Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 23
Filter
Add more filters










Publication year range
1.
Nat Commun ; 14(1): 60, 2023 01 04.
Article in English | MEDLINE | ID: mdl-36599823

ABSTRACT

Realising the promise of genomics to revolutionise identification and surveillance of antimicrobial resistance (AMR) has been a long-standing challenge in clinical and public health microbiology. Here, we report the creation and validation of abritAMR, an ISO-certified bioinformatics platform for genomics-based bacterial AMR gene detection. The abritAMR platform utilises NCBI's AMRFinderPlus, as well as additional features that classify AMR determinants into antibiotic classes and provide customised reports. We validate abritAMR by comparing with PCR or reference genomes, representing 1500 different bacteria and 415 resistance alleles. In these analyses, abritAMR displays 99.9% accuracy, 97.9% sensitivity and 100% specificity. We also compared genomic predictions of phenotype for 864 Salmonella spp. against agar dilution results, showing 98.9% accuracy. The implementation of abritAMR in our institution has resulted in streamlined bioinformatics and reporting pathways, and has been readily updated and re-verified. The abritAMR tool and validation datasets are publicly available to assist laboratories everywhere harness the power of AMR genomics in professional practice.


Subject(s)
Anti-Bacterial Agents , Drug Resistance, Bacterial , Anti-Bacterial Agents/pharmacology , Workflow , Drug Resistance, Bacterial/genetics , Genomics , Computational Biology , Microbial Sensitivity Tests
2.
Microb Genom ; 6(12)2020 12.
Article in English | MEDLINE | ID: mdl-33180013

ABSTRACT

Complete genomes of microbial pathogens are essential for the phylogenomic analyses that increasingly underpin core public health laboratory activities. Here, we announce a BioProject (PRJNA556438) dedicated to sharing complete genomes chosen to represent a range of pathogenic bacteria with regional importance to Australia and the Southwest Pacific; enriching the catalogue of globally available complete genomes for public health while providing valuable strains to regional public health microbiology laboratories. In this first step, we present 26 complete high-quality bacterial genomes. Additionally, we describe here a framework for reconstructing complete microbial genomes and highlight some of the challenges and considerations for accurate and reproducible genome reconstruction.


Subject(s)
Bacteria/classification , Genome, Bacterial , Whole Genome Sequencing/methods , Australia , Bacteria/genetics , Databases, Genetic , High-Throughput Nucleotide Sequencing , Humans , Phylogeny , Public Health
3.
Antimicrob Agents Chemother ; 64(12)2020 11 17.
Article in English | MEDLINE | ID: mdl-33020158

ABSTRACT

In Australia, cases of shigellosis usually occur in returned travelers from regions of shigellosis endemicity or in men who have sex with men. Resistance to multiple antibiotics has significantly increased in Shigella sonnei isolates and represents a significant public health concern. We investigate an outbreak of multidrug-resistant S. sonnei in Victoria, Australia. We undertook whole-genome sequencing of 54 extended-spectrum-beta-lactamase (ESBL)-producing S. sonnei isolates received at the Microbiological Diagnostic Unit Public Health Laboratory between January 2019 and March 2020. The population structure and antimicrobial resistance profiles were identified by genomic analyses, with 73 previously characterized Australian S. sonnei isolates providing context. Epidemiological data, including age and sex of the shigellosis cases, were also collected. There was a significant increase in cases of ESBL S. sonnei from July 2019. Most of the ESBL S. sonnei isolates (65%) fell within a single cluster that was predominantly comprised of male cases that were characterized by the presence of the blaCTX-M-27 gene conferring resistance to extended-spectrum cephalosporins. These isolates were also multidrug resistant, including resistance to azithromycin and co-trimoxazole and reduced susceptibility to ciprofloxacin. Our data uncovered a prolonged clonal outbreak of ESBL S. sonnei infection that was likely first introduced by returned travelers and has subsequently been circulating locally in Australia. The emergence of a local outbreak of ESBL S. sonnei with a multidrug-resistant profile, including reduced susceptibility to ciprofloxacin, represents a significant public health threat.


Subject(s)
Dysentery, Bacillary , Sexual and Gender Minorities , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/therapeutic use , Disease Outbreaks , Dysentery, Bacillary/drug therapy , Dysentery, Bacillary/epidemiology , Homosexuality, Male , Humans , Male , Microbial Sensitivity Tests , Shigella sonnei/genetics , Victoria/epidemiology , beta-Lactamases/genetics
4.
J Cell Biol ; 216(1): 247-263, 2017 Jan 02.
Article in English | MEDLINE | ID: mdl-27998989

