Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 25
1.
Sci Adv ; 9(18): eadd0141, 2023 05 05.
Article En | MEDLINE | ID: mdl-37146151

Bats have been identified as natural reservoir hosts of several zoonotic viruses, prompting suggestions that they have unique immunological adaptations. Among bats, Old World fruit bats (Pteropodidae) have been linked to multiple spillovers. To test for lineage-specific molecular adaptations in these bats, we developed a new assembly pipeline to generate a reference-quality genome of the fruit bat Cynopterus sphinx and used this in comparative analyses of 12 bat species, including six pteropodids. Our results reveal that immunity-related genes have higher evolutionary rates in pteropodids than in other bats. Several lineage-specific genetic changes were shared across pteropodids, including the loss of NLRP1, duplications of PGLYRP1 and C5AR2, and amino acid replacements in MyD88. We introduced MyD88 transgenes containing Pteropodidae-specific residues into bat and human cell lines and found evidence of dampened inflammatory responses. By uncovering distinct immune adaptations, our results could help explain why pteropodids are frequently identified as viral hosts.


Chiroptera , Viruses , Animals , Humans , Chiroptera/genetics , Phylogeny , Evolution, Molecular , Myeloid Differentiation Factor 88/genetics , Myeloid Differentiation Factor 88/metabolism , Genome , Viruses/genetics
2.
Autophagy ; 19(7): 1916-1933, 2023 07.
Article En | MEDLINE | ID: mdl-36588386

Mitophagy is a form of autophagy that plays a key role in maintaining the homeostasis of functional mitochondria in the cell. Viruses have evolved various strategies to manipulate mitophagy to escape host immune responses and promote virus replication. In this study, the nucleoprotein (NP) of H1N1 virus (PR8 strain) was identified as a regulator of mitophagy. We revealed that NP-mediated mitophagy leads to the degradation of the mitochondria-anchored protein MAVS, thereby blocking MAVS-mediated antiviral signaling and promoting virus replication. The NP-mediated mitophagy is dependent on the interaction of NP with MAVS and the cargo receptor TOLLIP. Moreover, Y313 of NP is a key residue for the MAVS-NP interaction and NP-mediated mitophagy. The NPY313F mutation significantly attenuates the virus-induced mitophagy and the virus replication in vitro and in vivo. Taken together, our findings uncover a novel mechanism by which the NP of influenza virus induces mitophagy to attenuate innate immunity.Abbreviations: ACTB: actin beta; ATG7: autophagy related 7; ATG12: autophagy related 12; CCCP: carbonyl cyanide 3-chlorophenyl hydrazone; co-IP: co-immunoprecipitation; COX4/COXIV: cytochrome c oxidase subunit 4; DAPI: 4',6-diamidino-2-phenylindole, dihydrochloride; EID50: 50% egg infective dose; GAPDH: glyceraldehyde-3-phosphate dehydrogenase; GFP: green fluorescent protein; HEK: human embryonic kidney; hpi: hours post-infection; IAV: influenza A virus; IFN: interferon; IP: immunoprecipitation; LAMP1: lysosomal associated membrane protein 1; MAP1LC3/LC3: microtubule associated protein 1 light chain 3; MAVS: mitochondrial antiviral signaling protein; Mdivi-1: mitochondrial division inhibitor 1; MLD50: 50% mouse lethal dose; MOI: multiplicity of infection; NBR1: NBR1 autophagy cargo receptor; NP: nucleoprotein; PB1: basic polymerase 1; RFP: red fluorescent protein; RIGI: RNA sensor RIG-I; RIGI-N: RIGI-CARD; SeV: Sendai virus; SQSTM1/p62: sequestosome 1; TIMM23: translocase of inner mitochondrial membrane 23; TOLLIP: toll interacting protein; TOMM20: translocase of outer mitochondrial membrane 20; TUBA: tubulin alpha; Vec: empty vector; vRNP: viral ribonucleoprotein.


