Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 9 de 9
Filtrer
Plus de filtres










Base de données
Gamme d'année
1.
Gene ; 927: 148649, 2024 Jun 07.
Article de Anglais | MEDLINE | ID: mdl-38852697

RÉSUMÉ

During the birch pollen season an enhanced incidence of virus infections is noticed, raising the question whether pollen can affect anti-viral responses independent of allergic reactions. We previously showed that birch pollen-treatment of monocyte-derived dendritic cells (moDC) enhances human cytomegalovirus (HCMV) infection. Here we addressed how in moDC the relatively weak pollen response can affect the comparably strong response to HCMV. To this end, moDC were stimulated with aqueous birch pollen extract (APE), HCMV, and APE with HCMV, and transcriptomic signatures were determined after 6 and 24 h of incubation. Infection was monitored upon exposure of moDC to GFP expressing HCMV by flow cytometric analysis of GFP expressing cells. Principle component analysis of RNA sequencing data revealed close clustering of mock and APE treated moDC, whereas HCMV as well as APE with HCMV treated moDC clustered separately after 6 and 24 h of incubation, respectively. Communally induced genes were detected in APE, HCMV and APE with HCMV treated moDC. In APE with HCMV treated moDC, the comparably weak APE induced signatures were maintained after HCMV exposure. In particular, NF-κB/RELA and PI3K/AKT/MAPK signaling were altered upon APE with HCMV exposure. Earlier, we discovered that NF-κB inhibition alleviated APE induced enhancement of HCMV infection. Here we additionally found that impairment of PI3K signaling reduced HCMV infection in HCMV and APE with HCMV treated moDC. APE treated moDC that were exposed to HCMV show a unique host gene signature, which to a large extent is regulated by NF-κB activation and PI3K/AKT/MAPK signaling.

2.
Nat Commun ; 15(1): 1745, 2024 Feb 26.
Article de Anglais | MEDLINE | ID: mdl-38409141

RÉSUMÉ

Human cytomegalovirus (HCMV) is a widespread pathogen that in immunocompromised hosts can cause life-threatening disease. Studying HCMV-exposed monocyte-derived dendritic cells by single-cell RNA sequencing, we observe that most cells are entered by the virus, whereas less than 30% of them initiate viral gene expression. Increased viral gene expression is associated with activation of the stimulator of interferon genes (STING) that usually induces anti-viral interferon responses, and with the induction of several pro- (RHOB, HSP1A1, DNAJB1) and anti-viral (RNF213, TNFSF10, IFI16) genes. Upon progression of infection, interferon-beta but not interferon-lambda transcription is inhibited. Similarly, interferon-stimulated gene expression is initially induced and then shut off, thus further promoting productive infection. Monocyte-derived dendritic cells are composed of 3 subsets, with one being especially susceptible to HCMV. In conclusion, HCMV permissiveness of monocyte-derived dendritic cells depends on complex interactions between virus sensing, regulation of the interferon response, and viral gene expression.


Sujet(s)
Cytomegalovirus , Interférons , Humains , Cytomegalovirus/physiologie , Transduction du signal/génétique , Antiviraux/métabolisme , Cellules dendritiques/métabolisme , Protéines du choc thermique HSP40/métabolisme , Adenosine triphosphatases/métabolisme , Ubiquitin-protein ligases/métabolisme
3.
Microbiol Spectr ; : e0219223, 2023 Sep 14.
Article de Anglais | MEDLINE | ID: mdl-37707204

