Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 25
Filter
Add more filters










Publication year range
1.
Elife ; 122023 08 25.
Article in English | MEDLINE | ID: mdl-37622993

ABSTRACT

Adaptation of the functional proteome is essential to counter pathogens during infection, yet precisely timed degradation of these response proteins after pathogen clearance is likewise key to preventing autoimmunity. Interferon regulatory factor 1 (IRF1) plays an essential role as a transcription factor in driving the expression of immune response genes during infection. The striking difference in functional output with other IRFs is that IRF1 also drives the expression of various cell cycle inhibiting factors, making it an important tumor suppressor. Thus, it is critical to regulate the abundance of IRF1 to achieve a 'Goldilocks' zone in which there is sufficient IRF1 to prevent tumorigenesis, yet not too much which could drive excessive immune activation. Using genetic screening, we identified the E3 ligase receptor speckle type BTB/POZ protein (SPOP) to mediate IRF1 proteasomal turnover in human and mouse cells. We identified S/T-rich degrons in IRF1 required for its SPOP MATH domain-dependent turnover. In the absence of SPOP, elevated IRF1 protein levels functionally increased IRF1-dependent cellular responses, underpinning the biological significance of SPOP in curtailing IRF1 protein abundance.


Subject(s)
Gene Expression Regulation , Genes, Regulator , Humans , Animals , Mice , Interferon Regulatory Factor-1/genetics , Acclimatization , Immunologic Factors
2.
Elife ; 122023 03 24.
Article in English | MEDLINE | ID: mdl-36961408

ABSTRACT

Tristetraprolin (TTP) is a critical negative immune regulator. It binds AU-rich elements in the untranslated-regions of many mRNAs encoding pro-inflammatory mediators, thereby accelerating their decay. A key but poorly understood mechanism of TTP regulation is its timely proteolytic removal: TTP is degraded by the proteasome through yet unidentified phosphorylation-controlled drivers. In this study, we set out to identify factors controlling TTP stability. Cellular assays showed that TTP is strongly lysine-ubiquitinated, which is required for its turnover. A genetic screen identified the ubiquitin E3 ligase HUWE1 as a strong regulator of TTP proteasomal degradation, which we found to control TTP stability indirectly by regulating its phosphorylation. Pharmacological assessment of multiple kinases revealed that HUWE1-regulated TTP phosphorylation and stability was independent of the previously characterized effects of MAPK-mediated S52/S178 phosphorylation. HUWE1 function was dependent on phosphatase and E3 ligase binding sites identified in the TTP C-terminus. Our findings indicate that while phosphorylation of S52/S178 is critical for TTP stabilization at earlier times after pro-inflammatory stimulation, phosphorylation of the TTP C-terminus controls its stability at later stages.


Subject(s)
Tristetraprolin , Ubiquitin-Protein Ligases , Phosphorylation , Tristetraprolin/metabolism , Ubiquitin-Protein Ligases/metabolism , Proteolysis , Ubiquitin/metabolism , RNA Stability/genetics
3.
Science ; 379(6637): 1117-1123, 2023 03 17.
Article in English | MEDLINE | ID: mdl-36758105

ABSTRACT

Inhibitor of apoptosis proteins (IAPs) bind to pro-apoptotic proteases, keeping them inactive and preventing cell death. The atypical ubiquitin ligase BIRC6 is the only essential IAP, additionally functioning as a suppressor of autophagy. We performed a structure-function analysis of BIRC6 in complex with caspase-9, HTRA2, SMAC, and LC3B, which are critical apoptosis and autophagy proteins. Cryo-electron microscopy structures showed that BIRC6 forms a megadalton crescent shape that arcs around a spacious cavity containing receptor sites for client proteins. Multivalent binding of SMAC obstructs client binding, impeding ubiquitination of both autophagy and apoptotic substrates. On the basis of these data, we discuss how the BIRC6/SMAC complex can act as a stress-induced hub to regulate apoptosis and autophagy drivers.


