Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 54
Filter
1.
J Cell Physiol ; 238(10): 2348-2360, 2023 Oct.
Article in English | MEDLINE | ID: mdl-37565597

ABSTRACT

Innate immunity is the first line of defense against infections, which functions as a significant role in resisting pathogen invasion. Rapid immune response is initiated by pattern recognition receptors (PRRs) quickly distinguishing "self" and "non-self." Upon evolutionarily conserved pathogen-associated molecular pattern (PAMP) is recognized by PRRs, innate immune response against infection is triggered via an orchestration of molecular interaction, cytokines cascades, and immune cells. RIG-I plays a critical role in type I interferon (IFN-I) production by direct recognition of cytoplasmic double-stranded viral RNA. However, the activation mechanism of RIG-I is incompletely understood. In this study, we reported RNA-binding protein ZFP36 as a positive regulator of RIG-I-mediated IFN-I production. ZFP36 is a member of Zinc finger proteins (ZFPs) characterized by the zinc finger (ZnF) motif, which broadly involved gene transcription and signal transduction. However, its role in regulating antiviral innate immune signaling is still unclear. We found that ZFP36 associates with RIG-I and potentiates the FN-ß production induced by SeV. Mechanistically, ZFP36 promotes K63-linked polyubiquitination of RIG-I, mostly at K154/K164/K172, thereby facilitating the activation of RIG-I during infection. While the mutant ZFP36 (C118S/C162S) failed to increase polyubiquitination of RIG-I and SeV induced FN-ß. Our findings collectively demonstrated that ZFP36 acts as a positive regulator in antiviral innate immunity by targeting RIG-I for K63-linked ubiquitination, thus improving our understanding of the activation mechanism of RIG-I.

2.
Acta Biochim Biophys Sin (Shanghai) ; 55(10): 1582-1591, 2023 10 25.
Article in English | MEDLINE | ID: mdl-37580950

ABSTRACT

Retinoic acid-inducible gene I (RIG-I) is a cytosolic viral RNA receptor. Upon viral infection, the protein recognizes and then recruits adapter protein mitochondrial antiviral signaling (MAVS) protein, initiating the production of interferons and proinflammatory cytokines to establish an antiviral state. In the present study, we identify zinc finger protein 205 (ZNF205) which associates with RIG-I and promotes the Sendai virus (SeV)-induced antiviral innate immune response. Overexpression of ZNF205 facilitates interferon-beta (IFN-ß) introduction, whereas ZNF205 deficiency restricts its introduction. Mechanistically, the C-terminal zinc finger domain of ZNF205 interacts with the N-terminal tandem caspase recruitment domains (CARDs) of RIG-I; this interaction markedly promotes K63 ubiquitin-linked polyubiquitination of RIG-I, which is crucial for RIG-I activation. Thus, our results demonstrate that ZNF205 is a positive regulator of the RIG-I-mediated innate antiviral immune signaling pathway.


Subject(s)
Immunity, Innate , Signal Transduction , DEAD Box Protein 58/genetics , DEAD Box Protein 58/metabolism , Immunity, Innate/genetics , Interferon-beta/genetics , Interferons/genetics , Ubiquitin-Protein Ligases/metabolism , Ubiquitination , Sendai virus/immunology
3.
Virol Sin ; 38(3): 419-428, 2023 Jun.
Article in English | MEDLINE | ID: mdl-37086853

ABSTRACT

TANK-binding kinase 1 (TBK1) is a nodal protein involved in multiple signal transduction pathways. In RNA virus-mediated innate immunity, TBK1 is recruited to the prion-like platform formed by MAVS and subsequently activates the transcription factors IRF3/7 and NF-κB to produce type I interferon (IFN) and proinflammatory cytokines for the signaling cascade. In this study, TRAF7 was identified as a negative regulator of innate immune signaling. TRAF7 interacts with TBK1 and promotes K48-linked polyubiquitination and degradation of TBK1 through its RING domain, impairing the activation of IRF3 and the production of IFN-ß. In addition, we found that the conserved cysteine residues at position 131 of TRAF7 are necessary for its function toward TBK1. Knockout of TRAF7 could facilitate the activation of IRF3 and increase the transcript levels of downstream antiviral genes. These data suggest that TRAF7 negatively regulates innate antiviral immunity by promoting the K48-linked ubiquitination of TBK1.