RÉSUMÉ

The flavivirus non-structural protein 1 (NS1) is secreted from infected cells into the circulation and the serum levels correlate with disease severity. The effect of secreted NS1 (sNS1) on non-infected mammalian immune cells is largely unknown. Here, we expressed recombinant sNS1 proteins of tick-borne encephalitis virus (TBEV) and West Nile virus (WNV) and investigated their effects on dendritic cell (DC) effector functions. Murine bone marrow-derived DCs (BMDCs) showed reduced surface expression of co-stimulatory molecules and decreased release of pro-inflammatory cytokines when treated with sNS1 of TBEV or WNV prior to poly(I:C) stimulation. Transcriptional profiles of BMDCs that were sNS1-exposed prior to poly(I:C) stimulation showed two gene clusters that were downregulated by TBEV or WNV sNS1 and that were associated with innate and adaptive immune responses. Functionally, both sNS1 proteins modulated the capacity for BMDCs to induce specific T-cell responses as indicated by reduced IFN-γ levels in both CD4+ and CD8+ T cells after BMDC co-cultivation. In human monocyte-derived DCs, poly(I:C)-induced upregulation of co-stimulatory molecules and cytokine responses were even more strongly impaired by TBEV sNS1 or WNV sNS1 pretreatment than in the murine system. Our findings indicate that exogenous flaviviral sNS1 proteins interfere with DC-mediated stimulation of T cells, which is crucial for the initiation of cell-mediated adaptive immune responses in human flavivirus infections. Collectively, our data determine soluble flaviviral NS1 as a virulence factor responsible for a dampened immune response to flavivirus infections. IMPORTANCE The effective initiation of protective host immune responses controls the outcome of infection, and dysfunctional T-cell responses have previously been associated with symptomatic human flavivirus infections. We demonstrate that secreted flavivirus NS1 proteins modulate innate immune responses of uninfected bystander cells. In particular, sNS1 markedly reduced the capacity of dendritic cells to stimulate T-cell responses upon activation. Hence, by modulating cellular host responses that are required for effective antigen presentation and initiation of adaptive immunity, sNS1 proteins may contribute to severe outcomes of flavivirus disease.

4.
Glia ; 71(11): 2573-2590, 2023 11.
Article de Anglais | MEDLINE | ID: mdl-37455566

RÉSUMÉ

Demyelination in the central nervous system (CNS) is a hallmark of many neurodegenerative diseases such as multiple sclerosis (MS) and others. Here, we studied astrocytes during de- and remyelination in the cuprizone mouse model. To this end, we exploited the ribosomal tagging (RiboTag) technology that is based on Cre-mediated cell type-selective HA-tagging of ribosomes. Analyses were performed in the corpus callosum of GFAP-Cre+/- Rpl22HA/wt mice 5 weeks after cuprizone feeding, at the peak of demyelination, and 0.5 and 2 weeks after cuprizone withdrawal, when remyelination and tissue repair is initiated. After 5 weeks of cuprizone feeding, reactive astrocytes showed inflammatory signatures with enhanced expression of genes that modulate leukocyte migration (Tlr2, Cd86, Parp14) and they produced the chemokine CXCL10, as verified by histology. Furthermore, demyelination-induced reactive astrocytes expressed numerous ligands including Cx3cl1, Csf1, Il34, and Gas6 that act on homeostatic as well as activated microglia and thus potentially mediate activation and recruitment of microglia and enhancement of their phagocytotic activity. During early remyelination, HA-tagged cells displayed reduced inflammatory response signatures, as indicated by shutdown of CXCL10 production, and enhanced expression of osteopontin (SPP1) as well as of factors that are relevant for tissue remodeling (Timp1), regeneration and axonal repair. During late remyelination, the signatures shifted towards resolving inflammation by active suppression of lymphocyte activation and differentiation and support of glia cell differentiation. In conclusion, we detected highly dynamic astroglial transcriptomic signatures in the cuprizone model, which reflects excessive communication among glia cells and highlights different astrocyte functions during neurodegeneration and regeneration.