Subject(s)
Apoptosis Regulatory Proteins , Apoptosis , Inhibitor of Apoptosis Proteins , Mitochondrial Proteins , Humans , Apoptosis/physiology , Apoptosis Regulatory Proteins/chemistry , Apoptosis Regulatory Proteins/metabolism , Autophagy , Cryoelectron Microscopy , Inhibitor of Apoptosis Proteins/chemistry , Inhibitor of Apoptosis Proteins/metabolism , Mitochondrial Proteins/chemistry , Mitochondrial Proteins/metabolism , Ubiquitination , Protein Multimerization , High-Temperature Requirement A Serine Peptidase 2/chemistry , High-Temperature Requirement A Serine Peptidase 2/metabolism
4.
Nature ; 599(7885): 491-496, 2021 11.
Article in English | MEDLINE | ID: mdl-34711951

ABSTRACT

Protein expression and turnover are controlled through a complex interplay of transcriptional, post-transcriptional and post-translational mechanisms to enable spatial and temporal regulation of cellular processes. To systematically elucidate such gene regulatory networks, we developed a CRISPR screening assay based on time-controlled Cas9 mutagenesis, intracellular immunostaining and fluorescence-activated cell sorting that enables the identification of regulatory factors independent of their effects on cellular fitness. We pioneered this approach by systematically probing the regulation of the transcription factor MYC, a master regulator of cell growth1-3. Our screens uncover a highly conserved protein, AKIRIN2, that is essentially required for nuclear protein degradation. We found that AKIRIN2 forms homodimers that directly bind to fully assembled 20S proteasomes to mediate their nuclear import. During mitosis, proteasomes are excluded from condensing chromatin and re-imported into newly formed daughter nuclei in a highly dynamic, AKIRIN2-dependent process. Cells undergoing mitosis in the absence of AKIRIN2 become devoid of nuclear proteasomes, rapidly causing accumulation of MYC and other nuclear proteins. Collectively, our study reveals a dedicated pathway controlling the nuclear import of proteasomes in vertebrates and establishes a scalable approach to decipher regulators in essential cellular processes.


Subject(s)
Cell Nucleus/metabolism , DNA-Binding Proteins/metabolism , Nuclear Proteins/metabolism , Proteasome Endopeptidase Complex/metabolism , Transcription Factors/metabolism , Active Transport, Cell Nucleus , CRISPR-Cas Systems , Cell Line, Tumor , Female , Genes, myc , Humans , Male , Mitosis , Proteasome Endopeptidase Complex/chemistry , Protein Binding , Proteolysis
5.
Viruses ; 13(4)2021 03 30.
Article in English | MEDLINE | ID: mdl-33808506

ABSTRACT

The rapid and dynamic activation of the innate immune system is achieved through complex signaling networks regulated by post-translational modifications modulating the subcellular localization, activity, and abundance of signaling molecules. Many constitutively expressed signaling molecules are present in the cell in inactive forms, and become functionally activated once they are modified with ubiquitin, and, in turn, inactivated by removal of the same post-translational mark. Moreover, upon infection resolution a rapid remodeling of the proteome needs to occur, ensuring the removal of induced response proteins to prevent hyperactivation. This review discusses the current knowledge on the negative regulation of innate immune signaling pathways by deubiquitinating enzymes, and through degradative ubiquitination. It focusses on spatiotemporal regulation of deubiquitinase and E3 ligase activities, mechanisms for re-establishing proteostasis, and degradation through immune-specific feedback mechanisms vs. general protein quality control pathways.


Subject(s)
Gene Expression Regulation/immunology , Immunity, Innate/genetics , Proteasome Endopeptidase Complex/metabolism , Ubiquitination/physiology , Humans , Proteasome Endopeptidase Complex/genetics , Proteasome Endopeptidase Complex/immunology , Protein Binding , Protein Processing, Post-Translational , Protein Transport , Signal Transduction/genetics , Signal Transduction/immunology , Ubiquitination/genetics , Ubiquitination/immunology
6.
PLoS Pathog ; 17(4): e1009487, 2021 04.
Article in English | MEDLINE | ID: mdl-33905460

