Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
1.
Euro Surveill ; 28(39)2023 09.
Article in English | MEDLINE | ID: mdl-37768562

ABSTRACT

BackgroundNeisseria meningitidis is a commensal bacterium which can cause invasive disease. Colonisation studies are important to guide vaccination strategies.AimThe study's aim was to determine the prevalence of meningococcal colonisation, duration of carriage and distribution of genogroups in Iceland.MethodsWe collected samples from 1 to 6-year-old children, 15-16-year-old adolescents and 18-20-year-old young adults. Carriers were sampled at regular intervals until the first negative swab. Conventional culture methods and qPCR were applied to detect meningococci and determine the genogroup. Whole genome sequencing was done on groupable meningococci.ResultsNo meningococci were detected among 460 children, while one of 197 (0.5%) adolescents and 34 of 525 young adults (6.5 %) carried meningococci. Non-groupable meningococci were most common (62/77 isolates from 26/35 carriers), followed by genogroup B (MenB) (12/77 isolates from 6/35 carriers). Genogroup Y was detected in two individuals and genogroup W in one. None carried genogroup C (MenC). The longest duration of carriage was at least 21 months. Serial samples from persistent carriers were closely related in WGS.ConclusionsCarriage of pathogenic meningococci is rare in young Icelanders. Non-groupable meningococci were the most common colonising meningococci in Iceland, followed by MenB. No MenC were found. Whole genome sequencing suggests prolonged carriage of the same strains in persistent carriers.


Subject(s)
Neisseria meningitidis , Adolescent , Humans , Child , Young Adult , Longitudinal Studies , Cross-Sectional Studies , Iceland/epidemiology , Genotype , Neisseria meningitidis/genetics
2.
Vaccine ; 41(34): 4927-4932, 2023 07 31.
Article in English | MEDLINE | ID: mdl-37423800

ABSTRACT

Carriage of Neisseria meningitidisis an accepted endpoint in monitoring meningococcal vaccine effects. We applied molecular methods to assess the impact of menACWY vaccine implementation on meningococcal carriage and genogroup-specific prevalence in young adults in Fall of 2022, four years after the introduction of the tetravalent vaccine in the Netherlands. The overall carriage rate of genogroupable meningococci was not significantly different compared to a pre-menACWY cohort investigated in 2018 (20.8 % or 125 of 601 versus 17.4 % or 52 of 299 individuals, p = 0.25). Of 125 carriers of genogroupable meningococci, 122 (97.6 %) were positive for either vaccine-types menC, menW, menY or genogroups, menB, menE, and menX, which are not targeted by the menACWY vaccine. Compared with a pre-vaccine-implementation cohort, there was 3.8-fold reduction (p < 0.001) in vaccine-type carriage rates and 9.0-fold increase (p < 0.0001) in non-vaccine type menE prevalence. We observe a reduction in menW and menY and an increase in menE, which suggest that implementation of menACWY vaccine affected carriage.


Subject(s)
Meningococcal Infections , Meningococcal Vaccines , Neisseria meningitidis , Young Adult , Humans , Neisseria meningitidis/genetics , Netherlands/epidemiology , Meningococcal Infections/epidemiology , Meningococcal Infections/prevention & control , Genotype , Vaccines, Combined
3.
PLoS Pathog ; 16(7): e1008702, 2020 07.
Article in English | MEDLINE | ID: mdl-32667958