Influenza A Virus, H1N1 Subtype , Influenza A virus , Mice , Humans , Animals , Mitophagy/genetics , Autophagy , Nucleoproteins/pharmacology , Immunity, Innate , Antiviral Agents/pharmacology
3.
Proc Natl Acad Sci U S A ; 119(26): e2122805119, 2022 06 28.
Article En | MEDLINE | ID: mdl-35733260

During viral infection, sensing of viral RNA by retinoic acid-inducible gene-I-like receptors (RLRs) initiates an antiviral innate immune response, which is mediated by the mitochondrial adaptor protein VISA (virus-induced signal adaptor; also known as mitochondrial antiviral signaling protein [MAVS]). VISA is regulated by various posttranslational modifications (PTMs), such as polyubiquitination, phosphorylation, O-linked ß-d-N-acetylglucosaminylation (O-GlcNAcylation), and monomethylation. However, whether other forms of PTMs regulate VISA-mediated innate immune signaling remains elusive. Here, we report that Poly(ADP-ribosyl)ation (PARylation) is a PTM of VISA, which attenuates innate immune response to RNA viruses. Using a biochemical purification approach, we identified tankyrase 1 (TNKS1) as a VISA-associated protein. Viral infection led to the induction of TNKS1 and its homolog TNKS2, which translocated from cytosol to mitochondria and interacted with VISA. TNKS1 and TNKS2 catalyze the PARylation of VISA at Glu137 residue, thereby priming it for K48-linked polyubiquitination by the E3 ligase Ring figure protein 146 (RNF146) and subsequent degradation. Consistently, TNKS1, TNKS2, or RNF146 deficiency increased the RNA virus-triggered induction of downstream effector genes and impaired the replication of the virus. Moreover, TNKS1- or TNKS2-deficient mice produced higher levels of type I interferons (IFNs) and proinflammatory cytokines after virus infection and markedly reduced virus loads in the brains and lungs. Together, our findings uncover an essential role of PARylation of VISA in virus-triggered innate immune signaling, which represents a mechanism to avoid excessive harmful immune response.


Adaptor Proteins, Signal Transducing , Immunity, Innate , RNA Virus Infections , RNA Viruses , Tankyrases , Ubiquitin-Protein Ligases , Adaptor Proteins, Signal Transducing/metabolism , Animals , HEK293 Cells , Humans , Immunity, Innate/genetics , Mice , RNA Virus Infections/immunology , RNA Viruses/immunology , Tankyrases/genetics , Tankyrases/metabolism , Ubiquitin-Protein Ligases/metabolism , Ubiquitination
5.
Cell Discov ; 7(1): 46, 2021 Jun 22.
Article En | MEDLINE | ID: mdl-34155193

MITA (also known as STING) is an ER-located adaptor protein, which mediates DNA-triggered innate immune response and is critically involved in autoimmune diseases and tumorigenesis. MITA is regulated by post-translational modifications, but how post-transcriptional mechanisms are involved in the regulation of MITA is still largely unknown. Here, we identified the RNA-binding protein LUC7L2 as a negative regulator of DNA virus-triggered innate immune response. LUC7L2-deficient mice exhibited resistance to lethal herpes simplex virus 1 (HSV-1) infection and reduced HSV-1 loads in the brain. Mechanistically, LUC7L2 directly bound to intron 3 of MITA precursor messenger RNA, inhibited its splicing and promoted its nonsense-mediated decay, leading to its downregulation at protein level. LUC7L2-deficient cells had markedly increased MITA level, leading to heightened innate antiviral response. Finally, LUC7L2 was induced following HSV-1 infection. Our findings reveal a feedback negative post-transcriptional regulatory mechanism for regulation of MITA-mediated innate immune response to viral and aberrant cellular DNA.

6.
PLoS Pathog ; 17(2): e1009300, 2021 02.
Article En | MEDLINE | ID: mdl-33577621

Influenza A virus (IAV) has evolved various strategies to counteract the innate immune response using different viral proteins. However, the mechanism is not fully elucidated. In this study, we identified the PB1 protein of H7N9 virus as a new negative regulator of virus- or poly(I:C)-stimulated IFN induction and specifically interacted with and destabilized MAVS. A subsequent study revealed that PB1 promoted E3 ligase RNF5 to catalyze K27-linked polyubiquitination of MAVS at Lys362 and Lys461. Moreover, we found that PB1 preferentially associated with a selective autophagic receptor neighbor of BRCA1 (NBR1) that recognizes ubiquitinated MAVS and delivers it to autophagosomes for degradation. The degradation cascade mediated by PB1 facilitates H7N9 virus infection by blocking the RIG-I-MAVS-mediated innate signaling pathway. Taken together, these data uncover a negative regulatory mechanism involving the PB1-RNF5-MAVS-NBR1 axis and provide insights into an evasion strategy employed by influenza virus that involves selective autophagy and innate signaling pathways.


