Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 50
1.
Cell Chem Biol ; 29(7): 1113-1125.e6, 2022 07 21.
Article En | MEDLINE | ID: mdl-35728599

The increasingly frequent outbreaks of pathogenic viruses have underlined the urgent need to improve our arsenal of antivirals that can be deployed for future pandemics. Innate immunity is a powerful first line of defense against pathogens, and compounds that boost the innate response have high potential to act as broad-spectrum antivirals. Here, we harnessed localization-dependent protein-complementation assays (called Alpha Centauri) to measure the nuclear translocation of interferon regulatory factors (IRFs), thus providing a readout of innate immune activation following viral infection that is applicable to high-throughput screening of immunomodulatory molecules. As proof of concept, we screened a library of kinase inhibitors on severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection and identified Gilteritinib as a powerful enhancer of innate responses to viral infection. This immunostimulatory activity of Gilteritinib was found to be dependent on the AXL-IRF7 axis and results in a broad and potent antiviral activity against unrelated RNA viruses.


COVID-19 , Virus Diseases , Antiviral Agents/pharmacology , Humans , Immunity, Innate , SARS-CoV-2 , Virus Diseases/drug therapy
2.
J Virol ; 96(10): e0187521, 2022 05 25.
Article En | MEDLINE | ID: mdl-35475668

Persistent infection with some mucosal α-genus human papillomaviruses (HPVs; the most prevalent one being HPV16) can induce cervical carcinoma, anogenital cancers, and a subset of head and neck squamous cell carcinoma (HNSCC). Cutaneous ß-genus HPVs (such as HPV5 and HPV8) associate with skin lesions that can progress into squamous cell carcinoma with sun exposure in Epidermodysplasia verruciformis patients and immunosuppressed patients. Here, we analyzed mechanisms used by E6 proteins from the α- and ß-genus to inhibit the interferon-ß (IFNB1) response. HPV16 E6 mediates this effect by a strong direct interaction with interferon regulatory factor 3 (IRF3). The binding site of E6 was localized within a flexible linker between the DNA-binding domain and the IRF-activation domain of IRF3 containing an LxxLL motif. The crystallographic structure of the complex between HPV16 E6 and the LxxLL motif of IRF3 was solved and compared with the structure of HPV16 E6 interacting with the LxxLL motif of the ubiquitin ligase E6AP. In contrast, cutaneous HPV5 and HPV8 E6 proteins bind to the IRF3-binding domain (IBiD) of the CREB-binding protein (CBP), a key transcriptional coactivator in IRF3-mediated IFN-ß expression. IMPORTANCE Persistent HPV infections can be associated with the development of several cancers. The ability to persist depends on the ability of the virus to escape the host immune system. The type I interferon (IFN) system is the first-line antiviral defense strategy. HPVs carry early proteins that can block the activation of IFN-I. Among mucosal α-genus HPV types, the HPV16 E6 protein has a remarkable property to strongly interact with the transcription factor IRF3. Instead, cutaneous HPV5 and HPV8 E6 proteins bind to the IRF3 cofactor CBP. These results highlight the versatility of E6 proteins to interact with different cellular targets. The interaction between the HPV16 E6 protein and IRF3 might contribute to the higher prevalence of HPV16 than that of other high-risk mucosal HPV types in HPV-associated cancers.


Interferon Regulatory Factor-3 , Interferon-beta , Oncogene Proteins, Viral , Papillomavirus Infections , Repressor Proteins , Human papillomavirus 16/metabolism , Humans , Interferon Regulatory Factor-3/genetics , Interferon Regulatory Factor-3/metabolism , Interferon-beta/metabolism , Mucous Membrane/virology , Oncogene Proteins, Viral/genetics , Oncogene Proteins, Viral/metabolism , Papillomaviridae/metabolism , Repressor Proteins/genetics , Repressor Proteins/metabolism , Skin/virology
3.
J Immunol ; 207(10): 2589-2597, 2021 11 15.
Article En | MEDLINE | ID: mdl-34625522

