Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
1.
Immunity ; 55(1): 174-184.e5, 2022 01 11.
Article in English | MEDLINE | ID: mdl-35021055

ABSTRACT

Human immune responses to viral infections are highly variable, but the genetic factors that contribute to this variability are not well characterized. We used VirScan, a high-throughput epitope scanning technology, to analyze pan-viral antibody reactivity profiles of twins and SNP-genotyped individuals. Using these data, we determined the heritability and genomic loci associated with antibody epitope selection, response breadth, and control of Epstein-Barr virus (EBV) viral load. 107 EBV peptide reactivities were heritable and at least two Epstein-Barr nuclear antigen 2 (EBNA-2) reactivities were associated with variants in the MHC class II locus. We identified an EBV serosignature that predicted viral load in peripheral blood mononuclear cells and was associated with variants in the MHC class I locus. Our study illustrates the utility of epitope profiling to investigate the genetics of pathogen immunity, reports heritable features of the antibody response to viruses, and identifies specific HLA loci important for EBV epitope selection.


Subject(s)
Antibodies, Viral/metabolism , Epitopes/metabolism , Epstein-Barr Virus Infections/genetics , Epstein-Barr Virus Nuclear Antigens/metabolism , Genotype , Herpesvirus 4, Human/physiology , Immunodominant Epitopes/metabolism , Viral Proteins/metabolism , Adolescent , Adult , Aged , Cohort Studies , Epitope Mapping , Epitopes/genetics , Epstein-Barr Virus Infections/immunology , Epstein-Barr Virus Nuclear Antigens/genetics , Female , HLA Antigens/genetics , HLA Antigens/metabolism , Humans , Immunity, Humoral , Immunodominant Epitopes/genetics , Male , Middle Aged , Peptides/genetics , Polymorphism, Single Nucleotide , Quantitative Trait, Heritable , Seroepidemiologic Studies , Viral Load , Viral Proteins/genetics , Young Adult
2.
Immunity ; 55(6): 1051-1066.e4, 2022 06 14.
Article in English | MEDLINE | ID: mdl-35649416

ABSTRACT

Microbial exposures are crucial environmental factors that impact healthspan by sculpting the immune system and microbiota. Antibody profiling via Phage ImmunoPrecipitation Sequencing (PhIP-Seq) provides a high-throughput, cost-effective approach for detecting exposure and response to microbial protein products. We designed and constructed a library of 95,601 56-amino acid peptide tiles spanning 14,430 proteins with "toxin" or "virulence factor" keyword annotations. We used PhIP-Seq to profile the antibodies of ∼1,000 individuals against this "ToxScan" library. In addition to enumerating immunodominant antibody epitopes, we studied the age-dependent stability of the ToxScan profile and used a genome-wide association study to find that the MHC-II locus modulates bacterial epitope selection. We detected previously described anti-flagellin antibody responses in a Crohn's disease cohort and identified an association between anti-flagellin antibodies and juvenile dermatomyositis. PhIP-Seq with the ToxScan library is thus an effective tool for studying the environmental determinants of health and disease at cohort scale.


Subject(s)
Bacteriophages , Peptide Library , Amino Acid Sequence , Antibodies , Antibody Formation , Bacteriophages/genetics , Genome-Wide Association Study , Humans , Immunodominant Epitopes , Prevalence , Virulence Factors/genetics
3.
J Virol ; 91(12)2017 06 15.
Article in English | MEDLINE | ID: mdl-28404843

ABSTRACT

Severe acute respiratory syndrome coronavirus (SARS-CoV) is a highly pathogenic respiratory virus that causes morbidity and mortality in humans. After infection with SARS-CoV, the acute lung injury caused by the virus must be repaired to regain lung function. A dysregulation in this wound healing process leads to fibrosis. Many survivors of SARS-CoV infection develop pulmonary fibrosis (PF), with higher prevalence in older patients. Using mouse models of SARS-CoV pathogenesis, we have identified that the wound repair pathway, controlled by the epidermal growth factor receptor (EGFR), is critical to recovery from SARS-CoV-induced tissue damage. In mice with constitutively active EGFR [EGFR(DSK5) mice], we find that SARS-CoV infection causes enhanced lung disease. Importantly, we show that during infection, the EGFR ligands amphiregulin and heparin-binding EGF-like growth factor (HB-EGF) are upregulated, and exogenous addition of these ligands during infection leads to enhanced lung disease and altered wound healing dynamics. Our data demonstrate a key role of EGFR in the host response to SARS-CoV and how it may be implicated in lung disease induced by other highly pathogenic respiratory viruses.IMPORTANCE PF has many causative triggers, including severe respiratory viruses such as SARS-CoV. Currently there are no treatments to prevent the onset or limit the progression of PF, and the molecular pathways underlying the development of PF are not well understood. In this study, we identified a role for the balanced control of EGFR signaling as a key factor in progression to PF. These data demonstrate that therapeutic treatment modulating EGFR activation could protect against PF development caused by severe respiratory virus infection.