Adaptor Proteins, Signal Transducing/metabolism , Autophagy , DNA-Binding Proteins/metabolism , Immunity, Innate/immunology , Influenza, Human/immunology , Intracellular Signaling Peptides and Proteins/metabolism , Ubiquitin-Protein Ligases/metabolism , Viral Proteins/metabolism , Adaptor Proteins, Signal Transducing/genetics , DNA-Binding Proteins/genetics , HEK293 Cells , Humans , Influenza A Virus, H7N9 Subtype/physiology , Influenza, Human/metabolism , Influenza, Human/pathology , Influenza, Human/virology , Intracellular Signaling Peptides and Proteins/genetics , Mitochondria/metabolism , Signal Transduction , Ubiquitin/metabolism , Ubiquitin-Protein Ligases/genetics , Ubiquitination , Viral Proteins/genetics , Virus Replication
7.
Cell Mol Immunol ; 18(10): 2334-2343, 2021 10.
Article En | MEDLINE | ID: mdl-32415261

Cyclic GMP-AMP synthase (cGAS) is a key sensor critical for the recognition of DNA in the cytosol and catalyzes the synthesis of the second messenger cyclic GMP-AMP (cGAMP), which binds to the adapter protein MITA (also known as STING, MPYS, and ERIS) to initiate the innate immune response. How the binding of DNA to and the activation of cGAS are regulated remains poorly understood. Using a biochemical purification approach, we identified poly(rC)-binding protein 1 (PCBP1) as a cGAS-associated protein. PCBP1 was recruited to cGAS in a viral infection-dependent manner. PCBP1 directly bound to DNA and enhanced cGAS binding to its ligands, which was important for cGAS activation. Consistently, PCBP1 deficiency inhibited cytosolic DNA- and DNA virus-triggered transcription of downstream effector genes. These findings suggest that PCBP1 plays an important role in the cGAS-mediated innate immune response to DNA virus infection by promoting the binding of cGAS to viral DNA.


DNA-Binding Proteins , Immunity, Innate , Nucleotidyltransferases , DNA, Viral/genetics , Phosphorylation , Signal Transduction
8.
J Virol ; 94(24)2020 11 23.
Article En | MEDLINE | ID: mdl-33028715

H7N9 influenza A virus (IAV) is an emerged contagious pathogen that may cause severe human infections, even death. Understanding the precise cross talk between virus and host is vital for the development of effective vaccines and therapeutics. In the present study, we identified the nucleoprotein (NP) of H7N9 IAV as a positive regulator of RIG-I like receptor (RLR)-mediated signaling. Based on a loss-of-function strategy, we replaced H1N1 (mouse-adapted PR8 strain) NP with H7N9 NP, by using reverse genetics, and found that the replication and pathogenicity of recombinant PR8-H7N9NP (rPR8-H7N9NP) were significantly attenuated in cells and mice. Biochemical and cellular analyses revealed that H7N9 NP specifically interacts with tumor necrosis factor receptor (TNFR)-associated factor 3 (TRAF3) after viral infection. Subsequently, we identified a PXXQXS motif in the H7N9 NP that may be a determinant for the NP and TRAF3 interaction. Furthermore, H7N9 NP stabilized TRAF3 expression via competitively binding to TRAF3 with cellular inhibitor of apoptosis 2 (cIAP2), leading to the inhibition of the Lys48-linked polyubiquitination and degradation of TRAF3. Taken together, these data uncover a novel mechanism by which the NP of H7N9 IAV positively regulates TRAF3-mediated type I interferon signaling. Our findings provide insights into virus and host survival strategies that involve a specific viral protein that modulates an appropriate immune response in hosts.IMPORTANCE The NS1, PB2, PA-X, and PB1-F2 proteins of influenza A virus (IAV) are known to employ various strategies to counteract and evade host defenses. However, the viral components responsible for the activation of innate immune signaling remain elusive. Here, we demonstrate for the first time that the NP of H7N9 IAV specifically associates with and stabilizes the important adaptor molecule TRAF3, which potentiates RLR-mediated type I interferon induction. Moreover, we reveal that this H7N9 NP protein prevents the interaction between TRAF3 and cIAP2 that mediates Lys48-linked polyubiquitination of TRAF3 for degradation. The current study revealed a novel mechanism by which H7N9 NP upregulates TRAF3-mediated type I interferon production, leading to attenuation of viral replication and pathogenicity in cells and mice. Our finding provides a possible explanation for virus and host commensalism via viral manipulation of the host immune system.