ABSTRACT

Human ciliopathies, including Joubert syndrome (JBTS), arise from cilia dysfunction. The inositol polyphosphate 5-phosphatase INPP5E localizes to cilia and is mutated in JBTS. Murine Inpp5e ablation is embryonically lethal and recapitulates JBTS, including neural tube defects and polydactyly; however, the underlying defects in cilia signaling and the function of INPP5E at cilia are still emerging. We report Inpp5e-/- embryos exhibit aberrant Hedgehog-dependent patterning with reduced Hedgehog signaling. Using mouse genetics, we show increasing Hedgehog signaling via Smoothened M2 expression rescues some Inpp5e-/- ciliopathy phenotypes and "normalizes" Hedgehog signaling. INPP5E's phosphoinositide substrates PI(4,5)P2 and PI(3,4,5)P3 accumulated at the transition zone (TZ) in Hedgehog-stimulated Inpp5e-/- cells, which was associated with reduced recruitment of TZ scaffolding proteins and reduced Smoothened levels at cilia. Expression of wild-type, but not 5-phosphatase-dead, INPP5E restored TZ molecular organization and Smoothened accumulation at cilia. Therefore, we identify INPP5E as an essential point of convergence between Hedgehog and phosphoinositide signaling at cilia that maintains TZ function and Hedgehog-dependent embryonic development.


Subject(s)
Abnormalities, Multiple/enzymology , Cerebellum/abnormalities , Cilia/enzymology , Embryo, Mammalian/enzymology , Eye Abnormalities/enzymology , Kidney Diseases, Cystic/enzymology , Phosphatidylinositol 4,5-Diphosphate/metabolism , Phosphatidylinositol Phosphates/metabolism , Phosphoric Monoester Hydrolases/metabolism , Retina/abnormalities , Retinal Pigment Epithelium/enzymology , Second Messenger Systems , Abnormalities, Multiple/genetics , Animals , Cell Line , Cerebellum/enzymology , Disease Models, Animal , Eye Abnormalities/genetics , Gene Expression Regulation, Developmental , Genetic Predisposition to Disease , Hedgehog Proteins/genetics , Hedgehog Proteins/metabolism , Humans , Kidney Diseases, Cystic/genetics , Kruppel-Like Transcription Factors/genetics , Kruppel-Like Transcription Factors/metabolism , Mice, Inbred C57BL , Mice, Knockout , Phenotype , Phosphoric Monoester Hydrolases/deficiency , Phosphoric Monoester Hydrolases/genetics , Retina/enzymology , Smoothened Receptor/genetics , Smoothened Receptor/metabolism , Time Factors , Transfection , Zinc Finger Protein Gli2
5.
J Immunol ; 194(4): 1819-31, 2015 Feb 15.
Article in English | MEDLINE | ID: mdl-25595793

ABSTRACT

Herpesviruses are DNA viruses harboring the capacity to establish lifelong latent-recurrent infections. There is limited knowledge about viruses targeting the innate DNA-sensing pathway, as well as how the innate system impacts on the latent reservoir of herpesvirus infections. In this article, we report that murine gammaherpesvirus 68 (MHV68), in contrast to α- and ß-herpesviruses, induces very limited innate immune responses through DNA-stimulated pathways, which correspondingly played only a minor role in the control of MHV68 infections in vivo. Similarly, Kaposi's sarcoma-associated herpesvirus also did not stimulate immune signaling through the DNA-sensing pathways. Interestingly, an MHV68 mutant lacking deubiquitinase (DUB) activity, embedded within the large tegument protein open reading frame (ORF)64, gained the capacity to stimulate the DNA-activated stimulator of IFN genes (STING) pathway. We found that ORF64 targeted a step in the DNA-activated pathways upstream of the bifurcation into the STING and absent in melanoma 2 pathways, and lack of the ORF64 DUB was associated with impaired delivery of viral DNA to the nucleus, which, instead, localized to the cytoplasm. Correspondingly, the ORF64 DUB active site mutant virus exhibited impaired ability to establish latent infection in wild-type, but not STING-deficient, mice. Thus, gammaherpesviruses evade immune activation by the cytosolic DNA-sensing pathway, which, in the MHV68 model, facilitates establishment of infections.


Subject(s)
DNA, Viral/immunology , Gammaherpesvirinae/immunology , Herpesviridae Infections/immunology , Immunity, Innate/immunology , Virus Latency/immunology , Animals , Cytosol/immunology , Disease Models, Animal , Enzyme-Linked Immunosorbent Assay , Flow Cytometry , Humans , Macrophages/immunology , Macrophages/virology , Mice , Mice, Inbred C57BL , Mice, Knockout , Microscopy, Confocal , Real-Time Polymerase Chain Reaction
6.
EMBO J ; 33(15): 1654-66, 2014 Aug 01.
Article in English | MEDLINE | ID: mdl-24970844

ABSTRACT

Listeria monocytogenes is a gram-positive facultative intracellular bacterium, which replicates in the cytoplasm of myeloid cells. Interferon ß (IFNß) has been reported to play an important role in the mechanisms underlying Listeria disease. Although studies in murine cells have proposed the bacteria-derived cyclic-di-AMP to be the key bacterial immunostimulatory molecule, the mechanism for IFNß expression during L. monocytogenes infection in human myeloid cells remains unknown. Here we report that in human macrophages, Listeria DNA rather than cyclic-di-AMP is stimulating the IFN response via a pathway dependent on the DNA sensors IFI16 and cGAS as well as the signalling adaptor molecule STING. Thus, Listeria DNA is a major trigger of IFNß expression in human myeloid cells and is sensed to activate a pathway dependent on IFI16, cGAS and STING.