Respiratory syncytial virus (RSV) infection in infancy is associated with increased risk of asthma, except in those with allergic disease at the time of infection. Using house dust mite allergen, we examined the effect of pre-existing atopy on postviral airway disease using Sendai virus in mice, which models RSV infection in humans. Sendai virus drives postviral airway disease in nonatopic mice; however, pre-existing atopy protected against the development of airway disease. This protection depended upon neutrophils, as depletion of neutrophils at the time of infection restored the susceptibility of atopic mice to postviral airway disease. Associated with development of atopy was an increase in polymorphonuclear neutrophil-dendritic cell hybrid cells that develop in Th2 conditions and demonstrated increased viral uptake. Systemic inhibition of IL-4 reversed atopic protection against postviral airway disease, suggesting that increased virus uptake by neutrophils was IL-4 dependent. Finally, human neutrophils from atopic donors were able to reduce RSV infection of human airway epithelial cells in vitro, suggesting these findings could apply to the human. Collectively our data support the idea that pre-existing atopy derives a protective neutrophil response via potential interaction with IL-4, preventing development of postviral airway disease.


Hypersensitivity, Immediate/immunology , Neutrophils/immunology , Respiratory Syncytial Virus Infections/immunology , Respirovirus Infections/immunology , Animals , Humans , Mice , Mice, Inbred BALB C , Respiratory Syncytial Viruses/immunology , Sendai virus/immunology
4.
J Virol ; 95(9)2021 04 12.
Article En | MEDLINE | ID: mdl-33536170

N6-Methyladenosine (m6A) is the most abundant internal RNA modification catalyzed by host RNA methyltransferases. As obligate intracellular parasites, many viruses acquire m6A methylation in their RNAs. However, the biological functions of viral m6A methylation are poorly understood. Here, we found that viral m6A methylation serves as a molecular marker for host innate immunity to discriminate self from nonself RNA and that this novel biological function of viral m6A methylation is universally conserved in several families in nonsegmented negative-sense (NNS) RNA viruses. Using m6A methyltransferase (METTL3) knockout cells, we produced m6A-deficient virion RNAs from the representative members of the families Pneumoviridae, Paramyxoviridae, and Rhabdoviridae and found that these m6A-deficient viral RNAs triggered significantly higher levels of type I interferon compared to the m6A-sufficient viral RNAs, in a RIG-I-dependent manner. Reconstitution of the RIG-I pathway revealed that m6A-deficient virion RNA induced higher expression of RIG-I, bound to RIG-I more efficiently, enhanced RIG-I ubiquitination, and facilitated RIG-I conformational rearrangement and oligomerization. Furthermore, the m6A binding protein YTHDF2 is essential for suppression of the type I interferon signaling pathway, including by virion RNA. Collectively, our results suggest that several families in NNS RNA viruses acquire m6A in viral RNA as a common strategy to evade host innate immunity.IMPORTANCE The nonsegmented negative-sense (NNS) RNA viruses share many common replication and gene expression strategies. There are no vaccines or antiviral drugs for many of these viruses. We found that representative members of the families Pneumoviridae, Paramyxoviridae, and Rhabdoviridae among the NNS RNA viruses acquire m6A methylation in their genome and antigenome as a means to escape recognition by host innate immunity via a RIG-I-dependent signaling pathway. Viral RNA lacking m6A methylation induces a significantly higher type I interferon response than m6A-sufficient viral RNA. In addition to uncovering m6A methylation as a common mechanism for many NNS RNA viruses to evade host innate immunity, this study discovered a novel strategy to enhance type I interferon responses, which may have important applications in vaccine development, as robust innate immunity will likely promote the subsequent adaptive immunity.


Adenosine/analogs & derivatives , Host Microbial Interactions/immunology , Interferon Type I/immunology , Negative-Sense RNA Viruses , RNA Virus Infections , RNA, Viral/genetics , A549 Cells , Adenosine/genetics , Gene Expression Regulation, Viral , Gene Knockout Techniques , Humans , Immunity, Innate , Methyltransferases/genetics , Negative-Sense RNA Viruses/genetics , Negative-Sense RNA Viruses/immunology , Negative-Sense RNA Viruses/pathogenicity , RNA Processing, Post-Transcriptional , RNA Virus Infections/immunology , RNA Virus Infections/virology
5.
Eur J Immunol ; 50(12): 1959-1975, 2020 12.
Article En | MEDLINE | ID: mdl-32644192