Sujet(s)
Cuprizone , Maladies démyélinisantes , Souris , Animaux , Cuprizone/toxicité , Astrocytes/métabolisme , Maladies démyélinisantes/anatomopathologie , Névroglie/métabolisme , Corps calleux/anatomopathologie , Modèles animaux de maladie humaine , Souris de lignée C57BL , Gaine de myéline/métabolisme , Oligodendroglie/métabolisme
6.
Viruses ; 13(10)2021 10 14.
Article de Anglais | MEDLINE | ID: mdl-34696494

RÉSUMÉ

Viral encephalitis is a rare but serious syndrome. In addition to DNA-encoded herpes viruses, such as herpes simplex virus and varicella zoster virus, RNA-encoded viruses from the families of Flaviviridae, Rhabdoviridae and Paramyxoviridae are important neurotropic viruses. Whereas in the periphery, the role of Toll-like receptors (TLR) during immune stimulation is well understood, TLR functions within the CNS are less clear. On one hand, TLRs can affect the physiology of neurons during neuronal progenitor cell differentiation and neurite outgrowth, whereas under conditions of infection, the complex interplay between TLR stimulated neurons, astrocytes and microglia is just on the verge of being understood. In this review, we summarize the current knowledge about which TLRs are expressed by cell subsets of the CNS. Furthermore, we specifically highlight functional implications of TLR stimulation in neurons, astrocytes and microglia. After briefly illuminating some examples of viral evasion strategies from TLR signaling, we report on the current knowledge of primary immunodeficiencies in TLR signaling and their consequences for viral encephalitis. Finally, we provide an outlook with examples of TLR agonist mediated intervention strategies and potentiation of vaccine responses against neurotropic virus infections.


Sujet(s)
Encéphalite virale/immunologie , Récepteurs de type Toll/immunologie , Récepteurs de type Toll/métabolisme , Animaux , Astrocytes/virologie , Système nerveux central/immunologie , Système nerveux central/métabolisme , Herpès/immunologie , Interactions hôte-microbes , Humains , Immunité innée , Microglie/virologie , Neurones , Transduction du signal , Simplexvirus
7.
J Neurosci Res ; 99(10): 2478-2492, 2021 10.
Article de Anglais | MEDLINE | ID: mdl-34296786

RÉSUMÉ

Tick-borne encephalitis virus (TBEV), a member of the Flaviviridae family, is typically transmitted upon tick bite and can cause meningitis and encephalitis in humans. In TBEV-infected mice, mitochondrial antiviral-signaling protein (MAVS), the downstream adaptor of retinoic acid-inducible gene-I (RIG-I)-like receptor (RLR) signaling, is needed to induce early type I interferon (IFN) responses and to confer protection. To characterize the brain-resident cell subset that produces protective IFN-ß in TBEV-infected mice, we isolated neurons, astrocytes, and microglia from mice and exposed these cell types to TBEV in vitro. Under such conditions, neurons showed the highest percentage of infected cells, whereas astrocytes and microglia were infected to a lesser extent. In the supernatant (SN) of infected neurons, IFN-ß was not detectable, while infected astrocytes showed high and microglia low IFN-ß expression. Transcriptome analyses of astrocytes implied that MAVS signaling was needed early after TBEV infection. Accordingly, MAVS-deficient astrocytes showed enhanced TBEV infection and significantly reduced early IFN-ß responses. Nevertheless, at later time points, moderate amounts of IFN-ß were detected in the SN of infected MAVS-deficient astrocytes. Transcriptome analyses indicated that MAVS deficiency negatively affected the induction of early anti-viral responses, which resulted in significantly increased TBEV replication. Treatment with MyD88 and TRIF inhibiting peptides reduced only late IFN-ß responses of TBEV-infected WT astrocytes and blocked entirely IFN-ß responses of infected MAVS-deficient astrocytes. Thus, upon TBEV exposure of brain-resident cells, astrocytes are important IFN-ß producers showing biphasic IFN-ß induction that initially depends on MAVS and later on MyD88/TRIF signaling.