Subject(s)
Interferon Type I , Signal Transduction , Humans , Ubiquitination , Immunity, Innate , Antiviral Agents , HEK293 Cells , Protein Serine-Threonine Kinases/genetics , Tumor Necrosis Factor Receptor-Associated Peptides and Proteins
4.
Front Immunol ; 13: 972184, 2022.
Article in English | MEDLINE | ID: mdl-36045679

ABSTRACT

The virus-induced signaling adaptor protein VISA (also known as MAVS, ISP-1, Cardif) is a critical adaptor protein in the innate immune response to RNA virus infection. Upon viral infection, VISA self-aggregates to form a sizeable prion-like complex and recruits downstream signal components for signal transduction. Here, we discover that BAG6 (BCL2-associated athanogene 6, formerly BAT3 or Scythe) is an essential negative regulator in the RIG-I-like receptor signaling pathway. BAG6 inhibits the aggregation of VISA by promoting the K48-linked ubiquitination and specifically attenuates the recruitment of TRAF2 by VISA to inhibit RLR signaling. The aggregation of VISA and the interaction of VISA and TRAF2 are enhanced in BAG6-deficient cell lines after viral infection, resulting in the enhanced transcription level of downstream antiviral genes. Our research shows that BAG6 is a critical regulating factor in RIG-I/VISA-mediated innate immune response by targeting VISA.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Molecular Chaperones/metabolism , Nuclear Proteins/metabolism , Signal Transduction , Virus Diseases , Animals , Humans , Mice , Molecular Chaperones/genetics , TNF Receptor-Associated Factor 2/metabolism , Ubiquitin-Protein Ligases/metabolism , Ubiquitination
5.
Virus Genes ; 58(2): 122-132, 2022 Apr.
Article in English | MEDLINE | ID: mdl-35103914

ABSTRACT

Mitochondrial virus-induced signal adaptor (MAVS), also known as VISA, IPS-1, and Cardif, is a crucial adaptor protein in the RIG-I-like receptor (RLR) signaling pathway. Upon viral infection, RIG-I recognizes viral dsRNA and further transfers it to mitochondria, where it binds to MAVS through its CARD domain, generating a series of signal cascades. Transduction through this signaling cascade leads to phosphorylation and nuclear translocation of interferon regulatory factor 3/7 (IRF3/IRF7) and activation of NF-κB, which ultimately produces type I interferon (IFN) and proinflammatory cytokines. Here, our experiments demonstrated that overexpression of SRY-related high-mobility group protein 9 (SOX9) significantly inhibited Sendai virus (SeV)-induced and MAVS-mediated activation of the IFN-ß promoter and ISRE. However, knocking out the expression of SOX9 in cells promoted SeV-induced IFN-ß promoter and ISRE activation. Further studies have shown that SOX9 interacts with MAVS and targets MAVS to inhibit the association of MAVS-TRAF2, thereby inhibiting MAVS-mediated TRAF2 ubiquitination. Taken together, these results indicate that SOX9 downregulates IFN-ß expression and antiviral signal transduction by targeting MAVS.


Subject(s)
Antiviral Agents , SOX9 Transcription Factor , Signal Transduction , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/metabolism , Mitochondria/metabolism , SOX9 Transcription Factor/metabolism , Ubiquitination
6.
Microbiol Spectr ; 9(3): e0145821, 2021 12 22.
Article in English | MEDLINE | ID: mdl-34908452

ABSTRACT

TANK-binding kinase 1 (TBK1)/IκB kinase-ε (IKKε) mediates robust production of type I interferons (IFN-I) and proinflammatory cytokines in response to acute viral infection. However, excessive or prolonged production of IFN-I is harmful and even fatal to the host by causing autoimmune disorders. In this study, we identified mitogen-activated protein kinase kinase kinase kinase 1 (MAP4K1) as a negative regulator in the RIG-I-like receptor (RLR) signaling pathway. MAP4K1, a member of Ste20-like serine/threonine kinases, was previously known as a prominent regulator in adaptive immunity by downregulating T-cell receptor (TCR) signaling and B-cell receptor (BCR) signaling. However, its role in regulating antiviral innate immune signaling is still unclear. This study reports an undiscovered role of MAP4K1, which inhibits RLR signaling by targeting TBK1/IKKε for proteasomal degradation via the ubiquitin ligase DTX4. We initially identify MAP4K1 as an interacting partner of TBK1 by yeast two-hybrid screens and subsequently investigate its function in RLR-mediated antiviral signaling pathways. Overexpression of MAP4K1 significantly inhibits RNA virus-triggered activation of IFN-ß and the production of proinflammatory cytokines. Consistently, knockdown or knockout experiments show opposite effects. Furthermore, MAP4K1 promotes the degradation of TBK1/IKKε by K48-linked ubiquitination via DTX4. Knockdown of DTX4 abrogated the ubiquitination and degradation of TBK1/IKKε. Collectively, our results identify that MAP4K1 acts as a negative regulator in antiviral innate immunity by targeting TBK1/IKKε, discover a novel TBK1 inhibitor, and extend a novel functional role of MAP4K1 in immunity. IMPORTANCE TANK-binding kinase 1 (TBK1)/IκB kinase-ε (IKKε) mediates robust production of type I interferons (IFN-I) and proinflammatory cytokines to restrict the spread of invading viruses. However, excessive or prolonged production of IFN-I is harmful to the host by causing autoimmune disorders. In this study, we identified that mitogen-activated protein kinase kinase kinase kinase 1 (MAP4K1) is a negative regulator in the RLR signaling pathway. Notably, MAP4K1 promotes the degradation of TBK1/IKKε by K48-linked ubiquitination via the ubiquitin ligase DTX4, leading to the negative regulation of the IFN signaling pathway. Previous studies showed that MAP4K1 has a pivotal function in adaptive immune responses. This study identifies that MAP4K1 also plays a vital role in innate immunity and outlines a novel mechanism by which the IFN signaling pathway is tightly controlled to avoid excessive inflammation. Our study documents a novel TBK1 inhibitor, which serves as a potential therapeutic target for autoimmune diseases, and elucidated a significant function for MAP4K1 linked to innate immunity in addition to subsequent adaptive immunity.


