Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 39
Filter
1.
Methods Mol Biol ; 2682: 261-279, 2023.
Article in English | MEDLINE | ID: mdl-37610588

ABSTRACT

Diseases caused by henipaviruses feature incubation periods of up to 16 days, during which infected animals may show no apparent signs of disease yet be capable of transmitting the virus to humans. This risk has prompted research into host-derived biomarkers for early disease detection. Here, we describe a methodology for the assaying of host microRNAs (miRs), small non-coding RNAs that show promise as biomarkers for several human diseases and are responsive during early-stage henipavirus infection. In addition to their potential as disease biomarkers, miRNA profiling of henipavirus infections provides insight into cellular and immune pathways associated with disease pathogenesis.


Subject(s)
Henipavirus Infections , MicroRNAs , Animals , Humans , Biological Assay , MicroRNAs/genetics
2.
Immunobiology ; 228(3): 152380, 2023 05.
Article in English | MEDLINE | ID: mdl-37031606

ABSTRACT

Inflammation and lipid regulator with UBA-like and NBR1-like domains (ILRUN) is a protein-encoding gene associated with innate immune signaling, lipid metabolism and cancer. In the context of innate immunity, ILRUN inhibits IRF3-mediated transcription of antimicrobial and proinflammatory cytokines by inducing degradation of the transcriptional coactivators CBP and p300. There remains a paucity of information, however, regarding the innate immune roles of ILRUN beyond in vitro analyses. To address this, we utilize a knockout mouse model to investigate the effect of ILRUN on cytokine expression in splenocytes and on the development of immune cell populations in the spleen and thymus. We show elevated production of tumor necrosis factor and interleukin-6 cytokines in ILRUN-deficient splenocytes following stimulation with the innate immune ligands polyinosinic:polycytidylic acid or lipopolysaccharide. Differences were also observed in the populations of several T cell subsets, including regulatory, mucosal-associated invariant and natural killer. These data identify novel functions for ILRUN in the development of certain immune cell populations and support previous in vitro findings that ILRUN negatively regulates the synthesis of pathogen-stimulated cytokines. This establishes the ILRUN knockout mouse model as a valuable resource for further study of the functions of ILRUN in health and disease.


Subject(s)
Cytokines , T-Lymphocyte Subsets , Mice , Animals , Cytokines/metabolism , Immunity, Innate , Immunologic Factors/metabolism , Adjuvants, Immunologic/metabolism , Mice, Knockout
3.
Pathogens ; 11(11)2022 Nov 03.
Article in English | MEDLINE | ID: mdl-36365042

ABSTRACT

In Australia, there is a paucity of data about the extent and impact of zoonotic tick-related illnesses. Even less is understood about a multifaceted illness referred to as Debilitating Symptom Complexes Attributed to Ticks (DSCATT). Here, we describe a research plan for investigating the aetiology, pathophysiology, and clinical outcomes of human tick-associated disease in Australia. Our approach focuses on the transmission of potential pathogens and the immunological responses of the patient after a tick bite. The protocol is strengthened by prospective data collection, the recruitment of two external matched control groups, and sophisticated integrative data analysis which, collectively, will allow the robust demonstration of associations between a tick bite and the development of clinical and pathological abnormalities. Various laboratory analyses are performed including metagenomics to investigate the potential transmission of bacteria, protozoa and/or viruses during tick bite. In addition, multi-omics technology is applied to investigate links between host immune responses and potential infectious and non-infectious disease causations. Psychometric profiling is also used to investigate whether psychological attributes influence symptom development. This research will fill important knowledge gaps about tick-borne diseases. Ultimately, we hope the results will promote improved diagnostic outcomes, and inform the safe management and treatment of patients bitten by ticks in Australia.