ABSTRACT

The type I interferon response is an important innate antiviral pathway. Recognition of viral RNA by RIG-I-like receptors (RLRs) activates a signaling cascade that leads to type I interferon (IFN-α/ß) gene transcription. Multiple proteins in this signaling pathway (e.g. RIG-I, MDA5, MAVS, TBK1, IRF3) are regulated by (de)ubiquitination events. Most viruses have evolved mechanisms to counter this antiviral response. The leader protease (Lpro) of foot-and-mouth-disease virus (FMDV) has been recognized to reduce IFN-α/ß gene transcription; however, the exact mechanism is unknown. The proteolytic activity of Lpro is vital for releasing itself from the viral polyprotein and for cleaving and degrading specific host cell proteins, such as eIF4G and NF-κB. In addition, Lpro has been demonstrated to have deubiquitination/deISGylation activity. Lpro's deubiquitination/deISGylation activity and the cleavage/degradation of signaling proteins have both been postulated to be important for reduced IFN-α/ß gene transcription. Here, we demonstrate that TBK1, the kinase that phosphorylates and activates the transcription factor IRF3, is cleaved by Lpro in FMDV-infected cells as well as in cells infected with a recombinant EMCV expressing Lpro. In vitro cleavage experiments revealed that Lpro cleaves TBK1 at residues 692-694. We also observed cleavage of MAVS in HeLa cells infected with EMCV-Lpro, but only observed decreasing levels of MAVS in FMDV-infected porcine LFPK αVß6 cells. We set out to dissect Lpro's ability to cleave RLR signaling proteins from its deubiquitination/deISGylation activity to determine their relative contributions to the reduction of IFN-α/ß gene transcription. The introduction of specific mutations, of which several were based on the recently published structure of Lpro in complex with ISG15, allowed us to identify specific amino acid substitutions that separate the different proteolytic activities of Lpro. Characterization of the effects of these mutations revealed that Lpro's ability to cleave RLR signaling proteins but not its deubiquitination/deISGylation activity correlates with the reduced IFN-ß gene transcription.


Subject(s)
DEAD Box Protein 58/metabolism , Endopeptidases/metabolism , Foot-and-Mouth Disease Virus/metabolism , Interferon Type I/biosynthesis , Animals , Cell Line , Endopeptidases/genetics , Foot-and-Mouth Disease/immunology , Foot-and-Mouth Disease/metabolism , Foot-and-Mouth Disease Virus/immunology , Humans , Proteolysis
4.
Nat Microbiol ; 5(11): 1361-1373, 2020 11.
Article in English | MEDLINE | ID: mdl-32690955

ABSTRACT

Eukaryotic cells, when exposed to environmental or internal stress, activate the integrated stress response (ISR) to restore homeostasis and promote cell survival. Specific stress stimuli prompt dedicated stress kinases to phosphorylate eukaryotic initiation factor 2 (eIF2). Phosphorylated eIF2 (p-eIF2) in turn sequesters the eIF2-specific guanine exchange factor eIF2B to block eIF2 recycling, thereby halting translation initiation and reducing global protein synthesis. To circumvent stress-induced translational shutdown, viruses encode ISR antagonists. Those identified so far prevent or reverse eIF2 phosphorylation. We now describe two viral proteins-one from a coronavirus and the other from a picornavirus-that have independently acquired the ability to counteract the ISR at its very core by acting as a competitive inhibitor of p-eIF2-eIF2B interaction. This allows continued formation of the eIF2-GTP-Met-tRNAi ternary complex and unabated global translation at high p-eIF2 levels that would otherwise cause translational arrest. We conclude that eIF2 and p-eIF2 differ in their interaction with eIF2B to such effect that p-eIF2-eIF2B association can be selectively inhibited.


Subject(s)
Eukaryotic Initiation Factor-2B/antagonists & inhibitors , Eukaryotic Initiation Factor-2/antagonists & inhibitors , Stress, Physiological/physiology , Viral Proteins/metabolism , Animals , Binding Sites , Chlorocebus aethiops , Eukaryotic Cells/metabolism , Eukaryotic Initiation Factor-2/metabolism , Eukaryotic Initiation Factor-2B/metabolism , Gene Knockout Techniques , HEK293 Cells , HeLa Cells , Humans , Phosphorylation , Picornaviridae/metabolism , Protein Binding , Vero Cells
5.
J Virol ; 93(10)2019 05 15.
Article in English | MEDLINE | ID: mdl-30867299

