Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
Add more filters










Publication year range
1.
Front Immunol ; 12: 642715, 2021.
Article in English | MEDLINE | ID: mdl-33815396

ABSTRACT

A systematic and flexible immunoregulatory network is required to ensure the proper outcome of antiviral immune signaling and maintain homeostasis during viral infection. Tumor necrosis factor-α-induced protein 8-like 2 (TIPE2), a novel immunoregulatory protein, has been extensively studied in inflammatory response, apoptosis, and cancer. However, the function of TIPE2 in antiviral innate immunity is poorly clarified. In this study, we reported that the expression of TIPE2 declined at the early period and then climbed up in macrophages under RNA virus stimulation. Knockout of TIPE2 in the macrophages enhanced the antiviral capacity and facilitated type I interferon (IFN) signaling after RNA viral infection both in vitro and in vivo. Consistently, overexpression of TIPE2 inhibited the production of type I IFNs and pro-inflammatory cytokines, and thus promoted the viral infection. Moreover, TIPE2 restrained the activation of TBK1 and IRF3 in the retinoic acid inducible gene-I (RIG-I)-like receptors (RLR) signaling pathway by directly interacting with retinoic acid inducible gene-I (RIG-I). Taken together, our results suggested that TIPE2 suppresses the type I IFN response induced by RNA virus by targeting RIG-I and blocking the activation of downstream signaling. These findings will provide new insights to reveal the immunological function of TIPE2 and may help to develop new strategies for the clinical treatment of RNA viral infections.


Subject(s)
DEAD Box Protein 58/physiology , Intracellular Signaling Peptides and Proteins/physiology , Macrophages/immunology , RNA Virus Infections/immunology , Receptors, Immunologic/physiology , Tumor Necrosis Factor-alpha/pharmacology , Animals , Cells, Cultured , Humans , Immunity, Innate , Interferon Type I/antagonists & inhibitors , Interferon Type I/biosynthesis , Mice , Mice, Inbred C57BL , Signal Transduction/drug effects , Virus Replication
2.
J Infect Dev Ctries ; 15(1): 1-8, 2021 01 31.
Article in English | MEDLINE | ID: mdl-33571140

ABSTRACT

An innate immune response is essential to mobilize protective immunity upon the infection of respiratory epithelial cells with influenza A virus (IAV). The response is classified as early (nonspecific effectors), local systematic (effector cells recruitment) and late (antigen to lymphoid organ transport, naive B and T cells recognition, effector cells clonal expansion and differentiation). Virus particles are detected by the host cells as non-self by various sensors that are present on the cell surface, endosomes and cytosol. These sensors are collectively termed as pattern recognition receptors (PRRs). The PRRs distinguish unique molecular signatures known as pathogen-associated molecular pattern, which are present either on the cell surface or within intracellular compartments. PRRs have been classified into five major groups: C-Type Lectin Receptor (CLR), Toll-like receptor (TLR), Nod-like receptor (NLR), Retinoic acid-inducible gene-I-like receptor (RLR), which play a role in innate immunity to IAV infection, and the pyrin and hematopoietic interferon-inducible nuclear (PYHIN) domain protein. Here, we discuss the role of PRRs in cellular infectivity of IAV and highlight the recent progress.


Subject(s)
Influenza A virus/physiology , Influenza A virus/pathogenicity , Influenza, Human/immunology , Influenza, Human/virology , Receptors, Pattern Recognition/physiology , Signal Transduction , Animals , DEAD Box Protein 58/physiology , Host Microbial Interactions , Humans , Immunity, Innate , Lectins, C-Type/physiology , Nod1 Signaling Adaptor Protein/physiology , Nuclear Proteins/physiology , Receptors, Immunologic/physiology , Receptors, Virus/physiology , Toll-Like Receptors/physiology
3.
Biochem J ; 478(3): 493-510, 2021 02 12.
Article in English | MEDLINE | ID: mdl-33459340