4.
PLoS One ; 17(4): e0265670, 2022.
Article in English | MEDLINE | ID: mdl-35381016

ABSTRACT

Host biomarkers are increasingly being considered as tools for improved COVID-19 detection and prognosis. We recently profiled circulating host-encoded microRNA (miRNAs) during SARS-CoV-2 infection, revealing a signature that classified COVID-19 cases with 99.9% accuracy. Here we sought to develop a signature suited for clinical application by analyzing specimens collected using minimally invasive procedures. Eight miRNAs displayed altered expression in anterior nasal tissues from COVID-19 patients, with miR-142-3p, a negative regulator of interleukin-6 (IL-6) production, the most strongly upregulated. Supervised machine learning analysis revealed that a three-miRNA signature (miR-30c-2-3p, miR-628-3p and miR-93-5p) independently classifies COVID-19 cases with 100% accuracy. This study further defines the host miRNA response to SARS-CoV-2 infection and identifies candidate biomarkers for improved COVID-19 detection.


Subject(s)
COVID-19 , MicroRNAs , Biomarkers , COVID-19/diagnosis , Gene Expression Profiling , Humans , MicroRNAs/genetics , MicroRNAs/metabolism , Respiratory System/metabolism , SARS-CoV-2/genetics
5.
PLoS Pathog ; 17(7): e1009759, 2021 07.
Article in English | MEDLINE | ID: mdl-34320031

ABSTRACT

The host response to SARS-CoV-2 infection provide insights into both viral pathogenesis and patient management. The host-encoded microRNA (miRNA) response to SARS-CoV-2 infection, however, remains poorly defined. Here we profiled circulating miRNAs from ten COVID-19 patients sampled longitudinally and ten age and gender matched healthy donors. We observed 55 miRNAs that were altered in COVID-19 patients during early-stage disease, with the inflammatory miR-31-5p the most strongly upregulated. Supervised machine learning analysis revealed that a three-miRNA signature (miR-423-5p, miR-23a-3p and miR-195-5p) independently classified COVID-19 cases with an accuracy of 99.9%. In a ferret COVID-19 model, the three-miRNA signature again detected SARS-CoV-2 infection with 99.7% accuracy, and distinguished SARS-CoV-2 infection from influenza A (H1N1) infection and healthy controls with 95% accuracy. Distinct miRNA profiles were also observed in COVID-19 patients requiring oxygenation. This study demonstrates that SARS-CoV-2 infection induces a robust host miRNA response that could improve COVID-19 detection and patient management.


Subject(s)
COVID-19 Testing/methods , COVID-19/diagnosis , COVID-19/genetics , MicroRNAs/genetics , SARS-CoV-2 , Adult , Aged , Animals , COVID-19/blood , Case-Control Studies , Diagnosis, Differential , Disease Models, Animal , Female , Ferrets , Gene Expression , Host Microbial Interactions/genetics , Humans , Influenza A Virus, H1N1 Subtype , Longitudinal Studies , Male , MicroRNAs/blood , Middle Aged , Orthomyxoviridae Infections/diagnosis , Orthomyxoviridae Infections/genetics , Pandemics , Supervised Machine Learning
6.
NPJ Vaccines ; 6(1): 67, 2021 May 10.
Article in English | MEDLINE | ID: mdl-33972565

ABSTRACT

Vaccines against SARS-CoV-2 are likely to be critical in the management of the ongoing pandemic. A number of candidates are in Phase III human clinical trials, including ChAdOx1 nCoV-19 (AZD1222), a replication-deficient chimpanzee adenovirus-vectored vaccine candidate. In preclinical trials, the efficacy of ChAdOx1 nCoV-19 against SARS-CoV-2 challenge was evaluated in a ferret model of infection. Groups of ferrets received either prime-only or prime-boost administration of ChAdOx1 nCoV-19 via the intramuscular or intranasal route. All ChAdOx1 nCoV-19 administration combinations resulted in significant reductions in viral loads in nasal-wash and oral swab samples. No vaccine-associated adverse events were observed associated with the ChAdOx1 nCoV-19 candidate, with the data from this study suggesting it could be an effective and safe vaccine against COVID-19. Our study also indicates the potential for intranasal administration as a way to further improve the efficacy of this leading vaccine candidate.