Subject(s)
Host-Pathogen Interactions , Interferon-beta/metabolism , Listeria monocytogenes/pathogenicity , Membrane Proteins/metabolism , Nuclear Proteins/metabolism , Nucleotidyltransferases/metabolism , Phosphoproteins/metabolism , Animals , Cells, Cultured , Cytosol/metabolism , DNA, Bacterial/metabolism , Gene Knockdown Techniques , Humans , Listeria monocytogenes/genetics , Listeria monocytogenes/metabolism , Listeriosis/metabolism , Listeriosis/microbiology , Macrophages/metabolism , Macrophages/microbiology , Membrane Proteins/genetics , Mice , Mice, Inbred C57BL , Nuclear Proteins/genetics , Nucleotidyltransferases/genetics , Phosphoproteins/genetics , Protein Serine-Threonine Kinases/metabolism , Signal Transduction
7.
Biosci Rep ; 34(2)2014 04 01.
Article in English | MEDLINE | ID: mdl-27919038

ABSTRACT

Oncogenic mutations in PIK3CA lead to an increase in intrinsic phosphoinositide kinase activity, but it is thought that increased access of PI3Kα (phosphoinositide 3-kinase α) to its PM (plasma membrane) localized substrate is also required for increased levels of downstream PIP3/Akt [phosphoinositide-3,4,5-trisphosphate/also called PKB (protein kinase B)] signalling. We have studied the subcellular localization of wild-type and the two most common oncogenic mutants of PI3Kα in cells maintained in growth media, and starved or stimulated cells using a novel method in which PI3Kα is pre-formed as a 1:1 p110α:p85α complex in vitro then introduced into live cells by microinjection. Oncogenic E545K and H1047R mutants did not constitutively interact with membrane lipids in vitro or in cells maintained in 10% (v/v) FBS. Following stimulation of RTKs (receptor tyrosine kinases), microinjected PI3Kα was recruited to the PM, but oncogenic forms of PI3Kα were not recruited to the PM to a greater extent and did not reside at the PM longer than the wild-type PI3Kα. Instead, the E545K mutant specifically bound activated Cdc42 in vitro and microinjection of E545K was associated with the formation of cellular protrusions, providing some preliminary evidence that changes in protein-protein interactions may play a role in the oncogenicity of the E545K mutant in addition to the well-known changes in lipid kinase activity.


Subject(s)
Class I Phosphatidylinositol 3-Kinases/metabolism , Mutation, Missense , Proto-Oncogene Proteins c-akt/metabolism , cdc42 GTP-Binding Protein/metabolism , Amino Acid Substitution , Animals , Cell Membrane/enzymology , Cell Membrane/genetics , Class I Phosphatidylinositol 3-Kinases/genetics , Dogs , Humans , Madin Darby Canine Kidney Cells , Membrane Lipids/genetics , Membrane Lipids/metabolism , Protein Transport/genetics , Proto-Oncogene Proteins c-akt/genetics , cdc42 GTP-Binding Protein/genetics
8.
J Immunol ; 190(5): 2311-9, 2013 Mar 01.
Article in English | MEDLINE | ID: mdl-23345332

ABSTRACT

The innate immune system is important for control of infections, including herpesvirus infections. Intracellular DNA potently stimulates antiviral IFN responses. It is known that plasmacytoid dendritic cells sense herpesvirus DNA in endosomes via TLR9 and that nonimmune tissue cells can sense herpesvirus DNA in the nucleus. However, it remains unknown how and where myeloid cells, such as macrophages and conventional dendritic cells, detect infections with herpesviruses. In this study, we demonstrate that the HSV-1 capsid was ubiquitinated in the cytosol and degraded by the proteasome, hence releasing genomic DNA into the cytoplasm for detection by DNA sensors. In this context, the DNA sensor IFN-γ-inducible 16 is important for induction of IFN-ß in human macrophages postinfection with HSV-1 and CMV. Viral DNA localized to the same cytoplasmic regions as did IFN-γ-inducible 16, with DNA sensing being independent of viral nuclear entry. Thus, proteasomal degradation of herpesvirus capsids releases DNA to the cytoplasm for recognition by DNA sensors.