ABSTRACT

An integral aspect of innate immunity is the ability to detect foreign molecules of viral origin to initiate antiviral signaling via pattern recognition receptors (PRRs). One such receptor is the RNA helicase retinoic acid inducible gene 1 (RIG-I), which detects and is activated by 5'triphosphate uncapped double stranded RNA (dsRNA) as well as the cytoplasmic viral mimic dsRNA polyI:C. Once activated, RIG-I's CARD domains oligomerize and initiate downstream signaling via mitochondrial antiviral signaling protein (MAVS), ultimately inducing interferon (IFN) production. Another dsRNA binding protein PACT, originally identified as the cellular protein activator of dsRNA-activated protein kinase (PKR), is known to enhance RIG-I signaling in response to polyI:C treatment, in part by stimulating RIG-I's ATPase and helicase activities. TAR-RNA-binding protein (TRBP), which is ∼45% homologous to PACT, inhibits PKR signaling by binding to PKR as well as by sequestration of its' activators, dsRNA and PACT. Despite the extensive homology and similar structure of PACT and TRBP, the role of TRBP has not been explored much in RIG-I signaling. This work focuses on the effect of TRBP on RIG-I signaling and IFN production. Our results indicate that TRBP acts as an inhibitor of RIG-I signaling in a PACT- and PKR-independent manner. Surprisingly, this inhibition is independent of TRBP's post-translational modifications that are important for other signaling functions of TRBP, but TRBP's dsRNA-binding ability is essential. Our work has major implications on viral susceptibility, disease progression, and antiviral immunity as it demonstrates the regulatory interplay between PACT and TRBP IFN production.


Subject(s)
Carrier Proteins/physiology , DEAD Box Protein 58/physiology , RNA-Binding Proteins/physiology , Receptors, Immunologic/physiology , Signal Transduction/physiology , Active Transport, Cell Nucleus , Adenosine Triphosphate/metabolism , Animals , Fibroblasts , Genes, Reporter , HEK293 Cells , Humans , Immunity, Innate , Interferon Regulatory Factor-3/genetics , Interferon Regulatory Factor-3/metabolism , Interferons/physiology , Mice , Models, Biological , Mutation , Phosphorylation , Poly I-C/pharmacology , Protein Binding , Protein Domains , Protein Processing, Post-Translational , RNA, Double-Stranded/metabolism , Recombinant Fusion Proteins/metabolism , Recombinant Proteins/metabolism , Sequence Homology, Amino Acid
4.
Mol Immunol ; 130: 69-76, 2021 02.
Article in English | MEDLINE | ID: mdl-33360745

ABSTRACT

Ubiquitin specific protease 14 (USP14) is a regulator of protein deubiquitination and proteasome activation, and has been implicated in negative regulation of type I IFN signaling pathway. However, the effect of USP14 on RNA virus-related inflammatory response has not been studied. Retinoic acid-inducible gene I (RIG-I) is the important pattern recognition receptor of the innate immunity to detect RNA viruses or intracellular Poly(I:C)-LMW. Here, we reported that USP14 knockdown increased pro-inflammatory cytokines production in macrophages upon VSV infection or intracellular Poly(I:C)-LMW stimulation. USP14-overexpressed HeLa cells exhibited a decrease in RIG-I-mediated IL-6 and TNF-α expression. IU1, USP14 inhibitor, significantly promotes pro-inflammatory cytokines production in VSV-infected mice in vivo. Furthermore, USP14 was also found to inhibit the RIG-I-triggered NF-κB activation by deubiquitinating K63-linked RIG-I. Thus, our results demonstrate that USP14 is a negative regulator of RIG-I-mediated inflammatory response.