ABSTRACT

Most viruses have acquired mechanisms to suppress antiviral alpha/beta interferon (IFN-α/ß) and stress responses. Enteroviruses (EVs) actively counteract the induction of IFN-α/ß gene transcription and stress granule (SG) formation, which are increasingly implicated as a platform for antiviral signaling, but the underlying mechanisms remain poorly understood. Both viral proteases (2Apro and 3Cpro) have been implicated in the suppression of these responses, but these conclusions predominantly rely on ectopic overexpression of viral proteases or addition of purified viral proteases to cell lysates. Here, we present a detailed and comprehensive comparison of the effect of individual enterovirus proteases on the formation of SGs and the induction of IFN-α/ß gene expression in infected cells for representative members of the enterovirus species EV-A to EV-D. First, we show that SG formation and IFN-ß induction are suppressed in cells infected with EV-A71, coxsackie B3 virus (CV-B3), CV-A21, and EV-D68. By introducing genes encoding CV-B3 proteases in a recombinant encephalomyocarditis virus (EMCV) that was designed to efficiently activate antiviral responses, we show that CV-B3 2Apro, but not 3Cpro, is the major antagonist that counters SG formation and IFN-ß gene transcription and that 2Apro's proteolytic activity is essential for both functions. 2Apro efficiently suppressed SG formation despite protein kinase R (PKR) activation and α subunit of eukaryotic translation initiation factor 2 phosphorylation, suggesting that 2Apro antagonizes SG assembly or promotes its disassembly. Finally, we show that the ability to suppress SG formation and IFN-ß gene transcription is conserved in the 2Apro of EV-A71, CV-A21, and EV-D68. Collectively, our results indicate that enterovirus 2Apro plays a key role in inhibiting innate antiviral cellular responses.IMPORTANCE Enteroviruses are important pathogens that can cause a variety of diseases in humans, including aseptic meningitis, myocarditis, hand-foot-and-mouth disease, conjunctivitis, and acute flaccid paralysis. Like many other viruses, enteroviruses must counteract antiviral cellular responses to establish an infection. It has been suggested that enterovirus proteases cleave cellular factors to perturb antiviral pathways, but the exact contribution of viral proteases 2Apro and 3Cpro remains elusive. Here, we show that 2Apro, but not 3Cpro, of all four human EV species (EV-A to EV-D) inhibits SG formation and IFN-ß gene transcription. Our observations suggest that enterovirus 2Apro has a conserved function in counteracting antiviral host responses and thereby is the main enterovirus "security protein." Understanding the molecular mechanisms of enterovirus immune evasion strategies may help to develop countermeasures to control infections with these viruses.


Subject(s)
Cytoplasmic Granules/metabolism , Enterovirus A, Human/metabolism , Peptide Hydrolases/metabolism , Antigens, Viral/metabolism , Antiviral Agents/pharmacology , Cell Line , Cysteine Endopeptidases/metabolism , Cytoplasmic Granules/virology , Encephalomyocarditis virus/genetics , Enterovirus/metabolism , Enterovirus A, Human/genetics , Enterovirus A, Human/pathogenicity , Enterovirus B, Human/genetics , Enterovirus Infections/virology , HeLa Cells , Host-Pathogen Interactions/drug effects , Humans , Immune Evasion/drug effects , Interferon Type I/metabolism , Interferon-beta/metabolism , Phosphorylation , Proteolysis , Signal Transduction/drug effects , Stress, Physiological/physiology , Viral Proteins/metabolism
6.
Proc Natl Acad Sci U S A ; 116(6): 2097-2102, 2019 02 05.
Article in English | MEDLINE | ID: mdl-30674674

ABSTRACT

Activation of the integrated stress response (ISR) by a variety of stresses triggers phosphorylation of the α-subunit of translation initiation factor eIF2. P-eIF2α inhibits eIF2B, the guanine nucleotide exchange factor that recycles inactive eIF2•GDP to active eIF2•GTP. eIF2 phosphorylation thereby represses translation. Persistent activation of the ISR has been linked to the development of several neurological disorders, and modulation of the ISR promises new therapeutic strategies. Recently, a small-molecule ISR inhibitor (ISRIB) was identified that rescues translation in the presence of P-eIF2α by facilitating the assembly of more active eIF2B. ISRIB enhances cognitive memory processes and has therapeutic effects in brain-injured mice without displaying overt side effects. While using ISRIB to investigate the ISR in picornavirus-infected cells, we observed that ISRIB rescued translation early in infection when P-eIF2α levels were low, but not late in infection when P-eIF2α levels were high. By treating cells with varying concentrations of poly(I:C) or arsenite to induce the ISR, we provide additional proof that ISRIB is unable to inhibit the ISR when intracellular P-eIF2α concentrations exceed a critical threshold level. Together, our data demonstrate that the effects of pharmacological activation of eIF2B are tuned by P-eIF2α concentration. Thus, ISRIB can mitigate undesirable outcomes of low-level ISR activation that may manifest neurological disease but leaves the cytoprotective effects of acute ISR activation intact. The insensitivity of cells to ISRIB during acute ISR may explain why ISRIB does not cause overt toxic side effects in vivo.