Subject(s)
ErbB Receptors/metabolism , Lung/pathology , Pulmonary Fibrosis/virology , Severe Acute Respiratory Syndrome/metabolism , Severe Acute Respiratory Syndrome/pathology , Severe acute respiratory syndrome-related coronavirus/pathogenicity , Amphiregulin/administration & dosage , Amphiregulin/metabolism , Animals , Disease Models, Animal , Humans , Lung/virology , Mice , Pulmonary Fibrosis/metabolism , Pulmonary Fibrosis/pathology , Severe acute respiratory syndrome-related coronavirus/physiology , Severe Acute Respiratory Syndrome/virology , Signal Transduction , Wound Healing/drug effects
4.
J Virol ; 91(1)2017 Jan 01.
Article in English | MEDLINE | ID: mdl-27795435

ABSTRACT

Middle East respiratory syndrome coronavirus (MERS-CoV) is an important emerging pathogen that was first described in 2012. While the cell surface receptor for MERS-CoV has been identified as dipeptidyl peptidase 4 (DPP4), the mouse DPP4 homologue does not allow virus entry into cells. Therefore, development of mouse models of MERS-CoV has been hampered by the fact that MERS-CoV does not replicate in commonly available mouse strains. We have previously described a mouse model in which mDPP4 was replaced with hDPP4 such that hDPP4 is expressed under the endogenous mDPP4 promoter. In this study, we used this mouse model to analyze the host response to MERS-CoV infection using immunological assays and transcriptome analysis. Depletion of CD4+ T cells, CD8+ T cells, or macrophages has no effect on MERS-CoV replication in the lungs of infected mice. However, we found that depletion of CD8+ T cells protects and depletion of macrophages exacerbates MERS-CoV-induced pathology and clinical symptoms of disease. Overall, we demonstrate an important role for the inflammatory response in regulating MERS-CoV pathogenesis in vivo IMPORTANCE: The Middle East respiratory syndrome coronavirus (MERS-CoV) is a highly pathogenic respiratory virus that emerged from zoonotic sources in 2012. Human infections are still occurring throughout Saudi Arabia at a 38% case fatality rate, with the potential for worldwide spread via air travel. In this work, we identify the host response to the virus and identify inflammatory pathways and cell populations that are critical for protection from severe lung disease. By understanding the immune response to MERS-CoV we can develop targeted therapies to inhibit pathogenesis in the future.


Subject(s)
CD8-Positive T-Lymphocytes/virology , Coronavirus Infections/immunology , Dipeptidyl Peptidase 4/genetics , Macrophages/virology , Middle East Respiratory Syndrome Coronavirus/pathogenicity , Receptors, Virus/genetics , Animals , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/virology , CD8-Positive T-Lymphocytes/immunology , Coronavirus Infections/genetics , Coronavirus Infections/pathology , Coronavirus Infections/virology , Dipeptidyl Peptidase 4/immunology , Disease Models, Animal , Gene Expression Profiling , Gene Expression Regulation , Host-Pathogen Interactions , Humans , Lung/immunology , Lung/virology , Lymphocyte Depletion , Macrophages/immunology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Middle East Respiratory Syndrome Coronavirus/genetics , Middle East Respiratory Syndrome Coronavirus/immunology , Promoter Regions, Genetic , Receptors, Virus/immunology , Transcriptome , Transgenes , Virus Internalization , Virus Replication
5.
J Virol ; 89(23): 11820-33, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26378163

