing to its role as a tumor suppressor gene. However, the molecular mechanism of tumor suppression is not yet understood. Using plasmid-directed cystatin E/M gene overexpression, a lentivirus-mediated tetracycline-inducible vector system, and human papillomavirus 16 (HPV 16) E6 and E7 gene-immortalized normal human epidermal keratinocytes, we demonstrated intracellular and non-cell-autonomous apoptotic growth inhibition of tumor cell lines and that growth inhibition is associated with cytoplasmic retention of NF-κB. We further demonstrated decreased phosphorylation of IκB kinase (IKKß) and IκBα in the presence of tumor necrosis factor alpha (TNF-α), confirming the role of cystatin E/M in the regulation of the NF-κB signaling pathway. Growth suppression of nude mouse xenograft tumors carrying a tetracycline-inducible vector system was observed with the addition of doxycycline in drinking water, confirming that the cystatin E/M gene is a tumor suppressor gene. Finally, immunohistochemical analyses of cervical carcinoma in situ and primary tumors have shown a statistically significant inverse relationship between the expression of cystatin E/M and cathepsin L and a direct relationship between the loss of cystatin E/M expression and nuclear expression of NF-κB. We therefore propose that the cystatin E/M suppressor gene plays an important role in the regulation of NF-κB.


Subject(s)
Cystatin M/metabolism , Cytoplasm/metabolism , I-kappa B Proteins/metabolism , NF-kappa B/metabolism , Tumor Necrosis Factor-alpha/metabolism , Uterine Cervical Neoplasms/pathology , Animals , Cathepsin L/metabolism , Cell Line, Tumor , Cell Proliferation , Cystatin M/genetics , Doxycycline/administration & dosage , Female , Gene Expression Regulation, Neoplastic , Genetic Vectors/pharmacology , HeLa Cells , Humans , Lentivirus/genetics , Mice , Mice, Nude , Neoplasm Transplantation , Phosphorylation , Signal Transduction , Uterine Cervical Neoplasms/genetics , Uterine Cervical Neoplasms/metabolism
12.
PLoS One ; 8(1): e53717, 2013.
Article in English | MEDLINE | ID: mdl-23308279

ABSTRACT

A key feature of the innate antiviral immune response is a rapid nonspecific response to virus infection largely mediated by the induction and extracellular secretion of type I interferons (IFNs) that restrict virus replication. Cytoplasmic sensors such as RIG-I recognize viral RNA and trigger antiviral signaling pathways that upregulate IFN transcription. However, it remains largely unknown how antiviral signaling is negatively regulated to maintain homeostasis after the elimination of virus. In this report, we have identified the RING domain-containing protein RING finger 11 (RNF11) as a novel negative regulator of innate antiviral signaling. Overexpression of RNF11 downregulated IFN-ß expression and enhanced viral replication whereas siRNA-mediated knockdown of RNF11 suppressed viral replication. RNF11 interacted with the noncanonical IKK kinases TBK1/IKKi and attenuated their Lys63-linked polyubiquitination by blocking interactions with the E3 ligase TRAF3. The inhibitory function of RNF11 was dependent on the ubiquitin-binding adaptor molecule TAX1BP1 which was required for RNF11 to target TBK1/IKKi. Collectively, these results indicate that RNF11 functions together with TAX1BP1 to restrict antiviral signaling and IFN-ß production.