Subject(s)
Cytokines/biosynthesis , I-kappa B Kinase/metabolism , Interferon-beta/biosynthesis , Protein Serine-Threonine Kinases/metabolism , Virus Diseases/immunology , DEAD Box Protein 58/metabolism , Humans , Immunity, Innate/immunology , Protein Serine-Threonine Kinases/genetics , RNA Viruses/immunology , Receptors, Immunologic/metabolism , Signal Transduction/immunology , Ubiquitination
7.
Front Microbiol ; 12: 770600, 2021.
Article in English | MEDLINE | ID: mdl-34880843

ABSTRACT

Mitochondrial antiviral signaling protein (MAVS), an adaptor protein, is activated by RIG-I, which is critical for an effective innate immune response to infection by various RNA viruses. Viral infection causes the RIG-I-like receptor (RLR) to recognize pathogen-derived dsRNA and then becomes activated to promote prion-like aggregation and activation of MAVS. Subsequently, through the recruitment of TRAF proteins, MAVS activates two signaling pathways mediated by TBK1-IRF3 and IKK- NF-κb, respectively, and turns on type I interferon and proinflammatory cytokines. This study discovered that NEDD4 binding protein 3 (N4BP3) is a positive regulator of the RLR signaling pathway by targeting MAVS. Overexpression of N4BP3 promoted virus-induced activation of the interferon-ß (IFN-ß) promoter and interferon-stimulated response element (ISRE). Further experiments showed that knockdown or knockout N4BP3 impaired RIG-I-like receptor (RLR)-mediated innate immune response, induction of downstream antiviral genes, and cellular antiviral responses. We also detected that N4BP3 could accelerate the interaction between MAVS and TRAF2. Related experiments revealed that N4BP3 could facilitate the ubiquitination modification of MAVS. These findings suggest that N4BP3 is a critical component of the RIG-I-like receptor (RLR)-mediated innate immune response by targeting MAVS, which also provided insight into the mechanisms of innate antiviral responses.

8.
Mol Immunol ; 134: 62-71, 2021 06.
Article in English | MEDLINE | ID: mdl-33713958

ABSTRACT

Retinoic acid-inducible gene I (RIG-I) plays a critical role in the recognition of intracytoplasmic viral RNA. Upon binding to the RNA of invading viruses, the activated RIG-I translocates to mitochondria, where it recruits adapter protein MAVS, causing a series of signaling cascades. In this study, we demonstrated that Hsp70 binding protein 1 (HSPBP1) promotes RIG-I-mediated signal transduction. The overexpression of HSPBP1 can increase the stability of RIG-I protein by inhibiting its K48-linked ubiquitination, and promote the activation of IRF3 and the production of IFN-ß induced by Sendai virus. Knockdown and knockout of HSPBP1 leads to down-regulation of virus-induced RIG-I expression, inhibits IRF3 activation, and reduces the production of IFNB1. These results indicate that HSPBP1 positively regulates the antiviral signal pathway induced by inhibiting the K48-linked ubiquitination of RIG-I.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , DEAD Box Protein 58/metabolism , Immunity, Innate/immunology , Receptors, Immunologic/metabolism , Signal Transduction/immunology , Adaptor Proteins, Signal Transducing/immunology , DEAD Box Protein 58/immunology , HEK293 Cells , Humans , Receptors, Immunologic/immunology , Respirovirus Infections/immunology , Sendai virus/immunology , Ubiquitination
9.
Mol Immunol ; 132: 117-125, 2021 04.
Article in English | MEDLINE | ID: mdl-33582548