Influenza A Virus, H7N9 Subtype/immunology , Nucleoproteins/metabolism , Signal Transduction/physiology , TNF Receptor-Associated Factor 3/genetics , TNF Receptor-Associated Factor 3/metabolism , A549 Cells , Animals , Apoptosis , Baculoviral IAP Repeat-Containing 3 Protein/metabolism , DEAD Box Protein 58 , Disease Models, Animal , Female , Gene Expression , Humans , Immunity, Innate , Influenza A Virus, H1N1 Subtype/immunology , Interferon Type I/metabolism , Lung/pathology , Mice , Mice, Inbred BALB C , Ubiquitination , Virulence , Virus Replication
9.
PLoS Pathog ; 16(4): e1008457, 2020 04.
Article En | MEDLINE | ID: mdl-32251420

The retinoic acid-inducible gene-I (RIG-I)-like receptors (RLRs), including RIG-I and melanoma differentiation-associated gene 5 (MDA5), sense cytoplasmic viral RNA and initiate innate antiviral responses. How RIG-I and MDA5 are differentially regulated remains enigmatic. In this study, we identified the guanylate-binding protein (GBP) and zinc-finger FYVE domain-containing protein ZFYVE1 as a negative regulator of MDA5- but not RIG-I-mediated innate antiviral responses. ZFYVE1-deficiency promoted MDA5- but not RIG-I-mediated transcription of downstream antiviral genes. Comparing to wild-type mice, Zfyve1-/- mice were significantly protected from lethality induced by encephalomyocarditis virus (EMCV) that is sensed by MDA5, whereas Zfyve1-/- and Zfyve1+/+ mice were comparable to death induced by vesicular stomatitis virus (VSV) that is sensed by RIG-I. Mechanistically, ZFYVE1 interacted with MDA5 but not RIG-I. ZFYVE1 bound to viral RNA and decreased the ligand binding and oligomerization of MDA5. These findings suggest that ZFYVE1 acts as a specific negative regulator of MDA5-mediated innate immune responses by inhibiting its ligand binding and oligomerization.


Cardiovirus Infections/immunology , DEAD Box Protein 58/immunology , Encephalomyocarditis virus/physiology , Interferon-Induced Helicase, IFIH1/immunology , Membrane Proteins/immunology , Animals , Cardiovirus Infections/genetics , Cardiovirus Infections/virology , DEAD Box Protein 58/genetics , Encephalomyocarditis virus/genetics , Humans , Immunity, Innate , Interferon-Induced Helicase, IFIH1/genetics , Membrane Proteins/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout
10.
Front Immunol ; 11: 608976, 2020.
Article En | MEDLINE | ID: mdl-33469458

Transforming growth factor-ß (TGF-ß)-activated kinase 1 (TAK1) is a member of the MAPK kinase kinase (MAPKKK) family and has been implicated in the regulation of a wide range of physiological and pathological processes. TAK1 functions through assembling with its binding partners TAK1-binding proteins (TAB1, TAB2, and TAB3) and can be activated by a variety of stimuli such as tumor necrosis factor α (TNFα), interleukin-1ß (IL-1ß), and toll-like receptor ligands, and they play essential roles in the activation of NF-κB and MAPKs. Numerous studies have demonstrated that post-translational modifications play important roles in properly controlling the activity, stability, and assembly of TAK1-TABs complex according to the indicated cellular environment. This review focuses on the recent advances in TAK1-TABs-mediated signaling and the regulations of TAK1-TABs complex by post-translational modifications.


Adaptor Proteins, Signal Transducing/metabolism , Inflammation/metabolism , MAP Kinase Kinase Kinases/metabolism , Tumor Necrosis Factor-alpha/metabolism , Animals , Humans , Interleukin-1beta/metabolism , Signal Transduction/physiology
11.
Cell Mol Immunol ; 17(7): 741-752, 2020 07.
Article En | MEDLINE | ID: mdl-31388100

Recognition of viral dsRNA by Toll-like receptor 3 (TLR3) leads to the induction of downstream antiviral effectors and the innate antiviral immune response. Here, we identified the zinc-finger FYVE domain-containing protein ZFYVE1, a guanylate-binding protein (GBP), as a positive regulator of TLR3-mediated signaling. Overexpression of ZFYVE1 promoted the transcription of downstream antiviral genes upon stimulation with the synthetic TLR3 ligand poly(I:C). Conversely, ZFYVE1 deficiency had the opposite effect. Zfyve1-/- mice were less susceptible than wild-type mice to inflammatory death induced by poly(I:C) but not LPS. ZFYVE1 was associated with TLR3, and the FYVE domain of ZFYVE1 and the ectodomain of TLR3 were shown to be responsible for their interaction. ZFYVE1 was bound to poly(I:C) and increased the binding affinity of TLR3 to poly(I:C). These findings suggest that ZFYVE1 plays an important role in the TLR3-mediated innate immune and inflammatory responses by promoting the ligand binding of TLR3.