ABSTRACT

Lipocalin 2 (LCN2) is a secreted glycoprotein with roles in multiple biological processes. It contributes to host defense by interference with bacterial iron uptake and exerts immunomodulatory functions in various diseases. Here, we aimed to characterize the function of LCN2 in lung macrophages and dendritic cells (DCs) using Lcn2-/- mice. Transcriptome analysis revealed strong LCN2-related effects in CD103+ DCs during homeostasis, with differential regulation of antigen processing and presentation and antiviral immunity pathways. We next validated the relevance of LCN2 in a mouse model of influenza infection, wherein LCN2 protected from excessive weight loss and improved survival. LCN2-deficiency was associated with enlarged mediastinal lymph nodes and increased lung T cell numbers, indicating a dysregulated immune response to influenza infection. Depletion of CD8+ T cells equalized weight loss between WT and Lcn2-/- mice, proving that LCN2 protects from excessive disease morbidity by dampening CD8+ T cell responses. In vivo T cell chimerism and in vitro T cell proliferation assays indicated that improved antigen processing by CD103+ DCs, rather than T cell intrinsic effects of LCN2, contribute to the exacerbated T cell response. Considering the antibacterial potential of LCN2 and that commensal microbes can modulate antiviral immune responses, we speculated that LCN2 might cause the observed influenza phenotype via the microbiome. Comparing the lung and gut microbiome of WT and Lcn2-/- mice by 16S rRNA gene sequencing, we observed profound effects of LCN2 on gut microbial composition. Interestingly, antibiotic treatment or co-housing of WT and Lcn2-/- mice prior to influenza infection equalized lung CD8+ T cell counts, suggesting that the LCN2-related effects are mediated by the microbiome. In summary, our results highlight a novel regulatory function of LCN2 in the modulation of antiviral immunity.


Subject(s)
Influenza, Human/immunology , Lipocalin-2/metabolism , Microbiota/immunology , Transcriptome , Animals , Antigen Presentation , CD8-Positive T-Lymphocytes/immunology , Dendritic Cells/immunology , Dendritic Cells/virology , Female , Gastrointestinal Microbiome , Homeostasis , Humans , Immunity , Influenza, Human/virology , Lipocalin-2/genetics , Lung/immunology , Lung/virology , Lymphocyte Activation , Macrophages/immunology , Male , Mice , Mice, Inbred C57BL , Specific Pathogen-Free Organisms
7.
Sci Rep ; 9(1): 7901, 2019 05 27.
Article in English | MEDLINE | ID: mdl-31133683

ABSTRACT

Tripartite motif protein 52 (TRIM52) is a non-canonical TRIM family member harbouring the largest RING domain encoded in the human genome. In humans TRIM52 is conserved and has been under positive selection pressure, yet it has been lost in many non-primates. Competitive cell fitness assays demonstrated that TRIM52 ablation reduces cellular fitness in multiple different cell types. To better understand how this cell-essential factor is controlled, we investigated how expression of this non-canonical protein is regulated. Here, we show that TRIM52 mRNA is constitutively expressed from an intergenic region preceding the TRIM52 gene. Yet, TRIM52 protein is rapidly turned-over by the proteasome with a 3.5-minute half-life, one of the shortest in the human proteome. Consistent with this extremely rapid degradation rate, all three TRIM52 domains were identified to contribute to its instability. Intriguingly, a repetitive acidic loop in the RING domain was identified as one of the main destabilizing regions, which was unexpected given the prevailing notion that these sequences are poor proteasome substrates. This work indicates that the effect of such repetitive acidic regions on proteasomal degradation depends on the protein context, and it identifies TRIM52 as an attractive model protein to study what these contextual properties are.


Subject(s)
Proteasome Endopeptidase Complex/metabolism , Proteolysis , Repetitive Sequences, Amino Acid , Tripartite Motif Proteins/metabolism , Cell Line, Tumor , Gene Expression Regulation , HEK293 Cells , Half-Life , Humans , Hydrogen-Ion Concentration , Protein Domains , RNA, Messenger/metabolism , RNA-Seq , Single-Cell Analysis , Time Factors , Tripartite Motif Proteins/chemistry
8.
Curr Biol ; 29(2): R42-R44, 2019 01 21.
Article in English | MEDLINE | ID: mdl-30668943

ABSTRACT

Vunjak and Versteeg introduce the TRIM family of post-translational modifiers and the roles of these proteins in viral restriction, immune signaling and autophagy.


