Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 52
Filter
1.
PLoS Biol ; 22(2): e3002544, 2024 Feb.
Article in English | MEDLINE | ID: mdl-38422166

ABSTRACT

Ebolavirus (EBOV) belongs to a family of highly pathogenic viruses that cause severe hemorrhagic fever in humans. EBOV replication requires the activity of the viral polymerase complex, which includes the cofactor and Interferon antagonist VP35. We previously showed that the covalent ubiquitination of VP35 promotes virus replication by regulating interactions with the polymerase complex. In addition, VP35 can also interact non-covalently with ubiquitin (Ub); however, the function of this interaction is unknown. Here, we report that VP35 interacts with free (unanchored) K63-linked polyUb chains. Ectopic expression of Isopeptidase T (USP5), which is known to degrade unanchored polyUb chains, reduced VP35 association with Ub and correlated with diminished polymerase activity in a minigenome assay. Using computational methods, we modeled the VP35-Ub non-covalent interacting complex, identified the VP35-Ub interacting surface, and tested mutations to validate the interface. Docking simulations identified chemical compounds that can block VP35-Ub interactions leading to reduced viral polymerase activity. Treatment with the compounds reduced replication of infectious EBOV in cells and in vivo in a mouse model. In conclusion, we identified a novel role of unanchored polyUb in regulating Ebola virus polymerase function and discovered compounds that have promising anti-Ebola virus activity.


Subject(s)
Ebolavirus , Hemorrhagic Fever, Ebola , Nucleocapsid Proteins , Ubiquitin , Virus Replication , Animals , Humans , Mice , Ebolavirus/genetics , Ubiquitin/metabolism , Viral Regulatory and Accessory Proteins , Virus Replication/genetics
2.
Nature ; 585(7825): 414-419, 2020 09.
Article in English | MEDLINE | ID: mdl-32641828

ABSTRACT

Zika virus (ZIKV) belongs to the family Flaviviridae, and is related to other viruses that cause human diseases. Unlike other flaviviruses, ZIKV infection can cause congenital neurological disorders and replicates efficiently in reproductive tissues1-3. Here we show that the envelope protein (E) of ZIKV is polyubiquitinated by the E3 ubiquitin ligase TRIM7 through Lys63 (K63)-linked polyubiquitination. Accordingly, ZIKV replicates less efficiently in the brain and reproductive tissues of Trim7-/- mice. Ubiquitinated E is present on infectious virions of ZIKV when they are released from specific cell types, and enhances virus attachment and entry into cells. Specifically, K63-linked polyubiquitin chains directly interact with the TIM1 (also known as HAVCR1) receptor of host cells, which enhances virus entry in cells as well as in brain tissue in vivo. Recombinant ZIKV mutants that lack ubiquitination are attenuated in human cells and in wild-type mice, but not in live mosquitoes. Monoclonal antibodies against K63-linked polyubiquitin specifically neutralize ZIKV and reduce viraemia in mice. Our results demonstrate that the ubiquitination of ZIKV E is an important determinant of virus entry, tropism and pathogenesis.


Subject(s)
Ubiquitination , Viral Envelope Proteins/chemistry , Viral Envelope Proteins/metabolism , Virus Internalization , Zika Virus/metabolism , Zika Virus/pathogenicity , Animals , Antibodies, Monoclonal/immunology , Antibodies, Neutralizing/immunology , Brain/metabolism , Cell Line , Culicidae/cytology , Culicidae/virology , Endosomes/metabolism , Female , Hepatitis A Virus Cellular Receptor 1/metabolism , Humans , Male , Membrane Fusion , Mice , Organ Specificity , Polyubiquitin/immunology , Polyubiquitin/metabolism , Tripartite Motif Proteins/metabolism , Ubiquitin-Protein Ligases/metabolism , Viral Tropism , Viremia/immunology , Viremia/prevention & control , Viremia/virology , Virus Replication , Zika Virus/chemistry , Zika Virus/genetics , Zika Virus Infection/prevention & control , Zika Virus Infection/virology
3.
Proc Natl Acad Sci U S A ; 120(23): e2220005120, 2023 06 06.
Article in English | MEDLINE | ID: mdl-37252973

ABSTRACT

Dengue virus (DENV) is the most important human virus transmitted by mosquitos. Dengue pathogenesis is characterized by a large induction of proinflammatory cytokines. This cytokine induction varies among the four DENV serotypes (DENV1 to 4) and poses a challenge for live DENV vaccine design. Here, we identify a viral mechanism to limit NF-κB activation and cytokine secretion by the DENV protein NS5. Using proteomics, we found that NS5 binds and degrades the host protein ERC1 to antagonize NF-κB activation, limit proinflammatory cytokine secretion, and reduce cell migration. We found that ERC1 degradation involves unique properties of the methyltransferase domain of NS5 that are not conserved among the four DENV serotypes. By obtaining chimeric DENV2 and DENV4 viruses, we map the residues in NS5 for ERC1 degradation, and generate recombinant DENVs exchanging serotype properties by single amino acid substitutions. This work uncovers a function of the viral protein NS5 to limit cytokine production, critical to dengue pathogenesis. Importantly, the information provided about the serotype-specific mechanism for counteracting the antiviral response can be applied to improve live attenuated vaccines.