Subject(s)
Carrier Proteins/genetics , Fibroblasts/immunology , Gene Expression Regulation , Immunity, Innate , Intracellular Signaling Peptides and Proteins/genetics , Neoplasm Proteins/genetics , Signal Transduction , Animals , Carrier Proteins/antagonists & inhibitors , Carrier Proteins/immunology , Cell Line , DNA-Binding Proteins , Fibroblasts/virology , HEK293 Cells , Humans , Interferon-beta/biosynthesis , Interferon-beta/immunology , Intracellular Signaling Peptides and Proteins/immunology , Mice , Neoplasm Proteins/immunology , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/immunology , RNA, Small Interfering/genetics , TNF Receptor-Associated Factor 3/genetics , TNF Receptor-Associated Factor 3/immunology , Ubiquitination , Vesiculovirus/immunology , Virus Replication
13.
Nat Immunol ; 13(12): 1155-61, 2012 Dec.
Article in English | MEDLINE | ID: mdl-23142775

ABSTRACT

The induction of type I interferons by the bacterial secondary messengers cyclic di-GMP (c-di-GMP) or cyclic di-AMP (c-di-AMP) is dependent on a signaling axis that involves the adaptor STING, the kinase TBK1 and the transcription factor IRF3. Here we identified the heliase DDX41 as a pattern-recognition receptor (PRR) that sensed both c-di-GMP and c-di-AMP. DDX41 specifically and directly interacted with c-di-GMP. Knockdown of DDX41 via short hairpin RNA in mouse or human cells inhibited the induction of genes encoding molecules involved in the innate immune response and resulted in defective activation of STING, TBK1 and IRF3 in response to c-di-GMP or c-di-AMP. Our results suggest a mechanism whereby c-di-GMP and c-di-AMP are detected by DDX41, which forms a complex with STING to signal to TBK1-IRF3 and activate the interferon response.


Subject(s)
Cyclic GMP/analogs & derivatives , DEAD-box RNA Helicases/metabolism , Dinucleoside Phosphates/metabolism , Interferon Type I/immunology , Listeria monocytogenes/immunology , Listeria monocytogenes/metabolism , Receptors, Pattern Recognition/metabolism , Animals , Cell Line , Cyclic GMP/metabolism , DEAD-box RNA Helicases/genetics , Humans , Immunity, Innate , Interferon Regulatory Factor-3/metabolism , Macrophages/immunology , Membrane Proteins/metabolism , Mice , Protein Serine-Threonine Kinases/metabolism , RNA Interference , RNA, Small Interfering , Receptors, Pattern Recognition/genetics , Second Messenger Systems , Signal Transduction
14.
Immunity ; 36(6): 1073-86, 2012 Jun 29.
Article in English | MEDLINE | ID: mdl-22579474

ABSTRACT

STING is an essential signaling molecule for DNA and cyclic di-GMP (c-di-GMP)-mediated type I interferon (IFN) production via TANK-binding kinase 1 (TBK1) and interferon regulatory factor 3 (IRF3) pathway. It contains an N-terminal transmembrane region and a cytosolic C-terminal domain (CTD). Here, we describe crystal structures of STING CTD alone and complexed with c-di-GMP in a unique binding mode. The strictly conserved aa 153-173 region was shown to be cytosolic and participated in dimerization via hydrophobic interactions. The STING CTD functions as a dimer and the dimerization was independent of posttranslational modifications. Binding of c-di-GMP enhanced interaction of a shorter construct of STING CTD (residues 139-344) with TBK1. This suggests an extra TBK1 binding site, other than serine 358. This study provides a glimpse into the unique architecture of STING and sheds light on the mechanism of c-di-GMP-mediated TBK1 signaling.


Subject(s)
Cyclic GMP/analogs & derivatives , Membrane Proteins/chemistry , Amino Acid Sequence , Conserved Sequence , Crystallography, X-Ray , Cyclic GMP/metabolism , Dimerization , HEK293 Cells , Humans , Hydrophobic and Hydrophilic Interactions , Membrane Proteins/metabolism , Models, Molecular , Molecular Sequence Data , Peptide Fragments/chemistry , Peptide Fragments/metabolism , Protein Binding , Protein Conformation , Protein Interaction Mapping , Protein Serine-Threonine Kinases/metabolism , Recombinant Fusion Proteins/chemistry , Sequence Alignment , Sequence Homology, Amino Acid , Structure-Activity Relationship
15.
Immunity ; 34(6): 821-2, 2011 Jun 24.
Article in English | MEDLINE | ID: mdl-21703534

ABSTRACT

In this issue of Immunity, Allen et al. (2011) and Xia et al. (2011) provide in vivo and biochemical evidence that NLRX1, a member of the nucleotide binding domain and leucine-rich-repeat-containing protein family, functions as a negative regulator of RIG-I and Toll-like receptors.