Sujet(s)
Protéines adaptatrices de la transduction du signal/métabolisme , Protéines adaptatrices du transport vésiculaire/métabolisme , Astrocytes/métabolisme , Virus de l'encéphalite à tiques (sous-groupe)/métabolisme , Encéphalites à tiques/métabolisme , Facteur de différenciation myéloïde-88/métabolisme , Animaux , Astrocytes/virologie , Encéphalites à tiques/prévention et contrôle , Souris , Souris de lignée C57BL , Souris transgéniques , Transduction du signal/physiologie
8.
Sci Immunol ; 6(60)2021 06 25.
Article de Anglais | MEDLINE | ID: mdl-34172587

RÉSUMÉ

Viral encephalitis initiates a series of immunological events in the brain that can lead to brain damage and death. Astrocytes express IFN-ß in response to neurotropic infection, whereas activated microglia produce proinflammatory cytokines and accumulate at sites of infection. Here, we observed that neurotropic vesicular stomatitis virus (VSV) infection causes recruitment of leukocytes into the central nervous system (CNS), which requires MyD88, an adaptor of Toll-like receptor and interleukin-1 receptor signaling. Infiltrating leukocytes, and in particular CD8+ T cells, protected against lethal VSV infection of the CNS. Reconstitution of MyD88, specifically in neurons, restored chemokine production in the olfactory bulb as well as leukocyte recruitment into the infected CNS and enhanced survival. Comparative analysis of the translatome of neurons and astrocytes verified neurons as the critical source of chemokines, which regulated leukocyte infiltration of the infected brain and affected survival.


Sujet(s)
Lymphocytes T CD8+/immunologie , Chimiokines/métabolisme , Encéphalite virale/immunologie , Facteur de différenciation myéloïde-88/métabolisme , Infections à Rhabdoviridae/immunologie , Protéines adaptatrices du transport vésiculaire/génétique , Protéines adaptatrices du transport vésiculaire/métabolisme , Animaux , Modèles animaux de maladie humaine , Encéphalite virale/anatomopathologie , Encéphalite virale/virologie , Femelle , Humains , Mâle , Souris , Souris knockout , Facteur de différenciation myéloïde-88/génétique , Neurones/métabolisme , Bulbe olfactif/cytologie , Bulbe olfactif/immunologie , Bulbe olfactif/anatomopathologie , Bulbe olfactif/virologie , Infections à Rhabdoviridae/anatomopathologie , Infections à Rhabdoviridae/virologie , Transduction du signal/immunologie , Vesiculovirus/immunologie
9.
Am J Trop Med Hyg ; 95(5): 1077-1085, 2016 Nov 02.
Article de Anglais | MEDLINE | ID: mdl-27601522

RÉSUMÉ

Understanding the genetic structure and transmission dynamics of Plasmodium falciparum parasites in malaria-endemic regions is crucial before the implementation of interventions. Located in a high-transmission region of western Kenya where P. falciparum is the predominant species, the Lake Victoria islands are ideal for feasibility of malaria elimination studies. We analyzed genetic variation in eight microsatellite loci to examine parasite population structure and gene flow patterns across five sites. High levels of genetic diversity were measured throughout the region (mean heterozygosity index = 0.84). The overall fixation index value between the sites was 0.044, indicating that approximately 5% of the overall allelic variation is due to differences between the populations. Based on these results, we concluded that parasite population structure in the studied islands is shaped by human migration patterns that maintain extensive parasite gene flow between the sites. Consequently, any malaria elimination and interventions strategies in the study area will have to be carried out broadly on all four islands and adjoining mainland region.


Sujet(s)
Variation génétique , Plasmodium falciparum/génétique , Allèles , Études transversales , ADN des protozoaires/génétique , Flux des gènes , Locus génétiques , Techniques de génotypage , Humains , Iles , Kenya , Lacs , Paludisme à Plasmodium falciparum/épidémiologie , Répétitions microsatellites , Plasmodium falciparum/isolement et purification
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE
...