Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 20
Filtrer
1.
Antiviral Res ; 225: 105844, 2024 May.
Article de Anglais | MEDLINE | ID: mdl-38428749

RÉSUMÉ

The Third International Conference on Crimean-Congo Hemorrhagic Fever (CCHF) was held in Thessaloniki, Greece, September 19-21, 2023, bringing together a diverse group of international partners, including public health professionals, clinicians, ecologists, epidemiologists, immunologists, and virologists. The conference was attended by 118 participants representing 24 countries and the World Health Organization (WHO). Meeting sessions covered the epidemiology of CCHF in humans; Crimean-Congo hemorrhagic fever virus (CCHFV) in ticks; wild and domestic animal hosts; molecular virology; pathogenesis and animal models; immune response related to therapeutics; and CCHF prevention in humans. The concluding session focused on recent WHO recommendations regarding disease prevention, control strategies, and innovations against CCHFV outbreaks. This meeting report summarizes lectures by the invited speakers and highlights advances in the field.


Sujet(s)
Virus de la fièvre hémorragique de Crimée-Congo , Fièvre hémorragique de Crimée-Congo , Tiques , Animaux , Humains , Fièvre hémorragique de Crimée-Congo/épidémiologie , Grèce , Épidémies de maladies
2.
Nat Microbiol ; 9(6): 1499-1512, 2024 Jun.
Article de Anglais | MEDLINE | ID: mdl-38548922

RÉSUMÉ

Climate change and population densities accelerated transmission of highly pathogenic viruses to humans, including the Crimean-Congo haemorrhagic fever virus (CCHFV). Here we report that the Low Density Lipoprotein Receptor (LDLR) is a critical receptor for CCHFV cell entry, playing a vital role in CCHFV infection in cell culture and blood vessel organoids. The interaction between CCHFV and LDLR is highly specific, with other members of the LDLR protein family failing to bind to or neutralize the virus. Biosensor experiments demonstrate that LDLR specifically binds the surface glycoproteins of CCHFV. Importantly, mice lacking LDLR exhibit a delay in CCHFV-induced disease. Furthermore, we identified the presence of Apolipoprotein E (ApoE) on CCHFV particles. Our findings highlight the essential role of LDLR in CCHFV infection, irrespective of ApoE presence, when the virus is produced in tick cells. This discovery holds profound implications for the development of future therapies against CCHFV.


Sujet(s)
Apolipoprotéines E , Virus de la fièvre hémorragique de Crimée-Congo , Fièvre hémorragique de Crimée-Congo , Récepteurs aux lipoprotéines LDL , Pénétration virale , Animaux , Humains , Souris , Apolipoprotéines E/métabolisme , Apolipoprotéines E/génétique , Virus de la fièvre hémorragique de Crimée-Congo/génétique , Virus de la fièvre hémorragique de Crimée-Congo/physiologie , Fièvre hémorragique de Crimée-Congo/virologie , Fièvre hémorragique de Crimée-Congo/métabolisme , Souris knockout , Récepteurs aux lipoprotéines LDL/métabolisme , Récepteurs aux lipoprotéines LDL/génétique , Récepteurs viraux/métabolisme , Tiques/virologie , Tiques/métabolisme
3.
Nat Commun ; 14(1): 6785, 2023 10 25.
Article de Anglais | MEDLINE | ID: mdl-37880247

RÉSUMÉ

Marburg and Ebola filoviruses are two of the deadliest infectious agents and several outbreaks have occurred in the last decades. Although several receptors and co-receptors have been reported for Ebola virus, key host factors remain to be elucidated. In this study, using a haploid cell screening platform, we identify the guanine nucleotide exchange factor CCZ1 as a key host factor in the early stage of filovirus replication. The critical role of CCZ1 for filovirus infections is validated in 3D primary human hepatocyte cultures and human blood-vessel organoids, both critical target sites for Ebola and Marburg virus tropism. Mechanistically, CCZ1 controls early to late endosomal trafficking of these viruses. In addition, we report that CCZ1 has a role in the endosomal trafficking of endocytosis-dependent SARS-CoV-2 infections, but not in infections by Lassa virus, which enters endo-lysosomal trafficking at the late endosome stage. Thus, we have identified an essential host pathway for filovirus infections in cell lines and engineered human target tissues. Inhibition of CCZ1 nearly completely abolishes Marburg and Ebola infections. Thus, targeting CCZ1 could potentially serve as a promising drug target for controlling infections caused by various viruses, such as SARS-CoV-2, Marburg, and Ebola.


Sujet(s)
Ebolavirus , Fièvre hémorragique à virus Ebola , Maladie de Marbourg , Marburgvirus , Protéines du transport vésiculaire , Animaux , Humains , Ebolavirus/métabolisme , Lysosomes , Maladie de Marbourg/génétique , Maladie de Marbourg/métabolisme , Marburgvirus/métabolisme , Protéines du transport vésiculaire/métabolisme
4.
Life Sci Alliance ; 6(7)2023 07.
Article de Anglais | MEDLINE | ID: mdl-37072184

RÉSUMÉ

Viruses with an RNA genome are often the cause of zoonotic infections. In order to identify novel pro-viral host cell factors, we screened a haploid insertion-mutagenized mouse embryonic cell library for clones that are resistant to Rift Valley fever virus (RVFV). This screen returned the low-density lipoprotein receptor-related protein 1 (LRP1) as a top hit, a plasma membrane protein involved in a wide variety of cell activities. Inactivation of LRP1 in human cells reduced RVFV RNA levels already at the attachment and entry stages of infection. Moreover, the role of LRP1 in promoting RVFV infection was dependent on physiological levels of cholesterol and on endocytosis. In the human cell line HuH-7, LRP1 also promoted early infection stages of sandfly fever Sicilian virus and La Crosse virus, but had a minor effect on late infection by vesicular stomatitis virus, whereas encephalomyocarditis virus was entirely LRP1-independent. Moreover, siRNA experiments in human Calu-3 cells demonstrated that also SARS-CoV-2 infection benefitted from LRP1. Thus, we identified LRP1 as a host factor that supports infection by a spectrum of RNA viruses.


Sujet(s)
COVID-19 , Virus de la fièvre de la vallée du Rift , Animaux , Humains , Souris , Protéine-1 apparentée au récepteur des LDL/génétique , Protéine-1 apparentée au récepteur des LDL/métabolisme , SARS-CoV-2/génétique , Virus de la fièvre de la vallée du Rift/génétique , Virus de la fièvre de la vallée du Rift/métabolisme , Petit ARN interférent/génétique , Petit ARN interférent/métabolisme , Lipoprotéines LDL/métabolisme
5.
EMBO Mol Med ; 14(8): e15230, 2022 08 08.
Article de Anglais | MEDLINE | ID: mdl-35781796

RÉSUMÉ

The recent emergence of multiple SARS-CoV-2 variants has caused considerable concern due to both reduced vaccine efficacy and escape from neutralizing antibody therapeutics. It is, therefore, paramount to develop therapeutic strategies that inhibit all known and future SARS-CoV-2 variants. Here, we report that all SARS-CoV-2 variants analyzed, including variants of concern (VOC) Alpha, Beta, Gamma, Delta, and Omicron, exhibit enhanced binding affinity to clinical grade and phase 2 tested recombinant human soluble ACE2 (APN01). Importantly, soluble ACE2 neutralized infection of VeroE6 cells and human lung epithelial cells by all current VOC strains with markedly enhanced potency when compared to reference SARS-CoV-2 isolates. Effective inhibition of infections with SARS-CoV-2 variants was validated and confirmed in two independent laboratories. These data show that SARS-CoV-2 variants that have emerged around the world, including current VOC and several variants of interest, can be inhibited by soluble ACE2, providing proof of principle of a pan-SARS-CoV-2 therapeutic.


Sujet(s)
Angiotensin-converting enzyme 2 , Traitements médicamenteux de la COVID-19 , Humains , Peptidyl-Dipeptidase A/génétique , Peptidyl-Dipeptidase A/métabolisme , SARS-CoV-2
6.
J Virol ; 96(13): e0059922, 2022 07 13.
Article de Anglais | MEDLINE | ID: mdl-35695578

RÉSUMÉ

Crimean-Congo hemorrhagic fever virus (CCHFV) is a tick-borne orthonairovirus that causes a severe, often fatal, hemorrhagic disease throughout Africa, Asia, and Southeast Europe. A wide variety of strains are circulating in the field which broadly correlate to their geographic distribution. The viral determinants of pathogenicity remain unclear, as does the contribution of strain-specific differences to pathology. Aigai virus (AIGV) is a closely related virus (formally designated CCHFV genotype VI, Europe II, or AP92-like virus), which has been proposed to be less virulent than CCHFV. However, the molecular details leading to potential differences in virulence are unknown. To explore if differences exist, life cycle modeling systems, including both a minigenome and a transcriptionally competent virus-like particle assay, were developed for AIGV to allow the comparison with the CCHFV reference IbAr10200 strain. Using this approach, we could demonstrate that AIGV exhibits lower viral gene expression than the reference strain of CCHFV. Subsequent systematic exchange of viral components revealed that the L protein is responsible for the observed differences in gene expression and that the interferon (IFN) antagonistic activity of the ovarian tumor-type protease domain is not responsible for this effect. IMPORTANCE Crimean-Congo hemorrhagic fever virus (CCHFV) is the cause of severe hemorrhagic disease, which is often fatal. Present throughout Africa, Asia, and Southeast Europe, a diverse number of viral genotypes exist. However, the viral determinants of pathogenicity remain unclear. It has been proposed that the closely related Aigai virus (AIGV) may be a less virulent virus. Here, using newly developed and improved life cycle modeling systems we have examined potential differences between the CCHFV reference strain, IbAr10200, and AIGV. Using this approach, we identified lower viral gene expression driven by the AIGV viral polymerase as a major difference which may be indicative of lower virulence.


Sujet(s)
Virus de la fièvre hémorragique de Crimée-Congo , Virulence , Afrique , Animaux , Modèles animaux de maladie humaine , Europe , Régulation de l'expression des gènes viraux , Génotype , Virus de la fièvre hémorragique de Crimée-Congo/classification , Virus de la fièvre hémorragique de Crimée-Congo/génétique , Virus de la fièvre hémorragique de Crimée-Congo/pathogénicité , Fièvre hémorragique de Crimée-Congo/virologie , Humains , Spécificité d'espèce , Virulence/génétique
7.
Nat Microbiol ; 6(2): 187-195, 2021 02.
Article de Anglais | MEDLINE | ID: mdl-33257849

RÉSUMÉ

There is currently no specific prophylaxis or vaccine against Crimean-Congo haemorrhagic fever virus (CCHFV). Crimean-Congo haemorrhagic fever (CCHF) is a severe febrile illness transmitted by Hyalomma ticks in endemic areas, handling of infected livestock or care of infected patients. We report here the successful protection against CCHFV-mediated disease in a non-human primate disease model. Cynomolgus macaques were vaccinated with a DNA-based vaccine using in vivo electroporation-assisted delivery. The vaccine contained two plasmids encoding the glycoprotein precursor (GPC) and the nucleoprotein (NP) of CCHFV. Animals received three vaccinations and we recorded potent antibody and T cell responses after vaccination. While all sham-vaccinated animals developed viraemia, high tissue viral loads and CCHF-induced disease, the NP + GPC vaccinated animals were significantly protected. In conclusion, this is evidence of a vaccine that can protect against CCHFV-induced disease in a non-human primate model. This supports clinical development of the vaccine to protect groups at risk for contracting the infection.


Sujet(s)
Virus de la fièvre hémorragique de Crimée-Congo/immunologie , Fièvre hémorragique de Crimée-Congo/prévention et contrôle , Vaccins à ADN/immunologie , Vaccins antiviraux/immunologie , Animaux , Modèles animaux de maladie humaine , Virus de la fièvre hémorragique de Crimée-Congo/génétique , Macaca fascicularis , Protéines nucléocapside/génétique , Protéines nucléocapside/immunologie , Précurseurs de protéines/génétique , Précurseurs de protéines/immunologie , Vaccins à ADN/usage thérapeutique , Vaccins antiviraux/usage thérapeutique
8.
PLoS Negl Trop Dis ; 14(12): e0008942, 2020 12.
Article de Anglais | MEDLINE | ID: mdl-33275608

RÉSUMÉ

BACKGROUND: Many ruminant diseases of viral aetiology can be effectively prevented using appropriate vaccination measures. For diseases such as Rift Valley fever (RVF) the long inter-epizootic periods make routine vaccination programs unfeasible. Coupling RVF prophylaxis with seasonal vaccination programmes by means of multivalent vaccine platforms would help to reduce the risk of new RVF outbreaks. METHODOLOGY/PRINCIPAL FINDINGS: In this work we generated recombinant attenuated Rift Valley fever viruses (RVFVs) encoding in place of the virulence factor NSs either the VP2 capsid protein or a truncated form of the non-structural NS1 protein of bluetongue virus serotype 4 (BTV-4). The recombinant viruses were able to carry and express the heterologous BTV genes upon consecutive passages in cell cultures. In murine models, a single immunization was sufficient to protect mice upon RVFV challenge and to elicit a specific immune response against BTV-4 antigens that was fully protective after a BTV-4 boost. In sheep, a natural host for RVFV and BTV, both vaccines proved immunogenic although conferred only partial protection after a virulent BTV-4 reassortant Morocco strain challenge. CONCLUSIONS/SIGNIFICANCE: Though additional optimization will be needed to improve the efficacy data against BTV in sheep, our findings warrant further developments of attenuated RVFV as a dual vaccine platform carrying heterologous immune relevant antigens for ruminant diseases in RVF risk areas.


Sujet(s)
Anticorps antiviraux/immunologie , Antigènes viraux/immunologie , Virus de la langue bleue/immunologie , Fièvre catarrhale du mouton/prévention et contrôle , Fièvre de la Vallée du Rift/prévention et contrôle , Vaccination/médecine vétérinaire , Vaccins antiviraux/immunologie , Animaux , Fièvre catarrhale du mouton/virologie , Virus de la langue bleue/génétique , Protéines de capside/génétique , Protéines de capside/immunologie , Femelle , Immunité , Souris , Virus recombinants , Fièvre de la Vallée du Rift/virologie , Virus de la fièvre de la vallée du Rift/génétique , Sérogroupe , Ovis , Vaccins atténués/immunologie , Protéines virales non structurales/génétique , Protéines virales non structurales/immunologie
9.
PLoS Negl Trop Dis ; 14(9): e0008610, 2020 09.
Article de Anglais | MEDLINE | ID: mdl-32931521

RÉSUMÉ

Crimean-Congo Hemorrhagic Fever virus (CCHFV; family Nairoviridae) is an extremely pathogenic member of the Bunyavirales order. Previous studies have shown that the N-terminal domain of the CCHFV polymerase (L) contains an ovarian tumor-type protease (OTU) domain with the capability to remove both ubiquitin and ISG15 molecules from proteins. The approximately 200 amino acids-long OTU domain, if ectopically expressed, can interfere with both the induction of antiviral type I interferons (IFN) as well as the IFN-stimulated signaling. A OTU protease mutant (C40A), by contrast, was inactive in that respect. However, the effect of the OTU protease activity in the context of the full-length L protein (approximately 4000 amino acids) is only poorly characterized, and recombinant CCHFV with the C40A mutation could not be rescued. Here, we employed transcriptionally active virus-like particles (tc-VLPs) to investigate the interaction between the L-embedded OTU protease and the IFN system. Our data show a cis requirement of the OTU protease for optimal CCHFV polymerase activity in human HuH-7 cells. The L-embedded OTU did not influence IFN signaling, the sensitivity to IFN, or IFN induction. Moreover, the attenuation of OTU C40A-mutated L could not be relieved by inactivating the IFN response, but after overexpression of conjugation-competent ISG15 the polymerase activity recovered to wild-type levels. Consequently, ISG15 was used to produce OTU-deficient tc-VLPs, a potential vaccine candidate. Our data thus indicate that in the context of full-length L the OTU domain is important for the regulation of CCHFV polymerase by ISG15.


Sujet(s)
Cytokines/génétique , DNA-directed RNA polymerases/génétique , Virus de la fièvre hémorragique de Crimée-Congo/génétique , Virus de la fièvre hémorragique de Crimée-Congo/immunologie , Peptide hydrolases/génétique , Ubiquitines/génétique , Cellules A549 , Animaux , Lignée cellulaire tumorale , Chlorocebus aethiops , Cytokines/métabolisme , DNA-directed RNA polymerases/métabolisme , Fièvre hémorragique de Crimée-Congo/anatomopathologie , Humains , Interféron de type I/immunologie , Domaines protéiques/génétique , Maturation post-traductionnelle des protéines/génétique , Ubiquitines/métabolisme , Cellules Vero
10.
PLoS Pathog ; 16(9): e1008850, 2020 09.
Article de Anglais | MEDLINE | ID: mdl-32956404

RÉSUMÉ

Crimean-Congo hemorrhagic fever virus (CCHFV) is a tick-borne orthonairovirus that has become a serious threat to the public health. CCHFV has a single-stranded, tripartite RNA genome composed of L, M, and S segments. Cleavage of the M polyprotein precursor generates the two envelope glycoproteins (GPs) as well as three secreted nonstructural proteins GP38 and GP85 or GP160, representing GP38 only or GP38 linked to a mucin-like protein (MLD), and a double-membrane-spanning protein called NSm. Here, we examined the relevance of each M-segment non-structural proteins in virus assembly, egress and infectivity using a well-established CCHFV virus-like-particle system (tc-VLP). Deletion of MLD protein had no impact on infectivity although it reduced by 60% incorporation of GPs into particles. Additional deletion of GP38 abolished production of infectious tc-VLPs. The loss of infectivity was associated with impaired Gc maturation and exclusion from the Golgi, showing that Gn is not sufficient to target CCHFV GPs to the site of assembly. Consistent with this, efficient complementation was achieved in cells expressing MLD-GP38 in trans with increased levels of preGc to Gc conversion, co-targeting to the Golgi, resulting in particle incorporation and restored infectivity. Contrastingly, a MLD-GP38 variant retained in the ER allowed preGc cleavage but failed to rescue miss-localization or infectivity. NSm deletion, conversely, did not affect trafficking of Gc but interfered with Gc processing, particle formation and secretion. NSm expression affected N-glycosylation of different viral proteins most likely due to increased speed of trafficking through the secretory pathway. This highlights a potential role of NSm in overcoming Golgi retention and facilitating CCHFV egress. Thus, deletions of GP38 or NSm demonstrate their important role on CCHFV particle production and infectivity. GP85 is an essential viral factor for preGc cleavage, trafficking and Gc incorporation into particles, whereas NSm protein is involved in CCHFV assembly and virion secretion.


Sujet(s)
Virus de la fièvre hémorragique de Crimée-Congo/physiologie , Protéines virales structurales , Assemblage viral , Lignée cellulaire tumorale , Délétion de gène , Humains , Protéines virales structurales/génétique , Protéines virales structurales/métabolisme
11.
Nucleic Acids Res ; 47(20): 10914-10930, 2019 11 18.
Article de Anglais | MEDLINE | ID: mdl-31584100

RÉSUMÉ

Toscana virus (TOSV) is an arthropod-borne human pathogen responsible for seasonal outbreaks of fever and meningoencephalitis in the Mediterranean basin. TOSV is a segmented negative-strand RNA virus (sNSV) that belongs to the genus phlebovirus (family Phenuiviridae, order Bunyavirales), encompassing other important human pathogens such as Rift Valley fever virus (RVFV). Here, we carried out a structural and functional characterization of the TOSV cap-snatching endonuclease, an N terminal domain of the viral polymerase (L protein) that provides capped 3'OH primers for transcription. We report TOSV endonuclease crystal structures in the apo form, in complex with a di-ketoacid inhibitor (DPBA) and in an intermediate state of inhibitor release, showing details on substrate binding and active site dynamics. The structure reveals substantial folding rearrangements absent in previously reported cap-snatching endonucleases. These include the relocation of the N terminus and the appearance of new structural motifs important for transcription and replication. The enzyme shows high activity rates comparable to other His+ cap-snatching endonucleases. Moreover, the activity is dependent on conserved residues involved in metal ion and substrate binding. Altogether, these results bring new light on the structure and function of cap-snatching endonucleases and pave the way for the development of specific and broad-spectrum antivirals.


Sujet(s)
Endonucleases/composition chimique , Endonucleases/métabolisme , Coiffes des ARN/métabolisme , Virus de la fièvre à phlébotomes de Naples/enzymologie , Protéines virales/composition chimique , Protéines virales/métabolisme , Biocatalyse , Domaine catalytique , Cations divalents/pharmacologie , Séquence conservée , Réplication de l'ADN/effets des médicaments et des substances chimiques , Antienzymes/pharmacologie , Stabilité enzymatique/effets des médicaments et des substances chimiques , Modèles moléculaires , Protéines mutantes/métabolisme , Domaines protéiques , Électricité statique , Sulfates/métabolisme , Transcription génétique/effets des médicaments et des substances chimiques
13.
J Virol ; 91(10)2017 05 15.
Article de Anglais | MEDLINE | ID: mdl-28250124

RÉSUMÉ

Crimean-Congo hemorrhagic fever virus (CCHFV) is a bunyavirus causing severe hemorrhagic fever disease in humans, with high mortality rates. The requirement of a high-containment laboratory and the lack of an animal model hampered the study of the immune response and protection of vaccine candidates. Using the recently developed interferon alpha receptor knockout (IFNAR-/-) mouse model, which replicates human disease, we investigated the immunogenicity and protection of two novel CCHFV vaccine candidates: a DNA vaccine encoding a ubiquitin-linked version of CCHFV Gc, Gn, and N and one using transcriptionally competent virus-like particles (tc-VLPs). In contrast to most studies that focus on neutralizing antibodies, we measured both humoral and cellular immune responses. We demonstrated a clear and 100% efficient preventive immunity against lethal CCHFV challenge with the DNA vaccine. Interestingly, there was no correlation with the neutralizing antibody titers alone, which were higher in the tc-VLP-vaccinated mice. However, the animals with a lower neutralizing titer, but a dominant cell-mediated Th1 response and a balanced Th2 response, resisted the CCHFV challenge. Moreover, we found that in challenged mice with a Th1 response (immunized by DNA/DNA and boosted by tc-VLPs), the immune response changed to Th2 at day 9 postchallenge. In addition, we were able to identify new linear B-cell epitope regions that are highly conserved between CCHFV strains. Altogether, our results suggest that a predominantly Th1-type immune response provides the most efficient protective immunity against CCHFV challenge. However, we cannot exclude the importance of the neutralizing antibodies as the surviving immunized mice exhibited substantial amounts of them.IMPORTANCE Crimean-Congo hemorrhagic fever virus (CCHFV) is responsible for hemorrhagic diseases in humans, with a high mortality rate. There is no FDA-approved vaccine, and there are still gaps in our knowledge of the immune responses to infection. The recently developed mouse models mimic human CCHF disease and are useful to study the immunogenicity and the protection by vaccine candidates. Our study shows that mice vaccinated with a specific DNA vaccine were fully protected. Importantly, we show that neutralizing antibodies are not sufficient for protection against CCHFV challenge but that an extra Th1-specific cellular response is required. Moreover, we describe the identification of five conserved B-cell epitopes, of which only one was previously known, that could be of great importance for the development of diagnostics tools and the improvement of vaccine candidates.


Sujet(s)
Protéines de capside/immunologie , Fièvre hémorragique de Crimée-Congo/immunologie , Fièvre hémorragique de Crimée-Congo/prévention et contrôle , Plasmides/génétique , Vaccins à ADN/immunologie , Vaccins à pseudo-particules virales/immunologie , Protéines de l'enveloppe virale/immunologie , Animaux , Anticorps neutralisants/sang , Protéines de capside/génétique , Modèles animaux de maladie humaine , Déterminants antigéniques des lymphocytes B/immunologie , Virus de la fièvre hémorragique de Crimée-Congo/composition chimique , Virus de la fièvre hémorragique de Crimée-Congo/génétique , Virus de la fièvre hémorragique de Crimée-Congo/immunologie , Fièvre hémorragique de Crimée-Congo/virologie , Humains , Immunité cellulaire , Immunisation , Immunogénicité des vaccins , Interféron alpha/déficit , Interféron alpha/génétique , Souris , Souris knockout , Plasmides/administration et posologie , Lymphocytes auxiliaires Th1 , Lymphocytes auxiliaires Th2 , Vaccins à ADN/administration et posologie , Vaccins à ADN/génétique , Vaccins à pseudo-particules virales/administration et posologie , Protéines de l'enveloppe virale/génétique
14.
Sci Rep ; 6: 26637, 2016 05 24.
Article de Anglais | MEDLINE | ID: mdl-27217069

RÉSUMÉ

Crimean Congo hemorrhagic fever virus (CCHFV) is a highly virulent tick-borne pathogen that causes hemorrhagic fever in humans. The geographic range of human CCHF cases largely reflects the presence of ticks. However, highly similar CCHFV lineages occur in geographically distant regions. Tick-infested migratory birds have been suggested, but not confirmed, to contribute to the dispersal. Bats have recently been shown to carry nairoviruses distinct from CCHFV. In order to assess the presence of CCHFV in a wide range of bat species over a wide geographic range, we analyzed 1,135 sera from 16 different bat species collected in Congo, Gabon, Ghana, Germany, and Panama. Using a CCHFV glycoprotein-based indirect immunofluorescence test (IIFT), we identified reactive antibodies in 10.0% (114/1,135) of tested bats, pertaining to 12/16 tested species. Depending on the species, 3.6%-42.9% of cave-dwelling bats and 0.6%-7.1% of foliage-living bats were seropositive (two-tailed t-test, p = 0.0447 cave versus foliage). 11/30 IIFT-reactive sera from 10 different African bat species had neutralizing activity in a virus-like particle assay. Neutralization of full CCHFV was confirmed in 5 of 7 sera. Widespread infection of cave-dwelling bats may indicate a role for bats in the life cycle and geographic dispersal of CCHFV.


Sujet(s)
Chiroptera , Virus de la fièvre hémorragique de Crimée-Congo , Fièvre hémorragique de Crimée-Congo , Afrique centrale/épidémiologie , Animaux , Chiroptera/sang , Chiroptera/virologie , Allemagne/épidémiologie , Fièvre hémorragique de Crimée-Congo/sang , Fièvre hémorragique de Crimée-Congo/épidémiologie , Humains , Panama/épidémiologie
15.
J Virol ; 89(11): 5957-67, 2015 Jun.
Article de Anglais | MEDLINE | ID: mdl-25810550

RÉSUMÉ

UNLABELLED: Crimean-Congo hemorrhagic fever virus (CCHFV; genus Nairovirus) is an extremely pathogenic member of the Bunyaviridae family. Since handling of the virus requires a biosafety level 4 (BSL-4) facility, little is known about pathomechanisms and host interactions. Here, we describe the establishment of a transcriptionally competent virus-like particle (tc-VLP) system for CCHFV. Recombinant polymerase (L), nucleocapsid protein (N) and a reporter minigenome expressed in human HuH-7 cells resulted in formation of transcriptionally active nucleocapsids that could be packaged by coexpressed CCHFV glycoproteins into tc-VLPs. The tc-VLPs resembled authentic virus particles in their protein composition and neutralization sensitivity to anti-CCHFV antibodies and could recapitulate all steps of the viral replication cycle. Particle attachment, entry, and primary transcription were modeled by infection of naive cells. The subsequent steps of genome replication, secondary transcription, and particle assembly and release can be obtained upon passaging the tc-VLPs on cells expressing CCHFV structural proteins. The utility of the VLP system was demonstrated by showing that the endonuclease domain of L is located around amino acid D693, as was predicted in silico by B. Morin et al. (PLoS Pathog 6:e1001038, 2010, http://dx.doi.org/10.1371/journal.ppat.1001038). The tc-VLP system will greatly facilitate studies and diagnostics of CCHFV under non-BSL-4 conditions. IMPORTANCE: Crimean-Congo hemorrhagic fever virus (CCHFV) is an extremely virulent pathogen of humans. Since the virus can be handled only at the highest biosafety level, research is restricted to a few specialized laboratories. We developed a plasmid-based system to produce virus-like particles with the ability to infect cells and transcribe a reporter genome. Due to the absence of viral genes, the virus-like particles are unable to spread or cause disease, thus allowing study of aspects of CCHFV biology under relaxed biosafety conditions.


Sujet(s)
Endonucleases/métabolisme , Virus de la fièvre hémorragique de Crimée-Congo/enzymologie , Virus de la fièvre hémorragique de Crimée-Congo/physiologie , Protéines virales/métabolisme , Virosomes/métabolisme , Lignée cellulaire , DNA-directed RNA polymerases/génétique , Endonucleases/génétique , Expression des gènes , Gènes rapporteurs , Virus de la fièvre hémorragique de Crimée-Congo/génétique , Hépatocytes/virologie , Humains , Protéines nucléocapside/génétique , Structure tertiaire des protéines , Transcription génétique , Protéines virales/génétique , Virosomes/génétique , Assemblage viral , Attachement viral , Pénétration virale
16.
Nucleic Acids Res ; 42(9): 6025-37, 2014 May.
Article de Anglais | MEDLINE | ID: mdl-24688060

RÉSUMÉ

Toscana virus is an emerging bunyavirus in Mediterranean Europe where it accounts for 80% of pediatric meningitis cases during the summer. The negative-strand ribonucleic acid (RNA) genome of the virus is wrapped around the virally encoded nucleoprotein N to form the ribonucleoprotein complex (RNP). We determined crystal structures of hexameric N alone (apo) and in complex with a nonameric single-stranded RNA. RNA is sequestered in a sequence-independent fashion in a deep groove inside the hexamer. At the junction between two adjacent copies of Ns, RNA binding induced an inter-subunit rotation, which opened the RNA-binding tunnel and created a new assembly interface at the outside of the hexamer. Based on these findings, we suggest a structural model for how binding of RNA to N promotes the formation of helical RNPs, which are a characteristic hallmark of many negative-strand RNA viruses.


Sujet(s)
Protéines nucléocapside/composition chimique , ARN viral/composition chimique , Virus de la fièvre à phlébotomes de Naples/physiologie , Sites de fixation , Cristallographie aux rayons X , Modèles moléculaires , Liaison aux protéines , Multimérisation de protéines , Structure quaternaire des protéines , Structure secondaire des protéines , Assemblage viral
17.
Cell Host Microbe ; 13(3): 336-46, 2013 Mar 13.
Article de Anglais | MEDLINE | ID: mdl-23498958

RÉSUMÉ

Host defense to RNA viruses depends on rapid intracellular recognition of viral RNA by two cytoplasmic RNA helicases: RIG-I and MDA5. RNA transfection experiments indicate that RIG-I responds to naked double-stranded RNAs (dsRNAs) with a triphosphorylated 5' (5'ppp) terminus. However, the identity of the RIG-I stimulating viral structures in an authentic infection context remains unresolved. We show that incoming viral nucleocapsids containing a 5'ppp dsRNA "panhandle" structure trigger antiviral signaling that commences with RIG-I, is mediated through the adaptor protein MAVS, and terminates with transcription factor IRF-3. Independent of mammalian cofactors or viral polymerase activity, RIG-I bound to viral nucleocapsids, underwent a conformational switch, and homo-oligomerized. Enzymatic probing and superresolution microscopy suggest that RIG-I interacts with the panhandle structure of the viral nucleocapsids. These results define cytoplasmic entry of nucleocapsids as the proximal RIG-I-sensitive step during infection and establish viral nucleocapsids with a 5'ppp dsRNA panhandle as a RIG-I activator.


Sujet(s)
DEAD-box RNA helicases/immunologie , Nucléocapside/immunologie , Infections à virus à ARN/enzymologie , Infections à virus à ARN/immunologie , Virus à ARN/immunologie , Protéines adaptatrices de la transduction du signal/génétique , Protéines adaptatrices de la transduction du signal/immunologie , Protéine-58 à domaine DEAD , DEAD-box RNA helicases/composition chimique , DEAD-box RNA helicases/génétique , Génome viral , Interactions hôte-pathogène , Humains , Nucléocapside/composition chimique , Nucléocapside/génétique , Polyphosphates/métabolisme , Infections à virus à ARN/génétique , Infections à virus à ARN/virologie , Virus à ARN/composition chimique , Virus à ARN/génétique , ARN viral/composition chimique , ARN viral/génétique , ARN viral/immunologie , Récepteurs immunologiques , Transduction du signal
18.
J Virol ; 86(22): 12294-303, 2012 Nov.
Article de Anglais | MEDLINE | ID: mdl-22951837

RÉSUMÉ

Crimean-Congo hemorrhagic fever, a severe hemorrhagic disease found throughout Africa, Europe, and Asia, is caused by the tick-borne Crimean-Congo hemorrhagic fever virus (CCHFV). CCHFV is a negative-sense single-stranded RNA (ssRNA) virus belonging to the Nairovirus genus of the Bunyaviridae family. Its genome of three single-stranded RNA segments is encapsidated by the nucleocapsid protein (CCHFV N) to form the ribonucleoprotein complex. This ribonucleoprotein complex is required during replication and transcription of the viral genomic RNA. Here, we present the crystal structures of the CCHFV N in two distinct forms, an oligomeric form comprised of double antiparallel superhelices and a monomeric form. The head-to-tail interaction of the stalk region of one CCHFV N subunit with the base of the globular body of the adjacent subunit stabilizes the helical organization of the oligomeric form of CCHFV N. It also masks the conserved caspase-3 cleavage site present at the tip of the stalk region from host cell caspase-3 interaction and cleavage. By incubation with primer-length ssRNAs, we also obtained the crystal structure of CCHFV N in its monomeric form, which is similar to a recently published structure. The conformational change of CCHFV N upon deoligomerization results in the exposure of the caspase-3 cleavage site and subjects CCHFV N to caspase-3 cleavage. Mutations of this cleavage site inhibit cleavage by caspase-3 and result in enhanced viral polymerase activity. Thus, cleavage of CCHFV N by host cell caspase-3 appears to be crucial for controlling viral RNA synthesis and represents an important host defense mechanism against CCHFV infection.


Sujet(s)
Caspase-3/métabolisme , Virus de la fièvre hémorragique de Crimée-Congo/métabolisme , Nucléoprotéines/composition chimique , Sites de fixation , Clonage moléculaire , Cristallographie aux rayons X/méthodes , Génome viral , Virus de la fièvre hémorragique de Crimée-Congo/composition chimique , Humains , Modèles moléculaires , Conformation moléculaire , Liaison aux protéines , Conformation des protéines , Structure tertiaire des protéines , ARN/métabolisme , ARN viral/métabolisme , Transcription génétique
19.
PLoS One ; 5(7): e11671, 2010 Jul 20.
Article de Anglais | MEDLINE | ID: mdl-20652028

RÉSUMÉ

BACKGROUND: Deciphering host responses contributing to dengue shock syndrome (DSS), the life-threatening form of acute viral dengue infections, is required to improve both the differential prognosis and the treatments provided to DSS patients, a challenge for clinicians. METHODOLOGY/PRINCIPAL FINDINGS: Based on a prospective study, we analyzed the genome-wide expression profiles of whole blood cells from 48 matched Cambodian children: 19 progressed to DSS while 16 and 13 presented respectively classical dengue fever (DF) or dengue hemorrhagic fever grades I/II (DHF). Using multi-way analysis of variance (ANOVA) and adjustment of p-values to control the False Discovery Rate (FDR<10%), we identified a signature of 2959 genes differentiating DSS patients from both DF and DHF, and showed a strong association of this DSS-gene signature with the dengue disease phenotype. Using a combined approach to analyse the molecular patterns associated with the DSS-gene signature, we provide an integrative overview of the transcriptional responses altered in DSS children. In particular, we show that the transcriptome of DSS children blood cells is characterized by a decreased abundance of transcripts related to T and NK lymphocyte responses and by an increased abundance of anti-inflammatory and repair/remodeling transcripts. We also show that unexpected pro-inflammatory gene patterns at the interface between innate immunity, inflammation and host lipid metabolism, known to play pathogenic roles in acute and chronic inflammatory diseases associated with systemic vascular dysfunction, are transcriptionnally active in the blood cells of DSS children. CONCLUSIONS/SIGNIFICANCE: We provide a global while non exhaustive overview of the molecular mechanisms altered in of DSS children and suggest how they may interact to lead to final vascular homeostasis breakdown. We suggest that some mechanisms identified should be considered putative therapeutic targets or biomarkers of progression to DSS.


Sujet(s)
Analyse de profil d'expression de gènes , Immunité innée/immunologie , Dengue sévère/immunologie , Adolescent , Analyse de variance , Enfant , Enfant d'âge préscolaire , Biologie informatique , Femelle , Humains , Nourrisson , Inflammation/immunologie , Métabolisme lipidique , Mâle , Séquençage par oligonucléotides en batterie , Réaction de polymérisation en chaîne , Études prospectives
20.
Emerg Infect Dis ; 15(9): 1385-94, 2009 Sep.
Article de Anglais | MEDLINE | ID: mdl-19788805

RÉSUMÉ

Chlamydia trachomatis is a global cause of blinding trachoma and sexually transmitted infections (STIs). We used comparative genomics of the family Chlamydiaceae to select conserved housekeeping genes for C. trachomatis multilocus sequencing, characterizing 19 reference and 68 clinical isolates from 6 continental/subcontinental regions. There were 44 sequence types (ST). Identical STs for STI isolates were recovered from different regions, whereas STs for trachoma isolates were restricted by continent. Twenty-nine of 52 alleles had nonuniform distributions of frequencies across regions (p<0.001). Phylogenetic analysis showed 3 disease clusters: invasive lymphogranuloma venereum strains, globally prevalent noninvasive STI strains (ompA genotypes D/Da, E, and F), and nonprevalent STI strains with a trachoma subcluster. Recombinant strains were observed among STI clusters. Single nucleotide polymorphisms (SNPs) were predictive of disease specificity. Multilocus and SNP typing can now be used to detect diverse and emerging C. trachomatis strains for epidemiologic and evolutionary studies of trachoma and STI populations worldwide.


Sujet(s)
Protéines bactériennes/génétique , Techniques de typage bactérien , Infections à Chlamydia/microbiologie , Chlamydia trachomatis/classification , Maladies transmissibles émergentes/microbiologie , Lymphogranulomatose vénérienne/microbiologie , Trachome/microbiologie , Infections à Chlamydia/anatomopathologie , Chlamydia trachomatis/génétique , Chlamydia trachomatis/isolement et purification , Maladies transmissibles émergentes/anatomopathologie , ADN bactérien/analyse , Génomique , Humains , Lymphogranulomatose vénérienne/anatomopathologie , Phénotype , Phylogenèse , Polymorphisme de nucléotide simple , Valeur prédictive des tests , Recombinaison génétique , Analyse de séquence d'ADN , Maladies sexuellement transmissibles/microbiologie , Maladies sexuellement transmissibles/anatomopathologie , Spécificité d'espèce , Trachome/anatomopathologie
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE
...