Plasmacytoid dendritic cells (DCs) are reported to induce robust type-I interferon (IFN) response, whereas cDC1 DCs develop moderate type-I IFN response upon TLR9 stimulation. It is very interesting to understand how this signaling under TLR9 is tightly regulated for the induction of type-I IFNs. Here, we report co-repressor protein NCoR1 as the major factor fine-tuning the signaling pathways regulating IFN-ß expression under TLR9 in cDC1 DCs. We found that NCoR1 knockdown induced a robust IFN-ß-mediated antiviral response upon TLR9 activation in cDC1 DCs. At the molecular level, we showed that NCoR1 directly repressed MyD88-IRF7 signaling axis in cDC1 cells. Therefore, NCoR1 depletion enhanced pIRF7 levels, IFN-ß secretion, and downstream pSTAT1-pSTAT2 signaling, leading to sustained induction of IFN stimulatory genes. Integrative genomic analysis depicted strong enrichment of an antiviral gene-module in CpG-activated NCoR1 knockdown DCs upon TLR9 activation. Moreover, we confirmed our findings in primary DCs derived from splenocytes of WT and NCoR1 DC-/- animals, which showed protection from Sendai and Vesicular Stomatitis viruses upon CpG activation. Ultimately, we identified that NCoR1-HDAC3 complex is involved in repressing the type-I IFN response in cDC1 DCs.


Dendritic Cells/metabolism , Interferon Regulatory Factor-7/metabolism , Interferon Type I/metabolism , Myeloid Differentiation Factor 88/metabolism , Nuclear Receptor Co-Repressor 1/metabolism , Toll-Like Receptor 9/metabolism , Animals , Cells, Cultured , Female , Mice , Mice, Inbred C57BL , Mice, Knockout , Signal Transduction/physiology
6.
Vaccines (Basel) ; 8(2)2020 Apr 29.
Article En | MEDLINE | ID: mdl-32365702

Small ruminant lentiviruses (SRLVs) are widely spread in the ovine and caprine populations, causing an incurable disease affecting animal health and production. Vaccine development is hindered owing to the high genetic heterogeneity of lentiviruses and the selection of T-cell and antibody escape mutants, requiring antigen delivery optimization. Sendai virus (SeV) is a respiratory paramyxovirus in mice that has been recognized as a potent inducer of innate immune responses in several species, including mouse and human. The aim of this study was to stimulate an innate antiviral response in ovine cells and evaluate the potential inhibitory effect upon small ruminant lentivirus (SRLV) infections. Ovine alveolar macrophages (AMs), blood-derived macrophages (BDMs), and skin fibroblasts (OSFs) were stimulated through infection with SeV encoding green fluorescent protein (GFP). SeV efficiently infected ovine cells, inducing an antiviral state in AM from SRLV naturally-infected animals, as well as in in vitro SRLV-infected BDM and OSF from non-infected animals. Supernatants from SeV-infected AM induced an antiviral state when transferred to fresh cells challenged with SRLV. Similar to SRLV, infectivity of an HIV-1-GFP lentiviral vector was also restricted in ovine cells infected with SeV. In myeloid cells, an M1-like proinflammatory polarization was observed together with an APOBEC3Z1 induction, among other lentiviral restriction factors. Our observations may boost new approximations in ameliorating the SRLV burden by stimulation of the innate immune response using SeV-based vaccine vectors.

7.
Arch Virol ; 165(8): 1899-1903, 2020 Aug.
Article En | MEDLINE | ID: mdl-32462284

Tacaribe virus (TCRV) is the prototype of the New World arenaviruses (also known as TCRV serocomplex viruses). While TCRV is not itself a human pathogen, many closely related members of this group cause hemorrhagic fever, and thus TCRV has long served as an important BSL2 system for research into diverse areas of arenavirus biology. Due to its widespread use, a coding-complete sequence for both the S and L segments of the bipartite genome has been publically available for almost 30 years. However, more recently, this sequence has been found to contain significant discrepancies compared to other samples of the same original strain (i.e., TRVL-11573). Further, it is incomplete with respect to the genome ends, which contain critical regulatory elements for RNA synthesis. In order to rectify these issues we now present the first complete genome sequence for this important prototype arenavirus. In addition to completing the S segment 5' end, we identified an apparent error in the L segment 3' end as well as substantial discrepancies in the S segment intergenic region likely to affect folding. Comparison of this sequence with existing partial sequences confirmed a 12-amino-acid deletion in GP, including putative glycosylation sites, and a 4-amino-acid exchange flanking the exonuclease domain of NP. Accounting for these corrections, the TRVL-11573 strain appears to be nearly identical to that isolated in Florida in 2012. The availability of this information provides a solid basis for future molecular and genetic work on this important prototype arenavirus.