Subject(s)
Autophagy , Immunity, Innate/genetics , Signal Transduction/immunology , Tripartite Motif Proteins/physiology , Virus Diseases/immunology , Animals , Tripartite Motif Proteins/genetics , Tripartite Motif Proteins/immunology
9.
Oncotarget ; 9(17): 13565-13581, 2018 Mar 02.
Article in English | MEDLINE | ID: mdl-29568378

ABSTRACT

Tripartite motif (TRIM) proteins have been shown to play important roles in cancer development and progression by modulating cell proliferation or resistance from cell death during non-homeostatic stress conditions found in tumor micro-environments. In this study, we set out to investigate the importance for cellular fitness of the virtually uncharacterized family member TRIM52. The human TRIM52 gene has arisen recently in evolution, making it unlikely that TRIM52 is required for basic cellular functions in normal cells. However, a recent genome-wide ablation screening study has suggested that TRIM52 may be essential for optimal proliferation or survival in certain genetic cancer backgrounds. Identifying genes which fit this concept of genetic context-dependent fitness in cancer cells is of interest as they are promising targets for tumor-specific therapy. We report here that TRIM52 ablation significantly diminished the proliferation of specific glioblastoma cell lines in cell culture and mouse xenografts by compromising their cell cycle progression in a p53-dependent manner. Together, our findings point to a non-redundant TRIM52 function that is required for optimal proliferation.

10.
Crit Rev Biochem Mol Biol ; 52(4): 425-460, 2017 08.
Article in English | MEDLINE | ID: mdl-28524749

ABSTRACT

Ubiquitination plays a central role in the regulation of various biological functions including immune responses. Ubiquitination is induced by a cascade of enzymatic reactions by E1 ubiquitin activating enzyme, E2 ubiquitin conjugating enzyme, and E3 ubiquitin ligase, and reversed by deubiquitinases. Depending on the enzymes, specific linkage types of ubiquitin chains are generated or hydrolyzed. Because different linkage types of ubiquitin chains control the fate of the substrate, understanding the regulatory mechanisms of ubiquitin enzymes is central. In this review, we highlight the most recent knowledge of ubiquitination in the immune signaling cascades including the T cell and B cell signaling cascades as well as the TNF signaling cascade regulated by various ubiquitin enzymes. Furthermore, we highlight the TRIM ubiquitin ligase family as one of the examples of critical E3 ubiquitin ligases in the regulation of immune responses.


Subject(s)
B-Lymphocytes/immunology , T-Lymphocytes/immunology , Ubiquitin-Protein Ligases/metabolism , Ubiquitin/metabolism , Humans , Signal Transduction , Ubiquitination
11.
Nat Commun ; 5: 4820, 2014 Sep 05.
Article in English | MEDLINE | ID: mdl-25190485

ABSTRACT

The TRIpartite Motif (TRIM) family of RING-domain-containing proteins participate in a variety of cellular functions. The ß-transducin repeat-containing protein (ß-TrCP), a component of the Skp-Cullin-F-box-containing (SCF) E3 ubiquitin ligase complex, recognizes the NF-κB inhibitor IκBα and precursor p100 for proteasomal degradation and processing, respectively. ß-TrCP thus plays a critical role in both canonical and non-canonical NF-κB activation. Here we report that TRIM9 is a negative regulator of NF-κB activation. Interaction between the phosphorylated degron motif of TRIM9 and the WD40 repeat region of ß-TrCP prevented ß-TrCP from binding its substrates, stabilizing IκBα and p100 and thereby blocking NF-κB activation. Consequently, expression or depletion of the TRIM9 gene significantly affected NF-κB-induced inflammatory cytokine production. This study not only elucidates a mechanism for TRIM9-mediated regulation of the ß-TrCP SCF complex activity but also identifies TRIM9 as a brain-specific negative regulator of the NF-κB pro-inflammatory signalling pathway.