ABSTRACT

UNLABELLED: Severe acute respiratory syndrome (SARS) emerged in November 2002 as a case of atypical pneumonia in China, and the causative agent of SARS was identified to be a novel coronavirus, severe acute respiratory syndrome coronavirus (SARS-CoV). Bone marrow stromal antigen 2 (BST-2; also known as CD317 or tetherin) was initially identified to be a pre-B-cell growth promoter, but it also inhibits the release of virions of the retrovirus human immunodeficiency virus type 1 (HIV-1) by tethering budding virions to the host cell membrane. Further work has shown that BST-2 restricts the release of many other viruses, including the human coronavirus 229E (hCoV-229E), and the genomes of many of these viruses encode BST-2 antagonists to overcome BST-2 restriction. Given the previous studies on BST-2, we aimed to determine if BST-2 has the ability to restrict SARS-CoV and if the SARS-CoV genome encodes any proteins that modulate BST-2's antiviral function. Through an in vitro screen, we identified four potential BST-2 modulators encoded by the SARS-CoV genome: the papain-like protease (PLPro), nonstructural protein 1 (nsp1), ORF6, and ORF7a. As the function of ORF7a in SARS-CoV replication was previously unknown, we focused our study on ORF7a. We found that BST-2 does restrict SARS-CoV, but the loss of ORF7a leads to a much greater restriction, confirming the role of ORF7a as an inhibitor of BST-2. We further characterized the mechanism of BST-2 inhibition by ORF7a and found that ORF7a localization changes when BST-2 is overexpressed and ORF7a binds directly to BST-2. Finally, we also show that SARS-CoV ORF7a blocks the restriction activity of BST-2 by blocking the glycosylation of BST-2. IMPORTANCE: The severe acute respiratory syndrome coronavirus (SARS-CoV) emerged from zoonotic sources in 2002 and caused over 8,000 infections and 800 deaths in 37 countries around the world. Identifying host factors that regulate SARS-CoV pathogenesis is critical to understanding how this lethal virus causes disease. We have found that BST-2 is capable of restricting SARS-CoV release from cells; however, we also identified a SARS-CoV protein that inhibits BST-2 function. We show that the SARS-CoV protein ORF7a inhibits BST-2 glycosylation, leading to a loss of BST-2's antiviral function.


Subject(s)
Antigens, CD/physiology , Glycosylation , Open Reading Frames/genetics , Severe Acute Respiratory Syndrome/virology , Severe acute respiratory syndrome-related coronavirus/physiology , Virion/physiology , Virus Attachment , Animals , Chlorocebus aethiops , Chromatography, Affinity , Cloning, Molecular , Coronavirus 3C Proteases , Cysteine Endopeptidases/genetics , DNA Primers/genetics , Flow Cytometry , GPI-Linked Proteins/physiology , HEK293 Cells , Humans , Immunoprecipitation , Microscopy, Confocal , Microscopy, Electron , Open Reading Frames/physiology , RNA-Dependent RNA Polymerase/genetics , Reverse Transcriptase Polymerase Chain Reaction , Vero Cells , Viral Nonstructural Proteins/genetics , Viral Proteins/genetics
6.
J Biol Chem ; 289(48): 33629-43, 2014 Nov 28.
Article in English | MEDLINE | ID: mdl-25301952

ABSTRACT

The cellular response to mitogens is tightly regulated via transcriptional and post-transcriptional mechanisms to rapidly induce genes that promote proliferation and efficiently attenuate their expression to prevent malignant growth. RNase L is an endoribonuclease that mediates diverse antiproliferative activities, and tristetraprolin (TTP) is a mitogen-induced RNA-binding protein that directs the decay of proliferation-stimulatory mRNAs. In light of their roles as endogenous proliferative constraints, we examined the mechanisms and functional interactions of RNase L and TTP to attenuate a mitogenic response. Mitogen stimulation of RNase L-deficient cells significantly increased TTP transcription and the induction of other mitogen-induced mRNAs. This regulation corresponded with elevated expression of serum-response factor (SRF), a master regulator of mitogen-induced transcription. RNase L destabilized the SRF transcript and formed a complex with SRF mRNA in cells providing a mechanism by which RNase L down-regulates SRF-induced genes. TTP and RNase L proteins interacted in cells suggesting that RNase L is directed to cleave TTP-bound RNAs as a mechanism of substrate specificity. Consistent with their concerted function in RNA turnover, the absence of either RNase L or TTP stabilized SRF mRNA, and a subset of established TTP targets was also regulated by RNase L. RNase L deficiency enhanced mitogen-induced proliferation demonstrating its functional role in limiting the mitogenic response. Our findings support a model of feedback regulation in which RNase L and TTP target SRF mRNA and SRF-induced transcripts. Accordingly, meta-analysis revealed an enrichment of RNase L and TTP targets among SRF-regulated genes suggesting that the RNase L/TTP axis represents a viable target to inhibit SRF-driven proliferation in neoplastic diseases.