Arenaviruses, New World/genetics , Florida , Humans , Regulatory Elements, Transcriptional/genetics , Virus Replication/genetics , Whole Genome Sequencing/methods
8.
Mol Cell ; 71(1): 6-7, 2018 07 05.
Article En | MEDLINE | ID: mdl-29979969

In a recent Cell paper, Jiang et al. (2018) have shown that lnc-Lsm3b, a long non-coding RNA induced by type I IFN late in the infection in mouse macrophages, prevents further activation of RIG-I acting as a decoy for RIG-I.


Immunity, Innate , RNA, Long Noncoding , Animals , Cell Communication , Mice
9.
PLoS Pathog ; 14(4): e1006962, 2018 04.
Article En | MEDLINE | ID: mdl-29630666

Despite their genetic similarities, enteric and respiratory enteroviruses (EVs) have highly heterogeneous biophysical properties and cause a vast diversity of human pathologies. In vitro differences include acid sensitivity, optimal growth temperature and tissue tropism, which reflect a preferential in vivo replication in the respiratory or gastrointestinal tract and are thus key determinants of EV virulence. To investigate the underlying cause of these differences, we generated chimeras at the capsid-level between EV-D68 (a respiratory EV) and EV-D94 (an enteric EV). Although some chimeras were nonfunctional, EV-D94 with both the capsid and 2A protease or the capsid only of EV-D68 were both viable. Using this latter construct, we performed several functional assays, which indicated that capsid proteins determine acid sensitivity and tropism in cell lines and in respiratory, intestinal and neural tissues. Additionally, capsid genes were shown to also participate in determining the optimal growth temperature, since EV-D94 temperature adaptation relied on single mutations in VP1, while constructs with EV-D68 capsid could not adapt to higher temperatures. Finally, we demonstrate that EV-D68 maintains residual binding-capacity after acid-treatment despite a loss of infectivity. In contrast, non-structural rather than capsid proteins modulate the innate immune response in tissues. These unique biophysical insights expose another layer in the phenotypic diversity of one of world's most prevalent pathogens and could aid target selection for vaccine or antiviral development.


Acids/chemistry , Capsid Proteins/metabolism , Enterovirus Infections/virology , Enterovirus/physiology , Intestines/virology , Neurons/virology , Respiratory System/virology , Capsid Proteins/genetics , Enterovirus/classification , Enterovirus Infections/genetics , Enterovirus Infections/metabolism , Humans , Temperature , Viral Tropism
10.
Elife ; 72018 04 13.
Article En | MEDLINE | ID: mdl-29651984

Host factors restricting the transmission of respiratory viruses are poorly characterized. We analyzed the contribution of type I and type III interferon (IFN) using a mouse model in which the virus is selectively administered to the upper airways, mimicking a natural respiratory virus infection. Mice lacking functional IFN-λ receptors (Ifnlr1-/-) no longer restricted virus dissemination from the upper airways to the lungs. Ifnlr1-/- mice shed significantly more infectious virus particles via the nostrils and transmitted the virus much more efficiently to naïve contacts compared with wild-type mice or mice lacking functional type I IFN receptors. Prophylactic treatment with IFN-α or IFN-λ inhibited initial virus replication in all parts of the respiratory tract, but only IFN-λ conferred long-lasting antiviral protection in the upper airways and blocked virus transmission. Thus, IFN-λ has a decisive and non-redundant function in the upper airways that greatly limits transmission of respiratory viruses to naïve contacts.