Subject(s)
Brain/metabolism , NF-kappa B/metabolism , Nerve Tissue Proteins/metabolism , Signal Transduction/physiology , Ubiquitin-Protein Ligases/metabolism , beta-Transducin Repeat-Containing Proteins/metabolism , Animals , DNA Primers/genetics , Enzyme-Linked Immunosorbent Assay , Gene Knockdown Techniques , HEK293 Cells , HeLa Cells , Humans , Immunoblotting , Immunoprecipitation , Mass Spectrometry , Microscopy, Confocal , Plasmids/genetics , Polymerase Chain Reaction , RNA, Small Interfering/genetics , Rats , Rats, Sprague-Dawley , Tripartite Motif Proteins
12.
Cytokine Growth Factor Rev ; 25(5): 563-76, 2014 Oct.
Article in English | MEDLINE | ID: mdl-25172371

ABSTRACT

During the immune response, striking the right balance between positive and negative regulation is critical to effectively mount an anti-microbial defense while preventing detrimental effects from exacerbated immune activation. Intra-cellular immune signaling is tightly regulated by various post-translational modifications, which allow for this dynamic response. One of the post-translational modifiers critical for immune control is ubiquitin, which can be covalently conjugated to lysines in target molecules, thereby altering their functional properties. This is achieved in a process involving E3 ligases which determine ubiquitination target specificity. One of the most prominent E3 ligase families is that of the tripartite motif (TRIM) proteins, which counts over 70 members in humans. Over the last years, various studies have contributed to the notion that many members of this protein family are important immune regulators. Recent studies into the mechanisms by which some of the TRIMs regulate the innate immune system have uncovered important immune regulatory roles of both covalently attached, as well as unanchored poly-ubiquitin chains. This review highlights TRIM evolution, recent findings in TRIM-mediated immune regulation, and provides an outlook to current research hurdles and future directions.


Subject(s)
Adaptor Proteins, Signal Transducing/immunology , Immunity, Innate , Signal Transduction , Amino Acid Motifs , Animals , Cytokines/immunology , Evolution, Molecular , Humans , Invertebrates/classification , Membrane Proteins/immunology , Ubiquitin/immunology
13.
Immunity ; 40(6): 880-95, 2014 Jun 19.
Article in English | MEDLINE | ID: mdl-24882218

ABSTRACT

Type I interferons (IFN-I) are essential antiviral cytokines produced upon microbial infection. IFN-I elicits this activity through the upregulation of hundreds of IFN-I-stimulated genes (ISGs). The full breadth of ISG induction demands activation of a number of cellular factors including the IκB kinase epsilon (IKKε). However, the mechanism of IKKε activation upon IFN receptor signaling has remained elusive. Here we show that TRIM6, a member of the E3-ubiquitin ligase tripartite motif (TRIM) family of proteins, interacted with IKKε and promoted induction of IKKε-dependent ISGs. TRIM6 and the E2-ubiquitin conjugase UbE2K cooperated in the synthesis of unanchored K48-linked polyubiquitin chains, which activated IKKε for subsequent STAT1 phosphorylation. Our work attributes a previously unrecognized activating role of K48-linked unanchored polyubiquitin chains in kinase activation and identifies the UbE2K-TRIM6-ubiquitin axis as critical for IFN signaling and antiviral response.


Subject(s)
I-kappa B Kinase/immunology , Interferon Type I/immunology , Polyubiquitin/biosynthesis , Ubiquitin-Protein Ligases/immunology , Animals , Antiviral Agents , Cells, Cultured , Enzyme Activation/immunology , Humans , Janus Kinase 1 , Mice , Phosphorylation/immunology , RNA Interference , RNA, Small Interfering , STAT1 Transcription Factor/immunology , Signal Transduction/immunology , Tripartite Motif Proteins , Ubiquitin-Conjugating Enzymes/immunology , Ubiquitin-Protein Ligases/genetics
14.
J Mol Biol ; 426(6): 1265-84, 2014 Mar 20.
Article in English | MEDLINE | ID: mdl-24333484

ABSTRACT

Tripartite motif (TRIM) proteins have been implicated in multiple cellular functions, including antiviral activity. Research efforts so far indicate that the antiviral activity of TRIMs relies, for the most part, on their function as E3-ubiquitin ligases. A substantial number of the TRIM family members have been demonstrated to mediate innate immune cell signal transduction and subsequent cytokine induction. In addition, a subset of TRIMs has been shown to restrict viral replication by directly targeting viral proteins. Although the body of work on the cellular roles of TRIM E3-ubiquitin ligases has rapidly grown over the last years, many aspects of their molecular workings and multi-functionality remain unclear. The antiviral function of many TRIMs seems to be conferred by specific isoforms, by sub-cellular localization and in cell-type-specific contexts. Here we review recent findings on TRIM antiviral functions, current limitations and an outlook for future research.