Subject(s)
Cell Proliferation/drug effects , Endoribonucleases/metabolism , Gene Expression Regulation/drug effects , Mitogens/pharmacology , RNA Stability/drug effects , Transcription, Genetic/drug effects , Animals , Cell Proliferation/physiology , Gene Expression Regulation/physiology , HEK293 Cells , HeLa Cells , Humans , Mice , Models, Biological , RNA Stability/physiology , Transcription, Genetic/physiology , Tristetraprolin/metabolism
7.
Antimicrob Agents Chemother ; 58(8): 4885-93, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24841273

ABSTRACT

Outbreaks of emerging infections present health professionals with the unique challenge of trying to select appropriate pharmacologic treatments in the clinic with little time available for drug testing and development. Typically, clinicians are left with general supportive care and often untested convalescent-phase plasma as available treatment options. Repurposing of approved pharmaceutical drugs for new indications presents an attractive alternative to clinicians, researchers, public health agencies, drug developers, and funding agencies. Given the development times and manufacturing requirements for new products, repurposing of existing drugs is likely the only solution for outbreaks due to emerging viruses. In the studies described here, a library of 290 compounds was screened for antiviral activity against Middle East respiratory syndrome coronavirus (MERS-CoV) and severe acute respiratory syndrome coronavirus (SARS-CoV). Selection of compounds for inclusion in the library was dependent on current or previous FDA approval or advanced clinical development. Some drugs that had a well-defined cellular pathway as target were included. In total, 27 compounds with activity against both MERS-CoV and SARS-CoV were identified. The compounds belong to 13 different classes of pharmaceuticals, including inhibitors of estrogen receptors used for cancer treatment and inhibitors of dopamine receptor used as antipsychotics. The drugs identified in these screens provide new targets for in vivo studies as well as incorporation into ongoing clinical studies.


Subject(s)
Antiviral Agents/pharmacology , Drug Repositioning , Middle East Respiratory Syndrome Coronavirus/drug effects , Severe acute respiratory syndrome-related coronavirus/drug effects , Small Molecule Libraries/pharmacology , Animals , Antipsychotic Agents/pharmacology , Chlorocebus aethiops , Drug Approval , Estrogen Antagonists/pharmacology , High-Throughput Screening Assays , Humans , Inhibitory Concentration 50 , Middle East Respiratory Syndrome Coronavirus/physiology , Severe acute respiratory syndrome-related coronavirus/physiology , Vero Cells , Virus Replication/drug effects
8.
Nat Commun ; 15(1): 1577, 2024 Feb 21.
Article in English | MEDLINE | ID: mdl-38383452

ABSTRACT

We investigate a relatively underexplored component of the gut-immune axis by profiling the antibody response to gut phages using Phage Immunoprecipitation Sequencing (PhIP-Seq). To cover large antigenic spaces, we develop Dolphyn, a method that uses machine learning to select peptides from protein sets and compresses the proteome through epitope-stitching. Dolphyn compresses the size of a peptide library by 78% compared to traditional tiling, increasing the antibody-reactive peptides from 10% to 31%. We find that the immune system develops antibodies to human gut bacteria-infecting viruses, particularly E.coli-infecting Myoviridae. Cost-effective PhIP-Seq libraries designed with Dolphyn enable the assessment of a wider range of proteins in a single experiment, thus facilitating the study of the gut-immune axis.