Subject(s)
Capsid/metabolism , Cytomegalovirus/metabolism , DNA, Viral/genetics , Herpesvirus 1, Human/metabolism , Macrophages/metabolism , Nuclear Proteins/metabolism , Phosphoproteins/metabolism , Proteasome Endopeptidase Complex/metabolism , Animals , Cell Line , Cell Nucleus/metabolism , Chlorocebus aethiops , Cytomegalovirus/genetics , Cytosol/metabolism , DNA, Viral/immunology , Dendritic Cells/metabolism , Dendritic Cells/virology , Gene Silencing , Herpesvirus 1, Human/genetics , Humans , Interferon-beta/biosynthesis , Interferon-beta/immunology , Macrophages/virology , Nuclear Proteins/antagonists & inhibitors , Nuclear Proteins/immunology , Phosphoproteins/antagonists & inhibitors , Phosphoproteins/immunology , RNA, Small Interfering/genetics , Ubiquitination , Vero Cells
9.
Nat Immunol ; 13(8): 737-43, 2012 Jun 17.
Article in English | MEDLINE | ID: mdl-22706339

ABSTRACT

The innate immune system senses infection by detecting either evolutionarily conserved molecules essential for the survival of microbes or the abnormal location of molecules. Here we demonstrate the existence of a previously unknown innate detection mechanism induced by fusion between viral envelopes and target cells. Virus-cell fusion specifically stimulated a type I interferon response with expression of interferon-stimulated genes, in vivo recruitment of leukocytes and potentiation of signaling via Toll-like receptor 7 (TLR7) and TLR9. The fusion-dependent response was dependent on the stimulator of interferon genes STING but was independent of DNA, RNA and viral capsid. We suggest that membrane fusion is sensed as a danger signal with potential implications for defense against enveloped viruses and various conditions of giant-cell formation.


Subject(s)
Cell Fusion , Herpesvirus 1, Human/immunology , Herpesvirus 1, Human/physiology , Immunity, Innate , Interferon Type I/biosynthesis , Membrane Fusion , Membrane Proteins/metabolism , Animals , Chemokine CXCL10/metabolism , HEK293 Cells , HeLa Cells , Humans , Leukocytes/immunology , Leukocytes/metabolism , Lymphocyte Activation , Macrophages/metabolism , Membrane Glycoproteins/metabolism , Mice , Mice, Knockout , Myeloid Differentiation Factor 88/genetics , Myeloid Differentiation Factor 88/metabolism , Signal Transduction , Toll-Like Receptor 7/metabolism , Toll-Like Receptor 9/metabolism , Virus Internalization
10.
J Clin Invest ; 122(4): 1368-76, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22426207

ABSTRACT

Herpes simplex viruses (HSVs) are highly prevalent neurotropic viruses. While they can replicate lytically in cells of the epithelial lineage, causing lesions on mucocutaneous surfaces, HSVs also establish latent infections in neurons, which act as reservoirs of virus for subsequent reactivation events. Immunological control of HSV involves activation of innate immune pattern-recognition receptors such as TLR3, which detects double-stranded RNA and induces type I IFN expression. Humans with defects in the TLR3/IFN pathway have an elevated susceptibility to HSV infections of the CNS. However, it is not known what cell type mediates the role of TLR3 in the immunological control of HSV, and it is not known whether TLR3 sensing occurs prior to or after CNS entry. Here, we show that in mice TLR3 provides early control of HSV-2 infection immediately after entry into the CNS by mediating type I IFN responses in astrocytes. Tlr3-/- mice were hypersusceptible to HSV-2 infection in the CNS after vaginal inoculation. HSV-2 exhibited broader neurotropism in Tlr3-/- mice than it did in WT mice, with astrocytes being most abundantly infected. Tlr3-/- mice did not exhibit a global defect in innate immune responses to HSV, but astrocytes were defective in HSV-induced type I IFN production. Thus, TLR3 acts in astrocytes to sense HSV-2 infection immediately after entry into the CNS, possibly preventing HSV from spreading beyond the neurons mediating entry into the CNS.


Subject(s)
Astrocytes/virology , Herpes Simplex/virology , Herpesvirus 2, Human/physiology , Myelitis/virology , Toll-Like Receptor 3/deficiency , Viral Tropism/physiology , Animals , Astrocytes/metabolism , Cerebellum/virology , Disease Susceptibility , Female , Herpes Simplex/complications , Herpes Simplex/immunology , Herpes Simplex/metabolism , Herpesvirus 2, Human/immunology , Immunity, Innate , Immunity, Mucosal , Interferon-beta/biosynthesis , Mice , Mice, Inbred C57BL , Mice, Knockout , Myelitis/complications , Myelitis/immunology , Myelitis/metabolism , Neurons/virology , Organ Specificity , Paraplegia/etiology , Receptors, Interferon/deficiency , Receptors, Interferon/genetics , Spinal Cord/virology , Toll-Like Receptor 3/genetics , Toll-Like Receptor 3/physiology , Urinary Retention/etiology , Vagina/immunology , Vagina/innervation , Vagina/virology , Virus Activation , Virus Latency , Interferon gamma Receptor
11.
Biochem Biophys Res Commun ; 418(4): 806-10, 2012 Feb 24.
Article in English | MEDLINE | ID: mdl-22310710