Subject(s)
Antiviral Agents/therapeutic use , Immunity, Innate/immunology , Ubiquitin-Protein Ligases/immunology , Virus Diseases/drug therapy , Virus Replication/immunology , Viruses/immunology , Animals , Humans , Virus Diseases/immunology , Virus Diseases/virology
15.
Immunity ; 38(2): 384-98, 2013 Feb 21.
Article in English | MEDLINE | ID: mdl-23438823

ABSTRACT

Innate immunity conferred by the type I interferon is critical for antiviral defense. To date only a limited number of tripartite motif (TRIM) proteins have been implicated in modulation of innate immunity and anti-microbial activity. Here we report the complementary DNA cloning and systematic analysis of all known 75 human TRIMs. We demonstrate that roughly half of the 75 TRIM-family members enhanced the innate immune response and that they do this at multiple levels in signaling pathways. Moreover, messenger RNA levels and localization of most of these TRIMs were found to be altered during viral infection, suggesting that their regulatory activities are highly controlled at both pre- and posttranscriptional levels. Taken together, our data demonstrate a very considerable dedication of this large protein family to the positive regulation of the antiviral response, which supports the notion that this family of proteins evolved as a component of innate immunity.


Subject(s)
Carrier Proteins/genetics , Immunity, Innate , Leukocytes, Mononuclear/metabolism , RNA, Messenger/genetics , Receptors, Pattern Recognition/genetics , Rhabdoviridae Infections/metabolism , Zinc Fingers/genetics , Alternative Splicing , Antiviral Restriction Factors , Carrier Proteins/antagonists & inhibitors , Carrier Proteins/immunology , Cell Line , Cloning, Molecular , Gene Expression Profiling , Gene Expression Regulation , Humans , Leukocytes, Mononuclear/immunology , Leukocytes, Mononuclear/virology , Protein Isoforms/antagonists & inhibitors , Protein Isoforms/genetics , Protein Isoforms/immunology , RNA, Messenger/immunology , RNA, Small Interfering/genetics , Receptors, Pattern Recognition/immunology , Rhabdoviridae Infections/immunology , Rhabdoviridae Infections/virology , Signal Transduction , Tripartite Motif Proteins , Ubiquitin-Protein Ligases , Vesiculovirus/immunology , Zinc Fingers/immunology
16.
PLoS Pathog ; 8(11): e1003059, 2012.
Article in English | MEDLINE | ID: mdl-23209422

ABSTRACT

Influenza A viruses can adapt to new host species, leading to the emergence of novel pathogenic strains. There is evidence that highly pathogenic viruses encode for non-structural 1 (NS1) proteins that are more efficient in suppressing the host immune response. The NS1 protein inhibits type-I interferon (IFN) production partly by blocking the TRIM25 ubiquitin E3 ligase-mediated Lys63-linked ubiquitination of the viral RNA sensor RIG-I, required for its optimal downstream signaling. In order to understand possible mechanisms of viral adaptation and host tropism, we examined the ability of NS1 encoded by human (Cal04), avian (HK156), swine (SwTx98) and mouse-adapted (PR8) influenza viruses to interact with TRIM25 orthologues from mammalian and avian species. Using co-immunoprecipitation assays we show that human TRIM25 binds to all tested NS1 proteins, whereas the chicken TRIM25 ortholog binds preferentially to the NS1 from the avian virus. Strikingly, none of the NS1 proteins were able to bind mouse TRIM25. Since NS1 can inhibit IFN production in mouse, we tested the impact of TRIM25 and NS1 on RIG-I ubiquitination in mouse cells. While NS1 efficiently suppressed human TRIM25-dependent ubiquitination of RIG-I 2CARD, NS1 inhibited the ubiquitination of full-length mouse RIG-I in a mouse TRIM25-independent manner. Therefore, we tested if the ubiquitin E3 ligase Riplet, which has also been shown to ubiquitinate RIG-I, interacts with NS1. We found that NS1 binds mouse Riplet and inhibits its activity to induce IFN-ß in murine cells. Furthermore, NS1 proteins of human but not swine or avian viruses were able to interact with human Riplet, thereby suppressing RIG-I ubiquitination. In conclusion, our results indicate that influenza NS1 protein targets TRIM25 and Riplet ubiquitin E3 ligases in a species-specific manner for the inhibition of RIG-I ubiquitination and antiviral IFN production.