Subject(s)
Dengue Virus , Dengue , Viral Nonstructural Proteins , Humans , Cytokines , NF-kappa B/metabolism , Serogroup , Viral Nonstructural Proteins/metabolism
4.
PLoS Pathog ; 18(5): e1010532, 2022 05.
Article in English | MEDLINE | ID: mdl-35533195

ABSTRACT

Ebola virus (EBOV) VP35 is a polyfunctional protein involved in viral genome packaging, viral polymerase function, and host immune antagonism. The mechanisms regulating VP35's engagement in different functions are not well-understood. We previously showed that the host E3 ubiquitin ligase TRIM6 ubiquitinates VP35 at lysine 309 (K309) to facilitate virus replication. However, how K309 ubiquitination regulates the function of VP35 as the viral polymerase co-factor and the precise stage(s) of the EBOV replication cycle that require VP35 ubiquitination are not known. Here, we generated recombinant EBOVs encoding glycine (G) or arginine (R) mutations at VP35/K309 (rEBOV-VP35/K309G/-R) and show that both mutations prohibit VP35/K309 ubiquitination. The K309R mutant retains dsRNA binding and efficient type-I Interferon (IFN-I) antagonism due to the basic residue conservation. The rEBOV-VP35/K309G mutant loses the ability to efficiently antagonize the IFN-I response, while the rEBOV-VP35/K309R mutant's suppression is enhanced. The replication of both mutants was significantly attenuated in both IFN-competent and -deficient cells due to impaired interactions with the viral polymerase. The lack of ubiquitination on VP35/K309 or TRIM6 deficiency disrupts viral transcription with increasing severity along the transcriptional gradient. This disruption of the transcriptional gradient results in unbalanced viral protein production, including reduced synthesis of the viral transcription factor VP30. In addition, lack of ubiquitination on K309 results in enhanced interactions with the viral nucleoprotein and premature nucleocapsid packaging, leading to dysregulation of virus assembly. Overall, we identified a novel role of VP35 ubiquitination in coordinating viral transcription and assembly.


Subject(s)
Ebolavirus , Hemorrhagic Fever, Ebola , Ebolavirus/metabolism , Humans , Lysine/genetics , Lysine/metabolism , Nucleocapsid Proteins/metabolism , Ubiquitination , Viral Regulatory and Accessory Proteins/genetics , Viral Regulatory and Accessory Proteins/metabolism , Viral Transcription
5.
Immunity ; 40(6): 880-95, 2014 Jun 19.
Article in English | MEDLINE | ID: mdl-24882218

ABSTRACT

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


Subject(s)
I-kappa B Kinase/immunology , Interferon Type I/immunology , Polyubiquitin/biosynthesis , Ubiquitin-Protein Ligases/immunology , Animals , Antiviral Agents , Cells, Cultured , Enzyme Activation/immunology , Humans , Janus Kinase 1 , Mice , Phosphorylation/immunology , RNA Interference , RNA, Small Interfering , STAT1 Transcription Factor/immunology , Signal Transduction/immunology , Tripartite Motif Proteins , Ubiquitin-Conjugating Enzymes/immunology , Ubiquitin-Protein Ligases/genetics
6.
Immunity ; 38(2): 384-98, 2013 Feb 21.
Article in English | MEDLINE | ID: mdl-23438823

ABSTRACT

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


Subject(s)
Carrier Proteins/genetics , Immunity, Innate , Leukocytes, Mononuclear/metabolism , RNA, Messenger/genetics , Receptors, Pattern Recognition/genetics , Rhabdoviridae Infections/metabolism , Zinc Fingers/genetics , Alternative Splicing , Antiviral Restriction Factors , Carrier Proteins/antagonists & inhibitors , Carrier Proteins/immunology , Cell Line , Cloning, Molecular , Gene Expression Profiling , Gene Expression Regulation , Humans , Leukocytes, Mononuclear/immunology , Leukocytes, Mononuclear/virology , Protein Isoforms/antagonists & inhibitors , Protein Isoforms/genetics , Protein Isoforms/immunology , RNA, Messenger/immunology , RNA, Small Interfering/genetics , Receptors, Pattern Recognition/immunology , Rhabdoviridae Infections/immunology , Rhabdoviridae Infections/virology , Signal Transduction , Tripartite Motif Proteins , Ubiquitin-Protein Ligases , Vesiculovirus/immunology , Zinc Fingers/immunology
7.
J Virol ; 94(2)2020 01 06.
Article in English | MEDLINE | ID: mdl-31694946