Subject(s)
Bacteriophages , Peptide Library , Humans , Epitopes , Amino Acid Sequence , Peptides/genetics , Antibodies , Bacteriophages/genetics , Epitope Mapping/methods
9.
Cell Rep Methods ; 3(10): 100619, 2023 Oct 23.
Article in English | MEDLINE | ID: mdl-37883924

ABSTRACT

High-dimensional flow cytometry is the gold standard to study the human immune system in large cohorts. However, large sample sizes increase inter-experimental variation because of technical and experimental inaccuracies introduced by batch variability. Our high-throughput sample processing pipeline in combination with 28-color flow cytometry focuses on increased throughput (192 samples/experiment) and high reproducibility. We implemented quality control checkpoints to reduce technical and experimental variation. Finally, we integrated FlowSOM clustering to facilitate automated data analysis and demonstrate the reproducibility of our pipeline in a study with 3,357 samples. We reveal age-associated immune dynamics in 2,300 individuals, signified by decreasing T and B cell subsets with age. In addition, by combining genetic analyses, our approach revealed unique immune signatures associated with a single nucleotide polymorphism (SNP) that abrogates CD45 isoform splicing. In summary, we provide a versatile and reliable high-throughput, flow cytometry-based pipeline for immune discovery and exploration in large cohorts.


Subject(s)
B-Lymphocyte Subsets , Leukocytes , Humans , Immunophenotyping , Reproducibility of Results , Flow Cytometry/methods
10.
bioRxiv ; 2023 Jul 31.
Article in English | MEDLINE | ID: mdl-37577562

ABSTRACT

We investigated a relatively underexplored component of the gut-immune axis by profiling the antibody response to gut phages using Phage Immunoprecipitation Sequencing (PhIP-Seq). To enhance this approach, we developed Dolphyn, a novel method that uses machine learning to select peptides from protein sets and compresses the proteome through epitope-stitching. Dolphyn improves the fraction of gut phage library peptides bound by antibodies from 10% to 31% in healthy individuals, while also reducing the number of synthesized peptides by 78%. In our study on gut phages, we discovered that the immune system develops antibodies to bacteria-infecting viruses in the human gut, particularly E.coli-infecting Myoviridae. Cost-effective PhIP-Seq libraries designed with Dolphyn enable the assessment of a wider range of proteins in a single experiment, thus facilitating the study of the gut-immune axis.

11.
Biology (Basel) ; 11(7)2022 Jul 13.
Article in English | MEDLINE | ID: mdl-36101433

ABSTRACT

Enteroviruses such as group B coxsackieviruses (CVB) are commonly suspected as causes of myocarditis that can lead to dilated cardiomyopathy (DCM), and the mouse model of CVB3 myocarditis is routinely used to understand DCM pathogenesis. Mechanistically, autoimmunity is suspected due to the presence of autoantibodies for select antigens. However, their role continues to be enigmatic, which also raises the question of whether the breadth of autoantibodies is sufficiently characterized. Here, we attempted to comprehensively analyze the autoantibody repertoire using Phage ImmunoPrecipitation Sequencing (PhIP-Seq), a versatile and high-throughput platform, in the mouse model of CVB3 myocarditis. First, PhIP-Seq analysis using the VirScan library revealed antibody reactivity only to CVB3 in the infected group but not in controls, thus validating the technique in this model. Second, using the mouse peptide library, we detected autoantibodies to 32 peptides from 25 proteins in infected animals that are ubiquitously expressed and have not been previously reported. Third, by using ELISA as a secondary assay, we confirmed antibody reactivity in sera from CVB3-infected animals to cytochrome c oxidase assembly factor 4 homolog (COA4) and phosphoinositide-3-kinase adaptor protein 1 (PIK3AP1), indicating the specificity of antibody detection by PhIP-Seq technology. Fourth, we noted similar antibody reactivity patterns in CVB3 and CVB4 infections, suggesting that the COA4- and PIK3AP1-reactive antibodies could be common to multiple CVB infections. The specificity of the autoantibodies was affirmed with influenza-infected animals that showed no reactivity to any of the antigens tested. Taken together, our data suggest that the autoantibodies identified by PhIP-Seq may have relevance to CVB pathogenesis, with a possibility that similar reactivity could be expected in human DCM patients.

12.
Cell Rep ; 41(12): 111754, 2022 12 20.
Article in English | MEDLINE | ID: mdl-36543141

ABSTRACT

Anelloviruses represent a major constituent of the commensal human virome; however, little is known about their immunobiology. Here, we present "AnelloScan," a T7 phage library representing the open reading frame 1 (ORF1), ORF2, ORF3, and torque teno virus (TTV)-derived apoptosis-inducing protein (TAIP) sequences of more than 800 human anelloviruses and profile the antibody reactivities of serum samples from a cross-sectional cohort of 156 subjects by using phage-immunoprecipitation sequencing (PhIP-Seq). A majority of anellovirus peptides are not reactive in any of the subjects tested (n = ∼28,000; ∼85% of the library). Antibody-reactive peptides are largely restricted to the C-terminal region of the capsid protein ORF1. Moreover, using a longitudinal cohort of matched blood-transfusion donors and recipients, we find that most transmitted anelloviruses do not elicit a detectable antibody reactivity in the recipient and that the remainder elicit delayed responses appearing ∼100-150 days after transfusion.


