Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 32
Filter
Add more filters










Publication year range
1.
ACS Infect Dis ; 6(10): 2783-2799, 2020 10 09.
Article in English | MEDLINE | ID: mdl-32870648

ABSTRACT

Marburg virus (MARV) causes sporadic outbreaks of severe disease with high case fatality rates in humans. To date, neither therapeutics nor prophylactic approaches have been approved for MARV disease. The MARV matrix protein VP40 (mVP40) plays central roles in virus assembly and budding. mVP40 also inhibits interferon signaling by inhibiting the function of Janus kinase 1. This suppression of host antiviral defenses likely contributes to MARV virulence and therefore is a potential therapeutic target. We developed and optimized a cell-based high-throughput screening (HTS) assay in 384-well format to measure mVP40 interferon (IFN) antagonist function such that inhibitors could be identified. We performed a pilot screen of 1280 bioactive compounds and identified 3 hits, azaguanine-8, tosufloxacin hydrochloride, and linezolid, with Z scores > 3 and no significant cytotoxicity. Of these, azaguanine-8 inhibited MARV growth at noncytotoxic concentrations. These data demonstrate the suitability of the HTS mVP40 assay for drug discovery and suggest potential directions for anti-MARV therapeutic development.


Subject(s)
Marburg Virus Disease , Marburgvirus , Animals , High-Throughput Screening Assays , Humans , Interferons , Virus Assembly
2.
Life Sci ; 260: 118482, 2020 Nov 01.
Article in English | MEDLINE | ID: mdl-32971105

ABSTRACT

Cardiovascular disease (CVD) is the most common co-morbidity associated with COVID-19 and the fatality rate in COVID-19 patients with CVD is higher compared to other comorbidities, such as hypertension and diabetes. Preliminary data suggest that COVID-19 may also cause or worsen cardiac injury in infected patients through multiple mechanisms such as 'cytokine storm', endotheliosis, thrombosis, lymphocytopenia etc. Autopsies of COVID-19 patients reveal an infiltration of inflammatory mononuclear cells in the myocardium, confirming the role of the immune system in mediating cardiovascular damage in response to COVID-19 infection and also suggesting potential causal mechanisms for the development of new cardiac pathologies and/or exacerbation of underlying CVDs in infected patients. In this review, we discuss the potential underlying molecular mechanisms that drive COVID-19-mediated cardiac damage, as well as the short term and expected long-term cardiovascular ramifications of COVID-19 infection in patients.


Subject(s)
Betacoronavirus/isolation & purification , Cardiovascular Diseases/etiology , Coronavirus Infections/complications , Inflammation/etiology , Pneumonia, Viral/complications , COVID-19 , Cardiovascular Diseases/pathology , Coronavirus Infections/transmission , Coronavirus Infections/virology , Humans , Inflammation/pathology , Pandemics , Pneumonia, Viral/transmission , Pneumonia, Viral/virology , Prognosis , SARS-CoV-2
3.
J Virol ; 94(13)2020 06 16.
Article in English | MEDLINE | ID: mdl-32295912

ABSTRACT

Menglà virus (MLAV), identified in Rousettus bats, is a phylogenetically distinct member of the family Filoviridae Because the filoviruses Ebola virus (EBOV) and Marburg virus (MARV) modulate host innate immunity, MLAV VP35, VP40, and VP24 proteins were compared with their EBOV and MARV homologs for innate immune pathway modulation. In human and Rousettus cells, MLAV VP35 behaved like EBOV and MARV VP35s, inhibiting virus-induced activation of the interferon beta (IFN-ß) promoter and interferon regulatory factor 3 (IRF3) phosphorylation. MLAV VP35 also interacted with PACT, a host protein engaged by EBOV VP35 to inhibit RIG-I signaling. MLAV VP35 also inhibits PKR activation. MLAV VP40 was demonstrated to inhibit type I IFN-induced gene expression in human and bat cells. It blocked STAT1 tyrosine phosphorylation induced either by type I IFN or overexpressed Jak1, paralleling MARV VP40. MLAV VP40 also inhibited virus-induced IFN-ß promoter activation, a property shared by MARV VP40 and EBOV VP24. A Jak kinase inhibitor did not recapitulate this inhibition in the absence of viral proteins. Therefore, inhibition of Jak-STAT signaling is insufficient to explain inhibition of IFN-ß promoter activation. MLAV VP24 did not inhibit IFN-induced gene expression or bind karyopherin α proteins, properties of EBOV VP24. MLAV VP24 differed from MARV VP24 in that it failed to interact with Keap1 or activate an antioxidant response element reporter gene due to the absence of a Keap1-binding motif. These functional observations support a closer relationship of MLAV to MARV than to EBOV but also are consistent with MLAV belonging to a distinct genus.IMPORTANCE EBOV and MARV, members of the family Filoviridae, are highly pathogenic zoonotic viruses that cause severe disease in humans. Both viruses use several mechanisms to modulate the host innate immune response, and these likely contribute to the severity of disease. Here, we demonstrate that MLAV, a filovirus newly discovered in a bat, suppresses antiviral type I interferon responses in both human and bat cells. Inhibitory activities are possessed by MLAV VP35 and VP40, which parallels how MARV blocks IFN responses. However, whereas MARV activates cellular antioxidant responses through an interaction between its VP24 protein and host protein Keap1, MLAV VP24 lacks a Keap1-binding motif and fails to activate this cytoprotective response. These data indicate that MLAV possesses immune-suppressing functions that could facilitate human infection. They also support the placement of MLAV in a different genus than either EBOV or MARV.