ABSTRACT

Reactive oxygen species (ROS) are crucial secondary messengers of signaling pathways. Redox-dependent signaling events have been previously described in the innate immune response. However, the mechanism by which ROS modulates anti-viral innate immune signaling is not fully clarified. Here, we report that mitochondria-derived ROS differentially regulate the innate response to DNA and RNA viruses (herpes simplex virus (HSV) and Sendai virus (SeV), respectively), with the cytokine response to HSV being negatively regulated by mitochondrial ROS. Importantly, specific activation of Toll-like receptors (TLRs) and DNA receptors (DNARs) but not retinoic acid inducible gene I (RIG-I)-like receptors (RLRs), led to signaling cascades that were inhibited by mitochondrial ROS production. Thus, localized mitochondrial ROS exerts negative modulation of innate immune responses to the DNA virus HSV-2 but not the RNA virus SeV.


Subject(s)
Herpesvirus 2, Human/immunology , Immunity, Innate , Mitochondria/metabolism , Reactive Oxygen Species/metabolism , Sendai virus/immunology , Animals , Cytokines/biosynthesis , DEAD Box Protein 58 , DEAD-box RNA Helicases/metabolism , DNA, Viral/immunology , Interferon Regulatory Factor-3/metabolism , Mice , Mice, Inbred C57BL , NF-kappa B/metabolism , RNA, Viral/immunology , Receptors, Cell Surface/metabolism , Signal Transduction
12.
PLoS One ; 7(1): e29291, 2012.
Article in English | MEDLINE | ID: mdl-22235281

ABSTRACT

BACKGROUND: Innate immune responses have recently been appreciated to play an important role in the pathogenesis of HIV infection. Whereas inadequate innate immune sensing of HIV during acute infection may contribute to failure to control and eradicate infection, persistent inflammatory responses later during infection contribute in driving chronic immune activation and development of immunodeficiency. However, knowledge on specific HIV PAMPs and cellular PRRs responsible for inducing innate immune responses remains sparse. METHODS/PRINCIPAL FINDINGS: Here we demonstrate a major role for RIG-I and the adaptor protein MAVS in induction of innate immune responses to HIV genomic RNA. We found that secondary structured HIV-derived RNAs induced a response similar to genomic RNA. In primary human peripheral blood mononuclear cells and primary human macrophages, HIV RNA induced expression of IFN-stimulated genes, whereas only low levels of type I IFN and tumor necrosis factor α were produced. Furthermore, secondary structured HIV-derived RNA activated pathways to NF-κB, MAP kinases, and IRF3 and co-localized with peroxisomes, suggesting a role for this organelle in RIG-I-mediated innate immune sensing of HIV RNA. CONCLUSIONS/SIGNIFICANCE: These results establish RIG-I as an innate immune sensor of cytosolic HIV genomic RNA with secondary structure, thereby expanding current knowledge on HIV molecules capable of stimulating the innate immune system.


Subject(s)
DEAD-box RNA Helicases/metabolism , Genome, Viral/immunology , HIV-1/genetics , Immunity, Innate , Nucleic Acid Conformation , RNA, Viral/chemistry , RNA, Viral/metabolism , Adaptor Proteins, Signal Transducing/metabolism , Animals , Cell Line, Tumor , DEAD Box Protein 58 , HIV-1/immunology , HIV-1/metabolism , Humans , Interferon Regulatory Factors/metabolism , Leukocytes, Mononuclear/cytology , Leukocytes, Mononuclear/immunology , Leukocytes, Mononuclear/metabolism , Leukocytes, Mononuclear/virology , Mice , Mice, Inbred C57BL , NF-kappa B/metabolism , Oligoribonucleotides/chemistry , Oligoribonucleotides/metabolism , Peroxisomes/metabolism , Peroxisomes/virology , Protein Transport , Receptors, Immunologic , Signal Transduction/immunology , Viral Proteins/metabolism , p38 Mitogen-Activated Protein Kinases/metabolism
13.
J Immunol ; 187(10): 5268-76, 2011 Nov 15.
Article in English | MEDLINE | ID: mdl-21998456

ABSTRACT

Autophagy has been established as a player in host defense against viruses. The mechanisms by which the host induces autophagy during infection are diverse. In the case of HSV type 1 (HSV-1), dsRNA-dependent protein kinase is essential for induction of autophagy in fibroblasts through phosphorylation of eukaryotic initiation factor 2α (eIF2α). HSV-1 counteracts autophagy via ICP34.5, which dephosphorylates eIF2α and inhibits Beclin 1. Investigation of autophagy during HSV-1 infection has largely been conducted in permissive cells, but recent work suggests the existence of a eIF2α-independent autophagy-inducing pathway in nonpermissive cells. To clarify and further characterize the existence of a novel autophagy-inducing pathway in nonpermissive cells, we examined different HSV and cellular components in murine myeloid cells for their role in autophagy. We demonstrate that HSV-1-induced autophagy does not correlate with phosphorylation of eIF2α, is independent of functional dsRNA-dependent protein kinase, and is not antagonized by ICP34.5. Autophagy was activated independent of viral gene expression, but required viral entry. Importantly, we found that the presence of genomic DNA in the virion was essential for induction of autophagy and, conversely, that transfection of HSV-derived DNA induced microtubule-associated protein 1 L chain II formation, a marker of autophagy. This occurred through a mechanism dependent on stimulator of IFN genes, an essential component for the IFN response to intracellular DNA. Finally, we observed that HSV-1 DNA was present in the cytosol devoid of capsid material following HSV-1 infection of dendritic cells. Thus, our data suggest that HSV-1 genomic DNA induces autophagy in nonpermissive cells in a stimulator of IFN gene-dependent manner.