7.
J Virol ; 95(15): e0032721, 2021 07 12.
Article in English | MEDLINE | ID: mdl-33963054

ABSTRACT

The human protein-coding gene ILRUN (inflammation and lipid regulator with UBA-like and NBR1-like domains; previously C6orf106) was identified as a proviral factor for Hendra virus infection and was recently characterized to function as an inhibitor of type I interferon expression. Here, we have utilized transcriptome sequencing (RNA-seq) to define cellular pathways regulated by ILRUN in the context of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection of Caco-2 cells. We find that inhibition of ILRUN expression by RNA interference alters transcription profiles of numerous cellular pathways, including upregulation of the SARS-CoV-2 entry receptor ACE2 and several other members of the renin-angiotensin aldosterone system. In addition, transcripts of the SARS-CoV-2 coreceptors TMPRSS2 and CTSL were also upregulated. Inhibition of ILRUN also resulted in increased SARS-CoV-2 replication, while overexpression of ILRUN had the opposite effect, identifying ILRUN as a novel antiviral factor for SARS-CoV-2 replication. This represents, to our knowledge, the first report of ILRUN as a regulator of the renin-angiotensin-aldosterone system (RAAS). IMPORTANCE There is no doubt that the current rapid global spread of COVID-19 has had significant and far-reaching impacts on our health and economy and will continue to do so. Research in emerging infectious diseases, such as severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), is growing rapidly, with new breakthroughs in the understanding of host-virus interactions to assist with the development of innovative and exciting therapeutic strategies. Here, we present the first evidence that modulation of the human protein-coding gene ILRUN functions as an antiviral factor for SARS-CoV-2 infection, likely through its newly identified role in regulating the expression of SARS-CoV-2 entry receptors ACE2, TMPRSS2, and CTSL. These data improve our understanding of biological pathways that regulate host factors critical to SARS-CoV-2 infection, contributing to the development of antiviral strategies to deal with the current SARS-CoV-2 pandemic.


Subject(s)
Angiotensin-Converting Enzyme 2/biosynthesis , COVID-19/metabolism , Down-Regulation , Gene Expression Regulation, Enzymologic , Neoplasm Proteins/metabolism , SARS-CoV-2/metabolism , Angiotensin-Converting Enzyme 2/genetics , Animals , COVID-19/genetics , Caco-2 Cells , Cathepsin L/biosynthesis , Cathepsin L/genetics , Chlorocebus aethiops , Humans , Neoplasm Proteins/genetics , Renin-Angiotensin System , SARS-CoV-2/genetics , Serine Endopeptidases/biosynthesis , Serine Endopeptidases/genetics , Vero Cells
8.
Int J Mol Sci ; 22(7)2021 Mar 25.
Article in English | MEDLINE | ID: mdl-33806254

ABSTRACT

The global COVID-19 pandemic caused by SARS-CoV-2 has resulted in over 2.2 million deaths. Disease outcomes range from asymptomatic to severe with, so far, minimal genotypic change to the virus so understanding the host response is paramount. Transcriptomics has become incredibly important in understanding host-pathogen interactions; however, post-transcriptional regulation plays an important role in infection and immunity through translation and mRNA stability, allowing tight control over potent host responses by both the host and the invading virus. Here, we apply ribosome profiling to assess post-transcriptional regulation of host genes during SARS-CoV-2 infection of a human lung epithelial cell line (Calu-3). We have identified numerous transcription factors (JUN, ZBTB20, ATF3, HIVEP2 and EGR1) as well as select antiviral cytokine genes, namely IFNB1, IFNL1,2 and 3, IL-6 and CCL5, that are restricted at the post-transcriptional level by SARS-CoV-2 infection and discuss the impact this would have on the host response to infection. This early phase restriction of antiviral transcripts in the lungs may allow high viral load and consequent immune dysregulation typically seen in SARS-CoV-2 infection.


Subject(s)
Cytokines/genetics , RNA Processing, Post-Transcriptional , Ribosomes/metabolism , Ribosomes/virology , SARS-CoV-2/immunology , Transcription Factors/genetics , Animals , Antiviral Agents/antagonists & inhibitors , Cell Line, Tumor , Chlorocebus aethiops , Computational Biology , Cytokines/metabolism , Epithelial Cells/immunology , Epithelial Cells/virology , Gene Expression Profiling , Host Microbial Interactions , Humans , Immunity, Innate/genetics , Lung/immunology , Lung/virology , RNA, Messenger/metabolism , RNA-Seq , Ribosomes/genetics , SARS-CoV-2/metabolism , Transcription Factors/metabolism , Transcriptome , Vero Cells
9.
J Virol Methods ; 286: 113977, 2020 12.
Article in English | MEDLINE | ID: mdl-32979405