ABSTRACT

Several members of the tripartite motif (TRIM) family of E3 ubiquitin ligases regulate immune pathways, including the antiviral type I interferon (IFN-I) system. Previously, we demonstrated that TRIM6 is involved in IFN-I induction and signaling. In the absence of TRIM6, optimal IFN-I signaling is reduced, allowing increased replication of interferon-sensitive viruses. Despite having evolved numerous mechanisms to restrict the vertebrate host's IFN-I response, West Nile virus (WNV) replication is sensitive to pretreatment with IFN-I. However, the regulators and products of the IFN-I pathway that are important in regulating WNV replication are incompletely defined. Consistent with WNV's sensitivity to IFN-I, we found that in TRIM6 knockout (TRIM6-KO) A549 cells, WNV replication is significantly increased and IFN-I induction and signaling are impaired compared to wild-type (wt) cells. IFN-ß pretreatment was more effective in protecting against subsequent WNV infection in wt cells than TRIM6-KO, indicating that TRIM6 contributes to the establishment of an IFN-induced antiviral response against WNV. Using next-generation sequencing, we identified VAMP8 as a potential factor involved in this TRIM6-mediated antiviral response. VAMP8 knockdown resulted in reduced JAK1 and STAT1 phosphorylation and impaired induction of several interferon-stimulated genes (ISGs) following WNV infection or IFN-ß treatment. Furthermore, VAMP8-mediated STAT1 phosphorylation required the presence of TRIM6. Therefore, the VAMP8 protein is a novel regulator of IFN-I signaling, and its expression and function are dependent on TRIM6 activity. Overall, these results provide evidence that TRIM6 contributes to the antiviral response against WNV and identify VAMP8 as a novel regulator of the IFN-I system.IMPORTANCE WNV is a mosquito-borne flavivirus that poses a threat to human health across large discontinuous areas throughout the world. Infection with WNV results in febrile illness, which can progress to severe neurological disease. Currently, there are no approved treatment options to control WNV infection. Understanding the cellular immune responses that regulate viral replication is important in diversifying the resources available to control WNV. Here, we show that the elimination of TRIM6 in human cells results in an increase in WNV replication and alters the expression and function of other components of the IFN-I pathway through VAMP8. Dissecting the interactions between WNV and host defenses both informs basic molecular virology and promotes the development of host- and virus-targeted antiviral strategies.


Subject(s)
Immunity, Innate , Interferon Type I/immunology , R-SNARE Proteins/immunology , Tripartite Motif Proteins/immunology , Ubiquitin-Protein Ligases/immunology , Virus Replication/immunology , West Nile Fever/immunology , West Nile virus/physiology , A549 Cells , Gene Deletion , HEK293 Cells , Humans , Janus Kinase 1/genetics , Janus Kinase 1/immunology , Phosphorylation/genetics , Phosphorylation/immunology , R-SNARE Proteins/genetics , STAT1 Transcription Factor/genetics , STAT1 Transcription Factor/immunology , Tripartite Motif Proteins/genetics , Ubiquitin-Protein Ligases/genetics , Virus Replication/genetics , West Nile Fever/genetics , West Nile Fever/pathology
8.
J Virol ; 94(23)2020 11 09.
Article in English | MEDLINE | ID: mdl-32938761

ABSTRACT

SARS-CoV-2, a novel coronavirus (CoV) that causes COVID-19, has recently emerged causing an ongoing outbreak of viral pneumonia around the world. While distinct from SARS-CoV, both group 2B CoVs share similar genome organization, origins to bat CoVs, and an arsenal of immune antagonists. In this report, we evaluate type I interferon (IFN-I) sensitivity of SARS-CoV-2 relative to the original SARS-CoV. Our results indicate that while SARS-CoV-2 maintains similar viral replication to SARS-CoV, the novel CoV is much more sensitive to IFN-I. In Vero E6 and in Calu3 cells, SARS-CoV-2 is substantially attenuated in the context of IFN-I pretreatment, whereas SARS-CoV is not. In line with these findings, SARS-CoV-2 fails to counteract phosphorylation of STAT1 and expression of ISG proteins, while SARS-CoV is able to suppress both. Comparing SARS-CoV-2 and influenza A virus in human airway epithelial cultures, we observe the absence of IFN-I stimulation by SARS-CoV-2 alone but detect the failure to counteract STAT1 phosphorylation upon IFN-I pretreatment, resulting in near ablation of SARS-CoV-2 infection. Next, we evaluated IFN-I treatment postinfection and found that SARS-CoV-2 was sensitive even after establishing infection. Finally, we examined homology between SARS-CoV and SARS-CoV-2 in viral proteins shown to be interferon antagonists. The absence of an equivalent open reading frame 3b (ORF3b) and genetic differences versus ORF6 suggest that the two key IFN-I antagonists may not maintain equivalent function in SARS-CoV-2. Together, the results identify key differences in susceptibility to IFN-I responses between SARS-CoV and SARS-CoV-2 that may help inform disease progression, treatment options, and animal model development.IMPORTANCE With the ongoing outbreak of COVID-19, differences between SARS-CoV-2 and the original SARS-CoV could be leveraged to inform disease progression and eventual treatment options. In addition, these findings could have key implications for animal model development as well as further research into how SARS-CoV-2 modulates the type I IFN response early during infection.