Antiviral Agents/pharmacology , Interferon-gamma/pharmacology , Lung/drug effects , Orthomyxoviridae Infections/prevention & control , Orthomyxoviridae/drug effects , Receptors, Interferon/physiology , Respiratory System/drug effects , Animals , Cytokines/metabolism , Lung/immunology , Lung/virology , Mice , Mice, Inbred C57BL , Mice, Knockout , Orthomyxoviridae Infections/transmission , Orthomyxoviridae Infections/virology , Respiratory System/immunology , Respiratory System/virology , Virus Replication
11.
Proc Natl Acad Sci U S A ; 114(31): 8342-8347, 2017 08 01.
Article En | MEDLINE | ID: mdl-28716935

Viral respiratory infections are usually mild and self-limiting; still they exceptionally result in life-threatening infections in previously healthy children. To investigate a potential genetic cause, we recruited 120 previously healthy children requiring support in intensive care because of a severe illness caused by a respiratory virus. Using exome and transcriptome sequencing, we identified and characterized three rare loss-of-function variants in IFIH1, which encodes an RIG-I-like receptor involved in the sensing of viral RNA. Functional testing of the variants IFIH1 alleles demonstrated that the resulting proteins are unable to induce IFN-ß, are intrinsically less stable than wild-type IFIH1, and lack ATPase activity. In vitro assays showed that IFIH1 effectively restricts replication of human respiratory syncytial virus and rhinoviruses. We conclude that IFIH1 deficiency causes a primary immunodeficiency manifested in extreme susceptibility to common respiratory RNA viruses.


Genetic Predisposition to Disease/genetics , Immunologic Deficiency Syndromes/genetics , Interferon-Induced Helicase, IFIH1/genetics , Interferon-beta/biosynthesis , Respiratory Syncytial Viruses/immunology , Respiratory Tract Infections/virology , Rhinovirus/immunology , Adenosine Triphosphatases/genetics , Child, Preschool , Critical Care , Female , Genetic Variation/genetics , Humans , Immunologic Deficiency Syndromes/immunology , Infant , Infant, Newborn , Interferon-beta/immunology , Male , Prospective Studies , Protein Isoforms/genetics , Respiratory Tract Infections/immunology , Virus Replication/immunology
12.
J Gen Virol ; 98(6): 1282-1293, 2017 Jun.
Article En | MEDLINE | ID: mdl-28631605

Retinoic acid inducible gene (RIG-I)-mediated innate immunity plays a pivotal role in defence against virus infections. Previously we have shown that Sendai virus (SeV) defective interfering (DI) RNA functions as an exclusive and potent RIG-I ligand in DI-RNA-rich SeV-Cantell infected cells. To further understand how RIG-I is activated during SeV infection, we used a different interferon (IFN)-inducing SeV strain, recombinant SeVΔC, which, in contrast to SeV-Cantell is believed to stimulate IFN production due to the lack of the SeV IFN antagonist protein C. Surprisingly, we found that in SevΔC-infected cells, DI RNAs also functioned as an exclusive RIG-I ligand. Infections with wild-type SeV failed to generate any RIG-I-associated immunostimulatory RNA and this correlated with the lack of DI genomes in infected cells, as well as with the absence of cellular innate immune responses. Supplementation of the C protein in the context of SeVΔC infection led to a reduction in the number of DI RNAs, further supporting the potential role of the C protein as a negative regulator of DI generation and/or accumulation. Our findings indicate that limiting DI genome production is an important function of viral IFN antagonist proteins.


DEAD Box Protein 58/metabolism , Gene Deletion , Gene Expression Regulation, Viral , RNA, Small Interfering/metabolism , Sendai virus/immunology , Viral Proteins/genetics , Adjuvants, Immunologic/metabolism , Genetic Complementation Test , HeLa Cells , Humans , RNA, Viral/metabolism , Receptors, Immunologic , Sendai virus/genetics
13.
RNA Biol ; 14(10): 1431-1443, 2017 10 03.
Article En | MEDLINE | ID: mdl-28277929

RNA decay and RNA maturation are important steps in the regulation of bacterial gene expression. RNase J, which is present in about half of bacterial species, has been shown to possess both endo- and 5' to 3' exo-ribonuclease activities. The exonucleolytic activity is clearly involved in the degradation of mRNA and in the maturation of at least the 5' end of 16S rRNA in the 2 Firmicutes Staphylococcus aureus and Bacillus subtilis. The endoribonuclease activity of RNase J from several species has been shown to be weak in vitro and 3-D structural data of different RNase J orthologs have not provided a clear explanation for the molecular basis of this activity. Here, we show that S. aureus RNase J1 is a manganese dependent homodimeric enzyme with strong 5' to 3' exo-ribonuclease as well as endo-ribonuclease activity. In addition, we demonstrated that SauJ1 can efficiently degrade 5' triphosphorylated RNA. Our results highlight RNase J1 as an important player in RNA turnover in S. aureus.