ABSTRACT

The development of medical countermeasures against severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) requires robust viral assays. Here we have adapted a protocol for polyethylene glycol (PEG)-mediated precipitation of SARS-CoV-2 stocks without the need for ultracentrifugation. Virus precipitation resulted in a ∼1.5 log10 increase in SARS-CoV-2 titres of virus prepared in VeroE6 cells and enabled the infection of several immortalized human cell lines (Caco-2 and Calu-3) at a high multiplicity of infection not practically achievable without virus concentration. This protocol underscores the utility of PEG-mediated precipitation for SARS-CoV-2 and provides a resource for a range of coronavirus research areas.


Subject(s)
Betacoronavirus/isolation & purification , Clinical Laboratory Techniques/methods , Coronavirus Infections/virology , Pneumonia, Viral/virology , Polyethylene Glycols/chemistry , Animals , COVID-19 , COVID-19 Testing , Caco-2 Cells , Chlorocebus aethiops , Coronavirus Infections/diagnosis , Humans , Pandemics , Pneumonia, Viral/diagnosis , Reverse Transcriptase Polymerase Chain Reaction , SARS-CoV-2 , Ultracentrifugation/methods , Vero Cells
10.
Front Microbiol ; 11: 1197, 2020.
Article in English | MEDLINE | ID: mdl-32582115

ABSTRACT

In the pursuit of improved diagnostic tests for infectious diseases, several classes of molecules have been scrutinized as prospective biomarkers. Small (18-22 nucleotide), non-coding RNA transcripts called microRNAs (miRNAs) have emerged as promising candidates with extensive diagnostic potential, due to their role in numerous diseases, previously established methods for quantitation and their stability within biofluids. Despite efforts to identify, characterize and apply miRNA signatures as diagnostic markers in a range of non-infectious diseases, their application in infectious disease has advanced relatively slowly. Here, we outline the benefits that miRNA biomarkers offer to the diagnosis, management, and treatment of infectious diseases. Investigation of these novel biomarkers could advance the use of personalized medicine in infectious disease treatment, which raises important considerations for validating their use as diagnostic or prognostic markers. Finally, we discuss new and emerging miRNA detection platforms, with a focus on rapid, point-of-care testing, to evaluate the benefits and obstacles of miRNA biomarkers for infectious disease.

11.
Heliyon ; 6(6): e04115, 2020 Jun.
Article in English | MEDLINE | ID: mdl-32518853

ABSTRACT

Regulation of type-I interferon (IFN) production is essential to the balance between antimicrobial defence and autoimmune disorders. The human protein-coding gene ILRUN (inflammation and lipid regulator with UBA-like and NBR1-like domains, previously C6orf106) was recently characterised as an inhibitor of antiviral and proinflammatory cytokine (interferon-alpha/beta and tumor necrosis factor alpha) transcription. Currently there is a paucity of information about the molecular characteristics of ILRUN, despite it being associated with several diseases including virus infection, coronary artery disease, obesity and cancer. Here, we characterise ILRUN as a highly phylogenetically conserved protein containing UBA-like and a NBR1-like domains that are both essential for inhibition of type-I interferon and tumor necrosis factor alpha) transcription in human cells. We also solved the crystal structure of the NBR1-like domain, providing insights into its potential role in ILRUN function. This study provides critical information for future investigations into the role of ILRUN in health and disease.