Subject(s)
DEAD-box RNA Helicases/metabolism , Influenza A virus/metabolism , Influenza, Human/metabolism , Interferons/biosynthesis , Ubiquitination , Viral Nonstructural Proteins/metabolism , Animals , Chlorocebus aethiops , DEAD Box Protein 58 , DEAD-box RNA Helicases/genetics , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Dogs , HeLa Cells , Humans , Influenza A virus/genetics , Influenza, Human/genetics , Interferons/genetics , Mice , Mice, Knockout , Receptors, Immunologic , Transcription Factors/genetics , Transcription Factors/metabolism , Tripartite Motif Proteins , Ubiquitin-Protein Ligases/genetics , Ubiquitin-Protein Ligases/metabolism , Vero Cells , Viral Nonstructural Proteins/genetics
17.
Cytokine Growth Factor Rev ; 23(6): 273-82, 2012 Dec.
Article in English | MEDLINE | ID: mdl-22964110

ABSTRACT

Detection of invading pathogens by pattern recognition receptors (PRRs) is crucial for the activation of the innate immune response. These sensors signal through intertwining signaling cascades which result in the expression of pro-inflammatory cytokines and type I interferons. Conjugation, or binding, of ubiquitin and ubiquitin-like modifiers (UBLs) to a plethora of immune signaling molecules forms a common theme in innate immune regulation. Numerous E3 ligases and deubiquitylating enzymes (DUBs) actively modify signaling components in order to achieve a balanced activation of the innate immune system. This review will discuss how this balance is achieved and which questions remain regarding innate immune regulation by ubiquitin and UBLs.


Subject(s)
Immunity, Innate/immunology , Ubiquitin/immunology , Animals , Humans , Receptors, Pattern Recognition/immunology
18.
PLoS One ; 7(1): e29870, 2012.
Article in English | MEDLINE | ID: mdl-22272257

ABSTRACT

Type I interferon (IFN) stimulates expression and conjugation of the ubiquitin-like modifier IFN-stimulated gene 15 (ISG15), thereby restricting replication of a wide variety of viruses. Conjugation of ISG15 is critical for its antiviral activity in mice. HECT domain and RCC1-like domain containing protein 5 (HerC5) mediates global ISGylation in human cells, whereas its closest relative, HerC6, does not. So far, the requirement of HerC5 for ISG15-mediated antiviral activity has remained unclear. One of the main obstacles to address this issue has been that no HerC5 homologue exists in mice, hampering the generation of a good knock-out model. However, mice do express a homologue of HerC6 that, in contrast to human HerC6, can mediate ISGylation.Here we report that the mouse HerC6 N-terminal RCC1-like domain (RLD) allows ISG15 conjugation when replacing the corresponding domain in the human HerC6 homologue. In addition, sequences in the C-terminal HECT domain of mouse HerC6 also appear to facilitate efficient ISGylation. Mouse HerC6 paralleled human HerC5 in localization and IFN-inducibility. Moreover, HerC6 knock-down in mouse cells abolished global ISGylation, whereas its over expression enhanced the IFNß promoter and conferred antiviral activity against vesicular stomatitis virus and Newcastle disease virus. Together these data indicate that HerC6 is likely the functional counterpart of human HerC5 in mouse cells, suggesting that HerC6(-/-) mice may provide a feasible model to study the role of human HerC5 in antiviral responses.