Membrane Proteins/deficiency , Signal Transduction , Toll-Like Receptor 3/metabolism , Zinc Fingers , Animals , Endosomes/drug effects , Endosomes/metabolism , Immunity, Innate/drug effects , Inflammation/pathology , Ligands , Membrane Proteins/metabolism , Mice, Inbred C57BL , Mice, Knockout , Poly I-C/pharmacology , Protein Binding/drug effects , Signal Transduction/drug effects
12.
J Immunol ; 203(1): 259-268, 2019 07 01.
Article En | MEDLINE | ID: mdl-31127032

The dynamic regulations of ubiquitination and deubiquitination play important roles in TGF-ß-activated kinase 1 (TAK1)-mediated NF-κB activation, which regulates various physiological and pathological events. We identified ubiquitin-specific protease (USP)19 as a negative regulator of TNF-α- and IL-1ß-triggered NF-κB activation by deubiquitinating TAK1. Overexpression of USP19 but not its enzymatic inactive mutant inhibited TNF-α- and IL-1ß-triggered NF-κB activation and transcription of downstream genes, whereas USP19 deficiency had the opposite effects. Usp19-/- mice produced higher levels of inflammatory cytokines and were more susceptible to TNF-α- and IL-1ß-triggered septicemia death compared with their wild-type littermates. Mechanistically, USP19 interacted with TAK1 in a TNF-α- or IL-1ß-dependent manner and specifically deconjugated K63- and K27-linked polyubiquitin chains from TAK1, leading to the impairment of TAK1 activity and the disruption of the TAK1-TAB2/3 complex. Our findings provide new insights to the complicated molecular mechanisms of the attenuation of the inflammatory response.


Endopeptidases/metabolism , Inflammation/immunology , MAP Kinase Kinase Kinases/metabolism , NF-kappa B/metabolism , Sepsis/immunology , Adaptor Proteins, Signal Transducing/metabolism , Animals , Disease Susceptibility , Endopeptidases/genetics , HEK293 Cells , Humans , Immune Tolerance , Interleukin-1beta/metabolism , Mice , Mice, Knockout , Protein Binding , RNA, Small Interfering/genetics , Tumor Necrosis Factor-alpha/metabolism , Ubiquitination
13.
Immunity ; 49(3): 438-448.e5, 2018 09 18.
Article En | MEDLINE | ID: mdl-30193849

Recognition of viral RNA by the retinoic acid-inducible gene-I (RIG-I)-like receptors (RLRs) initiates innate antiviral immune response. How the binding of viral RNA to and activation of the RLRs are regulated remains enigmatic. In this study, we identified ZCCHC3 as a positive regulator of the RLRs including RIG-I and MDA5. ZCCHC3 deficiency markedly inhibited RNA virus-triggered induction of downstream antiviral genes, and ZCCHC3-deficient mice were more susceptible to RNA virus infection. ZCCHC3 was associated with RIG-I and MDA5 and functions in two distinct processes for regulation of RIG-I and MDA5 activities. ZCCHC3 bound to dsRNA and enhanced the binding of RIG-I and MDA5 to dsRNA. ZCCHC3 also recruited the E3 ubiquitin ligase TRIM25 to the RIG-I and MDA5 complexes to facilitate its K63-linked polyubiquitination and activation. Thus, ZCCHC3 is a co-receptor for RIG-I and MDA5, which is critical for RLR-mediated innate immune response to RNA virus.


DEAD Box Protein 58/metabolism , RNA Virus Infections/immunology , RNA Viruses/physiology , RNA, Viral/metabolism , RNA-Binding Proteins/metabolism , Animals , Clustered Regularly Interspaced Short Palindromic Repeats , DNA-Binding Proteins/metabolism , Gene Expression Regulation, Viral , HEK293 Cells , Humans , Immunity, Innate , Interferon-Induced Helicase, IFIH1/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Protein Binding , RNA, Viral/immunology , RNA-Binding Proteins/genetics , THP-1 Cells , Transcription Factors/metabolism , Ubiquitination
14.
Nat Commun ; 9(1): 3349, 2018 08 22.
Article En | MEDLINE | ID: mdl-30135424