12.
mBio ; 11(1)2020 01 28.
Article in English | MEDLINE | ID: mdl-31992625

ABSTRACT

Activation of cyclic GMP-AMP (cGAMP) synthase (cGAS) plays a critical role in antiviral responses to many DNA viruses. Sensing of cytosolic DNA by cGAS results in synthesis of the endogenous second messenger cGAMP that activates stimulator of interferon genes (STING) in infected cells. Critically, cGAMP can also propagate antiviral responses to uninfected cells through intercellular transfer, although the modalities of this transfer between epithelial and immune cells remain poorly defined. We demonstrate here that cGAMP-producing epithelial cells can transactivate STING in cocultured macrophages through direct cGAMP transfer. cGAMP transfer was reliant upon connexin expression by epithelial cells and pharmacological inhibition of connexins blunted STING-dependent transactivation of the macrophage compartment. Macrophage transactivation by cGAMP contributed to a positive-feedback loop amplifying antiviral responses, significantly protecting uninfected epithelial cells against viral infection. Collectively, our findings constitute the first direct evidence of a connexin-dependent cGAMP transfer to macrophages by epithelial cells, to amplify antiviral responses.IMPORTANCE Recent studies suggest that extracellular cGAMP can be taken up by macrophages to engage STING through several mechanisms. Our work demonstrates that connexin-dependent communication between epithelial cells and macrophages plays a significant role in the amplification of antiviral responses mediated by cGAMP and suggests that pharmacological strategies aimed at modulating connexins may have therapeutic applications to control antiviral responses in humans.


Subject(s)
Connexins/metabolism , Host-Pathogen Interactions , Nucleotides, Cyclic/metabolism , Phagocytes/immunology , Phagocytes/metabolism , Virus Diseases/etiology , Virus Diseases/metabolism , Animals , Biomarkers , Cells, Cultured , Host-Pathogen Interactions/immunology , Humans , Immunomodulation , Mice
13.
Nat Commun ; 9(1): 3057, 2018 08 03.
Article in English | MEDLINE | ID: mdl-30076298

ABSTRACT

Recent studies indicate that nucleoli play critical roles in the DNA-damage response (DDR) via interaction of DDR machinery including NBS1 with nucleolar Treacle protein, a key mediator of ribosomal RNA (rRNA) transcription and processing. Here, using proteomics, confocal and single molecule super-resolution imaging, and infection under biosafety level-4 containment, we show that this nucleolar DDR pathway is targeted by infectious pathogens. We find that the matrix proteins of Hendra virus and Nipah virus, highly pathogenic viruses of the Henipavirus genus in the order Mononegavirales, interact with Treacle and inhibit its function, thereby silencing rRNA biogenesis, consistent with mimicking NBS1-Treacle interaction during a DDR. Furthermore, inhibition of Treacle expression/function enhances henipavirus production. These data identify a mechanism for viral modulation of host cells by appropriating the nucleolar DDR and represent, to our knowledge, the first direct intranucleolar function for proteins of any mononegavirus.


Subject(s)
Cell Nucleolus/physiology , Cell Nucleolus/virology , DNA Damage/physiology , Hendra Virus/physiology , Nipah Virus/physiology , Cell Cycle Proteins/metabolism , HEK293 Cells , HeLa Cells , Henipavirus/genetics , Henipavirus Infections , Host-Pathogen Interactions/physiology , Humans , Mononegavirales/genetics , Nuclear Proteins/metabolism , Nucleoproteins/metabolism , Proteomics , RNA, Ribosomal/biosynthesis , Viral Proteins/metabolism
14.
J Biol Chem ; 293(27): 10561-10573, 2018 07 06.
Article in English | MEDLINE | ID: mdl-29802199

ABSTRACT

Host recognition of intracellular viral RNA and subsequent induction of cytokine signaling are tightly regulated at the cellular level and are a target for manipulation by viruses and therapeutics alike. Here, we characterize chromosome 6 ORF 106 (C6orf106) as an evolutionarily conserved inhibitor of the innate antiviral response. C6orf106 suppresses the synthesis of interferon (IFN)-α/ß and proinflammatory tumor necrosis factor (TNF) α in response to the dsRNA mimic poly(I:C) and to Sendai virus infection. Unlike canonical inhibitors of antiviral signaling, C6orf106 blocks interferon-regulatory factor 3 (IRF3) and, to a lesser extent, NF-κB activity without modulating their activation, nuclear translocation, cellular expression, or degradation. Instead, C6orf106 interacts with IRF3 and inhibits IRF3 recruitment to type I IFN promoter sequences while also reducing the nuclear levels of the coactivator proteins p300 and CREB-binding protein (CBP). In summary, we have defined C6orf106 as a negative regulator of antiviral immunity that blocks IRF3-dependent cytokine production via a noncanonical and poorly defined mechanism. This work presents intriguing implications for antiviral immunity, autoimmune disorders, and cancer.