Subject(s)
DEAD Box Protein 58/genetics , Interleukin-6/metabolism , NF-kappa B/metabolism , Receptors, Immunologic/genetics , Tumor Necrosis Factor-alpha/metabolism , Ubiquitin Thiolesterase/physiology , Animals , Cells, Cultured , DEAD Box Protein 58/physiology , Down-Regulation/genetics , Down-Regulation/immunology , Female , Gene Expression Regulation , HEK293 Cells , HeLa Cells , Humans , Inflammation/genetics , Inflammation/metabolism , Macrophages, Peritoneal/immunology , Macrophages, Peritoneal/metabolism , Mice , Mice, Inbred C57BL , Receptors, Immunologic/physiology , Signal Transduction/genetics , Signal Transduction/immunology , THP-1 Cells
5.
PLoS Biol ; 18(12): e3000996, 2020 12.
Article in English | MEDLINE | ID: mdl-33264285

ABSTRACT

RNA interference (RNAi) is an antiviral pathway common to many eukaryotes that detects and cleaves foreign nucleic acids. In mammals, mitochondrially localized proteins such as mitochondrial antiviral signaling (MAVS), retinoic acid-inducible gene I (RIG-I), and melanoma differentiation-associated protein 5 (MDA5) mediate antiviral responses. Here, we report that mitochondrial dysfunction in Caenorhabditis elegans activates RNAi-directed silencing via induction of a pathway homologous to the mammalian RIG-I helicase viral response pathway. The induction of RNAi also requires the conserved RNA decapping enzyme EOL-1/DXO. The transcriptional induction of eol-1 requires DRH-1 as well as the mitochondrial unfolded protein response (UPRmt). Upon mitochondrial dysfunction, EOL-1 is concentrated into foci that depend on the transcription of mitochondrial RNAs that may form double-stranded RNA (dsRNA), as has been observed in mammalian antiviral responses. Enhanced RNAi triggered by mitochondrial dysfunction is necessary for the increase in longevity that is induced by mitochondrial dysfunction.


Subject(s)
DEAD Box Protein 58/metabolism , Mitochondria/metabolism , RNA Interference/physiology , Adaptor Proteins, Signal Transducing , Animals , Caenorhabditis elegans/genetics , Caenorhabditis elegans Proteins/metabolism , DEAD Box Protein 58/physiology , DEAD-box RNA Helicases/metabolism , Gene Expression Regulation/genetics , Interferon-Induced Helicase, IFIH1/metabolism , Mitochondria/physiology , RNA, Double-Stranded/genetics , RNA, Small Interfering/genetics , RNA, Viral/genetics , Signal Transduction/genetics
6.
Front Immunol ; 11: 678, 2020.
Article in English | MEDLINE | ID: mdl-32425931

ABSTRACT

The innate immune system, which senses invading pathogens, plays a critical role as the first line of host defense. After recognition of foreign RNA ligands (e.g., RNA viruses), host cells generate an innate immune or antiviral response via the interferon-mediated signaling pathway. Retinoic acid-inducible gene I (RIG-1) acts as a major sensor that recognizes a broad range of RNA ligands in mammals; however, chickens lack a RIG-1 homolog, meaning that RNA ligands should be recognized by other cellular sensors such as melanoma differentiation-associated protein 5 (MDA5) and toll-like receptors (TLRs). However, it is unclear which of these cellular sensors compensates for the loss of RIG-1 to act as the major sensor for RNA ligands. Here, we show that chicken MDA5 (cMDA5), rather than chicken TLRs (cTLRs), plays a pivotal role in the recognition of RNA ligands, including poly I:C and influenza virus. First, we used a knockdown approach to show that both cMDA5 and cTLR3 play roles in inducing interferon-mediated innate immune responses against RNA ligands in chicken DF-1 cells. Furthermore, targeted knockout of cMDA5 or cTLR3 in chicken DF-1 cells revealed that loss of cMDA5 impaired the innate immune responses against RNA ligands; however, the responses against RNA ligands were retained after loss of cTLR3. In addition, double knockout of cMDA5 and cTLR3 in chicken DF-1 cells abolished the innate immune responses against RNA ligands, suggesting that cMDA5 is the major sensor whereas cTLR3 is a secondary sensor. Taken together, these findings provide an understanding of the functional role of cMDA5 in the recognition of RNA ligands in chicken DF-1 cells and may facilitate the development of an innate immune-deficient cell line or chicken model.