Cyclic GMP-AMP synthase (cGAS) senses double-strand (ds) DNA in the cytosol and then catalyzes synthesis of the second messenger cGAMP, which activates the adaptor MITA/STING to initiate innate antiviral response. How cGAS activity is regulated remains enigmatic. Here, we identify ZCCHC3, a CCHC-type zinc-finger protein, as a positive regulator of cytosolic dsDNA- and DNA virus-triggered signaling. We show that ZCCHC3-deficiency inhibits dsDNA- and DNA virus-triggered induction of downstream effector genes, and that ZCCHC3-deficient mice are more susceptible to lethal herpes simplex virus type 1 or vaccinia virus infection. ZCCHC3 directly binds to dsDNA, enhances the binding of cGAS to dsDNA, and is important for cGAS activation following viral infection. Our results suggest that ZCCHC3 is a co-sensor for recognition of dsDNA by cGAS, which is important for efficient innate immune response to cytosolic dsDNA and DNA virus.


DNA/metabolism , Immunity, Innate/physiology , Nucleotidyltransferases/metabolism , RNA Nucleotidyltransferases/metabolism , Animals , DNA/genetics , Immunity, Innate/genetics , Mice , Mice, Knockout , Nucleotides, Cyclic/metabolism , Nucleotidyltransferases/genetics , RNA Nucleotidyltransferases/genetics , Signal Transduction/genetics , Signal Transduction/physiology
15.
J Virol ; 92(11)2018 06 01.
Article En | MEDLINE | ID: mdl-29563291

Nonstructural protein 1 (NS1) of influenza A virus regulates innate immune responses via various mechanisms. We previously showed that a naturally occurring deletion (the EALQR motif) in the NS1 effector domain of an H5N1 swine-origin avian influenza virus impairs the inhibition of type I interferon (IFN) in chicken fibroblasts and attenuates virulence in chickens. Here we found that the virus bearing this deletion in its NS1 effector domain showed diminished inhibition of IFN-related cytokine expression and attenuated virulence in mice. We further showed that deletion of the EALQR motif disrupted NS1 dimerization, impairing double-stranded RNA (dsRNA) sequestration and competitive binding with RIG-I. In addition, the EALQR-deleted NS1 protein could not bind to TRIM25, unlike full-length NS1, and was less able to block TRIM25 oligomerization and self-ubiquitination, further impairing the inhibition of TRIM25-mediated RIG-I ubiquitination compared to that with full-length NS1. Our data demonstrate that the EALQR deletion prevents NS1 from blocking RIG-I-mediated IFN induction via a novel mechanism to attenuate viral replication and virulence in mammalian cells and animals.IMPORTANCE H5 highly pathogenic avian influenza viruses have infected more than 800 individuals across 16 countries, with an overall case fatality rate of 53%. Among viral proteins, nonstructural protein 1 (NS1) of influenza virus is considered a key determinant for type I interferon (IFN) antagonism, pathogenicity, and host range. However, precisely how NS1 modulates virus-host interaction, facilitating virus survival, is not fully understood. Here we report that a naturally occurring deletion (of the EALQR motif) in the NS1 effector domain of an H5N1 swine-origin avian influenza virus disrupted NS1 dimerization, which diminished the blockade of IFN induction via the RIG-I signaling pathway, thereby impairing virus replication and virulence in the host. Our study demonstrates that the EALQR motif of NS1 regulates virus fitness to attain a virus-host compromise state in animals and identifies this critical motif as a potential target for the future development of small molecular drugs and attenuated vaccines.


Influenza A Virus, H5N1 Subtype/genetics , Influenza A Virus, H5N1 Subtype/immunology , Interferon Type I/immunology , Viral Nonstructural Proteins/genetics , A549 Cells , Animals , Cell Line, Tumor , Chick Embryo , Chlorocebus aethiops , DNA-Binding Proteins/metabolism , Female , HEK293 Cells , Humans , Immunity, Innate/immunology , Membrane Proteins/metabolism , Mice , Mice, Inbred BALB C , Nerve Tissue Proteins/metabolism , Protein Binding/genetics , Protein Domains/genetics , Receptors, Cell Surface , Sequence Deletion/genetics , THP-1 Cells , Transcription Factors/metabolism , Ubiquitination , Vero Cells , Viral Nonstructural Proteins/metabolism
16.
Cell Mol Immunol ; 15(9): 858-867, 2018 09.
Article En | MEDLINE | ID: mdl-28435159

Interferon-induced transmembrane protein 3 (IFITM3) is a restriction factor that can be induced by viral infection and interferons (IFNs). It inhibits the entry and replication of many viruses, which are independent of receptor usage but dependent on processes that occur in endosomes. In this study, we demonstrate that IFITM3 plays important roles in regulating the RNA-virus-triggered production of IFN-ß in a negative-feedback manner. Overexpression of IFITM3 inhibited Sendai virus-triggered induction of IFN-ß, whereas knockdown of IFITM3 had the opposite effect. We also showed that IFITM3 was constitutively associated with IRF3 and regulated the homeostasis of IRF3 by mediating the autophagic degradation of IRF3. These findings suggest a novel inhibitory function of IFITM3 on the RNA-virus-triggered production of type I IFNs and cellular antiviral responses.