Manganese/metabolism , Ribonucleases/metabolism , Staphylococcus aureus/enzymology , Bacterial Proteins/chemistry , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Cloning, Molecular , Gene Expression Regulation, Bacterial , Phosphorylation , Protein Structure, Quaternary , Ribonucleases/chemistry , Ribonucleases/genetics , Staphylococcus aureus/genetics , Staphylococcus aureus/growth & development
14.
Article En | MEDLINE | ID: mdl-28137809

Synthetic peptides derived from the heptad repeat (HR) of fusion (F) proteins can be used as dominant negative inhibitors to inhibit the fusion mechanism of class I viral F proteins. Here, we have performed a stapled-peptide scan across the HR2 domain of the respiratory syncytial virus (RSV) F protein with the aim to identify a minimal domain capable of disrupting the formation of the postfusion six-helix bundle required for viral cell entry. Constraining the peptides with a single staple was not sufficient to inhibit RSV infection. However, the insertion of double staples led to the identification of novel short stapled peptides that display nanomolar potency in HEp-2 cells and are exceptionally robust to proteolytic degradation. By replacing each amino acid of the peptides by an alanine, we found that the substitution of residues 506 to 509, located in a patch of polar contacts between HR2 and HR1, severely affected inhibition. Finally, we show that intranasal delivery of the most potent peptide to BALB/c mice significantly decreased RSV infection in upper and lower respiratory tracts. The discovery of this minimal HR2 sequence as a means for inhibition of RSV infection provides the basis for further medicinal chemistry efforts toward developing RSV fusion antivirals.


Antiviral Agents/pharmacology , Peptides/pharmacology , Respiratory Syncytial Virus Infections/drug therapy , Respiratory Syncytial Virus, Human/drug effects , Viral Fusion Proteins/chemistry , Virus Internalization/drug effects , Administration, Intranasal , Amino Acid Sequence , Amino Acid Substitution , Animals , Antiviral Agents/chemical synthesis , Binding Sites , Female , HeLa Cells , Humans , Mice , Mice, Inbred BALB C , Peptides/chemical synthesis , Protein Binding , Protein Conformation, alpha-Helical , Protein Interaction Domains and Motifs , Protein Stability , Proteolysis , Respiratory Syncytial Virus Infections/virology , Respiratory Syncytial Virus, Human/chemistry , Respiratory Syncytial Virus, Human/growth & development , Sequence Alignment , Sequence Homology, Amino Acid , Virus Replication/drug effects
15.
Cell Host Microbe ; 20(6): 698-700, 2016 Dec 14.
Article En | MEDLINE | ID: mdl-27978430

Viruses have evolved a remarkable array of strategies to escape the host's innate immune responses. In this issue of Cell Host & Microbe, Zhao et al. (2016b) reveal a viral strategy to inactivate RIG-I signaling that relies on deamidation of RIG-I.


DEAD Box Protein 58/immunology , Herpesvirus 1, Human/immunology , Immunity, Innate , Signal Transduction/immunology , Adenosine Triphosphate/metabolism , Herpes Simplex/immunology , Interferon Regulatory Factor-3/metabolism , Interferon Regulatory Factor-7/metabolism , NF-kappa B/metabolism , Polyubiquitin/pharmacology , Receptors, Immunologic , TNF Receptor-Associated Factor 6/metabolism , Viral Structural Proteins/immunology
16.
BMC Biol ; 14: 69, 2016 08 18.
Article En | MEDLINE | ID: mdl-27538435