Subject(s)
Antiviral Agents/pharmacology , Betacoronavirus/drug effects , Interferon Type I/pharmacology , Interferon-alpha/pharmacology , Severe acute respiratory syndrome-related coronavirus/drug effects , Animals , Antiviral Agents/antagonists & inhibitors , Antiviral Agents/metabolism , Betacoronavirus/immunology , Betacoronavirus/physiology , Cell Line , Cell Line, Tumor , Chlorocebus aethiops , Humans , Interferon Type I/antagonists & inhibitors , Interferon Type I/immunology , Interferon Type I/metabolism , Interferon-alpha/antagonists & inhibitors , Interferon-alpha/immunology , Interferon-alpha/metabolism , Phosphorylation , Recombinant Proteins/pharmacology , Severe acute respiratory syndrome-related coronavirus/immunology , Severe acute respiratory syndrome-related coronavirus/physiology , SARS-CoV-2 , STAT1 Transcription Factor/metabolism , Signal Transduction , Vero Cells , Viral Proteins/chemistry , Viral Proteins/genetics , Viral Proteins/metabolism , Virus Replication/drug effects
9.
J Virol ; 93(22)2019 11 15.
Article in English | MEDLINE | ID: mdl-31462558

ABSTRACT

Enteric viruses exploit bacterial components, including lipopolysaccharides (LPS) and peptidoglycan (PG), to facilitate infection in humans. Because of their origin in the bat enteric system, we wondered if severe acute respiratory syndrome coronavirus (SARS-CoV) or Middle East respiratory syndrome CoV (MERS-CoV) also use bacterial components to modulate infectivity. To test this question, we incubated CoVs with LPS and PG and evaluated infectivity, finding no change following LPS treatment. However, PG from Bacillus subtilis reduced infection >10,000-fold, while PG from other bacterial species failed to recapitulate this. Treatment with an alcohol solvent transferred inhibitory activity to the wash, and mass spectrometry revealed surfactin, a cyclic lipopeptide antibiotic, as the inhibitory compound. This antibiotic had robust dose- and temperature-dependent inhibition of CoV infectivity. Mechanistic studies indicated that surfactin disrupts CoV virion integrity, and surfactin treatment of the virus inoculum ablated infection in vivo Finally, similar cyclic lipopeptides had no effect on CoV infectivity, and the inhibitory effect of surfactin extended broadly to enveloped viruses, including influenza, Ebola, Zika, Nipah, chikungunya, Una, Mayaro, Dugbe, and Crimean-Congo hemorrhagic fever viruses. Overall, our results indicate that peptidoglycan-associated surfactin has broad viricidal activity and suggest that bacteria by-products may negatively modulate virus infection.IMPORTANCE In this article, we consider a role for bacteria in shaping coronavirus infection. Taking cues from studies of enteric viruses, we initially investigated how bacterial surface components might improve CoV infection. Instead, we found that peptidoglycan-associated surfactin is a potent viricidal compound that disrupts virion integrity with broad activity against enveloped viruses. Our results indicate that interactions with commensal bacterial may improve or disrupt viral infections, highlighting the importance of understanding these microbial interactions and their implications for viral pathogenesis and treatment.


Subject(s)
Lipopeptides/pharmacology , Peptides, Cyclic/pharmacology , Peptidoglycan/metabolism , RNA Viruses/drug effects , Animals , Cell Line , Chlorocebus aethiops , Coronavirus Infections/virology , Flaviviridae/drug effects , Lipopeptides/immunology , Lipopeptides/metabolism , Middle East Respiratory Syndrome Coronavirus/metabolism , Peptides, Cyclic/immunology , Peptides, Cyclic/metabolism , Peptidoglycan/genetics , Severe acute respiratory syndrome-related coronavirus/metabolism , Severe Acute Respiratory Syndrome/virology , Vero Cells , Virus Diseases/metabolism
10.
J Gen Virol ; 100(12): 1641-1662, 2019 12.
Article in English | MEDLINE | ID: mdl-31661051