Subject(s)
Acetamides/chemistry , Acetamides/pharmacology , Cyclohexylamines/chemistry , Cyclohexylamines/pharmacology , Stress, Physiological/drug effects , Animals , Arsenites/pharmacology , Cell Line , Eukaryotic Initiation Factor-2/antagonists & inhibitors , Eukaryotic Initiation Factor-2/metabolism , Humans , Phosphorylation , Picornaviridae , Picornaviridae Infections/metabolism , Picornaviridae Infections/virology , Poly I-C/pharmacology
7.
J Virol ; 93(2)2019 01 15.
Article in English | MEDLINE | ID: mdl-30404792

ABSTRACT

Like other viruses, the picornavirus foot-and-mouth disease virus (FMDV; genus Aphthovirus), one of the most notorious pathogens in the global livestock industry, needs to navigate antiviral host responses to establish an infection. There is substantial insight into how FMDV suppresses the type I interferon (IFN) response, but it is largely unknown whether and how FMDV modulates the integrated stress response. Here, we show that the stress response is suppressed during FMDV infection. Using a chimeric recombinant encephalomyocarditis virus (EMCV), in which we functionally replaced the endogenous stress response antagonist by FMDV leader protease (Lpro) or 3Cpro, we demonstrate an essential role for Lpro in suppressing stress granule (SG) formation. Consistently, infection with a recombinant FMDV lacking Lpro resulted in SG formation. Additionally, we show that Lpro cleaves the known SG scaffold proteins G3BP1 and G3BP2 but not TIA-1. We demonstrate that the closely related equine rhinitis A virus (ERAV) Lpro also cleaves G3BP1 and G3BP2 and also suppresses SG formation, indicating that these abilities are conserved among aphthoviruses. Neither FMDV nor ERAV Lpro interfered with phosphorylation of RNA-dependent protein kinase (PKR) or eIF2α, indicating that Lpro does not affect SG formation by inhibiting the PKR-triggered signaling cascade. Taken together, our data suggest that aphthoviruses actively target scaffolding proteins G3BP1 and G3BP2 and antagonize SG formation to modulate the integrated stress response.IMPORTANCE The picornavirus foot-and-mouth disease virus (FMDV) is a notorious animal pathogen that puts a major economic burden on the global livestock industry. Outbreaks have significant consequences for animal health and product safety. Like many other viruses, FMDV must manipulate antiviral host responses to establish infection. Upon infection, viral double-stranded RNA (dsRNA) is detected, which results in the activation of the RNA-dependent protein kinase (PKR)-mediated stress response, leading to a stop in cellular and viral translation and the formation of stress granules (SG), which are thought to have antiviral properties. Here, we show that FMDV can suppress SG formation via its leader protease (Lpro). Simultaneously, we observed that Lpro can cleave the SG scaffolding proteins G3BP1 and G3BP2. Understanding the molecular mechanisms of the antiviral host response evasion strategies of FMDV may help to develop countermeasures to control FMDV infections in the future.