16.
Microbes Infect ; 13(3): 209-15, 2011 Mar.
Article in English | MEDLINE | ID: mdl-21111841

ABSTRACT

The deubiquitinating enzyme A20 (also known as TNFAIP3) is essential for maintaining immune homeostasis. A20 is a key regulator of inflammatory, antiviral and apoptotic signaling pathways. Here, we review recent advances illustrating the role of A20 as an essential negative regulator of inflammatory and antiviral signaling.


Subject(s)
Inflammation/immunology , Intracellular Signaling Peptides and Proteins/physiology , Nuclear Proteins/physiology , Virus Diseases/immunology , Animals , Cell Death , DNA-Binding Proteins , Humans , Inflammation/virology , Intracellular Signaling Peptides and Proteins/immunology , Intracellular Signaling Peptides and Proteins/metabolism , Nuclear Proteins/immunology , Nuclear Proteins/metabolism , Signal Transduction , Tumor Necrosis Factor alpha-Induced Protein 3
18.
J Biol Chem ; 285(20): 14999-15009, 2010 May 14.
Article in English | MEDLINE | ID: mdl-20304918

ABSTRACT

Induction of type I interferons by the transcription factor IRF3 is essential in the initiation of antiviral innate immunity. Activation of IRF3 requires C-terminal phosphorylation by the upstream kinases TBK1-IKKi, where IRF3 phosphorylation promotes dimerization, and subsequent nuclear translocation to the IFNbeta promoter. Recent studies have described the ubiquitin-editing enzyme A20 as a negative regulator of IRF3 signaling by associating with TBK1-IKKi; however, the regulatory mechanism of A20 inhibition remains unclear. Here we describe the adaptor protein, TAX1BP1, as a key regulator of A20 function in terminating signaling to IRF3. Murine embryonic fibroblasts (MEFs) deficient in TAX1BP1 displayed increased amounts of IFNbeta production upon viral challenge compared with WT MEFs. TAX1BP1 inhibited virus-mediated activation of IRF3 at the level of TBK1-IKKi. TAX1BP1 and A20 blocked antiviral signaling by disrupting Lys(63)-linked polyubiquitination of TBK1-IKKi independently of the A20 deubiquitination domain. Furthermore, TAX1BP1 was required for A20 effector function because A20 was defective for the targeting and inactivation of TBK1 and IKKi in Tax1bp1(-)(/)(-) MEFs. Additionally, we found the E3 ubiquitin ligase TRAF3 to play a critical role in promoting TBK1-IKKi ubiquitination. Collectively, our results demonstrate TBK1-IKKi to be novel substrates for A20 and further identify a novel mechanism whereby A20 and TAX1BP1 restrict antiviral signaling by disrupting a TRAF3-TBK1-IKKi signaling complex.


Subject(s)
Intracellular Signaling Peptides and Proteins/physiology , Neoplasm Proteins/physiology , Nuclear Proteins/physiology , Protein Kinases/metabolism , Protein Serine-Threonine Kinases/metabolism , Signal Transduction , Vesiculovirus/pathogenicity , Animals , Cell Line , DNA-Binding Proteins , Enzyme-Linked Immunosorbent Assay , Humans , Mice , Tumor Necrosis Factor alpha-Induced Protein 3
19.
EMBO J ; 28(5): 513-22, 2009 Mar 04.
Article in English | MEDLINE | ID: mdl-19131965