ABSTRACT

The innate immune system responds rapidly to protect against viral infections, but an overactive response can cause harmful damage. To avoid this, the response is tightly regulated by post-translational modifications (PTMs). The ubiquitin system represents a powerful PTM machinery that allows for the reversible linkage of ubiquitin to activate and deactivate a target's function. A precise enzymatic cascade of ubiquitin-activating, conjugating and ligating enzymes facilitates ubiquitination. Viruses have evolved to take advantage of the ubiquitin pathway either by targeting factors to dampen the antiviral response or by hijacking the system to enhance their replication. The tripartite motif (TRIM) family of E3 ubiquitin ligases has garnered attention as a major contributor to innate immunity. Many TRIM family members limit viruses either indirectly as components in innate immune signalling, or directly by targeting viral proteins for degradation. In spite of this, TRIMs and other ubiquitin ligases can be appropriated by viruses and repurposed as valuable tools in viral replication. This duality of function suggests a new frontier of research for TRIMs and raises new challenges for discerning the subtleties of these pro-viral mechanisms. Here, we review current findings regarding the involvement of TRIMs in host-virus interactions. We examine ongoing developments in the field, including novel roles for unanchored ubiquitin in innate immunity, the direct involvement of ubiquitin ligases in promoting viral replication, recent controversies on the role of ubiquitin and TRIM25 in activation of the pattern recognition receptor RIG-I, and we discuss the implications these studies have on future research directions.


Subject(s)
Host-Pathogen Interactions , Tripartite Motif Proteins/metabolism , Ubiquitin/metabolism , Virus Diseases/metabolism , Virus Diseases/virology , Virus Physiological Phenomena , Disease Resistance/genetics , Disease Resistance/immunology , Host-Pathogen Interactions/genetics , Host-Pathogen Interactions/immunology , Humans , Immunity, Innate , Immunomodulation , Models, Biological , Proteasome Endopeptidase Complex , Protein Binding , Protein Processing, Post-Translational , Signal Transduction , Ubiquitination , Viral Proteins/metabolism , Virus Diseases/genetics , Virus Replication
11.
J Virol ; 91(18)2017 09 15.
Article in English | MEDLINE | ID: mdl-28679761

ABSTRACT

Ebola virus (EBOV), a member of the Filoviridae family, is a highly pathogenic virus that causes severe hemorrhagic fever in humans and is responsible for epidemics throughout sub-Saharan, central, and West Africa. The EBOV genome encodes VP35, an important viral protein involved in virus replication by acting as an essential cofactor of the viral polymerase as well as a potent antagonist of the host antiviral type I interferon (IFN-I) system. By using mass spectrometry analysis and coimmunoprecipitation assays, we show here that VP35 is ubiquitinated on lysine 309 (K309), a residue located on its IFN antagonist domain. We also found that VP35 interacts with TRIM6, a member of the E3-ubiquitin ligase tripartite motif (TRIM) family. We recently reported that TRIM6 promotes the synthesis of unanchored K48-linked polyubiquitin chains, which are not covalently attached to any protein, to induce efficient antiviral IFN-I-mediated responses. Consistent with this notion, VP35 also associated noncovalently with polyubiquitin chains and inhibited TRIM6-mediated IFN-I induction. Intriguingly, we also found that TRIM6 enhances EBOV polymerase activity in a minigenome assay and TRIM6 knockout cells have reduced replication of infectious EBOV, suggesting that VP35 hijacks TRIM6 to promote EBOV replication through ubiquitination. Our work provides evidence that TRIM6 is an important host cellular factor that promotes EBOV replication, and future studies will focus on whether TRIM6 could be targeted for therapeutic intervention against EBOV infection.IMPORTANCE EBOV belongs to a family of highly pathogenic viruses that cause severe hemorrhagic fever in humans and other mammals with high mortality rates (40 to 90%). Because of its high pathogenicity and lack of licensed antivirals and vaccines, EBOV is listed as a tier 1 select-agent risk group 4 pathogen. An important mechanism for the severity of EBOV infection is its suppression of innate immune responses. The EBOV VP35 protein contributes to pathogenesis, because it serves as an essential cofactor of the viral polymerase as well as a potent antagonist of innate immunity. However, how VP35 function is regulated by host cellular factors is poorly understood. Here, we report that the host E3-ubiquitin ligase TRIM6 promotes VP35 ubiquitination and is important for efficient virus replication. Therefore, our study identifies a new host factor, TRIM6, as a potential target in the development of antiviral drugs against EBOV.