Autophagosomes/metabolism , Interferon Regulatory Factor-3/immunology , Interferon Type I/immunology , Membrane Proteins/immunology , Proteolysis , RNA Virus Infections/immunology , RNA Viruses/immunology , RNA-Binding Proteins/immunology , HEK293 Cells , HeLa Cells , Humans , Interferon Regulatory Factor-3/genetics , Interferon Type I/genetics , Membrane Proteins/genetics , RNA Virus Infections/genetics , RNA Virus Infections/pathology , RNA Viruses/genetics , RNA-Binding Proteins/genetics
17.
J Immunol ; 199(5): 1856-1864, 2017 09 01.
Article En | MEDLINE | ID: mdl-28747347

TLR-mediated signaling pathways play critical roles in host defense against microbials. However, dysregulation of innate immune and inflammatory responses triggered by TLRs would result in harmful damage to the host. Using a Trim8 gene-knockout mouse model, we show that tripartite motif (TRIM) 8 negatively regulates TLR3- and TLR4-mediated innate immune and inflammatory responses. TRIM8 deficiency leads to increased polyinosinic-polycytidylic acid- and LPS-triggered induction of downstream anti-microbial genes including TNF, Il6, Rantes, and Ifnb, evaluated serum cytokine levels, and increased susceptibility of mice to polyinosinic-polycytidylic acid- and LPS-induced inflammatory death as well as Salmonella typhimurium infection-induced loss of body weight and septic shock. TRIM8 interacted with Toll/IL-1 receptor domain-containing adapter-inducing IFN-ß and mediated its K6- and K33-linked polyubiquitination, leading to disruption of the Toll/IL-1 receptor domain-containing adapter-inducing IFN-ß-TANK-binding kinase-1 association. Our findings uncover an additional mechanism on the termination of TLR3/4-mediated inflammatory and innate immune responses.


Adaptor Proteins, Vesicular Transport/metabolism , Carrier Proteins/metabolism , Inflammation/immunology , Nerve Tissue Proteins/metabolism , Protein Serine-Threonine Kinases/metabolism , Salmonella Infections/immunology , Salmonella typhimurium/immunology , Shock, Septic/immunology , Animals , Carrier Proteins/genetics , Cytokines/genetics , Cytokines/metabolism , HEK293 Cells , Humans , Immunity, Innate , Inflammation/microbiology , Inflammation Mediators/metabolism , Lipopolysaccharides/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout , Nerve Tissue Proteins/genetics , Poly I-C/immunology , Protein Binding , Signal Transduction , Toll-Like Receptor 3/metabolism , Toll-Like Receptor 4/metabolism , Ubiquitin-Protein Ligases
18.
J Immunol ; 197(12): 4704-4713, 2016 12 15.
Article En | MEDLINE | ID: mdl-27821666

Duck Tembusu virus (DTMUV) is an emergent infectious pathogen that has caused severe disease in ducks and huge economic losses to the poultry industry in China since 2009. Previously, we showed that DTMUV inhibits IFN-ß induction early in infection; however, the mechanisms of the inhibition of innate immune responses remain poorly understood. In this study, we screened DTMUV-encoded structural and nonstructural proteins using reporter assays and found that DTMUV NS1 markedly suppressed virus-triggered IFN-ß expression by inhibiting retinoic acid-inducible gene I-like receptor signaling. Moreover, we found that DTMUV NS1 specifically interacted with the C-terminal domain of virus-induced signaling adaptor and impaired the association of retinoic acid-inducible gene I or melanoma differentiation-associated gene 5 and virus-induced signaling adaptor, thereby downregulating the retinoic acid-inducible gene I-like receptor-mediated signal transduction and cellular antiviral responses, leading to evasion of the innate immune response. Together, our findings reveal a novel mechanism manipulated by DTMUV to circumvent the host antiviral immune response.