Subject(s)
Filoviridae Infections/physiopathology , Filoviridae/genetics , Animals , Chiroptera/immunology , Chiroptera/virology , Ebolavirus , Filoviridae/metabolism , Filoviridae/pathogenicity , HEK293 Cells , Humans , Immunity, Innate , Interferon Regulatory Factor-3/immunology , Interferon Regulatory Factor-3/metabolism , Interferon-beta/immunology , Kelch-Like ECH-Associated Protein 1/metabolism , Marburgvirus , NF-E2-Related Factor 2/metabolism , STAT1 Transcription Factor , Viral Proteins/metabolism , Viral Regulatory and Accessory Proteins/genetics , Viral Regulatory and Accessory Proteins/metabolism
4.
Cell Rep ; 28(12): 3032-3046.e6, 2019 09 17.
Article in English | MEDLINE | ID: mdl-31533029

ABSTRACT

Zaire ebolavirus (EBOV) VP35 protein is a suppressor of type I interferon (IFN) production, an inhibitor of dendritic cell maturation, and a putative virulence determinant. Here, a recombinant EBOV encoding a mutant VP35 virus (VP35m) is demonstrated to activate RIG-I-like receptor signaling and innate antiviral pathways. When inoculated into cynomolgus macaques, VP35m exhibits dramatic attenuation as compared to wild-type EBOV (wtEBOV), with 20 or 300 times the standard 100% lethal challenge dose not causing EBOV disease (EVD). Further, VP35m infection, despite limited replication in vivo, activates antigen presentation and innate immunity pathways and elicits increased frequencies of proliferating memory T cells and B cells and production of anti-EBOV antibodies. Upon wtEBOV challenge, VP35m-immunized animals survive, exhibiting host responses consistent with an orderly immune response and the absence of excessive inflammation. These data demonstrate that VP35 is a critical EBOV immune evasion factor and provide insights into immune mechanisms of EBOV control.


Subject(s)
B-Lymphocytes , Ebolavirus , Hemorrhagic Fever, Ebola , T-Lymphocytes , Viral Regulatory and Accessory Proteins , Virulence Factors , Animals , B-Lymphocytes/immunology , B-Lymphocytes/pathology , Chlorocebus aethiops , Ebolavirus/genetics , Ebolavirus/immunology , Ebolavirus/pathogenicity , Hemorrhagic Fever, Ebola/genetics , Hemorrhagic Fever, Ebola/immunology , Hemorrhagic Fever, Ebola/pathology , Hemorrhagic Fever, Ebola/prevention & control , Macaca fascicularis , T-Lymphocytes/immunology , T-Lymphocytes/pathology , Vero Cells , Viral Regulatory and Accessory Proteins/genetics , Viral Regulatory and Accessory Proteins/immunology , Virulence Factors/genetics , Virulence Factors/immunology
5.
J Exp Med ; 216(9): 2038-2056, 2019 09 02.
Article in English | MEDLINE | ID: mdl-31217193

ABSTRACT

Autosomal recessive IRF7 and IRF9 deficiencies impair type I and III IFN immunity and underlie severe influenza pneumonitis. We report three unrelated children with influenza A virus (IAV) infection manifesting as acute respiratory distress syndrome (IAV-ARDS), heterozygous for rare TLR3 variants (P554S in two patients and P680L in the third) causing autosomal dominant (AD) TLR3 deficiency. AD TLR3 deficiency can underlie herpes simplex virus-1 (HSV-1) encephalitis (HSE) by impairing cortical neuron-intrinsic type I IFN immunity to HSV-1. TLR3-mutated leukocytes produce normal levels of IFNs in response to IAV. In contrast, TLR3-mutated fibroblasts produce lower levels of IFN-ß and -λ, and display enhanced viral susceptibility, upon IAV infection. Moreover, the patients' iPSC-derived pulmonary epithelial cells (PECs) are susceptible to IAV. Treatment with IFN-α2b or IFN-λ1 rescues this phenotype. AD TLR3 deficiency may thus underlie IAV-ARDS by impairing TLR3-dependent, type I and/or III IFN-mediated, PEC-intrinsic immunity. Its clinical penetrance is incomplete for both IAV-ARDS and HSE, consistent with their typically sporadic nature.


Subject(s)
Influenza, Human/genetics , Inheritance Patterns/genetics , Pneumonia/genetics , Toll-Like Receptor 3/deficiency , Alleles , Child , Child, Preschool , Epithelial Cells/drug effects , Epithelial Cells/metabolism , Fatal Outcome , Female , Fibroblasts/drug effects , Fibroblasts/pathology , Heterozygote , Humans , Induced Pluripotent Stem Cells/drug effects , Induced Pluripotent Stem Cells/metabolism , Infant , Infant, Newborn , Influenza A virus/drug effects , Influenza A virus/physiology , Interferons/metabolism , Loss of Function Mutation/genetics , Lung/pathology , Male , Mutation, Missense/genetics , Poly I-C/pharmacology , Protein Transport
6.
ACS Infect Dis ; 5(8): 1385-1396, 2019 08 09.
Article in English | MEDLINE | ID: mdl-31120240

ABSTRACT

Marburg virus causes sporadic outbreaks of severe hemorrhagic fever with high case fatality rates. Approved, effective, and safe therapeutic or prophylactic countermeasures are lacking. To address this, we used phage display to engineer a synthetic antibody, sFab H3, which binds the Marburg virus VP35 protein (mVP35). mVP35 is a critical cofactor of the viral replication complex and a viral immune antagonist. sFab H3 displayed high specificity for mVP35 and not for the closely related Ebola virus VP35. sFab H3 inhibited viral-RNA synthesis in a minigenome assay, suggesting its potential use as an antiviral. We characterized sFab H3 by a combination of biophysical and biochemical methods, and a crystal structure of the complex solved to 1.7 Å resolution defined the molecular interface between the sFab H3 and mVP35 interferon inhibitory domain. Our study identifies mVP35 as a therapeutic target using an approach that provides a framework for generating engineered Fabs targeting other viral proteins.


Subject(s)
Antibodies, Viral/pharmacology , Immunoglobulin Fab Fragments/pharmacology , Marburgvirus/drug effects , RNA, Viral/biosynthesis , Viral Regulatory and Accessory Proteins/immunology , Binding Sites, Antibody , Cell Surface Display Techniques , Crystallization , Crystallography, X-Ray , Humans , Marburgvirus/genetics , Marburgvirus/physiology , Models, Molecular , Virus Replication/drug effects
7.
Cell ; 175(7): 1917-1930.e13, 2018 12 13.
Article in English | MEDLINE | ID: mdl-30550789

ABSTRACT

Ebola virus (EBOV) infection often results in fatal illness in humans, yet little is known about how EBOV usurps host pathways during infection. To address this, we used affinity tag-purification mass spectrometry (AP-MS) to generate an EBOV-host protein-protein interaction (PPI) map. We uncovered 194 high-confidence EBOV-human PPIs, including one between the viral transcription regulator VP30 and the host ubiquitin ligase RBBP6. Domain mapping identified a 23 amino acid region within RBBP6 that binds to VP30. A crystal structure of the VP30-RBBP6 peptide complex revealed that RBBP6 mimics the viral nucleoprotein (NP) binding to the same interface of VP30. Knockdown of endogenous RBBP6 stimulated viral transcription and increased EBOV replication, whereas overexpression of either RBBP6 or the peptide strongly inhibited both. These results demonstrate the therapeutic potential of biologics that target this interface and identify additional PPIs that may be leveraged for novel therapeutic strategies.


Subject(s)
Carrier Proteins , DNA-Binding Proteins , Ebolavirus/physiology , Hemorrhagic Fever, Ebola/metabolism , Transcription Factors , Viral Proteins , Virus Replication/physiology , Carrier Proteins/chemistry , Carrier Proteins/genetics , Carrier Proteins/metabolism , Crystallography, X-Ray , DNA-Binding Proteins/chemistry , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , HEK293 Cells , HeLa Cells , Hemorrhagic Fever, Ebola/genetics , Hemorrhagic Fever, Ebola/pathology , Humans , Protein Interaction Mapping , Transcription Factors/chemistry , Transcription Factors/genetics , Transcription Factors/metabolism , Ubiquitin-Protein Ligases , Viral Proteins/chemistry , Viral Proteins/genetics , Viral Proteins/metabolism
8.
Antiviral Res ; 158: 288-302, 2018 10.
Article in English | MEDLINE | ID: mdl-30144461

ABSTRACT

Specific host pathways that may be targeted therapeutically to inhibit the replication of Ebola virus (EBOV) and other emerging viruses remain incompletely defined. A screen of 200,000 compounds for inhibition of an EBOV minigenome (MG) assay that measures the function of the viral polymerase complex identified as hits several compounds with an amino-tetrahydrocarbazole scaffold. This scaffold was structurally similar to GSK983, a compound previously described as having broad-spectrum antiviral activity due to its impairing de novo pyrimidine biosynthesis through inhibition of dihydroorotate dehydrogenase (DHODH). We generated compound SW835, the racemic version of GSK983 and demonstrated that SW835 and brequinar, another DHODH inhibitor, potently inhibit the MG assay and the replication of EBOV, vesicular stomatitis virus (VSV) and Zika (ZIKV) in vitro. Nucleoside and deoxynucleoside supplementation studies demonstrated that depletion of pyrimidine pools contributes to antiviral activity of these compounds. As reported for other DHODH inhibitors, SW835 and brequinar also induced expression of interferon stimulated genes (ISGs). ISG induction was demonstrated to occur without production of IFNα/ß and independently of the IFNα receptor and was not blocked by EBOV-encoded suppressors of IFN signaling pathways. Furthermore, we demonstrated that transcription factor IRF1 is required for this ISG induction, and that IRF1 induction requires the DNA damage response kinase ATM. Therefore, de novo pyrimidine biosynthesis is critical for the replication of EBOV and other RNA viruses and inhibition of this pathway activates an ATM and IRF1-dependent innate immune response that subverts EBOV immune evasion functions.


Subject(s)
Ebolavirus/drug effects , Immunity, Innate/drug effects , Nucleosides/pharmacology , Pyrimidines/antagonists & inhibitors , Pyrimidines/biosynthesis , Virus Replication/drug effects , A549 Cells , Antiviral Agents/pharmacology , Biphenyl Compounds/chemistry , Biphenyl Compounds/pharmacology , Carbazoles/chemistry , Carbazoles/pharmacology , DNA Damage , Dihydroorotate Dehydrogenase , HEK293 Cells , Hemorrhagic Fever, Ebola/drug therapy , Hemorrhagic Fever, Ebola/virology , Host-Pathogen Interactions/drug effects , Humans , Immune Evasion , Immunity, Innate/genetics , Interferon Regulatory Factor-1/metabolism , Interferon Regulatory Factor-1/pharmacology , Interferon-alpha/metabolism , Interferon-beta/metabolism , Oxidoreductases Acting on CH-CH Group Donors/antagonists & inhibitors , RNA Viruses/drug effects , Signal Transduction/drug effects , Vesiculovirus/drug effects , Zika Virus/drug effects
9.
Cell Rep ; 24(4): 861-872.e6, 2018 07 24.
Article in English | MEDLINE | ID: mdl-30044983

ABSTRACT

Non-retroviral integrated RNA viral sequences (NIRVs) potentially encoding ∼280 amino acid homologs to filovirus VP35 proteins are present across the Myotis genus of bats. These are estimated to have been maintained for ∼18 million years, indicating their co-option. To address the reasons for co-option, 16 Myotis VP35s were characterized in comparison to VP35s from the extant filoviruses Ebola virus and Marburg virus, in which VP35s play critical roles in immune evasion and RNA synthesis. The Myotis VP35s demonstrated a conserved suppression of innate immune signaling, albeit with reduced potency, in either human or Myotis cells. Their attenuation reflects a lack of dsRNA binding that in the filoviral VP35s correlates with potent suppression of interferon responses. Despite divergent function, evolution has preserved in Myotis the structure of the filoviral VP35s, indicating that this structure is critical for co-opted function, possibly as a regulator of innate immune signaling.


Subject(s)
Chiroptera/genetics , Ebolavirus/immunology , Filoviridae/immunology , Viral Regulatory and Accessory Proteins/chemistry , Viral Regulatory and Accessory Proteins/immunology , Animals , Chick Embryo , Ebolavirus/genetics , Filoviridae/genetics , Genome , HEK293 Cells , Humans , Interferon-beta/biosynthesis , Interferon-beta/genetics , Interferon-beta/immunology , Marburgvirus/genetics , Marburgvirus/immunology , Models, Molecular , Protein Binding , RNA, Double-Stranded/metabolism , Signal Transduction , Structure-Activity Relationship , Viral Regulatory and Accessory Proteins/antagonists & inhibitors , Viral Regulatory and Accessory Proteins/genetics
10.
Sci Rep ; 8(1): 8989, 2018 06 12.
Article in English | MEDLINE | ID: mdl-29895962

ABSTRACT

There are many emerging and re-emerging globally prevalent viruses for which there are no licensed vaccines or antiviral medicines. Arbidol (ARB, umifenovir), used clinically for decades in several countries as an anti-influenza virus drug, inhibits many other viruses. In the current study, we show that ARB inhibits six different isolates of Zika virus (ZIKV), including African and Asian lineage viruses in multiple cell lines and primary human vaginal and cervical epithelial cells. ARB protects against ZIKV-induced cytopathic effects. Time of addition studies indicate that ARB is most effective at suppressing ZIKV when added to cells prior to infection. Moreover, ARB inhibits pseudoviruses expressing the ZIKV Envelope glycoprotein. Thus, ARB, a broadly acting anti-viral agent with a well-established safety profile, inhibits ZIKV, likely by blocking viral entry.


Subject(s)
Antiviral Agents/pharmacology , Indoles/pharmacology , Zika Virus Infection , Zika Virus/metabolism , A549 Cells , Animals , Chlorocebus aethiops , Cytopathogenic Effect, Viral/drug effects , HEK293 Cells , Humans , Vero Cells , Viral Envelope Proteins/metabolism , Zika Virus Infection/drug therapy , Zika Virus Infection/pathology
11.
Cell ; 172(5): 966-978.e12, 2018 02 22.
Article in English | MEDLINE | ID: mdl-29474922

ABSTRACT

Ebola virus nucleoprotein (eNP) assembles into higher-ordered structures that form the viral nucleocapsid (NC) and serve as the scaffold for viral RNA synthesis. However, molecular insights into the NC assembly process are lacking. Using a hybrid approach, we characterized the NC-like assembly of eNP, identified novel regulatory elements, and described how these elements impact function. We generated a three-dimensional structure of the eNP NC-like assembly at 5.8 Å using electron cryo-microscopy and identified a new regulatory role for eNP helices α22-α23. Biochemical, biophysical, and mutational analyses revealed that inter-eNP contacts within α22-α23 are critical for viral NC assembly and regulate viral RNA synthesis. These observations suggest that the N terminus and α22-α23 of eNP function as context-dependent regulatory modules (CDRMs). Our current study provides a framework for a structural mechanism for NC-like assembly and a new therapeutic target.


Subject(s)
Cryoelectron Microscopy , Ebolavirus/physiology , Ebolavirus/ultrastructure , Nucleocapsid/ultrastructure , Nucleoproteins/ultrastructure , Virus Assembly , Models, Biological , Mutant Proteins/chemistry , Mutation/genetics , Nucleoproteins/chemistry , Protein Multimerization , Protein Structure, Secondary , Protein Subunits/chemistry , Protein Subunits/metabolism , RNA, Viral/biosynthesis , RNA, Viral/chemistry , RNA, Viral/metabolism
12.
Antiviral Res ; 150: 193-201, 2018 02.
Article in English | MEDLINE | ID: mdl-29294299

ABSTRACT

Ebola virus (EBOV) is an enveloped negative-sense, single-stranded RNA virus of the filovirus family that causes severe disease in humans. Approved therapies for EBOV disease are lacking. EBOV RNA synthesis is carried out by a virus-encoded complex with RNA-dependent RNA polymerase activity that is required for viral propagation. This complex and its activities are therefore potential antiviral targets. To identify potential lead inhibitors of EBOV RNA synthesis, a library of small molecule compounds was screened against a previously established assay of EBOV RNA synthesis, the EBOV minigenome assay (MGA), in 384 well microplate format. The screen identified 56 hits that inhibited EBOV MGA activity by more than 70% while exhibiting less than 20% cell cytotoxicity. Inhibitory chemical scaffolds included angelicin derivatives, derivatives of the antiviral compound GSK983 and benzoquinolines. Structure-activity relationship (SAR) studies of the benzoquinoline scaffold produced ∼50 analogs and led to identification of an optimized compound, SW456, with a submicromolar IC50 in the EBOV MGA and antiviral activity against infectious EBOV in cell culture. The compound was also active against a MGA for another deadly filovirus, Marburg virus. It also exhibited antiviral activity towards a negative-sense RNA virus from the rhabdovirus family, vesicular stomatitis virus, and a positive-sense RNA virus, Zika virus. Overall, these data demonstrate the potential of the EBOV MGA to identify anti-EBOV compounds and identifies the benzoquinoline series as a broad-spectrum antiviral lead.


Subject(s)
Antiviral Agents/pharmacology , Ebolavirus/drug effects , High-Throughput Screening Assays , Quinolines/pharmacology , Virus Replication/drug effects , Antiviral Agents/chemistry , Dose-Response Relationship, Drug , Drug Discovery , Drug Evaluation, Preclinical , Ebolavirus/genetics , Humans , Quinolines/chemistry , Small Molecule Libraries
13.
Nat Microbiol ; 2: 17101, 2017 Jun 30.
Article in English | MEDLINE | ID: mdl-28665409

ABSTRACT

Human respiratory syncytial virus (hRSV) is a major cause of morbidity and mortality in the paediatric, elderly and immune-compromised populations1,2. A gap in our understanding of hRSV disease pathology is the interplay between virally encoded immune antagonists and host components that limit hRSV replication. hRSV encodes for non-structural (NS) proteins that are important immune antagonists3-6; however, the role of these proteins in viral pathogenesis is incompletely understood. Here, we report the crystal structure of hRSV NS1 protein, which suggests that NS1 is a structural paralogue of hRSV matrix (M) protein. Comparative analysis of the shared structural fold with M revealed regions unique to NS1. Studies on NS1 wild type or mutant alone or in recombinant RSVs demonstrate that structural regions unique to NS1 contribute to modulation of host responses, including inhibition of type I interferon responses, suppression of dendritic cell maturation and promotion of inflammatory responses. Transcriptional profiles of A549 cells infected with recombinant RSVs show significant differences in multiple host pathways, suggesting that NS1 may have a greater role in regulating host responses than previously appreciated. These results provide a framework to target NS1 for therapeutic development to limit hRSV-associated morbidity and mortality.


Subject(s)
Dendritic Cells/immunology , Host-Pathogen Interactions , Interferon Type I/immunology , Respiratory Syncytial Virus, Human/immunology , Respiratory Syncytial Virus, Human/physiology , Viral Nonstructural Proteins/chemistry , Viral Nonstructural Proteins/metabolism , A549 Cells , Animals , Chlorocebus aethiops , Dendritic Cells/metabolism , Humans , Interferon Type I/biosynthesis , Mutation , Protein Domains , Protein Folding , Protein Structure, Secondary , Transcriptome , Vero Cells , Viral Matrix Proteins/chemistry , Viral Nonstructural Proteins/genetics , Virus Replication
14.
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
15.
Nat Commun ; 8: 15576, 2017 06 08.
Article in English | MEDLINE | ID: mdl-28593988

ABSTRACT

Ebola virus (EBOV) is an enveloped negative-sense RNA virus that causes sporadic outbreaks with high case fatality rates. Ebola viral protein 30 (eVP30) plays a critical role in EBOV transcription initiation at the nucleoprotein (eNP) gene, with additional roles in the replication cycle such as viral assembly. However, the mechanistic basis for how eVP30 functions during the virus replication cycle is currently unclear. Here we define a key interaction between eVP30 and a peptide derived from eNP that is important to facilitate interactions leading to the recognition of the RNA template. We present crystal structures of the eVP30 C-terminus in complex with this eNP peptide. Functional analyses of the eVP30-eNP interface identify residues that are critical for viral RNA synthesis. Altogether, these results support a model where the eVP30-eNP interaction plays a critical role in transcription initiation and provides a novel target for the development of antiviral therapy.


Subject(s)
Ebolavirus/genetics , Nucleoproteins/metabolism , RNA, Viral/biosynthesis , Transcription Factors/metabolism , Transcription Initiation, Genetic/physiology , Viral Proteins/metabolism , Cell Line , Crystallography, X-Ray , HEK293 Cells , Humans , Protein Binding , Transcription, Genetic/genetics , Virus Replication/genetics
16.
PLoS One ; 12(6): e0178717, 2017.
Article in English | MEDLINE | ID: mdl-28636653

ABSTRACT

Ebola virus and Marburg virus are members of the Filovirdae family and causative agents of hemorrhagic fever with high fatality rates in humans. Filovirus virulence is partially attributed to the VP35 protein, a well-characterized inhibitor of the RIG-I-like receptor pathway that triggers the antiviral interferon (IFN) response. Prior work demonstrates the ability of VP35 to block potent RIG-I activators, such as Sendai virus (SeV), and this IFN-antagonist activity is directly correlated with its ability to bind RNA. Several structural studies demonstrate that VP35 binds short synthetic dsRNAs; yet, there are no data that identify viral immunostimulatory RNAs (isRNA) or host RNAs bound to VP35 in cells. Utilizing a SeV infection model, we demonstrate that both viral isRNA and host RNAs are bound to Ebola and Marburg VP35s in cells. By deep sequencing the purified VP35-bound RNA, we identified the SeV copy-back defective interfering (DI) RNA, previously identified as a robust RIG-I activator, as the isRNA bound by multiple filovirus VP35 proteins, including the VP35 protein from the West African outbreak strain (Makona EBOV). Moreover, RNAs isolated from a VP35 RNA-binding mutant were not immunostimulatory and did not include the SeV DI RNA. Strikingly, an analysis of host RNAs bound by wild-type, but not mutant, VP35 revealed that select host RNAs are preferentially bound by VP35 in cell culture. Taken together, these data support a model in which VP35 sequesters isRNA in virus-infected cells to avert RIG-I like receptor (RLR) activation.


Subject(s)
Ebolavirus/physiology , Hemorrhagic Fever, Ebola/immunology , High-Throughput Nucleotide Sequencing/methods , Host-Pathogen Interactions/immunology , RNA, Double-Stranded/metabolism , Viral Regulatory and Accessory Proteins/metabolism , Hemorrhagic Fever, Ebola/genetics , Hemorrhagic Fever, Ebola/virology , Humans , Interferons/antagonists & inhibitors , Sendai virus/genetics , Sendai virus/immunology , Signal Transduction , Viral Regulatory and Accessory Proteins/immunology
17.
Methods Mol Biol ; 1628: 133-142, 2017.
Article in English | MEDLINE | ID: mdl-28573616

ABSTRACT

Innate immunity is the first line of defense against virus infections and is marked by production of type I interferons (IFN), a family of cytokines that includes IFN-ß and several IFN-αs. For the filoviruses and many other RNA viruses that replicate in the cytoplasm, the RIG-I-like pattern recognition receptors (RLRs) are potential triggers of IFN production. To counteract such innate antiviral responses, many viruses encode proteins that antagonize RLR signaling. Ebola virus (EBOV) and other filoviruses produce VP35 proteins that block IFN induction via RLR signaling. We describe here cell-based reporter gene assays that quantify the IFN-antagonist function of filovirus VP35 proteins by assessing activation of the IFN-ß promoter.


Subject(s)
Filoviridae Infections/genetics , Filoviridae/genetics , Interferon Type I/genetics , Viral Regulatory and Accessory Proteins/genetics , Filoviridae/pathogenicity , Filoviridae Infections/pathology , Filoviridae Infections/virology , Host-Pathogen Interactions/genetics , Humans , Immunity, Innate/genetics , Interferon Type I/antagonists & inhibitors , Molecular Biology/methods , Signal Transduction/drug effects
18.
mBio ; 8(2)2017 04 04.
Article in English | MEDLINE | ID: mdl-28377530

ABSTRACT

Ebola virus (EBOV) protein VP35 inhibits production of interferon alpha/beta (IFN) by blocking RIG-I-like receptor signaling pathways, thereby promoting virus replication and pathogenesis. A high-throughput screening assay, developed to identify compounds that either inhibit or bypass VP35 IFN-antagonist function, identified five DNA intercalators as reproducible hits from a library of bioactive compounds. Four, including doxorubicin and daunorubicin, are anthracycline antibiotics that inhibit topoisomerase II and are used clinically as chemotherapeutic drugs. These compounds were demonstrated to induce IFN responses in an ATM kinase-dependent manner and to also trigger the DNA-sensing cGAS-STING pathway of IFN induction. These compounds also suppress EBOV replication in vitro and induce IFN in the presence of IFN-antagonist proteins from multiple negative-sense RNA viruses. These findings provide new insights into signaling pathways activated by important chemotherapy drugs and identify a novel therapeutic approach for IFN induction that may be exploited to inhibit RNA virus replication.IMPORTANCE Ebola virus and other emerging RNA viruses are significant but unpredictable public health threats. Therapeutic approaches with broad-spectrum activity could provide an attractive response to such infections. We describe a novel assay that can identify small molecules that overcome Ebola virus-encoded innate immune evasion mechanisms. This assay identified as hits cancer chemotherapeutic drugs, including doxorubicin. Follow-up studies provide new insight into how doxorubicin induces interferon (IFN) responses, revealing activation of both the DNA damage response kinase ATM and the DNA sensor cGAS and its partner signaling protein STING. The studies further demonstrate that the ATM and cGAS-STING pathways of IFN induction are a point of vulnerability not only for Ebola virus but for other RNA viruses as well, because viral innate immune antagonists consistently fail to block these signals. These studies thereby define a novel avenue for therapeutic intervention against emerging RNA viruses.


Subject(s)
Antiviral Agents/pharmacology , DNA Damage/immunology , Ebolavirus/physiology , Immune Evasion/drug effects , Interferons/metabolism , Topoisomerase II Inhibitors/pharmacology , Virus Replication/drug effects , Cell Line , Ebolavirus/immunology , Humans
19.
Nat Microbiol ; 2: 17037, 2017 Mar 27.
Article in English | MEDLINE | ID: mdl-28346446

ABSTRACT

During the last few decades, the global incidence of dengue virus (DENV) has increased dramatically, and it is now endemic in more than 100 countries. To establish a productive infection in humans, DENV uses different strategies to inhibit or avoid the host innate immune system. Several DENV proteins have been shown to strategically target crucial components of the type I interferon system. Here, we report that the DENV NS2B protease cofactor targets the DNA sensor cyclic GMP-AMP synthase (cGAS) for lysosomal degradation to avoid the detection of mitochondrial DNA during infection. Such degradation subsequently results in the inhibition of type I interferon production in the infected cell. Our data demonstrate a mechanism by which cGAS senses cellular damage upon DENV infection.


Subject(s)
DNA, Mitochondrial/physiology , Dengue Virus/genetics , Host-Pathogen Interactions , Nucleotidyltransferases/metabolism , Viral Nonstructural Proteins/metabolism , DNA, Mitochondrial/genetics , Dendritic Cells/virology , Dengue/immunology , Dengue/virology , Dengue Virus/chemistry , Dengue Virus/enzymology , Dengue Virus/immunology , HEK293 Cells , Humans , Immunity, Innate , Interferon Type I/immunology , Membrane Proteins/metabolism , Nucleotidyltransferases/deficiency , Nucleotidyltransferases/genetics , Nucleotidyltransferases/immunology , Signal Transduction , Viral Nonstructural Proteins/genetics
20.
Cell Rep ; 14(7): 1632-1640, 2016 Feb 23.
Article in English | MEDLINE | ID: mdl-26876165

ABSTRACT

Suppression of innate immune responses during filoviral infection contributes to disease severity. Ebola (EBOV) and Marburg (MARV) viruses each encode a VP35 protein that suppresses RIG-I-like receptor signaling and interferon-α/ß (IFN-α/ß) production by several mechanisms, including direct binding to double stranded RNA (dsRNA). Here, we demonstrate that in cell culture, MARV infection results in a greater upregulation of IFN responses as compared to EBOV infection. This correlates with differences in the efficiencies by which EBOV and MARV VP35s antagonize RIG-I signaling. Furthermore, structural and biochemical studies suggest that differential recognition of RNA elements by the respective VP35 C-terminal IFN inhibitory domain (IID) rather than affinity for RNA by the respective VP35s is critical for this observation. Our studies reveal functional differences in EBOV versus MARV VP35 RNA binding that result in unexpected differences in the host response to deadly viral pathogens.


Subject(s)
DEAD-box RNA Helicases/genetics , Ebolavirus/genetics , Interferon-alpha/immunology , Interferon-beta/immunology , Marburgvirus/genetics , RNA, Double-Stranded/genetics , Viral Regulatory and Accessory Proteins/genetics , Amino Acid Sequence , Cell Line , DEAD Box Protein 58 , DEAD-box RNA Helicases/chemistry , DEAD-box RNA Helicases/immunology , Ebolavirus/immunology , Gene Expression Regulation , Host-Pathogen Interactions , Humans , Interferon-alpha/antagonists & inhibitors , Interferon-alpha/biosynthesis , Interferon-beta/antagonists & inhibitors , Interferon-beta/biosynthesis , Marburgvirus/immunology , Models, Molecular , Molecular Sequence Data , Monocytes , Protein Binding , Protein Structure, Secondary , Protein Structure, Tertiary , RNA, Double-Stranded/chemistry , RNA, Double-Stranded/immunology , Receptors, Immunologic , Sequence Alignment , Signal Transduction , Species Specificity , Viral Regulatory and Accessory Proteins/chemistry , Viral Regulatory and Accessory Proteins/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...