Subject(s)
Cytokines/metabolism , Ubiquitin-Protein Ligases/metabolism , Ubiquitins/metabolism , Animals , Binding Sites , Blotting, Western , Cell Line , Cell Line, Tumor , Cytokines/genetics , Cytoplasm/drug effects , Cytoplasm/metabolism , Gene Expression/drug effects , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , HEK293 Cells , HeLa Cells , Humans , Interferon-beta/genetics , Interferon-beta/metabolism , Interferon-beta/pharmacology , Mice , Mice, Knockout , Microscopy, Confocal , NIH 3T3 Cells , Newcastle disease virus/genetics , Newcastle disease virus/metabolism , Promoter Regions, Genetic/genetics , Protein Binding , RNA Interference , Reverse Transcriptase Polymerase Chain Reaction , Ubiquitin-Protein Ligases/genetics , Ubiquitins/genetics
19.
Nat Chem Biol ; 7(10): 712-9, 2011 Sep 11.
Article in English | MEDLINE | ID: mdl-21909097

ABSTRACT

A chemical genetics approach was taken to identify inhibitors of NS1, a major influenza A virus virulence factor that inhibits host gene expression. A high-throughput screen of 200,000 synthetic compounds identified small molecules that reversed NS1-mediated inhibition of host gene expression. A counterscreen for suppression of influenza virus cytotoxicity identified naphthalimides that inhibited replication of influenza virus and vesicular stomatitis virus (VSV). The mechanism of action occurs through activation of REDD1 expression and concomitant inhibition of mammalian target of rapamycin complex 1 (mTORC1) via TSC1-TSC2 complex. The antiviral activity of naphthalimides was abolished in REDD1(-/-) cells. Inhibition of REDD1 expression by viruses resulted in activation of the mTORC1 pathway. REDD1(-/-) cells prematurely upregulated viral proteins via mTORC1 activation and were permissive to virus replication. In contrast, cells conditionally expressing high concentrations of REDD1 downregulated the amount of viral protein. Thus, REDD1 is a new host defense factor, and chemical activation of REDD1 expression represents a potent antiviral intervention strategy.


Subject(s)
Antiviral Agents/pharmacology , Naphthalimides/pharmacology , Orthomyxoviridae/drug effects , Transcription Factors/metabolism , Vesiculovirus/drug effects , Animals , Antiviral Agents/chemistry , Cell Line , Dogs , Dose-Response Relationship, Drug , Gene Expression Regulation/drug effects , Gene Expression Regulation/genetics , High-Throughput Screening Assays , Humans , Mice , Microbial Sensitivity Tests , Molecular Structure , Naphthalimides/chemistry , Orthomyxoviridae/genetics , Orthomyxoviridae/metabolism , Structure-Activity Relationship , Transcription Factors/deficiency , Vesiculovirus/genetics , Vesiculovirus/metabolism , Viral Nonstructural Proteins/antagonists & inhibitors , Viral Nonstructural Proteins/genetics , Viral Nonstructural Proteins/metabolism , Virus Replication/drug effects
20.
Curr Opin Microbiol ; 13(4): 508-16, 2010 Aug.
Article in English | MEDLINE | ID: mdl-20538505

ABSTRACT

Type I interferons (IFNs) play a crucial role in the innate immune avant-garde against viral infections. Virtually all viruses have developed means to counteract the induction, signaling, or antiviral actions of the IFN circuit. Over 170 different virus-encoded IFN antagonists from 93 distinct viruses have been described up to now, indicating that most viruses interfere with multiple stages of the IFN response. Although every viral IFN antagonist is unique in its own right, four main mechanisms are employed to circumvent innate immune responses: (i) general inhibition of cellular gene expression, (ii) sequestration of molecules in the IFN circuit, (iii) proteolytic cleavage, and (iv) proteasomal degradation of key components of the IFN system. The increasing understanding of how different viral IFN antagonists function has been translated to the generation of viruses with mutant IFN antagonists as potential live vaccine candidates. Moreover, IFN antagonists are attractive targets for inhibition by small-molecule compounds.


Subject(s)
Interferon Type I/immunology , Virus Diseases/immunology , Viruses/immunology , Animals , Humans , Immune Evasion , Interferon Type I/antagonists & inhibitors , Interferon Type I/genetics , Interferon Type I/metabolism , Viral Vaccines/immunology , Viral Vaccines/pharmacology , Virus Diseases/virology
SELECTION OF CITATIONS
SEARCH DETAIL
...