ABSTRACT

Upon recognition of intracytoplasmic viral RNA, activated RIG-I is recruited to the mitochondrion-located adaptor protein VISA (also known as MAVS, CARDIF, and IPS-1). VISA then acts as a central signaling platform for linking RIG-I and downstream signaling components, such as TRAF2, 5, and 6, TBK1, and IKK, leading to activation of the kinases TBK1 and IKK. These activated kinases further phosphorylate the transcription factors IRF3/7 and NF-κB, leading to the induction of downstream antiviral genes. Here, we report a mitochondrial isoform, deoxyuridine triphosphate nucleotidohydrolase (dUTPase), DUT-M, as a positive regulator in RLR-VISA-mediated antiviral signaling. DUT-M interacts with VISA and RIG-I to facilitate the assembly of the VISA-TRAF2 complex and to augment the polyubiquitination of TRAF2, leading to potentiated activation of IRF3 dimerization and phosphorylation of P65, and enhanced VISA-mediated innate immune response. RLR-VISA-mediated IRF3 dimerization and P65 phosphorylation, were inhibited in DUT-knockdown and DUT-deficient 293 cells. Thus, DUT-M is a positive regulator of the RIG-I-VISA-mediated innate immune response to RNA viruses.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Antiviral Agents/metabolism , Mitochondria/metabolism , Pyrophosphatases/metabolism , Signal Transduction/physiology , TNF Receptor-Associated Factor 2/metabolism , HEK293 Cells , Humans , Immunity, Innate/physiology , Interferon Regulatory Factor-3/metabolism , Phosphorylation/physiology , Ubiquitin-Protein Ligases/metabolism , Ubiquitination/physiology
11.
Am J Otolaryngol ; 41(4): 102472, 2020.
Article in English | MEDLINE | ID: mdl-32276733

ABSTRACT

OBJECTIVE: To observe the type of nystagmus in each position of posterior semicircular canal benign paroxysmal positional vertigo (BPPV) after treatment with the Epley maneuver and analyze the relationship between the type of nystagmus in the second and third positions of the Epley maneuver and the effect of treatment. Then, the role of orthotropic nystagmus in predicting the success of posterior semicircular canal BPPV treatment was explored. METHODS: Two hundred seventy-six patients diagnosed with posterior semicircular canal BPPV who were admitted from September 2018 to October 2019 to Zhejiang Hospital were included. All patients were treated with BPPV diagnosis and treatment system (Epley maneuver). During the treatment, we observed and recorded the type of nystagmus in the second and third positions, including the direction and duration of nystagmus. One hour after the first treatment, all patients were evaluated by both the Dix-Hallpike and Roll tests to determine whether the treatment was successful. The difference in the success rate of treatment between different types of nystagmus was compared, and the differences in sensitivity and specificity of orthotropic nystagmus in the second and third positions in predicting the effect of treatment were compared. RESULTS: Among the 234 patients who had successful repositioning for the first time, the proportion of orthotropic nystagmus during the third position of the Epley maneuver was 88.9%, which was significantly higher than 23% in the unsuccessful group (42 cases) (P < 0.05) The proportion of patients with reversed nystagmus (4.7% vs 33.3%, P < 0.05) and no nystagmus (6.4% vs 42.9%, P < 0.05) was lower in the successful group than in the unsuccessful group. The proportion of orthotropic nystagmus during the second position of the Epley maneuver was 50.9%, which was also higher than the 19% in the unsuccessful group (P < 0.05). The proportion of reversed nystagmus (13.7% vs 31%, P < 0.05) was lower in the successful group than in the unsuccessful group. Additionally, the proportion of no nystagmus (35.5% vs 50%, P = 0.074) was lower in the successful group than in the unsuccessful group, but the difference was not statistically significant. The sensitivity of orthotropic nystagmus in the third position (88.9%) of the Epley maneuver in predicting the efficacy of treatment was higher than that of orthotropic nystagmus in the second position (50.9%), but there was no significant difference in specificity between the two. CONCLUSION: Orthotropic nystagmus during the Epley maneuver, especially in the third position, has certain value in predicting the efficacy of posterior semicircular canal BPPV repositioning, which is better than its predictive effect in the second position, whereas reversed nystagmus or no nystagmus in the third position is suggestive of unsuccessful repositioning. Therefore, clinicians can carry out individualized treatments based on nystagmus types during repositioning to improve the effect of treatment.