ABSTRACT

The RING domain protein RNF11 is overexpressed in breast cancers and promotes tumour growth factor-beta (TGF-beta) signalling. RNF11 has been proposed to regulate TGF-beta signalling by interacting with HECT- and SCF-type E3 ligases; however, the role of RNF11 in other signalling pathways is poorly understood. Here, we demonstrate a novel function of RNF11 as a negative regulator of NF-kappaB and jun N-terminal kinase (JNK) signalling pathways. Knockdown of RNF11 with siRNA resulted in persistent tumour necrosis factor (TNF)- and lipopolysaccharide (LPS)-mediated NF-kappaB and JNK signalling. RNF11 interacted with the NF-kappaB inhibitor A20 and its regulatory protein TAX1BP1 in a stimulus-dependent manner. RNF11 negatively regulated RIP1 and TRAF6 ubiquitination upon stimulation with TNF and LPS, respectively. Furthermore, RNF11 was required for A20 to interact with and inactivate RIP1 to inhibit TNF-mediated NF-kappaB activation. Our studies reveal that RNF11, together with TAX1BP1 and Itch, is an essential component of an A20 ubiquitin-editing protein complex that ensures transient activation of inflammatory signalling pathways.


Subject(s)
Carrier Proteins/metabolism , Intracellular Signaling Peptides and Proteins/physiology , NF-kappa B/physiology , Nuclear Proteins/physiology , Ubiquitin-Protein Ligases/physiology , Carrier Proteins/genetics , Cell Line , DNA-Binding Proteins , Down-Regulation , Gene Knockdown Techniques , Humans , Intracellular Signaling Peptides and Proteins/metabolism , JNK Mitogen-Activated Protein Kinases/physiology , Lipopolysaccharides/pharmacology , Monocytes/metabolism , Neoplasm Proteins/metabolism , Nuclear Pore Complex Proteins/metabolism , Protein Binding , Protein Structure, Tertiary , RNA, Small Interfering/genetics , RNA-Binding Proteins/metabolism , Signal Transduction/physiology , TNF Receptor-Associated Factor 6/metabolism , Transforming Growth Factor beta/pharmacology , Tumor Necrosis Factor alpha-Induced Protein 3 , Ubiquitination
20.
Nat Immunol ; 9(3): 254-62, 2008 Mar.
Article in English | MEDLINE | ID: mdl-18246070

ABSTRACT

The ubiquitin-editing enzyme A20 is a critical negative regulator of inflammation and cytokine-mediated activation of the transcription factor NF-kappaB; however, little is known about the mechanisms of A20-mediated inactivation of signaling intermediates such as RIP1. Here we demonstrate that the regulatory molecule TAX1BP1 recruited the E3 ligase Itch to A20 via two 'PPXY' motifs. Itch was essential for the termination of tumor necrosis factor receptor signaling by controlling A20-mediated recruitment and inactivation of RIP1. Furthermore, the Tax oncoprotein of human T cell leukemia virus type I targeted this complex for inactivation by disrupting the interaction among TAX1BP1, A20 and Itch. Thus, our studies show a previously unappreciated complexity of A20 substrate recognition and inactivation whereby TAX1BP1 and Itch function as essential subunits of an A20 ubiquitin-editing complex.


Subject(s)
Down-Regulation/immunology , Intracellular Signaling Peptides and Proteins/physiology , Nuclear Proteins/physiology , Repressor Proteins/physiology , Signal Transduction/immunology , Ubiquitin-Protein Ligases/physiology , Animals , Cells, Cultured , DNA-Binding Proteins , Gene Deletion , Homeodomain Proteins/genetics , Humans , MAP Kinase Kinase 4/metabolism , Mice , Mice, Knockout , Neoplasm Proteins , Protein Serine-Threonine Kinases/metabolism , Tumor Necrosis Factor alpha-Induced Protein 3 , NF-kappaB-Inducing Kinase
SELECTION OF CITATIONS
SEARCH DETAIL
...