BACKGROUND: After viral infection and the stimulation of some pattern-recognition receptors, TANK-binding kinase I (TBK1) is activated by K63-linked polyubiquitination followed by trans-autophosphorylation. While the activated TBK1 induces type I interferon production by phosphorylating the transcription factor IRF3, the precise molecular mechanisms underlying TBK1 activation remain unclear. RESULTS: We report here the localization of the ubiquitinated and phosphorylated active form of TBK1 to the Golgi apparatus after the stimulation of RIG-I-like receptors (RLRs) or Toll-like receptor-3 (TLR3), due to TBK1 K63-linked ubiquitination on lysine residues 30 and 401. The ubiquitin-binding protein optineurin (OPTN) recruits ubiquitinated TBK1 to the Golgi apparatus, leading to the formation of complexes in which TBK1 is activated by trans-autophosphorylation. Indeed, OPTN deficiency in various cell lines and primary cells impairs TBK1 targeting to the Golgi apparatus and its activation following RLR or TLR3 stimulation. Interestingly, the Bluetongue virus NS3 protein binds OPTN at the Golgi apparatus, neutralizing its activity and thereby decreasing TBK1 activation and downstream signaling. CONCLUSIONS: Our results highlight an unexpected role of the Golgi apparatus in innate immunity as a key subcellular gateway for TBK1 activation after RNA virus infection.


Golgi Apparatus/virology , Immunity, Innate , Protein Serine-Threonine Kinases/metabolism , RNA Virus Infections/immunology , Cell Cycle Proteins , DEAD Box Protein 58/genetics , DEAD Box Protein 58/metabolism , Golgi Apparatus/metabolism , HEK293 Cells , HeLa Cells , Humans , Interferon Regulatory Factor-3/genetics , Interferon Regulatory Factor-3/metabolism , Membrane Transport Proteins , Phosphorylation , Protein Serine-Threonine Kinases/genetics , RNA Viruses , Receptors, Immunologic , Signal Transduction , Toll-Like Receptor 3/genetics , Toll-Like Receptor 3/metabolism , Transcription Factor TFIIIA/genetics , Transcription Factor TFIIIA/metabolism , Transfection , Ubiquitination , Viral Nonstructural Proteins/genetics , Viral Nonstructural Proteins/metabolism
17.
J Virol ; 90(1): 586-90, 2016 01 01.
Article En | MEDLINE | ID: mdl-26446607

Influenza virus RNA (vRNA) promoter panhandle structures are believed to be sensed by retinoic acid-inducible gene I (RIG-I). The occurrence of mismatches in this double-stranded RNA structure raises questions about their effect on innate sensing. Our results suggest that mismatches in vRNA promoters decrease binding to RIG-I in vivo, affecting RNA/RIG-I complex formation and preventing RIG-I activation. These results can be inferred to apply to other viruses and suggest that mismatches may represent a general viral strategy to escape RIG-I sensing.


Base Pair Mismatch , DEAD-box RNA Helicases/metabolism , Influenza A virus/immunology , RNA, Double-Stranded/genetics , RNA, Viral/genetics , RNA-Binding Proteins/metabolism , Cell Line , DEAD Box Protein 58 , Epithelial Cells/virology , Host-Pathogen Interactions , Humans , Immune Evasion , Immunity, Innate , Nucleic Acid Conformation , Protein Binding , RNA, Double-Stranded/chemistry , RNA, Viral/chemistry , Receptors, Immunologic
18.
J Immunol ; 195(3): 1025-33, 2015 Aug 01.
Article En | MEDLINE | ID: mdl-26101320

Because of their unique capacity to cross-present Ags to CD8(+) T cells, mouse lymphoid tissue-resident CD8(+) dendritic cells (DCs) and their migratory counterparts are critical for priming antiviral T cell responses. High expression of the dsRNA sensor TLR3 is a distinctive feature of these cross-presenting DC subsets. TLR3 engagement in CD8(+) DCs promotes cross-presentation and the acquisition of effector functions required for driving antiviral T cell responses. In this study, we performed a comprehensive analysis of the TLR3-induced antiviral program and cell-autonomous immunity in CD8(+) DC lines and primary CD8(+) DCs. We found that TLR3-ligand polyinosinic-polycytidylic acid and human rhinovirus infection induced a potent antiviral protection against Sendai and vesicular stomatitis virus in a TLR3 and type I IFN receptor-dependent manner. Polyinosinic-polycytidylic acid-induced antiviral genes were identified by mass spectrometry-based proteomics and transcriptomics in the CD8(+) DC line. Nanostring nCounter experiments confirmed that these antiviral genes were induced by TLR3 engagement in primary CD8(+) DCs, and indicated that many are secondary TLR3-response genes requiring autocrine IFN-ß stimulation. TLR3-activation thus establishes a type I IFN-dependent antiviral program in a DC subtype playing crucial roles in priming adaptive antiviral immune responses. This mechanism is likely to shield the priming of antiviral responses against inhibition or abrogation by the viral infection. It could be particularly relevant for viruses detected mainly by TLR3, which may not trigger type I IFN production by DCs that lack TLR3, such as plasmacytoid DCs or CD8(-) DCs.