Subject(s)
Antiviral Agents/pharmacology , Immunity, Innate/immunology , Interferon Regulatory Factor-3/antagonists & inhibitors , Neoplasm Proteins/pharmacology , Respirovirus Infections/prevention & control , Sendai virus/immunology , Animals , Antiviral Agents/administration & dosage , Chlorocebus aethiops , Gene Expression Regulation , HeLa Cells , Humans , Immunity, Innate/drug effects , Interferon Regulatory Factor-3/genetics , Interferon Regulatory Factor-3/metabolism , NF-kappa B/antagonists & inhibitors , NF-kappa B/genetics , NF-kappa B/metabolism , Neoplasm Proteins/administration & dosage , Respirovirus Infections/immunology , Respirovirus Infections/virology , Sendai virus/drug effects , Signal Transduction , Vero Cells
15.
Curr Top Microbiol Immunol ; 419: 191-213, 2018.
Article in English | MEDLINE | ID: mdl-28674944

ABSTRACT

Hendra and Nipah viruses (family Paramyxoviridae, genus Henipavirus) are zoonotic RNA viruses that cause lethal disease in humans and are designated as Biosafety Level 4 (BSL4) agents. Moreover, henipaviruses belong to the same group of viruses that cause disease more commonly in humans such as measles, mumps and respiratory syncytial virus. Due to the relatively recent emergence of the henipaviruses and the practical constraints of performing functional genomics studies at high levels of containment, our understanding of the henipavirus infection cycle is incomplete. In this chapter we describe recent loss-of-function (i.e. RNAi) functional genomics screens that shed light on the henipavirus-host interface at a genome-wide level. Further to this, we cross-reference RNAi results with studies probing host proteins targeted by henipavirus proteins, such as nuclear proteins and immune modulators. These functional genomics studies join a growing body of evidence demonstrating that nuclear and nucleolar host proteins play a crucial role in henipavirus infection. Furthermore these studies will underpin future efforts to define the role of nucleolar host-virus interactions in infection and disease.


Subject(s)
Genomics , Hendra Virus/immunology , Henipavirus Infections/genetics , Henipavirus Infections/immunology , Host-Pathogen Interactions , MicroRNAs/metabolism , Nipah Virus/immunology , Nuclear Proteins/metabolism , Henipavirus Infections/metabolism , Henipavirus Infections/virology , Humans , MicroRNAs/genetics , Nuclear Proteins/genetics
16.
mBio ; 8(5)2017 10 03.
Article in English | MEDLINE | ID: mdl-28974621

ABSTRACT

Inflammatory responses, while essential for pathogen clearance, can also be deleterious to the host. Chemical inhibition of topoisomerase 1 (Top1) by low-dose camptothecin (CPT) can suppress transcriptional induction of antiviral and inflammatory genes and protect animals from excessive and damaging inflammatory responses. We describe the unexpected finding that minor DNA damage from topoisomerase 1 inhibition with low-dose CPT can trigger a strong antiviral immune response through cyclic GMP-AMP synthase (cGAS) detection of cytoplasmic DNA. This argues against CPT having only anti-inflammatory activity. Furthermore, expression of the simian virus 40 (SV40) large T antigen was paramount to the proinflammatory antiviral activity of CPT, as it potentiated cytoplasmic DNA leakage and subsequent cGAS recruitment in human and mouse cell lines. This work suggests that the capacity of Top1 inhibitors to blunt inflammatory responses can be counteracted by viral oncogenes and that this should be taken into account for their therapeutic development.IMPORTANCE Recent studies suggest that low-dose DNA-damaging compounds traditionally used in cancer therapy can have opposite effects on antiviral responses, either suppressing (with the example of CPT) or potentiating (with the example of doxorubicin) them. Our work demonstrates that the minor DNA damage promoted by low-dose CPT can also trigger strong antiviral responses, dependent on the presence of viral oncogenes. Taken together, these results call for caution in the therapeutic use of low-dose chemotherapy agents to modulate antiviral responses in humans.