Subject(s)
Autophagy/immunology , Cytosol/virology , DNA, Viral , Herpesvirus 1, Human/immunology , Membrane Proteins/physiology , Myeloid Cells/immunology , Myeloid Cells/virology , Animals , Autophagy/genetics , Bone Marrow Cells/immunology , Bone Marrow Cells/virology , Cell Line , Cytosol/immunology , DNA, Viral/genetics , Dendritic Cells/immunology , Dendritic Cells/virology , Female , Membrane Proteins/deficiency , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Mutant Strains , Myeloid Cells/cytology
14.
PLoS Pathog ; 7(9): e1002250, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21949653

ABSTRACT

The innate immune response constitutes the first line of defense against infections. Pattern recognition receptors recognize pathogen structures and trigger intracellular signaling pathways leading to cytokine and chemokine expression. Reactive oxygen species (ROS) are emerging as an important regulator of some of these pathways. ROS directly interact with signaling components or induce other post-translational modifications such as S-glutathionylation, thereby altering target function. Applying live microscopy, we have demonstrated that herpes simplex virus (HSV) infection induces early production of ROS that are required for the activation of NF-κB and IRF-3 pathways and the production of type I IFNs and ISGs. All the known receptors involved in the recognition of HSV were shown to be dependent on the cellular redox levels for successful signaling. In addition, we provide biochemical evidence suggesting S-glutathionylation of TRAF family proteins to be important. In particular, by performing mutational studies we show that S-glutathionylation of a conserved cysteine residue of TRAF3 and TRAF6 is important for ROS-dependent activation of innate immune pathways. In conclusion, these findings demonstrate that ROS are essential for effective activation of signaling pathways leading to a successful innate immune response against HSV infection.


Subject(s)
Herpes Simplex/immunology , Herpes Simplex/metabolism , Immunity, Innate , Reactive Oxygen Species/metabolism , Receptors, Pattern Recognition/metabolism , Simplexvirus/immunology , TNF Receptor-Associated Factor 3/metabolism , Animals , Chemokines/metabolism , Cytokines/metabolism , Glutathione/metabolism , Interferon Regulatory Factor-3/metabolism , Interferon Type I/metabolism , Mice , Mice, Inbred C57BL , NF-kappa B/metabolism , RNA Interference , RNA, Small Interfering , Receptors, Pattern Recognition/genetics , Receptors, Pattern Recognition/immunology , Signal Transduction , Simplexvirus/pathogenicity , TNF Receptor-Associated Factor 3/genetics , TNF Receptor-Associated Factor 6/genetics , TNF Receptor-Associated Factor 6/metabolism
15.
Nat Rev Immunol ; 11(2): 143-54, 2011 02.
Article in English | MEDLINE | ID: mdl-21267015

ABSTRACT

Advances in innate immunity over the past decade have revealed distinct classes of pattern recognition receptors (PRRs) that detect pathogens at the cell surface and in intracellular compartments. This has shed light on how herpesviruses, which are large disease-causing DNA viruses that replicate in the nucleus, are initially recognized during cellular infection. Surprisingly, this involves multiple PRRs both on the cell surface and within endosomes and the cytosol. In this article we describe recent advances in our understanding of innate detection of herpesviruses, how this innate detection translates into anti-herpesvirus host defence, and how the viruses seek to evade this innate detection to establish persistent infections.


Subject(s)
Herpesviridae Infections/immunology , Herpesviridae/immunology , Host-Pathogen Interactions/immunology , Immunity, Innate , Receptors, Pattern Recognition/immunology , DNA, Viral/immunology , Humans , RNA, Viral/immunology , Signal Transduction , Toll-Like Receptors/immunology
16.
Nat Immunol ; 11(11): 997-1004, 2010 Nov.
Article in English | MEDLINE | ID: mdl-20890285

ABSTRACT

The detection of intracellular microbial DNA is critical to appropriate innate immune responses; however, knowledge of how such DNA is sensed is limited. Here we identify IFI16, a PYHIN protein, as an intracellular DNA sensor that mediates the induction of interferon-ß (IFN-ß). IFI16 directly associated with IFN-ß-inducing viral DNA motifs. STING, a critical mediator of IFN-ß responses to DNA, was recruited to IFI16 after DNA stimulation. Lowering the expression of IFI16 or its mouse ortholog p204 by RNA-mediated interference inhibited gene induction and activation of the transcription factors IRF3 and NF-κB induced by DNA and herpes simplex virus type 1 (HSV-1). IFI16 (p204) is the first PYHIN protein to our knowledge shown to be involved in IFN-ß induction. Thus, the PYHIN proteins IFI16 and AIM2 form a new family of innate DNA sensors we call 'AIM2-like receptors' (ALRs).