CD8-Positive T-Lymphocytes/immunology , Dendritic Cells/immunology , Interferon-beta/immunology , Toll-Like Receptor 3/immunology , Animals , Cross-Priming/immunology , Humans , Interferon-beta/genetics , Lymphocyte Activation/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout , Picornaviridae Infections/immunology , Picornaviridae Infections/virology , Poly I-C/immunology , Receptor, Interferon alpha-beta/immunology , Rhinovirus/immunology , Sendai virus/immunology , Vesicular stomatitis Indiana virus/immunology
19.
Mol Cell ; 58(1): 3-4, 2015 Apr 02.
Article En | MEDLINE | ID: mdl-25839430

In this issue, He et al. (2015) show how herpes virus usurps a cellular metabolic enzyme to induce RIG-I deamidation and RNA-independent activation, likely to better prevent further innate immune responses.


Carbon-Nitrogen Ligases with Glutamine as Amide-N-Donor/immunology , DEAD-box RNA Helicases/immunology , Gammaherpesvirinae/immunology , Immune Evasion/genetics , RNA, Viral/immunology , Viral Proteins/immunology , Animals , DEAD Box Protein 58 , Humans , Receptors, Immunologic
20.
mBio ; 6(2): e02349, 2015 Mar 03.
Article En | MEDLINE | ID: mdl-25736886

UNLABELLED: Many RNA viruses are detected by retinoic acid-inducible gene i (RIG-I), a cytoplasmic sensor that triggers an antiviral response upon binding non-self-RNA that contains a stretch of double-stranded RNA (dsRNA) bearing a base-paired 5' ppp nucleotide. To gain insight into how RIG-I discriminates between self-RNA and non-self-RNA, we used duplexes whose complementary bottom strand contained both ribo- and deoxynucleotides. These duplexes were examined for their binding to RIG-I and their relative abilities to stimulate ATPase activity, to induce RIG-I dimerization on the duplex, and to induce beta interferon (IFN-ß) expression. We show that the chemical nature of the bottom strand is not critical for RIG-I binding. However, two key ribonucleotides, at positions 2 and 5 on the bottom strand, are minimally required for the RIG-I ATPase activity, which is necessary but not sufficient for IFN-ß stimulation. We find that duplexes with shorter stretches of dsRNA, as model self-RNAs, bind less stably to RIG-I but nevertheless have an enhanced ability to stimulate the ATPase. Moreover, ATPase activity promotes RIG-I recycling on RIG-I/dsRNA complexes. Since pseudo-self-RNAs bind to RIG-I less stably, they are preferentially recycled by ATP hydrolysis that weakens the helicase domain binding of dsRNA. Our results suggest that one function of the ATPase is to restrict RIG-I signaling to its interaction with non-self-RNA. A model of how this discrimination occurs as a function of dsRNA length is presented. IMPORTANCE: The innate immune response to pathogens is based on the discrimination between self-RNA and non-self-RNA. The main determinants of this detection for RNA viruses are specific pathogen-associated molecular patterns (PAMPs) of RNA, which are detected by dedicated cytoplasmic pattern recognition receptors (PRRs). RIG-I is a PRR that specifically detects short viral dsRNAs amid a sea of cellular RNAs. Here we study the determinants of this discrimination and how RIG-I ATPase activity, the only enzymatic activity of this sensor, contributes to its activation in a manner restricted to its interaction with non-self-RNAs. We also show how the innate immune response evolves during infection via IFN expression, from a state in which discrimination of self-RNA from non-self-RNA is most important to one in which this discrimination is sacrificed for the effectiveness of the antiviral response.


Adenosine Triphosphatases/metabolism , DEAD-box RNA Helicases/metabolism , RNA/metabolism , DEAD Box Protein 58 , Gene Expression , Interferon-beta/biosynthesis , Protein Binding , Protein Multimerization , RNA, Double-Stranded/metabolism , Receptors, Immunologic , Substrate Specificity
...