Subject(s)
DNA Topoisomerases, Type I/drug effects , Immunity, Innate/drug effects , Nucleotides, Cyclic/metabolism , Simian virus 40/drug effects , Topoisomerase I Inhibitors/pharmacology , Animals , Antigens, Viral, Tumor/genetics , Antigens, Viral, Tumor/immunology , Antiviral Agents/pharmacology , Camptothecin/pharmacology , Cell Line , Coculture Techniques , DNA Damage , DNA Topoisomerases, Type I/metabolism , Fibroblasts/drug effects , Fibroblasts/virology , Humans , Inflammation , Mice , Simian virus 40/immunology , Simian virus 40/physiology , Virus Diseases/drug therapy , Virus Diseases/immunology , Virus Diseases/virology
17.
Sci Rep ; 7(1): 7431, 2017 08 07.
Article in English | MEDLINE | ID: mdl-28785041

ABSTRACT

Hendra virus (HeV) is an emerging zoonotic pathogen harbored by Australian mainland flying foxes. HeV infection can cause lethal disease in humans and horses, and to date all cases of human HeV disease have resulted from contact with infected horses. Currently, diagnosis of acute HeV infections in horses relies on the productive phase of infection when virus shedding may occur. An assay that identifies infected horses during the preclinical phase of infection would reduce the risk of zoonotic viral transmission during management of HeV outbreaks. Having previously shown that the host microRNA (miR)-146a is upregulated in the blood of HeV-infected horses days prior to the detection of viremia, we have profiled miRNAs at the transcriptome-wide level to comprehensively assess differences between infected and uninfected horses. Next-generation sequencing and the miRDeep2 algorithm identified 742 mature miRNA transcripts corresponding to 593 miRNAs in whole blood of six horses (three HeV-infected, three uninfected). Thirty seven miRNAs were differentially expressed in infected horses, two of which were validated by qRT-PCR. This study describes a methodology for the transcriptome-wide profiling of miRNAs in whole blood and supports the notion that measuring host miRNA expression levels may aid infectious disease diagnosis in the future.


Subject(s)
Circulating MicroRNA/genetics , Gene Expression Profiling/veterinary , Henipavirus Infections/veterinary , Horse Diseases/diagnosis , Horses/genetics , Animals , Australia , Circulating MicroRNA/blood , Early Diagnosis , Gene Expression Regulation , Hendra Virus/pathogenicity , Henipavirus Infections/blood , Henipavirus Infections/diagnosis , Henipavirus Infections/genetics , High-Throughput Nucleotide Sequencing/veterinary , Horse Diseases/blood , Horse Diseases/genetics , Horses/blood , Sequence Analysis, RNA/veterinary
18.
J Immunol Methods ; 444: 29-35, 2017 05.
Article in English | MEDLINE | ID: mdl-28216237

ABSTRACT

The ferret is an established animal model for a number of human respiratory viral infections, such as influenza virus and more recently, Ebola virus. However, a paucity of immunological reagents has hampered the study of cellular immune responses. Here we describe the development and characterisation of a novel monoclonal antibody (mAb) against the ferret CD4 antigen and the characterisation of ferret CD4 T lymphocytes. Recombinant production and purification of the ferret CD4 ectodomain soluble protein allowed hybridoma generation and the generation of a mAb (FeCD4) showing strong binding to ferret CD4 protein and lymphoid cells by flow cytometry. FeCD4 bound to its cognate antigen post-fixation with paraformaldehyde (PFA) which is routinely used to inactivate highly pathogenic viruses. We have also used FeCD4 in conjunction with other immune cell markers to characterise ferret T cells in both primary and secondary lymphoid organs. In summary, we have developed an important reagent for the study of cellular immunological responses in the ferret model of infectious disease.


Subject(s)
Antibodies, Monoclonal/immunology , CD4 Antigens/immunology , CD4-Positive T-Lymphocytes/immunology , Ferrets/immunology , Immunity, Cellular , Lymphoid Tissue/immunology , Animals , Antibodies, Monoclonal/metabolism , Antibody Specificity , CD4 Antigens/genetics , CD4 Antigens/metabolism , CD4-Positive T-Lymphocytes/drug effects , CD4-Positive T-Lymphocytes/metabolism , Cell Line , Cell Separation/methods , Concanavalin A/pharmacology , Enzyme-Linked Immunospot Assay , Ferrets/genetics , Ferrets/metabolism , Flow Cytometry , Hybridomas , Interferon-gamma/immunology , Interferon-gamma/metabolism , Lymphocyte Activation , Lymphoid Tissue/cytology , Lymphoid Tissue/metabolism , Phenotype , Protein Binding , Species Specificity , Transfection
19.
J Biol Chem ; 292(3): 826-836, 2017 01 20.
Article in English | MEDLINE | ID: mdl-27913620