Subject(s)
Anelloviridae , Torque teno virus , Humans , Antibody Formation , Cross-Sectional Studies , Torque teno virus/metabolism , Capsid Proteins/metabolism
13.
Cell Syst ; 11(4): 375-381.e4, 2020 10 21.
Article in English | MEDLINE | ID: mdl-33099407

ABSTRACT

Endopeptidases catalyze the internal cleavage of proteins, playing pivotal roles in protein turnover, substrate maturation, and the activation of signaling cascades. A broad range of biological functions in health and disease are controlled by proteases, yet assays to characterize their activities at a proteomic scale do not exist. To address this unmet need, we developed Sensing EndoPeptidase Activity via Release and recapture using flAnking Tag Epitopes (SEPARATE), which uses a monovalent phage display of the human proteome at a 90-aa peptide resolution. We demonstrate that SEPARATE is compatible with several human proteases from distinct catalytic classes, including caspase-1, ADAM17, and thrombin. Both well-characterized and newly identified substrates of these enzymes were detected in the assay. SEPARATE was used to discover a non-canonical caspase-1 substrate, the E3 ubiquitin ligase HUWE1, a key mediator of apoptotic cell death. SEPARATE enables efficient, unbiased assessment of endopeptidase activity by using a phage-displayed proteome. A record of this paper's Transparent Peer Review process is included in the Supplemental Information.


Subject(s)
Cell Surface Display Techniques/methods , Peptide Hydrolases/metabolism , Peptides/analysis , Proteomics/methods , ADAM17 Protein/metabolism , Caspase 1/metabolism , Humans , Peptide Library , Peptides/chemistry , Proteolysis , Proteome/metabolism , Substrate Specificity , THP-1 Cells , Thrombin/metabolism , Tumor Suppressor Proteins/metabolism , Ubiquitin-Protein Ligases/metabolism
14.
Antiviral Res ; 143: 142-150, 2017 07.
Article in English | MEDLINE | ID: mdl-28390872

ABSTRACT

Many survivors of the 2003 outbreak of severe acute respiratory syndrome (SARS) developed residual pulmonary fibrosis with increased severity seen in older patients. Autopsies of patients that died from SARS also showed fibrosis to varying extents. Pulmonary fibrosis can be occasionally seen as a consequence to several respiratory viral infections but is much more common after a SARS coronavirus (SARS-CoV) infection. Given the threat of future outbreaks of severe coronavirus disease, including Middle East respiratory syndrome (MERS), it is important to understand the mechanisms responsible for pulmonary fibrosis, so as to support the development of therapeutic countermeasures and mitigate sequelae of infection. In this article, we summarize pulmonary fibrotic changes observed after a SARS-CoV infection, discuss the extent to which other respiratory viruses induce fibrosis, describe available animal models to study the development of SARS-CoV induced fibrosis and review evidence that pulmonary fibrosis is caused by a hyperactive host response to lung injury mediated by epidermal growth factor receptor (EGFR) signaling. We summarize work from our group and others indicating that inhibiting EGFR signaling may prevent an excessive fibrotic response to SARS-CoV and other respiratory viral infections and propose directions for future research.


Subject(s)
ErbB Receptors/physiology , Pulmonary Fibrosis/etiology , Pulmonary Fibrosis/virology , Severe Acute Respiratory Syndrome/complications , Severe acute respiratory syndrome-related coronavirus/physiology , Severe acute respiratory syndrome-related coronavirus/pathogenicity , Signal Transduction/physiology , Animals , Disease Models, Animal , ErbB Receptors/metabolism , Host-Pathogen Interactions , Humans , Lung Injury/pathology , Lung Injury/virology , Mice , Pathology, Molecular , Pulmonary Fibrosis/metabolism , Pulmonary Fibrosis/pathology , Severe Acute Respiratory Syndrome/metabolism , Severe Acute Respiratory Syndrome/pathology , Severe Acute Respiratory Syndrome/virology , Wound Healing/physiology
15.
Vaccine ; 35(12): 1586-1589, 2017 03 14.
Article in English | MEDLINE | ID: mdl-28237499