Subject(s)
Cytoplasmic Granules/metabolism , Foot-and-Mouth Disease Virus/enzymology , Foot-and-Mouth Disease/metabolism , Peptide Hydrolases/metabolism , RNA Recognition Motif Proteins/metabolism , Animals , Aphthovirus/enzymology , Cell Line , Cricetinae , Encephalomyocarditis virus/enzymology , Foot-and-Mouth Disease/virology , HEK293 Cells , HeLa Cells , Humans , Stress, Physiological , Viral Proteins/metabolism
8.
Proc Natl Acad Sci U S A ; 115(10): 2371-2376, 2018 03 06.
Article in English | MEDLINE | ID: mdl-29463763

ABSTRACT

In response to viral infection, cells mount a potent inflammatory response that relies on ISG15 and ubiquitin posttranslational modifications. Many viruses use deubiquitinases and deISGylases that reverse these modifications and antagonize host signaling processes. We here reveal that the leader protease, Lbpro, from foot-and-mouth disease virus (FMDV) targets ISG15 and to a lesser extent, ubiquitin in an unprecedented manner. Unlike canonical deISGylases that hydrolyze the isopeptide linkage after the C-terminal GlyGly motif, Lbpro cleaves the peptide bond preceding the GlyGly motif. Consequently, the GlyGly dipeptide remains attached to the substrate Lys, and cleaved ISG15 is rendered incompetent for reconjugation. A crystal structure of Lbpro bound to an engineered ISG15 suicide probe revealed the molecular basis for ISG15 proteolysis. Importantly, anti-GlyGly antibodies, developed for ubiquitin proteomics, are able to detect Lbpro cleavage products during viral infection. This opens avenues for infection detection of FMDV based on an immutable, host-derived epitope.


Subject(s)
Cytokines , Endopeptidases , Ubiquitin/metabolism , Ubiquitins , Crystallography , Cytokines/chemistry , Cytokines/metabolism , Endopeptidases/chemistry , Endopeptidases/metabolism , HeLa Cells , Host-Pathogen Interactions , Humans , Models, Molecular , Protein Binding , Substrate Specificity , Ubiquitins/chemistry , Ubiquitins/metabolism
9.
Viruses ; 8(11)2016 11 15.
Article in English | MEDLINE | ID: mdl-27854284

ABSTRACT

Misfolded proteins from the endoplasmic reticulum (ER) are transported back into the cytosol for degradation via the ubiquitin-proteasome system. The human cytomegalovirus protein US11 hijacks this ER-associated protein degradation (ERAD) pathway to downregulate human leukocyte antigen (HLA) class I molecules in virus-infected cells, thereby evading elimination by cytotoxic T-lymphocytes. Recently, we identified the E3 ubiquitin ligase transmembrane protein 129 (TMEM129) as a key player in this process, where interference with TMEM129 activity in human cells completely abrogates US11-mediated class I degradation. Here, we set out to further characterize TMEM129. We show that TMEM129 is a non-glycosylated protein containing a non-cleaved signal anchor sequence. By glycosylation scanning mutagenesis, we show that TMEM129 is a tri-spanning ER-membrane protein that adopts an Nexo-Ccyto orientation. This insertion in the ER membrane positions the C-terminal really interesting new gene (RING) domain of TMEM129 in the cytosol, making it available to catalyze ubiquitination reactions that are required for cytosolic degradation of secretory proteins.


Subject(s)
Endoplasmic Reticulum/chemistry , Intracellular Membranes/chemistry , Membrane Proteins/analysis , Membrane Proteins/metabolism , Ubiquitin-Protein Ligases/analysis , Ubiquitin-Protein Ligases/metabolism , Humans , Models, Biological , Ubiquitination
10.
Sci Rep ; 6: 22064, 2016 Feb 24.
Article in English | MEDLINE | ID: mdl-26907999

ABSTRACT

Dendritic cells (DCs) present foreign antigen in major histocompatibility complex (MHC) class I molecules to cytotoxic T cells in a process called cross-presentation. An important step in this process is the release of antigen from the lumen of endosomes into the cytosol, but the mechanism of this step is still unclear. In this study, we show that reactive oxygen species (ROS) produced by the NADPH-oxidase complex NOX2 cause lipid peroxidation, a membrane disrupting chain-reaction, which in turn results in antigen leakage from endosomes. Antigen leakage and cross-presentation were inhibited by blocking ROS production or scavenging radicals and induced when using a ROS-generating photosensitizer. Endosomal antigen release was impaired in DCs from chronic granulomatous disease (CGD) patients with dysfunctional NOX2. Thus, NOX2 induces antigen release from endosomes for cross-presentation by direct oxidation of endosomal lipids. This constitutes a new cellular function for ROS in regulating immune responses against pathogens and cancer.