Subject(s)
Benign Paroxysmal Positional Vertigo/diagnosis , Benign Paroxysmal Positional Vertigo/therapy , Diagnostic Techniques, Otological , Nystagmus, Physiologic , Patient Positioning/methods , Physical Therapy Modalities , Adult , Aged , Aged, 80 and over , Benign Paroxysmal Positional Vertigo/physiopathology , Benign Paroxysmal Positional Vertigo/rehabilitation , Female , Humans , Male , Middle Aged , Predictive Value of Tests , Treatment Outcome , Young Adult
12.
Am J Otolaryngol ; 41(3): 102412, 2020.
Article in English | MEDLINE | ID: mdl-32057490

ABSTRACT

OBJECTIVES: To evaluate the efficacy of automatic benign paroxysmal positional vertigo (BPPV) diagnosis and treatment system for BPPV compared with the manual repositioning group. METHODS: Two hundred thirty patients diagnosed as idiopathic BPPV who were admitted from August 2018 to July 2019 in Zhejiang Hospital were included. Among them, 150 patients of posterior semicircular canal BPPV(pc-BPPV), 53 patients of horizontal semicircular canal BPPV(hc-BPPV), and 27 patients of horizontal semicircular canal calculus (hc-BPPV-cu) were randomly treated with BPPV diagnosis and treatment system(the experimental group) or manual repositioning (the control group). Resolution of vertigo and nystagmus on the Dix-Hallpike and Roll test on day 3,day 7,day 14 and day 28 follow-up after first treatment was the main outcome measure to assess the efficacy of treatment. RESULTS: At 3-day and 7-day follow-up after treatment with BPPV diagnosis and treatment system, 79%, 91%had complete resolution of vertigo and nystagmus, the effective rate in the experimental group were significantly higher than those in the control group, the differences were statistically significant(P < .05). On day 14, the effective rate in the experimental group (96%) was slightly higher than that in the control group(91%), but there was no significant difference between the two groups. And at 28-day after the first treatment, the effective rate was 100% in the experimental group and the control group. The repositioning efficiency of pc-BPPV (the first, second, third treatment), hc-BPPV (the first, second, third treatment), hc-BPPV-cu(the first, second treatment) in the experimental group were higher than the control group, and the secondary reposition of pc-BPPV in the experimental group was significantly higher than the control group(96%vs.84%; P < .05). While for the hc-BPPV-cu patients, the effective rate of the third treatment in the experimental group was slightly lower than that of the control group, but the differences were not statistically significant. CONCLUSIONS: BPPV diagnosis and treatment system is effective for the treatment of BPPV, with a better effective rate than those treated with manual maneuver, and is safe and easy to perform on patients.


Subject(s)
Benign Paroxysmal Positional Vertigo/diagnosis , Benign Paroxysmal Positional Vertigo/therapy , Moving and Lifting Patients/instrumentation , Adult , Aged , Aged, 80 and over , Female , Follow-Up Studies , Humans , Male , Middle Aged , Treatment Outcome , Young Adult
13.
Biochem Biophys Res Commun ; 522(4): 889-896, 2020 02 19.
Article in English | MEDLINE | ID: mdl-31806368

ABSTRACT

Upon invading the cell, the viral RNA is recognized by the RIG-I receptor located in the cytoplasm, causing the RIG-I receptor to be activated. The activated RIG-I receptor transmits downstream antiviral signals by interacting with the adaptor protein VISA located on the mitochondria, leading to the production of type Ⅰ interferons and crude inflammatory cytokine genes. Although there have been many studies on antiviral signal transduction of RIG-I receptors in recent years, the mechanism of RIG-I-VISA-mediated antiviral regulation is still not fully understood. In this study, we identified SNX5 as a negative regulator of RLR-mediated antiviral signaling. Our results show that overexpression of SNX5 inhibits viral-induced activation of the IFN-ß promoter, ISRE, NF-κB, and IRF3, whereas RNAi knockdown of SNX5 expression shows opposite results. We also found that overexpression of SNX5 enhanced RIG-I's K48 ubiquitination and attenuated its K63 ubiquitination, resulting in inhibition of virus-induced RIG-I expression. Besides, further studies show that SNX5 overexpression weakens the interaction between VISA and TRAF2/5. Our findings suggest that SNX5 negatively regulates RLR-mediated antiviral signaling by targeting the RIG-I-VISA signalosome and provide new evidence for the negative regulation of RIG-I-mediated innate immune response mechanisms.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Antiviral Agents/metabolism , DEAD Box Protein 58/metabolism , Signal Transduction , Sorting Nexins/metabolism , Gene Knockout Techniques , HEK293 Cells , Humans , Receptors, Immunologic , Sendai virus , Tumor Necrosis Factor Receptor-Associated Peptides and Proteins/metabolism , Ubiquitination
14.
J Med Virol ; 91(9): 1668-1678, 2019 09.
Article in English | MEDLINE | ID: mdl-31106867