Subject(s)
Immunity, Innate , Interferon-Induced Helicase, IFIH1/physiology , RNA, Double-Stranded/metabolism , Toll-Like Receptor 3/physiology , Animals , Cell Line , Chickens , DEAD Box Protein 58/physiology , Fibroblasts/immunology , Interferon-beta/genetics , Ligands , Orthomyxoviridae/physiology , Poly I-C/pharmacology , Promoter Regions, Genetic , Virus Replication
7.
Nat Rev Immunol ; 20(9): 537-551, 2020 09.
Article in English | MEDLINE | ID: mdl-32203325

ABSTRACT

Retinoic acid-inducible gene I (RIG-I)-like receptors (RLRs) are key sensors of virus infection, mediating the transcriptional induction of type I interferons and other genes that collectively establish an antiviral host response. Recent studies have revealed that both viral and host-derived RNAs can trigger RLR activation; this can lead to an effective antiviral response but also immunopathology if RLR activities are uncontrolled. In this Review, we discuss recent advances in our understanding of the types of RNA sensed by RLRs in the contexts of viral infection, malignancies and autoimmune diseases. We further describe how the activity of RLRs is controlled by host regulatory mechanisms, including RLR-interacting proteins, post-translational modifications and non-coding RNAs. Finally, we discuss key outstanding questions in the RLR field, including how our knowledge of RLR biology could be translated into new therapeutics.


Subject(s)
DEAD Box Protein 58/physiology , RNA/metabolism , Animals , Humans , Immune System/physiology , RNA, Viral/metabolism
8.
Front Immunol ; 10: 1069, 2019.
Article in English | MEDLINE | ID: mdl-31139191

ABSTRACT

The serine/threonine kinase phosphatase and tensin homolog (PTEN)-induced putative kinase 1(PINK1) controls mitochondrial quality and plays a vital role in the pathogenesis of early-onset Parkinson's disease. However, whether PINK1 has functions in innate antiviral immunity is largely unknown. Here, we report that viral infection down regulates PINK1 expression in macrophages. PINK1 knockdown results in decreased cytokine production and attenuated IRF3 and NF-κB activation upon viral infection. PINK1 promotes the retinoic-acid-inducible gene I (RIG-I)-like receptors (RLR)-triggered immune responses in a kinase domain-dependent manner. Furthermore, PINK1 associates with TRAF3 via the kinase domain and inhibits Parkin-mediated TRAF3 K48-linked proteasomal degradation. In addition, PINK1 interacts with Yes-associated protein 1 (YAP1) upon viral infection and impairs YAP1/IRF3 complex formation. Collectively, our results demonstrate that PINK1 positively regulates RIG-I triggered innate immune responses by inhibiting TRAF3 degradation and relieving YAP-mediated inhibition of the cellular antiviral response.


Subject(s)
DEAD Box Protein 58/physiology , Mitochondrial Proteins/physiology , Protein Kinases/physiology , Virus Diseases/immunology , Adaptor Proteins, Signal Transducing/physiology , Animals , Cytokines/biosynthesis , Female , HEK293 Cells , Humans , Infant , Interferon Regulatory Factor-3/physiology , Macrophages/immunology , Male , Mice , Mice, Inbred C57BL , NF-kappa B/physiology , RAW 264.7 Cells , Receptors, Immunologic , Signal Transduction/physiology , TNF Receptor-Associated Factor 3/physiology , Transcription Factors/physiology , Ubiquitination , YAP-Signaling Proteins
9.
Biomed J ; 41(4): 218-233, 2018 08.
Article in English | MEDLINE | ID: mdl-30348265

ABSTRACT

The respiratory epithelium is the major interface between the environment and the host. Sophisticated barrier, sensing, anti-microbial and immune regulatory mechanisms have evolved to help maintain homeostasis and to defend the lung against foreign substances and pathogens. During influenza virus infection, these specialised structural cells and populations of resident immune cells come together to mount the first response to the virus, one which would play a significant role in the immediate and long term outcome of the infection. In this review, we focus on the immune defence machinery of the respiratory epithelium and briefly explore how it repairs and regenerates after infection.