Subject(s)
Cross-Priming , Dendritic Cells/immunology , Granulomatous Disease, Chronic/immunology , Lipid Peroxidation/immunology , Membrane Glycoproteins/immunology , NADPH Oxidases/immunology , T-Lymphocytes, Cytotoxic/immunology , Animals , Antigen Presentation , Dendritic Cells/cytology , Dendritic Cells/drug effects , Endosomes/immunology , Endosomes/metabolism , Free Radical Scavengers/pharmacology , Gene Expression , Granulomatous Disease, Chronic/metabolism , Granulomatous Disease, Chronic/pathology , Histocompatibility Antigens Class I/genetics , Histocompatibility Antigens Class I/immunology , Humans , Jurkat Cells , Lipid Peroxidation/drug effects , Membrane Glycoproteins/genetics , Mice , Mice, Inbred C57BL , NADPH Oxidase 2 , NADPH Oxidases/genetics , Photosensitizing Agents/pharmacology , Primary Cell Culture , Reactive Oxygen Species/antagonists & inhibitors , Reactive Oxygen Species/immunology , Reactive Oxygen Species/metabolism , T-Lymphocytes, Cytotoxic/cytology , T-Lymphocytes, Cytotoxic/drug effects , alpha-Tocopherol/pharmacology
11.
Biomol Concepts ; 7(1): 17-28, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26741354

ABSTRACT

Immune responses are initiated by the interactions between antigen-presenting cells (APCs), such as dendritic cells (DCs), with responder cells, such as T cells, via a tight cellular contact interface called the immunological synapse. The immunological synapse is a highly organized subcellular structure that provides a platform for the presentation of antigen in major histocompatibility class I and II complexes (MHC class I and II) on the surface of the APC to receptors on the surface of the responder cells. In T cells, these contacts lead to highly polarized membrane trafficking that results in the local release of lytic granules and in the delivery and recycling of T cell receptors at the immunological synapse. Localized trafficking also occurs at the APC side of the immunological synapse, especially in DCs where antigen loaded in MHC class I and II is presented and cytokines are released specifically at the synapse. Whereas the molecular mechanisms underlying polarized membrane trafficking at the T cell side of the immunological synapse are increasingly well understood, these are still very unclear at the APC side. In this review, we discuss the organization of the APC side of the immunological synapse. We focus on the directional trafficking and release of membrane vesicles carrying MHC molecules and cytokines at the immunological synapses of DCs. We hypothesize that the specific delivery of MHC and the release of cytokines at the immunological synapse mechanistically resemble that of lytic granule release from T cells.


Subject(s)
Dendritic Cells/immunology , Immunological Synapses/immunology , Animals , Antigen Presentation , Humans , Protein Transport , Receptors, Antigen, T-Cell/immunology , T-Lymphocytes/immunology
12.
J Virol ; 89(21): 11169-73, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26311870

ABSTRACT

It is well known that plasmid DNA transfection, prior to virus infection, negatively affects infection efficiency. Here, we show that cytosolic plasmid DNA activates the cGAS/STING signaling pathway, which ultimately leads to the induction of an antiviral state of the cells. Using a transient one-plasmid clustered regularly interspaced short palindromic repeat (CRISPR)/Cas9 system, we generated cGAS/STING-knockout cells and show that these cells can be infected after plasmid DNA transfection as efficiently as nontransfected cells.


Subject(s)
Coxsackievirus Infections/metabolism , Cytosol/metabolism , Gene Transfer Techniques , Membrane Proteins/metabolism , Nucleotidyltransferases/metabolism , Plasmids/metabolism , Signal Transduction/physiology , CRISPR-Cas Systems , Flow Cytometry , Gene Knockout Techniques , Green Fluorescent Proteins , HeLa Cells , Humans , Luminescent Proteins , Membrane Proteins/genetics , Mengovirus/metabolism , Nucleotidyltransferases/genetics , Plasmids/genetics , Signal Transduction/genetics , Transfection/methods , Red Fluorescent Protein
SELECTION OF CITATIONS
SEARCH DETAIL
...