Avian Proteins/metabolism , Bird Diseases/immunology , Ducks/immunology , Flavivirus Infections/immunology , Flavivirus/immunology , Interferon-beta/metabolism , Viral Nonstructural Proteins/immunology , Animals , China , DEAD Box Protein 58/metabolism , Immune Evasion , Immunity, Cellular , Immunity, Innate , Interferon-Induced Helicase, IFIH1/metabolism , Signal Transduction
19.
Nat Immunol ; 17(3): 241-9, 2016 Mar.
Article En | MEDLINE | ID: mdl-26692175

The gene encoding PTEN is one of the most frequently mutated tumor suppressor-encoding genes in human cancer. While PTEN's function in tumor suppression is well established, its relationship to anti-microbial immunity remains unknown. Here we found a pivotal role for PTEN in the induction of type I interferon, the hallmark of antiviral innate immunity, that was independent of the pathway of the kinases PI(3)K and Akt. PTEN controlled the import of IRF3, a master transcription factor responsible for IFN-ß production, into the nucleus. We further identified a PTEN-controlled negative phosphorylation site at Ser97 of IRF3 and found that release from this negative regulation via the phosphatase activity of PTEN was essential for the activation of IRF3 and its import into the nucleus. Our study identifies crosstalk between PTEN and IRF3 in tumor suppression and innate immunity.


Immunity, Innate/immunology , Interferon Regulatory Factor-3/immunology , Interferon Type I/immunology , PTEN Phosphohydrolase/immunology , Respirovirus Infections/immunology , Rhabdoviridae Infections/immunology , Animals , Cell Line , Cell Line, Tumor , Cell Nucleus , Cell Proliferation , Cytokines/immunology , Dendritic Cells/immunology , Electrophoresis, Polyacrylamide Gel , Fluorescent Antibody Technique , Gene Transfer Techniques , HEK293 Cells , Humans , Immunoblotting , Immunoprecipitation , Interferon Regulatory Factor-3/genetics , Interferon Regulatory Factor-7/genetics , MCF-7 Cells , Macrophages/immunology , Mass Spectrometry , Mice , Microscopy, Confocal , Mutagenesis, Site-Directed , PTEN Phosphohydrolase/genetics , Phosphatidylinositol 3-Kinases/metabolism , Phosphorylation , Proto-Oncogene Proteins c-akt/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Sendai virus , Vesiculovirus
20.
EMBO Rep ; 16(4): 447-55, 2015 Apr.
Article En | MEDLINE | ID: mdl-25736436

Toll-like receptors (TLRs) are pattern recognition receptors that sense a variety of pathogens, initiate innate immune responses, and direct adaptive immunity. All TLRs except TLR3 recruit the adaptor MyD88 to ultimately elicit inflammatory gene expression, whereas TLR3 and internalized TLR4 use TIR-domain-containing adaptor TRIF for the induction of type I interferon and inflammatory cytokines. Here, we identify the WD repeat and FYVE-domain-containing protein WDFY1 as a crucial adaptor protein in the TLR3/4 signaling pathway. Overexpression of WDFY1 potentiates TLR3- and TLR4-mediated activation of NF-κB, interferon regulatory factor 3 (IRF3), and production of type I interferons and inflammatory cytokines. WDFY1 depletion has the opposite effect. WDFY1 interacts with TLR3 and TLR4 and mediates the recruitment of TRIF to these receptors. Our findings suggest a crucial role for WDFY1 in bridging the TLR-TRIF interaction, which is necessary for TLR signaling.


Adaptor Proteins, Vesicular Transport/genetics , Nuclear Proteins/genetics , Toll-Like Receptor 3/genetics , Toll-Like Receptor 4/genetics , Transcription Factors/genetics , Adaptor Proteins, Signal Transducing , Adaptor Proteins, Vesicular Transport/immunology , Amino Acid Motifs , Cytokines/genetics , Cytokines/immunology , Gene Expression Regulation , HEK293 Cells , Humans , Interferon Inducers/pharmacology , Interferon Regulatory Factor-3/genetics , Interferon Regulatory Factor-3/immunology , Interferon Type I/genetics , Interferon Type I/immunology , Molecular Sequence Data , Myeloid Differentiation Factor 88/genetics , Myeloid Differentiation Factor 88/immunology , NF-kappa B/genetics , NF-kappa B/immunology , Nuclear Proteins/antagonists & inhibitors , Nuclear Proteins/immunology , Plasmids/chemistry , Plasmids/immunology , Poly I-C/pharmacology , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Signal Transduction , Toll-Like Receptor 3/immunology , Toll-Like Receptor 4/immunology , Transcription Factors/antagonists & inhibitors , Transcription Factors/immunology , Transfection
...