Subject(s)
Influenza, Human/immunology , Respiratory Mucosa/immunology , Cell Polarity , Chemokines/physiology , Cytokines/physiology , DEAD Box Protein 58/physiology , Humans , Interferons/physiology , NLR Proteins/physiology , Tight Junctions/physiology , Toll-Like Receptors/physiology
10.
J Immunol ; 198(3): 1274-1284, 2017 02 01.
Article in English | MEDLINE | ID: mdl-28011935

ABSTRACT

Upon virus infection, host cells use retinoic-acid-inducible geneI I (RIG-I)-like receptors to recognize viral RNA and activate type I IFN expression. To investigate the role of protein methylation in the antiviral signaling pathway, we screened all the SET domain-containing proteins and identified TTLL12 as a negative regulator of RIG-I signaling. TTLL12 contains SET and TTL domains, which are predicted to have lysine methyltransferase and tubulin tyrosine ligase activities, respectively. Exogenous expression of TTLL12 represses IFN-ß expression induced by Sendai virus. TTLL12 deficiency by RNA interference and CRISPR-gRNA techniques increases the induced IFN-ß expression and inhibits virus replication in the cell. The global gene expression profiling indicated that TTLL12 specifically inhibits the expression of the downstream genes of innate immunity pathways. Cell fractionation and fluorescent staining indicated that TTLL12 is localized in the cytosol. The mutagenesis study suggested that TTLL12's ability to repress the RIG-I pathway is probably not dependent on protein modifications. Instead, TTLL12 directly interacts with virus-induced signaling adaptor (VISA), TBK1, and IKKε, and inhibits the interactions of VISA with other signaling molecules. Taken together, our findings demonstrate TTLL12 as a negative regulator of RNA-virus-induced type I IFN expression by inhibiting the interaction of VISA with other proteins.


Subject(s)
Adaptor Proteins, Signal Transducing/physiology , Carrier Proteins/physiology , Interferon Type I/physiology , Signal Transduction/physiology , Carrier Proteins/analysis , Cell Line , Cytosol/chemistry , DEAD Box Protein 58/physiology , Humans , I-kappa B Kinase/physiology , Immunity, Innate , Protein Serine-Threonine Kinases/physiology , Receptors, Immunologic , Virus Replication
11.
PLoS One ; 11(11): e0166088, 2016.
Article in English | MEDLINE | ID: mdl-27824940

ABSTRACT

Insects are not only major vectors of mammalian viruses, but are also host to insect-restricted viruses that can potentially be transmitted to mammals. While mammalian innate immune responses to arboviruses are well studied, less is known about how mammalian cells respond to viruses that are restricted to infect only invertebrates. Here we demonstrate that IIV-6, a DNA virus of the family Iridoviridae, is able to induce a type I interferon-dependent antiviral immune response in mammalian cells. Although IIV-6 is a DNA virus, we demonstrate that the immune response activated during IIV-6 infection is mediated by the RIG-I-like receptor (RLR) pathway, and not the canonical DNA sensing pathway via cGAS/STING. We further show that RNA polymerase III is required for maximal IFN-ß secretion, suggesting that viral DNA is transcribed by this enzyme into an RNA species capable of activating the RLR pathway. Finally, we demonstrate that the RLR-driven mammalian innate immune response to IIV-6 is functionally capable of protecting cells from subsequent infection with the arboviruses Vesicular Stomatitis virus and Kunjin virus. These results represent a novel example of an invertebrate DNA virus activating a canonically RNA sensing pathway in the mammalian innate immune response, which reduces viral load of ensuing arboviral infection.


Subject(s)
DEAD Box Protein 58/physiology , DNA Virus Infections/immunology , Immunity, Innate/immunology , Iridoviridae/immunology , Animals , Cell Line , Immunoblotting , Interferon-beta/metabolism , Mice , Mice, Inbred C57BL , Microscopy, Confocal , Microscopy, Electron, Transmission , RNA Interference , RNA Polymerase III/metabolism , Reverse Transcriptase Polymerase Chain Reaction
SELECTION OF CITATIONS
SEARCH DETAIL