Subject(s)
Ebolavirus/physiology , Host-Pathogen Interactions , Tripartite Motif Proteins/metabolism , Ubiquitin-Protein Ligases/metabolism , Ubiquitination , Viral Regulatory and Accessory Proteins/metabolism , Virus Replication , Animals , Cell Line , Humans , Immunoprecipitation , Mass Spectrometry
12.
PLoS Pathog ; 12(9): e1005880, 2016 09.
Article in English | MEDLINE | ID: mdl-27622505

ABSTRACT

For efficient replication, viruses have developed mechanisms to evade innate immune responses, including the antiviral type-I interferon (IFN-I) system. Nipah virus (NiV), a highly pathogenic member of the Paramyxoviridae family (genus Henipavirus), is known to encode for four P gene-derived viral proteins (P/C/W/V) with IFN-I antagonist functions. Here we report that NiV matrix protein (NiV-M), which is important for virus assembly and budding, can also inhibit IFN-I responses. IFN-I production requires activation of multiple signaling components including the IκB kinase epsilon (IKKε). We previously showed that the E3-ubiquitin ligase TRIM6 catalyzes the synthesis of unanchored K48-linked polyubiquitin chains, which are not covalently attached to any protein, and activate IKKε for induction of IFN-I mediated antiviral responses. Using co-immunoprecipitation assays and confocal microscopy we show here that the NiV-M protein interacts with TRIM6 and promotes TRIM6 degradation. Consequently, NiV-M expression results in reduced levels of unanchored K48-linked polyubiquitin chains associated with IKKε leading to impaired IKKε oligomerization, IKKε autophosphorylation and reduced IFN-mediated responses. This IFN antagonist function of NiV-M requires a conserved lysine residue (K258) in the bipartite nuclear localization signal that is found in divergent henipaviruses. Consistent with this, the matrix proteins of Ghana, Hendra and Cedar viruses were also able to inhibit IFNß induction. Live NiV infection, but not a recombinant NiV lacking the M protein, reduced the levels of endogenous TRIM6 protein expression. To our knowledge, matrix proteins of paramyxoviruses have never been reported to be involved in innate immune antagonism. We report here a novel mechanism of viral innate immune evasion by targeting TRIM6, IKKε and unanchored polyubiquitin chains. These findings expand the universe of viral IFN antagonism strategies and provide a new potential target for development of therapeutic interventions against NiV infections.


Subject(s)
Henipavirus Infections/immunology , I-kappa B Kinase/immunology , Immune Evasion , Interferon Type I/immunology , Nipah Virus/immunology , Tripartite Motif Proteins/immunology , Ubiquitin-Protein Ligases/immunology , Viral Proteins/immunology , A549 Cells , Animals , Chlorocebus aethiops , HeLa Cells , Henipavirus Infections/genetics , Humans , I-kappa B Kinase/genetics , Immunity, Innate , Interferon Type I/genetics , Nipah Virus/genetics , Polyubiquitin/genetics , Polyubiquitin/immunology , Protein Multimerization/genetics , Protein Multimerization/immunology , Tripartite Motif Proteins/genetics , Ubiquitin-Protein Ligases/genetics , Ubiquitination/genetics , Ubiquitination/immunology , Vero Cells , Viral Proteins/genetics
13.
J Virol ; 88(20): 12146-51, 2014 Oct.
Article in English | MEDLINE | ID: mdl-25078692

ABSTRACT

Although an effective interferon antagonist in human and avian cells, the novel H7N9 influenza virus NS1 protein is defective at inhibiting CPSF30. An I106M substitution in H7N9 NS1 can restore CPSF30 binding together with the ability to block host gene expression. Furthermore, a recombinant virus expressing H7N9 NS1-I106M replicates to higher titers in vivo, and is subtly more virulent, than the parental virus. Natural polymorphisms in H7N9 NS1 that enhance CPSF30 binding may be cause for concern.


Subject(s)
Amino Acids/genetics , Influenza A Virus, H7N9 Subtype/pathogenicity , Viral Nonstructural Proteins/metabolism , Viral Proteins/metabolism , Amino Acid Substitution , Animals , Chickens , Gene Expression , Humans , Influenza A Virus, H7N9 Subtype/metabolism , Influenza A Virus, H7N9 Subtype/physiology , Viral Nonstructural Proteins/genetics , Virulence
14.
PLoS Pathog ; 9(3): e1003265, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23555265

ABSTRACT

An estimated 50 million dengue virus (DENV) infections occur annually and more than forty percent of the human population is currently at risk of developing dengue fever (DF) or dengue hemorrhagic fever (DHF). Despite the prevalence and potential severity of DF and DHF, there are no approved vaccines or antiviral therapeutics available. An improved understanding of DENV immune evasion is pivotal for the rational development of anti-DENV therapeutics. Antagonism of type I interferon (IFN-I) signaling is a crucial mechanism of DENV immune evasion. DENV NS5 protein inhibits IFN-I signaling by mediating proteasome-dependent STAT2 degradation. Only proteolytically-processed NS5 can efficiently mediate STAT2 degradation, though both unprocessed and processed NS5 bind STAT2. Here we identify UBR4, a 600-kDa member of the N-recognin family, as an interacting partner of DENV NS5 that preferentially binds to processed NS5. Our results also demonstrate that DENV NS5 bridges STAT2 and UBR4. Furthermore, we show that UBR4 promotes DENV-mediated STAT2 degradation, and most importantly, that UBR4 is necessary for efficient viral replication in IFN-I competent cells. Our data underscore the importance of NS5-mediated STAT2 degradation in DENV replication and identify UBR4 as a host protein that is specifically exploited by DENV to inhibit IFN-I signaling via STAT2 degradation.