ABSTRACT

The emergence of avian H7N9 influenza A virus in humans with associated high mortality has highlighted the threat of a potential pandemic. Fatal H7N9 infections are characterized by hyperinflammation and increased cellular infiltrates in the lung. Currently there are limited therapies to address the pathologies associated with H7N9 infection and the virulence factors that contribute to these pathologies. We have found that PB1-F2 derived from H7N9 activates the NLRP3 inflammasome and induces lung inflammation and cellular recruitment that is NLRP3-dependent. We have also shown that H7N9 and A/Puerto Rico/H1N1 (PR8)PB1-F2 peptide treatment induces significant mitochondrial reactive oxygen production, which contributes to NLRP3 activation. Importantly, treatment of cells or mice with the specific NLRP3 inhibitor MCC950 significantly reduces IL-1ß maturation, lung cellular recruitment, and cytokine production. Together, these results suggest that PB1-F2 from H7N9 avian influenza A virus may be a major contributory factor to disease pathophysiology and excessive inflammation characteristic of clinical infections and that targeting the NLRP3 inflammasome may be an effective means to reduce the inflammatory burden associated with H7N9 infections.


Subject(s)
Influenza A Virus, H7N9 Subtype/immunology , NLR Family, Pyrin Domain-Containing 3 Protein/immunology , Orthomyxoviridae Infections/immunology , Peptides/immunology , Viral Proteins/immunology , Animals , Cell Line, Transformed , Furans , Heterocyclic Compounds, 4 or More Rings/pharmacology , Indenes , Inflammation/immunology , Influenza A Virus, H1N1 Subtype/immunology , Mice , Mitochondria/immunology , NLR Family, Pyrin Domain-Containing 3 Protein/antagonists & inhibitors , Reactive Oxygen Species/immunology , Sulfonamides , Sulfones/pharmacology
20.
PLoS Pathog ; 12(10): e1005974, 2016 Oct.
Article in English | MEDLINE | ID: mdl-27783670

ABSTRACT

Hendra and Nipah viruses (family Paramyxoviridae, genus Henipavirus) are bat-borne viruses that cause fatal disease in humans and a range of other mammalian species. Gaining a deeper understanding of host pathways exploited by henipaviruses for infection may identify targets for new anti-viral therapies. Here we have performed genome-wide high-throughput agonist and antagonist screens at biosafety level 4 to identify host-encoded microRNAs (miRNAs) impacting henipavirus infection in human cells. Members of the miR-181 and miR-17~93 families strongly promoted Hendra virus infection. miR-181 also promoted Nipah virus infection, but did not affect infection by paramyxoviruses from other genera, indicating specificity in the virus-host interaction. Infection promotion was primarily mediated via the ability of miR-181 to significantly enhance henipavirus-induced membrane fusion. Cell signalling receptors of ephrins, namely EphA5 and EphA7, were identified as novel negative regulators of henipavirus fusion. The expression of these receptors, as well as EphB4, were suppressed by miR-181 overexpression, suggesting that simultaneous inhibition of several Ephs by the miRNA contributes to enhanced infection and fusion. Immune-responsive miR-181 levels was also up-regulated in the biofluids of ferrets and horses infected with Hendra virus, suggesting that the host innate immune response may promote henipavirus spread and exacerbate disease severity. This study is the first genome-wide screen of miRNAs influencing infection by a clinically significant mononegavirus and nominates select miRNAs as targets for future anti-viral therapy development.


Subject(s)
Henipavirus Infections/genetics , MicroRNAs/genetics , Virus Internalization , Animals , Ferrets , Fluorescent Antibody Technique , Genome-Wide Association Study , Henipavirus , High-Throughput Nucleotide Sequencing , Horses , Humans , Real-Time Polymerase Chain Reaction
SELECTION OF CITATIONS
SEARCH DETAIL
...