ABSTRACT

Retinoic acid-inducible gene-I (RIG-I) belongs to the RIGI-like receptors (RLRs), a class of primary pattern recognition receptors. It senses viral double-strand RNA in the cytoplasm and delivers the activated signal to its adaptor virus-induced signaling adapter (VISA), which then recruits the downstream TNF receptor-associated factors and kinases, triggering a downstream signal cascade that leads to the production of proinflammatory cytokines and antiviral interferons (IFNs). However, the mechanism of RIG-I-mediated antiviral signaling is not fully understood. Here, we demonstrate that chitinase domain-containing 1 (CHID1), a member of the chitinase family, positively regulates the RLR antiviral signaling pathway by targeting the RIG-I/VISA signalosome. CHID1 overexpression enhances the activation of nuclear factor κB (NF-кB) and interferon regulatory factor 3 (IRF3) triggered by Sendai virus (SeV) by promoting the polyubiquitination of RIG-I and VISA, thereby potentiating IFN-ß production. CHID1 knockdown in human 239T cells inhibits SeV-induced activation of IRF3 and NF-κB and the induction of IFN-ß. These results indicate that CHID1 positively regulates RLR antiviral signal, revealing the novel mechanism of the RIG-I antiviral signaling pathway.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Carrier Proteins/metabolism , DEAD Box Protein 58/metabolism , Receptors, Pattern Recognition/metabolism , Signal Transduction , Carrier Proteins/genetics , Gene Expression , Gene Knockdown Techniques , HEK293 Cells , Humans , Interferon-beta/biosynthesis , Proteome , Proteomics/methods , Receptors, Immunologic , Ubiquitination
15.
Mol Immunol ; 109: 116-125, 2019 05.
Article in English | MEDLINE | ID: mdl-30927622

ABSTRACT

Interferon regulatory factor 7 (IRF7), a crucial regulator of type I interferons (IFNs), plays a crucial role in resistance to viral infection. The abnormal production of type I IFNs is associated with many types of disease, such as cancer and inflammatory disorders. Thus, understanding the post-translational modifications of IRF7 is essential to promoting an appropriate immune response. We have recently showed that the TAR RNA binding protein 2 (TARBP2) suppresses IFN-ß production and the innate antiviral response by targeting MAVS. Here, we further identified TARBP2 as a novel inhibitor of IRF7, which inhibits IRF7-mediated IFN-ß production triggered by the Sendai virus in 293 T cells. Overexpression of TARBP2 inhibits the phosphorylation as well as the K63-linked ubiquitination of IRF7, whilst TARBP2 also impairs the stability of endogenous TRAF6. Furthermore, TARBP2 participates in the interaction between IRF7 and TRAF6, thereby suppressing TRAF6-mediated K63-linked ubiquitination of IRF7, which is a prerequisite of IRF7 phosphorylation. Our findings further reveal the mechanism by which TARBP2 regulates the antiviral signaling pathways of the innate immune system.


Subject(s)
Interferon Regulatory Factor-7/metabolism , Lysine/metabolism , RNA-Binding Proteins/metabolism , TNF Receptor-Associated Factor 6/metabolism , Ubiquitination , HEK293 Cells , Humans , Interferon-beta/metabolism , Phosphorylation , Protein Binding , Proteolysis , Sendai virus/physiology , Signal Transduction
16.
Viruses ; 11(2)2019 02 15.
Article in English | MEDLINE | ID: mdl-30769920

ABSTRACT

RNA virus invasion induces a cytosolic RIG-I-like receptor (RLR) signaling pathway by promoting assembly of the Mitochondrial antiviral-signaling protein (MAVS) signalosome and triggers the rapid production of type I interferons (IFNs) and proinflammatory cytokines. During this process, the pivotal kinase TANK binding kinase 1 (TBK1) is recruited to the MAVS signalosome to transduce a robust innate antiviral immune response by phosphorylating transcription factors interferon regulatory factor 3 (IRF3) and nuclear factor (NF)-κB and promoting their nuclear translocation. However, the molecular mechanisms underlying the negative regulation of TBK1 are largely unknown. In the present study, we found that THO complex subunit 7 homolog (THOC7) negatively regulated the cellular antiviral response by promoting the proteasomal degradation of TBK1. THOC7 overexpression potently inhibited Sendai virus- or polyI:C-induced IRF3 dimerization and phosphorylation and IFN-ß production. In contrast, THOC7 knockdown had the opposite effects. Moreover, we simulated a node-activated pathway to show that THOC7 regulated the RIG-I-like receptors (RLR)-/MAVS-dependent signaling cascade at the TBK1 level. Furthermore, THOC7 was involved in the MAVS signalosome and promoted TBK1 degradation by increasing its K48 ubiquitin-associated polyubiquitination. Together, these findings suggest that THOC7 negatively regulates type I IFN production by promoting TBK1 proteasomal degradation, thus improving our understanding of innate antiviral immune responses.