ABSTRACT

The Middle East respiratory syndrome coronavirus (MERS-CoV) was first discovered in late 2012 and has gone on to cause over 1800 infections and 650 deaths. There are currently no approved therapeutics or vaccinations for MERS-CoV. The MERS-CoV spike (S) protein is responsible for receptor binding and virion entry to cells, is immunodominant and induces neutralizing antibodies in vivo, all of which, make the S protein an ideal target for anti-MERS-CoV vaccines. In this study, we demonstrate protection induced by vaccination with a recombinant MERS-CoV S nanoparticle vaccine and Matrix-M1 adjuvant combination in mice. The MERS-CoV S nanoparticle vaccine produced high titer anti-S neutralizing antibody and protected mice from MERS-CoV infection in vivo.


Subject(s)
Coronavirus Infections/prevention & control , Middle East Respiratory Syndrome Coronavirus/immunology , Spike Glycoprotein, Coronavirus/immunology , Vaccines, Virus-Like Particle/immunology , Viral Vaccines/immunology , Adjuvants, Immunologic/administration & dosage , Animals , Antibodies, Neutralizing/blood , Antibodies, Viral/blood , Disease Models, Animal , Mice, Inbred BALB C , Middle East Respiratory Syndrome Coronavirus/genetics , Spike Glycoprotein, Coronavirus/genetics , Vaccines, Synthetic/administration & dosage , Vaccines, Synthetic/genetics , Vaccines, Synthetic/immunology , Vaccines, Virus-Like Particle/administration & dosage , Vaccines, Virus-Like Particle/genetics , Viral Vaccines/administration & dosage , Viral Vaccines/genetics
16.
J Interferon Cytokine Res ; 33(1): 34-42, 2013 Jan.
Article in English | MEDLINE | ID: mdl-23113544

ABSTRACT

The endoribonuclease RNase-L is the terminal component of an interferon-regulated RNA decay pathway known as the 2'-5'-oligoadenylate (2-5A) system, whose established functions include antimicrobial and tumor suppressive activities. RNase-L activity requires binding of the small molecule 2-5A, leading to RNase-L dimerization and cleavage of single-stranded RNA. RNase-L expression is controlled post-transcriptionally by its 3'-untranslated region (3' UTR), which exerts a strong negative effect on RNase-L levels. MicroRNAs (miRNAs) are a class of small noncoding RNAs that repress expression of target genes by binding to regions of complementarity often in the 3' UTR. The miR-29 family acts as a tumor suppressor in several cancers, including acute and chronic myelogenous leukemia (CML), and has many oncogenic targets. We report that the miR-29 family represses RNase-L protein expression across several cell types. Using a luciferase reporter, we showed that miR-29 acts via 4 target sites within the RNASEL 3' UTR. Mutation of all sites is required for abrogation of miR-29 repression. In light of the reported tumor suppressive role of miR-29 in K562 CML cells and miR-29 repression of RNase-L in these cells, we generated K562 cells with stable RNase-L knockdown and demonstrated that loss of RNase-L inhibits proliferation in vitro as well as tumor growth in a xenograft model. Our findings identify a previously unknown miRNA regulator of RNase-L expression and support a novel oncogenic role for RNase-L in CML and potentially other hematopoietic malignancies.


Subject(s)
Endoribonucleases/genetics , Gene Expression Regulation, Leukemic , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/genetics , MicroRNAs/genetics , 3' Untranslated Regions/genetics , Animals , Binding Sites/genetics , Blotting, Western , Cell Line, Tumor , Cell Proliferation , Endoribonucleases/metabolism , Female , HEK293 Cells , HeLa Cells , Humans , K562 Cells , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/pathology , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/therapy , Luciferases/genetics , Luciferases/metabolism , Mice , Mice, Nude , MicroRNAs/metabolism , Mutation , RNA Interference , Reverse Transcriptase Polymerase Chain Reaction , Tumor Burden/genetics , Xenograft Model Antitumor Assays
18.
J Immunol ; 178(4): 2429-39, 2007 Feb 15.
Article in English | MEDLINE | ID: mdl-17277150