Subject(s)
DNA, Viral/immunology , Immunity, Innate , Intracellular Space/immunology , Nuclear Proteins/immunology , Phosphoproteins/immunology , Animals , Cell Line , DNA-Binding Proteins , Herpesvirus 1, Human/immunology , Humans , Interferon-beta/immunology , Interferon-beta/metabolism , Membrane Proteins/immunology , Mice , Monocytes/immunology , Signal Transduction
17.
J Virol ; 84(22): 11898-904, 2010 Nov.
Article in English | MEDLINE | ID: mdl-20844035

ABSTRACT

The 2'-5' oligoadenylate synthetase (OAS) proteins are traditionally considered intracellular antiviral proteins. However, several studies demonstrate a correlation between the concentration of freely circulating OAS protein in sera from hepatitis C patients and their clinical prognosis. Here we demonstrate that extracellular OAS1 enters into cells and possesses a strong antiviral activity, both in vitro and in vivo, which is independent of RNase L. The OAS protein directly inhibits viral proliferation and does not require the activation of known antiviral signaling pathways. We propose that OAS produced by cells infected with viruses is released to the extracellular space, where it acts as a paracrine antiviral agent. Thus, the OAS protein represents the first direct antiviral compound released by virus-infected cells.


Subject(s)
2',5'-Oligoadenylate Synthetase/immunology , Antiviral Agents/immunology , Endoribonucleases/immunology , Extracellular Space/enzymology , Host-Pathogen Interactions , Virus Diseases/enzymology , Virus Diseases/immunology , Viruses/immunology , 2',5'-Oligoadenylate Synthetase/genetics , Animals , Cell Line , Endoribonucleases/genetics , Extracellular Space/immunology , Humans , Immunity, Innate , Mice , Mice, Inbred C57BL , Virus Diseases/virology , Virus Physiological Phenomena
18.
J Virol ; 84(21): 11350-8, 2010 Nov.
Article in English | MEDLINE | ID: mdl-20739519

ABSTRACT

Innate recognition of viruses is mediated by pattern recognition receptors (PRRs) triggering expression of antiviral interferons (IFNs) and proinflammatory cytokines. In mice, Toll-like receptor 2 (TLR2) and TLR9 as well as intracellular nucleotide-sensing pathways have been shown to recognize herpes simplex virus (HSV). Here, we describe how human primary macrophages recognize early HSV infection via intracellular pathways. A number of inflammatory cytokines, IFNs, and IFN-stimulated genes were upregulated after HSV infection. We show that early recognition of HSV and induction of IFNs and inflammatory cytokines are independent of TLR2 and TLR9, since inhibition of TLR2 using TLR2 neutralizing antibodies did not affect virus-induced responses and the macrophages were unresponsive to TLR9 stimulation. Instead, HSV recognition involves intracellular recognition systems, since induction of tumor necrosis factor alpha (TNF-α) and IFNs was dependent on virus entry and replication. Importantly, expression of IFNs was strongly inhibited by small interfering RNA (siRNA) knockdown of MAVS, but this MAVS-dependent IFN induction occurred independently of the recently discovered polymerase III (Pol III)/RIG-I DNA sensing system. In contrast, induction of TNF-α was largely independent of MAVS, suggesting that induction of inflammatory cytokines during HSV infection proceeds via a novel pathway. Transfection with ODN2006, a broad inhibitor of intracellular nucleotide recognition, revealed that nucleotide-sensing systems are employed to induce both IFNs and TNF-α. Finally, using siRNA knockdown, we found that MDA5, but not RIG-I, was the primary mediator of HSV recognition. Thus, innate recognition of HSV by human primary macrophages occurs via two distinct intracellular nucleotide-sensing pathways responsible for induction of IFNs and inflammatory cytokine expression, respectively.