Subject(s)
Calmodulin-Binding Proteins/metabolism , Cytoskeletal Proteins/metabolism , Dengue Virus/physiology , Interferon Type I/metabolism , STAT2 Transcription Factor/metabolism , Animals , Cell Line , Chlorocebus aethiops , Dengue Virus/pathogenicity , Humans , Immune Evasion , Protein Binding , Signal Transduction , Ubiquitin-Protein Ligases , Vero Cells , Viral Nonstructural Proteins/metabolism
15.
J Immunol ; 191(4): 1732-43, 2013 Aug 15.
Article in English | MEDLINE | ID: mdl-23842752

ABSTRACT

Tuberculosis, caused by Mycobacterium tuberculosis, remains a leading cause of mortality and morbidity worldwide, causing ≈ 1.4 million deaths per year. Key immune components for host protection during tuberculosis include the cytokines IL-12, IL-1, and TNF-α, as well as IFN-γ and CD4(+) Th1 cells. However, immune factors determining whether individuals control infection or progress to active tuberculosis are incompletely understood. Excess amounts of type I IFN have been linked to exacerbated disease during tuberculosis in mouse models and to active disease in patients, suggesting tight regulation of this family of cytokines is critical to host resistance. In addition, the immunosuppressive cytokine IL-10 is known to inhibit the immune response to M. tuberculosis in murine models through the negative regulation of key proinflammatory cytokines and the subsequent Th1 response. We show in this study, using a combination of transcriptomic analysis, genetics, and pharmacological inhibitors, that the TPL-2-ERK1/2 signaling pathway is important in mediating host resistance to tuberculosis through negative regulation of type I IFN production. The TPL-2-ERK1/2 signaling pathway regulated production by macrophages of several cytokines important in the immune response to M. tuberculosis as well as regulating induction of a large number of additional genes, many in a type I IFN-dependent manner. In the absence of TPL-2 in vivo, excess type I IFN promoted IL-10 production and exacerbated disease. These findings describe an important regulatory mechanism for controlling tuberculosis and reveal mechanisms by which type I IFN may promote susceptibility to this important disease.


Subject(s)
Gene Expression Regulation/immunology , Interferon Type I/biosynthesis , MAP Kinase Kinase Kinases/immunology , MAP Kinase Signaling System , Proto-Oncogene Proteins/immunology , Tuberculosis/immunology , Animals , Bacterial Load , Cytokines/biosynthesis , Cytokines/genetics , Disease Resistance , Down-Regulation/immunology , Female , Gene Expression Profiling , Interferon Type I/genetics , Interleukin-10/immunology , Listeria monocytogenes/immunology , Listeria monocytogenes/isolation & purification , Listeriosis/immunology , MAP Kinase Kinase Kinases/deficiency , MAP Kinase Signaling System/drug effects , Macrophages/immunology , Macrophages/metabolism , Macrophages/microbiology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Monocytes/immunology , Mycobacterium tuberculosis/immunology , Mycobacterium tuberculosis/isolation & purification , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins/deficiency , Transcription, Genetic
16.
PLoS Pathog ; 8(11): e1003059, 2012.
Article in English | MEDLINE | ID: mdl-23209422

ABSTRACT

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


Subject(s)
DEAD-box RNA Helicases/metabolism , Influenza A virus/metabolism , Influenza, Human/metabolism , Interferons/biosynthesis , Ubiquitination , Viral Nonstructural Proteins/metabolism , Animals , Chlorocebus aethiops , DEAD Box Protein 58 , DEAD-box RNA Helicases/genetics , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Dogs , HeLa Cells , Humans , Influenza A virus/genetics , Influenza, Human/genetics , Interferons/genetics , Mice , Mice, Knockout , Receptors, Immunologic , Transcription Factors/genetics , Transcription Factors/metabolism , Tripartite Motif Proteins , Ubiquitin-Protein Ligases/genetics , Ubiquitin-Protein Ligases/metabolism , Vero Cells , Viral Nonstructural Proteins/genetics
17.
Adv Virus Res ; 119: 1-38, 2024.
Article in English | MEDLINE | ID: mdl-38897707

ABSTRACT

The ubiquitination process is a reversible posttranslational modification involved in many essential cellular functions, such as innate immunity, cell signaling, trafficking, protein stability, and protein degradation. Viruses can use the ubiquitin system to efficiently enter host cells, replicate and evade host immunity, ultimately enhancing viral pathogenesis. Emerging evidence indicates that enveloped viruses can carry free (unanchored) ubiquitin or covalently ubiquitinated viral structural proteins that can increase the efficiency of viral entry into host cells. Furthermore, viruses continuously evolve and adapt to take advantage of the host ubiquitin machinery, highlighting its importance during virus infection. This review discusses the battle between viruses and hosts, focusing on how viruses hijack the ubiquitination process at different steps of the replication cycle, with a specific emphasis on viral entry. We discuss how ubiquitination of viral proteins may affect tropism and explore emerging therapeutics strategies targeting the ubiquitin system for antiviral drug discovery.