Subject(s)
Immunity, Cellular , Immunity, Innate , Protein Serine-Threonine Kinases/metabolism , RNA-Binding Proteins/metabolism , Sendai virus/immunology , Gene Expression Regulation , HEK293 Cells , Humans , Interferon Regulatory Factor-3/metabolism , Interferon Type I/immunology , MCF-7 Cells , Phosphorylation , Proteasome Endopeptidase Complex/immunology , Protein Binding , Protein Serine-Threonine Kinases/genetics , RNA-Binding Proteins/genetics , Sendai virus/genetics , Signal Transduction , Ubiquitin/metabolism , Ubiquitination
17.
Biochem Biophys Res Commun ; 508(3): 667-674, 2019 01 15.
Article in English | MEDLINE | ID: mdl-30527812

ABSTRACT

Virus-induced signaling adaptor (VISA), which mediates the production of type I interferon, is crucial for the retinoic acid-inducible gene I (RIG-I)-like receptor (RLR) signaling pathway. Upon viral infection, RIG-I recognizes double-stranded viral RNA and interacts with VISA to mediate antiviral innate immunity. However, the mechanisms underlying RIG/VISA-mediated antiviral regulation remain unclear. In this study, we confirmed that receptor for activated C kinase 1 (RACK1) interacts with VISA and attenuates the RIG/VISA-mediated antiviral innate immune signaling pathway. Overexpression of RACK1 inhibited the interferon-ß (IFN-ß) promoter; interferon-stimulated response element (ISRE); nuclear factor kappa B (NF-κB) activation; and dimerization of interferon regulatory factor 3 (IRF3) mediated by RIG-I, VISA, and TANK-binding kinase 1 (TBK1). A reduction in RACK1 expression level upon small interfering RNA knockdown increased RIG/VISA-mediated antiviral transduction. Additionally, RACK1 disrupted formation of the VISA-tumor necrosis factor receptor-associated factor 2 (TRAF2), VISA-TRAF3, and VISA-TRAF6 complexes during RIG-I/VISA-mediated signal transduction. Additionally, RACK1 enhanced K48-linked ubiquitination of VISA, attenuated its K63-linked ubiquitination, and decreased VISA-mediated antiviral signal transduction. Together, these results indicate that RACK1 interacts with VISA to repress downstream signaling and downregulates virus-induced IFN-ß production in the RIG-I/VISA signaling pathway.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Antiviral Agents/metabolism , DEAD Box Protein 58/metabolism , Neoplasm Proteins/metabolism , Receptors for Activated C Kinase/metabolism , Signal Transduction , Gene Knockdown Techniques , Humans , Interferon-beta/biosynthesis , Interferon-beta/metabolism , Lysine/metabolism , Multiprotein Complexes/metabolism , Promoter Regions, Genetic/genetics , Protein Serine-Threonine Kinases/metabolism , RNA, Small Interfering/metabolism , Sendai virus/physiology , Tumor Necrosis Factor Receptor-Associated Peptides and Proteins/metabolism , Ubiquitination
18.
J Med Virol ; 91(3): 482-492, 2019 03.
Article in English | MEDLINE | ID: mdl-30267576

ABSTRACT

The mitochondrial antiviral signal protein mitochondrial antiviral signaling protein, also known as virus-induced signaling adaptor (VISA), plays a key role in regulating host innate immune signaling pathways. This study identifies FK506 binding protein 8 (FKBP8) as a candidate interacting protein of VISA through the yeast two-hybrid technique. The interaction of FKBP8 with VISA, retinoic acid inducible protein 1 (RIG-I), and IFN regulatory factor 3 (IRF3) was confirmed during viral infection in mammalian cells by coimmunoprecipitation. Overexpression of FKBP8 using a eukaryotic expression plasmid significantly attenuated Sendai virus-induced activation of the promoter interferons ß (IFN-ß), and transcription factors nuclear factor κ-light chain enhancer of activated B cells (NF-κB) and IFN-stimulated response element (ISRE). Overexpression of FKBP8 also decreased dimer-IRF3 activity, but enhanced virus replication. Conversely, knockdown of FKBP8 expression by RNA interference showed opposite effects. Further studies indicated that FKBP8 acts as a negative interacting partner to regulate RLR-VISA signaling by acting on VISA and TANK binding kinase 1 (TBK1). Additionally, FKBP8 played a negative role on virus-induced signaling by inhibiting the formation of TBK1-IRF3 and VISA-TRAF3 complexes. Notably, FKBP8 also promoted the degradation of TBK1, RIG-I, and TRAF3 resulting from FKBP8 reinforced Sendai virus-induced endogenous polyubiquitination of RIG-I, TBK1, and TNF receptor-associated factor 3 (TRAF3). Therefore, a novel function of FKBP8 in innate immunity antiviral signaling regulation was revealed in this study.