Subject(s)
DEAD-box RNA Helicases/immunology , Immunity, Innate , Macrophages/virology , Simplexvirus/immunology , Adaptor Proteins, Signal Transducing/metabolism , Cells, Cultured , Cytokines/biosynthesis , DEAD Box Protein 58 , DEAD-box RNA Helicases/metabolism , Humans , Interferon-Induced Helicase, IFIH1 , Interferons/biosynthesis , RNA Polymerase III/metabolism , Receptors, Immunologic , Signal Transduction/immunology
19.
Biochem J ; 419(1): 29-49, 2009 Apr 01.
Article in English | MEDLINE | ID: mdl-19272022

ABSTRACT

Phosphoinositides are membrane-bound signalling molecules that regulate cell proliferation and survival, cytoskeletal reorganization and vesicular trafficking by recruiting effector proteins to cellular membranes. Growth factor or insulin stimulation induces a canonical cascade resulting in the transient phosphorylation of PtdIns(4,5)P(2) by PI3K (phosphoinositide 3-kinase) to form PtdIns(3,4,5)P(3), which is rapidly dephosphorylated either by PTEN (phosphatase and tensin homologue deleted on chromosome 10) back to PtdIns(4,5)P(2), or by the 5-ptases (inositol polyphosphate 5-phosphatases), generating PtdIns(3,4)P(2). The 5-ptases also hydrolyse PtdIns(4,5)P(2), forming PtdIns4P. Ten mammalian 5-ptases have been identified, which share a catalytic mechanism similar to that of the apurinic/apyrimidinic endonucleases. Gene-targeted deletion of 5-ptases in mice has revealed that these enzymes regulate haemopoietic cell proliferation, synaptic vesicle recycling, insulin signalling, endocytosis, vesicular trafficking and actin polymerization. Several studies have revealed that the molecular basis of Lowe's syndrome is due to mutations in the 5-ptase OCRL (oculocerebrorenal syndrome of Lowe). Futhermore, the 5-ptases SHIP [SH2 (Src homology 2)-domain-containing inositol phosphatase] 2, SKIP (skeletal muscle- and kidney-enriched inositol phosphatase) and 72-5ptase (72 kDa 5-ptase)/Type IV/Inpp5e (inositol polyphosphate 5-phosphatase E) are implicated in negatively regulating insulin signalling and glucose homoeostasis in specific tissues. SHIP2 polymorphisms are associated with a predisposition to insulin resistance. Gene profiling studies have identified changes in the expression of various 5-ptases in specific cancers. In addition, 5-ptases such as SHIP1, SHIP2 and 72-5ptase/Type IV/Inpp5e regulate macrophage phagocytosis, and SHIP1 also controls haemopoietic cell proliferation. Therefore the 5-ptases are a significant family of signal-modulating enzymes that govern a plethora of cellular functions by regulating the levels of specific phosphoinositides. Emerging studies have implicated their loss or gain of function in human disease.


Subject(s)
Phosphoric Monoester Hydrolases/genetics , Phosphoric Monoester Hydrolases/metabolism , Diabetes Mellitus/genetics , Diabetes Mellitus/metabolism , Humans , Inositol Polyphosphate 5-Phosphatases , Neoplasms/genetics , Neoplasms/metabolism , Oculocerebrorenal Syndrome/genetics , Oculocerebrorenal Syndrome/metabolism , Phosphatidylinositol-3,4,5-Trisphosphate 5-Phosphatases , Phosphatidylinositols/metabolism , Phosphoric Monoester Hydrolases/physiology
20.
EMBO Rep ; 10(5): 487-93, 2009 May.
Article in English | MEDLINE | ID: mdl-19325558

ABSTRACT

Akt is a crucial phosphoinositide 3-kinase (PI(3)K) effector that regulates cell proliferation and survival. PI(3)K-generated signals, PtdIns(3,4,5)P(3) and PtdIns(3,4)P(2), direct Akt plasma membrane engagement. Pathological Akt plasma membrane association promotes oncogenesis. PtdIns(3,4)P(2) is degraded by inositol polyphosphate 4-phosphatase-1 (4-ptase-1) forming PtdIns(3)P; however, the role of 4-ptase-1 in regulating the activation and function of Akt is unclear. In mouse embryonic fibroblasts lacking 4-ptase-1 ((-/-)MEFs), the Akt-pleckstrin homology (PH) domain was constitutively membrane-associated both in serum-starved and agonist-stimulated cells, in contrast to (+/+)MEFs, in which it was detected only at the plasma membrane following serum stimulation. Epidermal growth factor (EGF) stimulation resulted in increased Ser(473) and Thr(308)-Akt phosphorylation and activation of Akt-dependent signalling in (-/-)MEFs, relative to (+/+)MEFs. Significantly, loss of 4-ptase-1 resulted in increased cell proliferation and decreased apoptosis. SV40-transformed (-/-)MEFs showed increased anchorage-independent cell growth and formed tumours in nude mice. This study provides the first evidence, to our knowledge, that 4-ptase-1 controls the activation of Akt and thereby cell proliferation, survival and tumorigenesis.


Subject(s)
Phosphatidylinositol 3-Kinases/metabolism , Phosphoric Monoester Hydrolases/physiology , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction/physiology , Animals , Apoptosis/drug effects , Cell Proliferation/drug effects , Cells, Cultured , Cycloheximide/pharmacology , Epidermal Growth Factor/pharmacology , Fibroblasts , Mice , Phosphoric Monoester Hydrolases/genetics , Signal Transduction/drug effects , Signal Transduction/genetics , Staurosporine/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...