ABSTRACT

The induction of type I (alphabeta) IFN following virus infection is necessary for the stimulation of effective antiviral host defense. In fibroblasts, a subset of primary genes (including those encoding IFN-beta and IFN-alpha4) are induced directly by intracellular dsRNA generated by the virus during its replication. These primary type I IFNs induce expression of IFN regulatory factor (IRF)-7, required for production of a second cascade of IFN-alpha subtypes and the further establishment of a complete antiviral state. Previously, we had reported on a role for Fas-associated death domain-containing protein (FADD) in the control of TLR-independent innate immune responses to virus infection. Our data in this study demonstrate that FADD is not only required for efficient primary gene induction, but is also essential for induction of Irf7 and effective expression of secondary IFN-alphas and other antiviral genes. Ectopic overexpression of IRF-7 partially rescued dsRNA responsiveness and IFN-alpha production, and a constitutively active variant of IRF-7 displayed normal activity in Fadd(-/-) murine embryonic fibroblasts. MC159, a FADD-interacting viral protein encoded by the molluscum contagiosum poxvirus was found to inhibit dsRNA-activated signaling events upstream of IRF-7. These data indicate that FADD's antiviral activity involves regulation of IRF-7-dependent production of IFN-alpha subtypes and consequent induction of secondary antiviral genes.


Subject(s)
Fas-Associated Death Domain Protein/immunology , Immunity, Innate , Interferon Regulatory Factor-7/immunology , Poxviridae Infections/immunology , Poxviridae/immunology , Signal Transduction/immunology , Animals , Cell Line , Fas-Associated Death Domain Protein/metabolism , Gene Expression Regulation/immunology , Interferon Regulatory Factor-7/biosynthesis , Interferon Type I/biosynthesis , Interferon Type I/immunology , Mice , Poxviridae Infections/metabolism , RNA, Double-Stranded/immunology , Transcriptional Activation , Virus Replication/immunology
19.
J Immunol ; 178(10): 6444-55, 2007 May 15.
Article in English | MEDLINE | ID: mdl-17475874

ABSTRACT

The DExD/H box RNA helicase retinoic acid-inducible gene I (RIG-I) and the melanoma differentiation-associated gene 5 (MDA5) are key intracellular receptors that recognize virus infection to produce type I IFN. A third helicase gene, Lgp2, is homologous to Rig-I and Mda5 but lacks a caspase activation and recruitment domain. We generated Lgp2-deficient mice and report that the loss of this gene greatly sensitizes cells to cytosolic polyinosinic/polycytidylic acid-mediated induction of type I IFN. However, negative feedback inhibition of IFN-beta transcription was found to be normal in the absence of LGP2, indicating that LGP2 is not the primary negative regulator of type I IFN production. Our data further indicate that Lgp2-/- mice exhibited resistance to lethal vesicular stomatitis virus infection, a virus whose replicative RNA intermediates are recognized specifically by RIG-I rather than by MDA5 to trigger the production of type I IFN. However, mice lacking LGP2 were observed to exhibit a defect in type I IFN production in response to infection by the encephalomyocarditis virus, the replication of which activates MDA5-dependent innate immune responses. Collectively, our data indicate a disparate regulatory role for LGP2 in the triggering of innate immune signaling pathways following RNA virus infection.


Subject(s)
Cardiovirus Infections/enzymology , Cardiovirus Infections/prevention & control , DEAD-box RNA Helicases/deficiency , DEAD-box RNA Helicases/genetics , Rhabdoviridae Infections/enzymology , Rhabdoviridae Infections/prevention & control , Animals , Cardiovirus Infections/genetics , Cardiovirus Infections/immunology , Cells, Cultured , DEAD Box Protein 58 , DEAD-box RNA Helicases/metabolism , DEAD-box RNA Helicases/physiology , Encephalomyocarditis virus/immunology , Female , Immunity, Innate/genetics , Interferon-Induced Helicase, IFIH1 , Male , Membrane Proteins/metabolism , Mice , Mice, Inbred C57BL , Mice, Transgenic , Nerve Tissue Proteins/metabolism , Receptors, Cell Surface , Rhabdoviridae Infections/genetics , Rhabdoviridae Infections/immunology , Signal Transduction/genetics , Signal Transduction/immunology , Vesicular stomatitis Indiana virus/immunology
SELECTION OF CITATIONS
SEARCH DETAIL