Subject(s)
Adaptor Proteins, Signal Transducing/immunology , Immunity, Innate , Sendai virus , Signal Transduction , Tacrolimus Binding Proteins/genetics , Adaptor Proteins, Signal Transducing/antagonists & inhibitors , DEAD Box Protein 58/genetics , DEAD Box Protein 58/immunology , HEK293 Cells , Humans , Interferon Regulatory Factor-3/genetics , Interferon Regulatory Factor-3/immunology , NF-kappa B/genetics , NF-kappa B/immunology , Protein Binding , Protein Serine-Threonine Kinases/immunology , Receptors, Immunologic , TNF Receptor-Associated Factor 3/genetics , TNF Receptor-Associated Factor 3/immunology , Two-Hybrid System Techniques , Ubiquitination
19.
Mol Immunol ; 104: 1-10, 2018 12.
Article in English | MEDLINE | ID: mdl-30390472

ABSTRACT

MAVS as an essential receptor protein for anti-virus innate immunity plays an important role in the production of virus-induced typeⅠ interferon and regulation of interferon regulatory factor 3/7. Understanding the MAVS-mediated antiviral signaling pathway can provide detailed insights. In this study, we identify transactivation response element RNA-binding protein (TARBP2), as an inhibitor of the cellular protein kinase PKR, negatively regulates virus -induced IFN-ß production by targets MAVS. Overexpression of TARBP2 inhibits virus-induced IFN-ß production as well as cellular antiviral response. Then knockdown of TARBP2 inhibited virus-induced IFN-ß signaling. Further studies demonstrated that TARBP2 interacted with MAVS and targeted MAVS to abrogate MAVS-RIG-I and MAVS-TRAF3 association. Our findings suggest that TARBP2 is an important non-redundant virus-mediated negative regulator of typeⅠ interferon.


Subject(s)
Adaptor Proteins, Signal Transducing/immunology , Immunity, Innate , Interferon-beta/immunology , RNA-Binding Proteins/immunology , Virus Diseases/immunology , Adaptor Proteins, Signal Transducing/genetics , HEK293 Cells , Humans , Interferon-beta/genetics , RNA-Binding Proteins/genetics , Virus Diseases/genetics
20.
Mol Med Rep ; 18(2): 2458-2466, 2018 Aug.
Article in English | MEDLINE | ID: mdl-29916539

ABSTRACT

Mitochondrial anti­viral signaling protein (VISA), additionally termed MAVS, IPS­1 and Cardif, is located at the outer membrane of mitochondria and is an essential adaptor in the Rig­like receptor (RLRs) signaling pathway. Upon viral infection, activated RLRs interact with VISA on mitochondria, forming a RLR­VISA platform, leading to the recruitment of different TRAF family members, including TRAF3, TRAF2 and TRAF6. This results in the phosphorylation and nuclear translocation of interferon regulatory factors 3 and 7 (IRF3/IRF7) by TANK binding kinase 1 (TBK1) and/or IKKε, as well as activation of NF­κB, to induce type I interferons (IFNs) and pro­inflammatory cytokines. It remains to be elucidated how VISA functions as a scaffold for protein complex assembly in mitochondria to regulate RLR­VISA antiviral signaling. In the present study, it was demonstrated that HAUS augmin like complex subunit 8 (HAUS8) augments the RLR­VISA­dependent antiviral signaling pathway by targeting the VISA complex. Co­immunoprecipitation verified that HAUS8 was associated with VISA and the VISA signaling complex components retinoic acid­inducible gene I (RIG­I) and TBK1 when the RLR­VISA signaling pathway was activated. The data demonstrated that overexpression of HAUS8 significantly promoted the activity of the transcription factors NF­κB, IRF3 and the IFN­ß promoter induced by Sendai virus­mediated RLR­VISA signaling. HAUS8 increased the polyubiquitination of VISA, RIG­I and TBK1. Knockdown of HAUS8 inhibited the activation of the transcription factors IRF­3, NF­κB and the IFN­ß promoter triggered by Sendai virus. Collectively, these results demonstrated that HAUS8 may function as a positive regulator of RLR­VISA dependent antiviral signaling by targeting the VISA complex, providing a novel regulatory mechanism of antiviral responses.


Subject(s)
Adaptor Proteins, Signal Transducing/genetics , Microtubule-Associated Proteins/genetics , Sendai virus/genetics , Virus Diseases/genetics , Antiviral Agents/chemistry , Antiviral Agents/therapeutic use , Humans , Interferon Regulatory Factor-3/genetics , Interferon Regulatory Factor-7/genetics , Interferon Type I/genetics , Interferon-beta/genetics , Intracellular Signaling Peptides and Proteins , Mitochondria , NF-kappa B/genetics , Sendai virus/pathogenicity , Signal Transduction/genetics , TNF Receptor-Associated Factor 2/genetics , TNF Receptor-Associated Factor 3/genetics , TNF Receptor-Associated Factor 6/genetics , Ubiquitination , Virus Diseases/prevention & control , Virus Diseases/virology
SELECTION OF CITATIONS
SEARCH DETAIL
...