Subject(s)
Ubiquitination , Virus Internalization , Virus Replication , Humans , Ubiquitin/metabolism , Viruses/metabolism , Host-Pathogen Interactions , Viral Proteins/metabolism , Viral Proteins/genetics , Virus Diseases/virology , Virus Diseases/immunology , Virus Diseases/metabolism , Animals , Protein Processing, Post-Translational
18.
bioRxiv ; 2024 Jun 17.
Article in English | MEDLINE | ID: mdl-38948778

ABSTRACT

SARS-CoV-2 is a highly transmissible virus that causes COVID-19 disease. Mechanisms of viral pathogenesis include excessive inflammation and viral-induced cell death, resulting in tissue damage. We identified the host E3-ubiquitin ligase TRIM7 as an inhibitor of apoptosis and SARS-CoV-2 replication via ubiquitination of the viral membrane (M) protein. Trim7 -/- mice exhibited increased pathology and virus titers associated with epithelial apoptosis and dysregulated immune responses. Mechanistically, TRIM7 ubiquitinates M on K14, which protects cells from cell death. Longitudinal SARS-CoV-2 sequence analysis from infected patients revealed that mutations on M-K14 appeared in circulating variants during the pandemic. The relevance of these mutations was tested in a mouse model. A recombinant M-K14/K15R virus showed reduced viral replication, consistent with the role of K15 in virus assembly, and increased levels of apoptosis associated with the loss of ubiquitination on K14. TRIM7 antiviral activity requires caspase-6 inhibition, linking apoptosis with viral replication and pathology.

19.
Annu Rev Pathol ; 18: 181-203, 2023 01 24.
Article in English | MEDLINE | ID: mdl-36151059

ABSTRACT

Zika virus (ZIKV) is an emerging virus from the Flaviviridae family that is transmitted to humans by mosquito vectors and represents an important health problem. Infections in pregnant women are of major concern because of potential devastating consequences during pregnancy and have been associated with microcephaly in newborns. ZIKV has a unique ability to use the host machinery to promote viral replication in a tissue-specific manner, resulting in characteristic pathological disorders. Recent studies have proposed that the host ubiquitin system acts as a major determinant of ZIKV tropism by providing the virus with an enhanced ability to enter new cells. In addition, ZIKV has developed mechanisms to evade the host immune response, thereby allowing the establishment of viral persistence and enhancing viral pathogenesis. We discuss recent reports on the mechanisms used by ZIKV to replicate efficiently, and we highlight potential new areas of research for the development of therapeutic approaches.


Subject(s)
Microcephaly , Zika Virus Infection , Zika Virus , Infant, Newborn , Animals , Female , Humans , Pregnancy , Zika Virus Infection/complications , Zika Virus Infection/drug therapy , Virus Replication
20.
Viruses ; 15(11)2023 Oct 25.
Article in English | MEDLINE | ID: mdl-38005825

ABSTRACT

Nipah virus (NiV; genus: Henipavirus; family: Paramyxoviridae) naturally infects Old World fruit bats (family Pteropodidae) without causing overt disease. Conversely, NiV infection in humans and other mammals can be lethal. Comparing bat antiviral responses with those of humans may illuminate the mechanisms that facilitate bats' tolerance. Tripartite motif proteins (TRIMs), a large family of E3-ubiquitin ligases, fine-tune innate antiviral immune responses, and two human TRIMs interact with Henipavirus proteins. We hypothesize that NiV infection induces the expression of an immunosuppressive TRIM in bat, but not human cells, to promote tolerance. Here, we show that TRIM40 is an interferon-stimulated gene (ISG) in pteropodid but not human cells. Knockdown of bat TRIM40 increases gene expression of IFNß, ISGs, and pro-inflammatory cytokines following poly(I:C) transfection. In Pteropus vampyrus, but not human cells, NiV induces TRIM40 expression within 16 h after infection, and knockdown of TRIM40 correlates with reduced NiV titers as compared to control cells. Bats may have evolved to express TRIM40 in response to viral infections to control immunopathogenesis.


Subject(s)
Chiroptera , DEAD Box Protein 58 , Henipavirus Infections , Tripartite Motif Proteins , Animals , Humans , Chiroptera/immunology , Chiroptera/virology , Immunity, Innate , Interferons/genetics , Nipah Virus/genetics , Tripartite Motif Proteins/metabolism , DEAD Box Protein 58/antagonists & inhibitors , DEAD Box Protein 58/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL