Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 51
Filter
Add more filters










Publication year range
1.
J Gen Virol ; 104(11)2023 11.
Article in English | MEDLINE | ID: mdl-37962188

ABSTRACT

Parainfluenza virus type 5 (PIV5) can either have a persistent or a lytic phenotype in cultured cells. We have previously shown that the phenotype is determined by the phosphorylation status of the phosphoprotein (P). Single amino acid substitutions at critical residues, including a serine-to-phenylalanine substitution at position 157 on P, result in a switch between persistent and lytic phenotypes. Here, using PIV5 vectors expressing either mCherry or GFP with persistent or lytic phenotypes, we show that in co-infections the persistent phenotype is dominant. Thus, in contrast to the cell death observed with cells infected solely with the lytic variant, in co-infected cells persistence is immediately established and both lytic and persistent genotypes persist. Furthermore, 10-20 % of virus released from dually infected cells contains both genotypes, indicating that PIV5 particles can package more than one genome. Co-infected cells continue to maintain both genotypes/phenotypes during cell passage, as do individual colonies of cells derived from a culture of persistently infected cells. A refinement of our model on how the dynamics of virus selection may occur in vivo is presented.


Subject(s)
Coinfection , Parainfluenza Virus 5 , Paramyxovirinae , Respirovirus Infections , Humans , Parainfluenza Virus 5/genetics , Phenotype
2.
mBio ; 14(3): e0010123, 2023 06 27.
Article in English | MEDLINE | ID: mdl-37097030

ABSTRACT

Infected hosts possess two alternative strategies to protect themselves against the negative impact of virus infections: resistance, used to abrogate virus replication, and disease tolerance, used to avoid tissue damage without controlling viral burden. The principles governing pathogen resistance are well understood, while less is known about those involved in disease tolerance. Here, we studied bluetongue virus (BTV), the cause of bluetongue disease of ruminants, as a model system to investigate the mechanisms of virus-host interactions correlating with disease tolerance. BTV induces clinical disease mainly in sheep, while cattle are considered reservoirs of infection, rarely exhibiting clinical symptoms despite sustained viremia. Using primary cells from multiple donors, we show that BTV consistently reaches higher titers in ovine cells than cells from cattle. The variable replication kinetics of BTV in sheep and cow cells were mostly abolished by abrogating the cell type I interferon (IFN) response. We identified restriction factors blocking BTV replication, but both the sheep and cow orthologues of these antiviral genes possess anti-BTV properties. Importantly, we demonstrate that BTV induces a faster host cell protein synthesis shutoff in primary sheep cells than cow cells, which results in an earlier downregulation of antiviral proteins. Moreover, by using RNA sequencing (RNA-seq), we also show a more pronounced expression of interferon-stimulated genes (ISGs) in BTV-infected cow cells than sheep cells. Our data provide a new perspective on how the type I IFN response in reservoir species can have overall positive effects on both virus and host evolution. IMPORTANCE The host immune response usually aims to inhibit virus replication in order to avoid cell damage and disease. In some cases, however, the infected host avoids the deleterious effects of infection despite high levels of viral replication. This strategy is known as disease tolerance, and it is used by animal reservoirs of some zoonotic viruses. Here, using a virus of ruminants (bluetongue virus [BTV]) as an experimental system, we dissected virus-host interactions in cells collected from species that are susceptible (sheep) or tolerant (cow) to disease. We show that (i) virus modulation of the host antiviral type I interferon (IFN) responses, (ii) viral replication kinetics, and (iii) virus-induced cell damage differ in tolerant and susceptible BTV-infected cells. Understanding the complex virus-host interactions in disease tolerance can allow us to disentangle the critical balance between protective and damaging host immune responses.


Subject(s)
Bluetongue , Interferon Type I , Female , Sheep , Animals , Cattle , Interferon Type I/genetics , Bluetongue/metabolism , Viremia , Antiviral Agents
3.
Biosensors (Basel) ; 12(12)2022 Dec 05.
Article in English | MEDLINE | ID: mdl-36551092

ABSTRACT

Immunodiagnostics have been widely used in the detection of disease biomarkers. The conventional immunological tests in central laboratories require expensive equipment and, for non-specialists, the tests are technically demanding and time-consuming, which has prevented their use by the public. Thus, point-of-care tests (POCT), such as lateral flow immunoassays, are being, or have been, developed as more convenient and low-cost methods for immunodiagnostics. However, the sensitivity of such tests is often a concern. Here, a fluorescence-linked immunosorbent assay (FLISA) using organic light-emitting diodes (OLEDs) as excitation light sources was investigated as a way forward for the development of compact and sensitive POCTs. Phycoerythrin (PE) was selected as the fluorescent dye, and OLEDs were designed with different emission spectra. The leakage light of different OLEDs for exciting PE was then investigated to reduce the background noise and improve the sensitivity of the system. Finally, as proof-of-principle that OLED-based technology can be successfully further developed for POCT, antibodies to severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) in human serum was detected by OLED-FLISA.


Subject(s)
COVID-19 , Immunosorbents , Humans , SARS-CoV-2 , Fluorescence , COVID-19/diagnosis , Antibodies, Viral
4.
Front Immunol ; 13: 888427, 2022.
Article in English | MEDLINE | ID: mdl-36159783

ABSTRACT

Purpose: Janus kinase-1 (JAK1) tyrosine kinase mediates signaling from multiple cytokine receptors, including interferon alpha/beta and gamma (IFN-α/ß and IFN-γ), which are important for viral and mycobacterial protection respectively. We previously reported autosomal recessive (AR) hypomorphic JAK1 mutations in a patient with recurrent atypical mycobacterial infections and relatively minor viral infections. This study tests the impact of partial JAK1 deficiency on cellular responses to IFNs and pathogen control. Methods: We investigated the role of partial JAK1 deficiency using patient cells and cell models generated with lentiviral vectors expressing shRNA. Results: Partial JAK1 deficiency impairs IFN-γ-dependent responses in multiple cell types including THP-1 macrophages, Epstein-Barr Virus (EBV)-transformed B cells and primary dermal fibroblasts. In THP-1 myeloid cells, partial JAK1 deficiency reduced phagosome acidification and apoptosis and resulted in defective control of mycobacterial infection with enhanced intracellular survival. Although both EBV-B cells and primary dermal fibroblasts with partial JAK1 deficiency demonstrate reduced IFN-α responses, control of viral infection was impaired only in patient EBV-B cells and surprisingly intact in patient primary dermal fibroblasts. Conclusion: Our data suggests that partial JAK1 deficiency predominantly affects susceptibility to mycobacterial infection through impact on the IFN-γ responsive pathway in myeloid cells. Susceptibility to viral infections as a result of reduced IFN-α responses is variable depending on cell type. Description of additional patients with inherited JAK1 deficiency will further clarify the spectrum of bacterial and viral susceptibility in this condition. Our results have broader relevance for anticipating infectious complications from the increasing use of selective JAK1 inhibitors.


Subject(s)
Epstein-Barr Virus Infections , Mycobacterium Infections , Mycobacterium , Herpesvirus 4, Human/genetics , Humans , Interferon-alpha/pharmacology , Interferon-beta , Interferon-gamma/genetics , Janus Kinase 1/genetics , Mycobacterium/genetics , Mycobacterium Infections/genetics , RNA, Small Interfering , Receptors, Cytokine
5.
Trends Genet ; 37(1): 46-58, 2021 01.
Article in English | MEDLINE | ID: mdl-32977999

ABSTRACT

The concept that type I interferons (IFN-I) are essential to antiviral immunity derives from studies on animal models and cell lines. Virtually all pathogenic viruses have evolved countermeasures to IFN-I restriction, and genetic loss of viral IFN-I antagonists leads to virus attenuation. But just how important is IFN-I to antiviral defence in humans? The recent discovery of genetic defects of IFN-I signalling illuminates this and other questions of IFN biology, including the role of the mucosa-restricted type III IFNs (IFN-III), informing our understanding of the place of the IFN system within the concerted antiviral response. Here we review monogenic lesions of IFN-I signalling pathways and summarise the organising principles which emerge.


Subject(s)
Antiviral Agents/immunology , Immunity, Innate/immunology , Interferon Type I/antagonists & inhibitors , Viruses/immunology , Animals , Antiviral Agents/pharmacology , Humans , Interferon Type I/genetics , Interferon Type I/metabolism , Signal Transduction , Viruses/drug effects
6.
J Immunol ; 205(1): 261-271, 2020 07 01.
Article in English | MEDLINE | ID: mdl-32423918

ABSTRACT

IFNs, produced during viral infections, induce the expression of hundreds of IFN-stimulated genes (ISGs). Some ISGs have specific antiviral activity, whereas others regulate the cellular response. Besides functioning as an antiviral effector, ISG15 is a negative regulator of IFN signaling, and inherited ISG15 deficiency leads to autoinflammatory IFNopathies, in which individuals exhibit elevated ISG expression in the absence of pathogenic infection. We have recapitulated these effects in cultured human A549-ISG15-/- cells and (using A549-UBA7-/- cells) confirmed that posttranslational modification by ISG15 (ISGylation) is not required for regulation of the type I IFN response. ISG15-deficient cells pretreated with IFN-α were resistant to paramyxovirus infection. We also showed that IFN-α treatment of ISG15-deficient cells led to significant inhibition of global protein synthesis, leading us to ask whether resistance was due to the direct antiviral activity of ISGs or whether cells were nonpermissive because of translation defects. We took advantage of the knowledge that IFN-induced protein with tetratricopeptide repeats 1 (IFIT1) is the principal antiviral ISG for parainfluenza virus 5. Knockdown of IFIT1 restored parainfluenza virus 5 infection in IFN-α-pretreated, ISG15-deficient cells, confirming that resistance was due to the direct antiviral activity of the IFN response. However, resistance could be induced if cells were pretreated with IFN-α for longer times, presumably because of inhibition of protein synthesis. These data show that the cause of virus resistance is 2-fold; ISG15 deficiency leads to the early overexpression of specific antiviral ISGs, but the later response is dominated by an unanticipated, ISG15-dependent loss of translational control.


Subject(s)
Cytokines/deficiency , Disease Resistance/genetics , Interferon-alpha/metabolism , Paramyxoviridae Infections/immunology , Signal Transduction/immunology , Ubiquitins/deficiency , A549 Cells , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/metabolism , Animals , Chlorocebus aethiops , Gene Knockdown Techniques , Gene Knockout Techniques , Humans , Parainfluenza Virus 2, Human/immunology , Parainfluenza Virus 3, Human/immunology , Parainfluenza Virus 5/immunology , Paramyxoviridae Infections/virology , Protein Processing, Post-Translational/immunology , RNA-Binding Proteins/genetics , RNA-Binding Proteins/metabolism , Signal Transduction/genetics , Ubiquitin-Activating Enzymes/genetics , Vero Cells
7.
J Virol ; 94(13)2020 06 16.
Article in English | MEDLINE | ID: mdl-32295916

ABSTRACT

During the replication of parainfluenza virus 5 (PIV5), copyback defective virus genomes (DVGs) are erroneously produced and are packaged into "infectious" virus particles. Copyback DVGs are the primary inducers of innate intracellular responses, including the interferon (IFN) response. While DVGs can interfere with the replication of nondefective (ND) virus genomes and activate the IFN-induction cascade before ND PIV5 can block the production of IFN, we demonstrate that the converse is also true, i.e., high levels of ND virus can block the ability of DVGs to activate the IFN-induction cascade. By following the replication and amplification of DVGs in A549 cells that are deficient in a variety of innate intracellular antiviral responses, we show that DVGs induce an uncharacterized IFN-independent innate response(s) that limits their replication. High-throughput sequencing was used to characterize the molecular structure of copyback DVGs. While there appears to be no sequence-specific break or rejoining points for the generation of copyback DVGs, our findings suggest there are region, size, and/or structural preferences selected for during for their amplification.IMPORTANCE Copyback defective virus genomes (DVGs) are powerful inducers of innate immune responses both in vitro and in vivo They impact the outcome of natural infections, may help drive virus-host coevolution, and promote virus persistence. Due to their potent interfering and immunostimulatory properties, DVGs may also be used therapeutically as antivirals and vaccine adjuvants. However, little is known of the host cell restrictions which limit their amplification. We show here that the generation of copyback DVGs readily occurs during parainfluenza virus 5 (PIV5) replication, but that their subsequent amplification is restricted by the induction of innate intracellular responses. Molecular characterization of PIV5 copyback DVGs suggests that while there are no genome sequence-specific breaks or rejoin points for the generation of copyback DVGs, genome region, size, and structural preferences are selected for during their evolution and amplification.


Subject(s)
Immunity, Innate/immunology , Parainfluenza Virus 5/genetics , Parainfluenza Virus 5/immunology , A549 Cells , Animals , Base Sequence/genetics , Cell Line , Chlorocebus aethiops , Cytoplasm , Defective Viruses/genetics , Genome, Viral/genetics , High-Throughput Nucleotide Sequencing/methods , Humans , Interferons/genetics , RNA, Viral/genetics , Vero Cells , Virion/genetics , Virus Diseases/genetics , Virus Replication/genetics
8.
Sci Immunol ; 4(42)2019 12 13.
Article in English | MEDLINE | ID: mdl-31836668

ABSTRACT

Excessive type I interferon (IFNα/ß) activity is implicated in a spectrum of human disease, yet its direct role remains to be conclusively proven. We investigated two siblings with severe early-onset autoinflammatory disease and an elevated IFN signature. Whole-exome sequencing revealed a shared homozygous missense Arg148Trp variant in STAT2, a transcription factor that functions exclusively downstream of innate IFNs. Cells bearing STAT2R148W in homozygosity (but not heterozygosity) were hypersensitive to IFNα/ß, which manifest as prolonged Janus kinase-signal transducers and activators of transcription (STAT) signaling and transcriptional activation. We show that this gain of IFN activity results from the failure of mutant STAT2R148W to interact with ubiquitin-specific protease 18, a key STAT2-dependent negative regulator of IFNα/ß signaling. These observations reveal an essential in vivo function of STAT2 in the regulation of human IFNα/ß signaling, providing concrete evidence of the serious pathological consequences of unrestrained IFNα/ß activity and supporting efforts to target this pathway therapeutically in IFN-associated disease.


Subject(s)
Immune System Diseases/genetics , Interferon Type I/immunology , STAT2 Transcription Factor/genetics , Germ-Line Mutation , Humans , Immune System Diseases/immunology , Infant , Male , Signal Transduction
9.
J Virol ; 93(17)2019 09 01.
Article in English | MEDLINE | ID: mdl-31189700

ABSTRACT

We have developed a high-throughput sequencing (HTS) workflow for investigating paramyxovirus transcription and replication. We show that sequencing of oligo(dT)-selected polyadenylated mRNAs, without considering the orientation of the RNAs from which they had been generated, cannot accurately be used to analyze the abundance of viral mRNAs because genomic RNA copurifies with the viral mRNAs. The best method is directional sequencing of infected cell RNA that has physically been depleted of ribosomal and mitochondrial RNA followed by bioinformatic steps to differentiate data originating from genomes from viral mRNAs and antigenomes. This approach has the advantage that the abundance of viral mRNA (and antigenomes) and genomes can be analyzed and quantified from the same data. We investigated the kinetics of viral transcription and replication during infection of A549 cells with parainfluenza virus type 2 (PIV2), PIV3, PIV5, or mumps virus and determined the abundances of individual viral mRNAs and readthrough mRNAs. We found that the mRNA abundance gradients differed significantly between all four viruses but that for each virus the pattern remained relatively stable throughout infection. We suggest that rapid degradation of non-poly(A) mRNAs may be primarily responsible for the shape of the mRNA abundance gradient in parainfluenza virus 3, whereas a combination of this factor and disengagement of RNA polymerase at intergenic sequences, particularly those at the NP:P and P:M gene boundaries, may be responsible in the other viruses.IMPORTANCE High-throughput sequencing (HTS) of virus-infected cells can be used to study in great detail the patterns of virus transcription and replication. For paramyxoviruses, and by analogy for all other negative-strand RNA viruses, we show that directional sequencing must be used to distinguish between genomic RNA and mRNA/antigenomic RNA because significant amounts of genomic RNA copurify with poly(A)-selected mRNA. We found that the best method is directional sequencing of total cell RNA, after the physical removal of rRNA (and mitochondrial RNA), because quantitative information on the abundance of both genomic RNA and mRNA/antigenomes can be simultaneously derived. Using this approach, we revealed new details of the kinetics of virus transcription and replication for parainfluenza virus (PIV) type 2, PIV3, PIV5, and mumps virus, as well as on the relative abundance of the individual viral mRNAs.


Subject(s)
Gene Expression Profiling/methods , Paramyxoviridae Infections/virology , Paramyxovirinae/physiology , RNA, Messenger/genetics , Whole Genome Sequencing/methods , A549 Cells , Gene Expression Regulation, Viral , Genome Size , High-Throughput Nucleotide Sequencing , Humans , Paramyxovirinae/classification , Paramyxovirinae/pathogenicity , RNA, Viral/genetics , Species Specificity , Virus Replication
10.
PLoS Pathog ; 15(2): e1007561, 2019 02.
Article in English | MEDLINE | ID: mdl-30742688

ABSTRACT

Paramyxoviruses can establish persistent infections both in vitro and in vivo, some of which lead to chronic disease. However, little is known about the molecular events that contribute to the establishment of persistent infections by RNA viruses. Using parainfluenza virus type 5 (PIV5) as a model we show that phosphorylation of the P protein, which is a key component of the viral RNA polymerase complex, determines whether or not viral transcription and replication becomes repressed at late times after infection. If the virus becomes repressed, persistence is established, but if not, the infected cells die. We found that single amino acid changes at various positions within the P protein switched the infection phenotype from lytic to persistent. Lytic variants replicated to higher titres in mice than persistent variants and caused greater infiltration of immune cells into infected lungs but were cleared more rapidly. We propose that during the acute phases of viral infection in vivo, lytic variants of PIV5 will be selected but, as the adaptive immune response develops, variants in which viral replication can be repressed will be selected, leading to the establishment of prolonged, persistent infections. We suggest that similar selection processes may operate for other RNA viruses.


Subject(s)
Paramyxoviridae Infections/genetics , Paramyxoviridae/genetics , Phosphoproteins/genetics , Viral Proteins/genetics , A549 Cells , Amino Acid Substitution/genetics , Animals , DNA-Directed RNA Polymerases/genetics , DNA-Directed RNA Polymerases/metabolism , Female , HEK293 Cells , Humans , Mice , Mice, Inbred BALB C , Parainfluenza Virus 5/genetics , Parainfluenza Virus 5/pathogenicity , Paramyxoviridae/pathogenicity , Paramyxoviridae Infections/metabolism , Phosphoproteins/metabolism , Phosphoproteins/physiology , Phosphorylation , RNA, Viral , Viral Proteins/metabolism , Viral Proteins/physiology , Virus Replication
11.
Nat Microbiol ; 3(11): 1234-1242, 2018 11.
Article in English | MEDLINE | ID: mdl-30224800

ABSTRACT

The molecular processes that determine the outcome of influenza virus infection in humans are multifactorial and involve a complex interplay between host, viral and bacterial factors1. However, it is generally accepted that a strong innate immune dysregulation known as 'cytokine storm' contributes to the pathology of infections with the 1918 H1N1 pandemic or the highly pathogenic avian influenza viruses of the H5N1 subtype2-4. The RNA sensor retinoic acid-inducible gene I (RIG-I) plays an important role in sensing viral infection and initiating a signalling cascade that leads to interferon expression5. Here, we show that short aberrant RNAs (mini viral RNAs (mvRNAs)), produced by the viral RNA polymerase during the replication of the viral RNA genome, bind to and activate RIG-I and lead to the expression of interferon-ß. We find that erroneous polymerase activity, dysregulation of viral RNA replication or the presence of avian-specific amino acids underlie mvRNA generation and cytokine expression in mammalian cells. By deep sequencing RNA samples from the lungs of ferrets infected with influenza viruses, we show that mvRNAs are generated during infection in vivo. We propose that mvRNAs act as the main agonists of RIG-I during influenza virus infection.


Subject(s)
DEAD Box Protein 58/metabolism , Immunity, Innate/immunology , Influenza A virus/physiology , Orthomyxoviridae Infections/immunology , RNA, Viral/metabolism , Animals , Cell Line , Cytokines/metabolism , DEAD Box Protein 58/genetics , Female , Ferrets , Influenza A virus/genetics , Interferon-beta/genetics , Interferon-beta/metabolism , Male , Mutation , Viral Proteins/genetics , Virus Replication
12.
Antiviral Res ; 150: 79-92, 2018 02.
Article in English | MEDLINE | ID: mdl-29037975

ABSTRACT

Viral interferon (IFN) antagonists are a diverse class of viral proteins that counteract the host IFN response, which is important for controlling viral infections. Viral IFN antagonists are often multifunctional proteins that perform vital roles in virus replication beyond IFN antagonism. The critical importance of viral IFN antagonists is highlighted by the fact that almost all viruses encode one of these proteins. Inhibition of viral IFN antagonists has the potential to exert pleiotropic antiviral effects and thus this important protein class represents a diverse plethora of novel therapeutic targets. To exploit this, we have successfully developed and executed a novel modular cell-based platform that facilitates the safe and rapid screening for inhibitors of a viral IFN antagonist of choice. The platform is based on two reporter cell-lines that provide a simple method to detect activation of IFN induction or signaling via an eGFP gene placed under the control of the IFNß or an ISRE-containing promoter, respectively. Expression of a target IFN antagonist in the appropriate reporter cell-line will block the IFN response and hence eGFP expression. We hypothesized that addition of a compound that inhibits IFN antagonist function will release the block imposed on the IFN response and hence restore eGFP expression, providing a measurable parameter for high throughput screening (HTS). We demonstrate assay proof-of-concept by (i) exploiting hepatitis C virus (HCV) protease inhibitors to inhibit NS3-4A's capacity to block IFN induction and (ii) successfully executing two HTS targeting viral IFN antagonists that block IFN signaling; NS2 and IE1 from human respiratory syncytial virus (RSV) and cytomegalovirus (CMV) respectively, two clinically important viruses for which vaccine development has thus far been unsuccessful and new antivirals are required. Both screens performed robustly and Z' Factor scores of >0.6 were achieved. We identified (i) four hit compounds that specifically inhibit RSV NS2's ability to block IFN signaling by mediating STAT2 degradation and exhibit modest antiviral activity and (ii) two hit compounds that interfere with IE1 transcription and significantly impair CMV replication. Overall, we demonstrate assay proof-of-concept as we target viral IFN antagonists from unrelated viruses and demonstrate its suitability for HTS.


Subject(s)
Antiviral Agents/pharmacology , Drug Discovery , Drug Evaluation, Preclinical , High-Throughput Screening Assays , Interferons/antagonists & inhibitors , Interferons/pharmacology , Viral Proteins/metabolism , Cell Line , Cell Survival/drug effects , Dose-Response Relationship, Drug , Drug Discovery/methods , Drug Evaluation, Preclinical/methods , Gene Expression Regulation/drug effects , Gene Expression Regulation, Viral/drug effects , Genes, Reporter , Humans , Protein Binding , Respiratory Syncytial Virus, Human/drug effects , Respiratory Syncytial Virus, Human/physiology , Signal Transduction , Virus Replication/drug effects
13.
J Gen Virol ; 98(9): 2267-2273, 2017 Sep.
Article in English | MEDLINE | ID: mdl-28869005

ABSTRACT

NS1 proteins of influenza A and B viruses share limited sequence homology, yet both are potent manipulators of host cell processes, particularly interferon (IFN) induction. Although many cellular partners are reported for A/NS1, only a few (e.g. PKR and ISG15) have been identified for B/NS1. Here, affinity-purification and mass spectrometry were used to expand the known host interactome of B/NS1. We identified 22 human proteins as new putative targets for B/NS1, validating several, including DHX9, ILF3, YBX1 and HNRNPC. Consistent with two RNA-binding domains in B/NS1, many of the identified factors bind RNA and some interact with B/NS1 in an RNA-dependent manner. Functional characterization of several B/NS1 interactors identified SNRNP200 as a potential positive regulator of host IFN responses, while ILF3 exhibited dual roles in both IFN induction and influenza B virus replication. These data provide a resource for future investigations into the mechanisms underpinning host cell modulation by influenza B virus NS1.


Subject(s)
Influenza B virus/isolation & purification , Influenza, Human/metabolism , Viral Nonstructural Proteins/metabolism , DEAD-box RNA Helicases/genetics , DEAD-box RNA Helicases/metabolism , Humans , Influenza B virus/genetics , Influenza B virus/metabolism , Influenza, Human/genetics , Influenza, Human/virology , Neoplasm Proteins/genetics , Neoplasm Proteins/metabolism , Nuclear Factor 90 Proteins/genetics , Nuclear Factor 90 Proteins/metabolism , Protein Binding , Viral Nonstructural Proteins/genetics , Y-Box-Binding Protein 1/genetics , Y-Box-Binding Protein 1/metabolism
14.
Viruses ; 9(7)2017 07 13.
Article in English | MEDLINE | ID: mdl-28703784

ABSTRACT

Modern vaccinology has increasingly focused on non-living vaccines, which are more stable than live-attenuated vaccines but often show limited immunogenicity. Immunostimulatory substances, known as adjuvants, are traditionally used to increase the magnitude of protective adaptive immunity in response to a pathogen-associated antigen. Recently developed adjuvants often include substances that stimulate pattern recognition receptors (PRRs), essential components of innate immunity required for the activation of antigen-presenting cells (APCs), which serve as a bridge between innate and adaptive immunity. Nearly all PRRs are potential targets for adjuvants. Given the recent success of toll-like receptor (TLR) agonists in vaccine development, molecules with similar, but additional, immunostimulatory activity, such as defective interfering particles (DIPs) of viruses, represent attractive candidates for vaccine adjuvants. This review outlines some of the recent advances in vaccine development related to the use of TLR agonists, summarizes the current knowledge regarding DIP immunogenicity, and discusses the potential applications of DIPs in vaccine adjuvantation.


Subject(s)
Adjuvants, Immunologic , Defective Viruses/immunology , Receptors, Pattern Recognition/agonists , Receptors, Pattern Recognition/immunology , Vaccines/chemistry , Adaptive Immunity , Animals , Antigen-Presenting Cells/immunology , Genome, Viral , Humans , Immunity, Innate , Immunogenicity, Vaccine , Mice , Toll-Like Receptors/agonists , Vaccines/administration & dosage
15.
Curr Opin Virol ; 23: 35-42, 2017 04.
Article in English | MEDLINE | ID: mdl-28319790

ABSTRACT

In a prototypical response to an acute viral infection it would be expected that the adaptive immune response would eliminate all virally infected cells within a few weeks of infection. However many (non-retrovirus) RNA viruses can establish 'within host' persistent infections that occasionally lead to chronic or reactivated disease. Despite the importance of 'within host' persistent RNA virus infections, much has still to be learnt about the molecular mechanisms by which RNA viruses establish persistent infections, why innate and adaptive immune responses fail to rapidly clear these infections, and the epidemiological and potential disease consequences of such infections.


Subject(s)
Chronic Disease , Host-Pathogen Interactions , RNA Virus Infections/immunology , RNA Virus Infections/virology , Virus Latency , Animals , Humans
16.
Genome Announc ; 4(4)2016 Jul 21.
Article in English | MEDLINE | ID: mdl-27445371

ABSTRACT

We have sequenced the parainfluenza virus 5 strain that persistently infects the commonly used AGS human cell line without causing cytopathology. This virus is most closely related to human strains, indicating that it may have originated from biopsy material or from laboratory contamination during generation of the cell line.

17.
J Biomol Screen ; 21(9): 978-88, 2016 Oct.
Article in English | MEDLINE | ID: mdl-27358388

ABSTRACT

Production of type I interferon (IFN) is an essential component of the innate immune response against invading pathogens. However, its production must be tightly regulated to avoid harmful effects. Compounds that modulate the IFN response are potentially valuable for a variety of applications due to IFN's beneficial and detrimental roles. We developed and executed a cell-based high-throughput screen (HTS) targeting components that participate in and/or regulate the IRF3 and nuclear factor (NF)-κB branches of the IFN induction pathway. The assay detects activation of the IFN induction pathway via an enhanced green fluorescent protein (eGFP) reporter gene under the control of the IFNß promoter and was optimized, miniaturized, and demonstrated suitable for HTS as robust Z' factor scores of >0.6 were consistently achieved. A diversity screening set of 15,667 small molecules was assayed and two novel hit compounds validated that specifically inhibit the IFN induction pathway. We demonstrate that one of these compounds acts at or upstream of IRF3 phosphorylation. A second cell-based assay to detect activation of the IFN signaling (Jak-Stat) pathway via an eGFP reporter gene under the control of an IFN-stimulated response element (ISRE) containing MxA promoter also performed well (robust Z' factor >0.7) and may therefore be similarly used to identify small molecules that modulate the IFN signaling pathway.


Subject(s)
High-Throughput Screening Assays/methods , Immunity, Innate/drug effects , Interferon Type I/antagonists & inhibitors , Small Molecule Libraries/isolation & purification , Green Fluorescent Proteins , Humans , Interferon Regulatory Factor-3/antagonists & inhibitors , NF-kappa B/antagonists & inhibitors , NF-kappa B/genetics , Signal Transduction/drug effects , Small Molecule Libraries/pharmacology
18.
J Virol ; 90(11): 5427-39, 2016 06 01.
Article in English | MEDLINE | ID: mdl-27009961

ABSTRACT

UNLABELLED: Bluetongue virus (BTV) is the causative agent of bluetongue, a major infectious disease of ruminants with serious consequences to both animal health and the economy. The clinical outcome of BTV infection is highly variable and dependent on a variety of factors related to both the virus and the host. In this study, we show that the BTV nonstructural protein NS4 favors viral replication in sheep, the animal species most affected by bluetongue. In addition, NS4 confers a replication advantage on the virus in interferon (IFN)-competent primary sheep endothelial cells and immortalized cell lines. We determined that in cells infected with an NS4 deletion mutant (BTV8ΔNS4), there is increased synthesis of type I IFN compared to cells infected with wild-type BTV-8. In addition, using RNA sequencing (RNA-seq), we show that NS4 modulates the host IFN response and downregulates mRNA levels of type I IFN and interferon-stimulated genes. Moreover, using reporter assays and protein synthesis assays, we show that NS4 downregulates the activities of a variety of promoters, such as the cytomegalovirus immediate-early promoter, the IFN-ß promoter, and a promoter containing interferon-stimulated response elements (ISRE). We also show that the NS4 inhibitory activity on gene expression is related to its nucleolar localization. Furthermore, NS4 does not affect mRNA splicing or cellular translation. The data obtained in this study strongly suggest that BTV NS4 is an IFN antagonist and a key determinant of viral virulence. IMPORTANCE: Bluetongue is one of the main infectious diseases of ruminants and is caused by bluetongue virus (BTV), an arthropod-borne virus transmitted from infected to susceptible animals by Culicoides biting midges. Bluetongue has a variable clinical outcome that can be related to both virus and host factors. It is therefore critical to understand the interplay between BTV and the host immune responses. In this study, we show that a nonstructural protein of BTV (NS4) is critical to counteract the innate immune response of the host. Infection of cells with a BTV mutant lacking NS4 results in increased synthesis of IFN-ß and upregulation of interferon-stimulated genes. In addition, we show that NS4 is a virulence factor for BTV by favoring viral replication in sheep, the animal species most susceptible to bluetongue.


Subject(s)
Bluetongue virus/chemistry , Bluetongue virus/pathogenicity , Bluetongue/virology , Interferon Type I/antagonists & inhibitors , Viral Nonstructural Proteins/chemistry , Viral Nonstructural Proteins/metabolism , Virulence Factors/metabolism , Animals , Bluetongue virus/genetics , Bluetongue virus/immunology , Cell Line , Endothelial Cells/virology , Immunity, Innate , Interferon Type I/biosynthesis , Interferon Type I/genetics , Interferon-beta/genetics , Promoter Regions, Genetic , Sequence Deletion , Sheep , Virulence , Virulence Factors/chemistry , Virulence Factors/isolation & purification , Virus Replication
19.
J Virol ; 90(5): 2616-27, 2015 Dec 23.
Article in English | MEDLINE | ID: mdl-26699638

ABSTRACT

UNLABELLED: Oropouche virus (OROV) is a midge-borne human pathogen with a geographic distribution in South America. OROV was first isolated in 1955, and since then, it has been known to cause recurring outbreaks of a dengue-like illness in the Amazonian regions of Brazil. OROV, however, remains one of the most poorly understood emerging viral zoonoses. Here we describe the successful recovery of infectious OROV entirely from cDNA copies of its genome and generation of OROV mutant viruses lacking either the NSm or the NSs coding region. Characterization of the recombinant viruses carried out in vitro demonstrated that the NSs protein of OROV is an interferon (IFN) antagonist as in other NSs-encoding bunyaviruses. Additionally, we demonstrate the importance of the nine C-terminal amino acids of OROV NSs in IFN antagonistic activity. OROV was also found to be sensitive to IFN-α when cells were pretreated; however, the virus was still capable of replicating at doses as high as 10,000 U/ml of IFN-α, in contrast to the family prototype BUNV. We found that OROV lacking the NSm protein displayed characteristics similar to those of the wild-type virus, suggesting that the NSm protein is dispensable for virus replication in the mammalian and mosquito cell lines that were tested. IMPORTANCE: Oropouche virus (OROV) is a public health threat in Central and South America, where it causes periodic outbreaks of dengue-like illness. In Brazil, OROV is the second most frequent cause of arboviral febrile illness after dengue virus, and with the current rates of urban expansion, more cases of this emerging viral zoonosis could occur. To better understand the molecular biology of OROV, we have successfully rescued the virus along with mutants. We have established that the C terminus of the NSs protein is important in interferon antagonism and that the NSm protein is dispensable for virus replication in cell culture. The tools described in this paper are important in terms of understanding this important yet neglected human pathogen.


Subject(s)
Host-Pathogen Interactions , Orthobunyavirus/genetics , Orthobunyavirus/physiology , Viral Nonstructural Proteins/deficiency , Viral Nonstructural Proteins/metabolism , Virus Replication , Animals , Cell Line , Humans , Interferons/antagonists & inhibitors , Orthobunyavirus/immunology , Reverse Genetics
20.
Sci Transl Med ; 7(307): 307ra154, 2015 Sep 30.
Article in English | MEDLINE | ID: mdl-26424569

ABSTRACT

Type I interferon (IFN-α/ß) is a fundamental antiviral defense mechanism. Mouse models have been pivotal to understanding the role of IFN-α/ß in immunity, although validation of these findings in humans has been limited. We investigated a previously healthy child with fatal encephalitis after inoculation of the live attenuated measles, mumps, and rubella (MMR) vaccine. By targeted resequencing, we identified a homozygous mutation in the high-affinity IFN-α/ß receptor (IFNAR2) in the proband, as well as a newborn sibling, that rendered cells unresponsive to IFN-α/ß. Reconstitution of the proband's cells with wild-type IFNAR2 restored IFN-α/ß responsiveness and control of IFN-attenuated viruses. Despite the severe outcome of systemic live vaccine challenge, the proband had previously shown no evidence of heightened susceptibility to respiratory viral pathogens. The phenotype of IFNAR2 deficiency, together with similar findings in STAT2-deficient patients, supports an essential but narrow role for IFN-α/ß in human antiviral immunity.


Subject(s)
Antiviral Agents/metabolism , Immunity , Receptor, Interferon alpha-beta/deficiency , Fatal Outcome , Genes, Recessive , Genetic Complementation Test , Humans , Infant , Interferons/metabolism , Receptor, Interferon alpha-beta/metabolism , Signal Transduction
SELECTION OF CITATIONS
SEARCH DETAIL
...