Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 1.383
Filtrer
1.
Methods Mol Biol ; 2824: 1-14, 2024.
Article de Anglais | MEDLINE | ID: mdl-39039402

RÉSUMÉ

Rift Valley fever virus (RVFV) is a pathogen transmitted to humans and livestock via mosquito bites. This virus, which was discovered in Kenya in 1930, is considered by the World Health Organization (WHO) and the World Organisation for Animal Health (WOAH) to be associated with a high risk of causing large-scale epidemics. However, means dedicated to fighting RVFV have been limited, and despite recent research efforts, the virus remains poorly understood at both the molecular and cellular levels as well as at a broader scale of research in the field and in animal and human populations. In this introductory chapter of a methods book, we aim to provide readers with a concise overview of RVFV, from its ecology and transmission to the structural and genomic organization of virions and its life cycle in host cells.


Sujet(s)
Fièvre de la Vallée du Rift , Virus de la fièvre de la vallée du Rift , Virus de la fièvre de la vallée du Rift/génétique , Fièvre de la Vallée du Rift/transmission , Fièvre de la Vallée du Rift/virologie , Animaux , Humains , Génome viral
2.
Methods Mol Biol ; 2824: 27-34, 2024.
Article de Anglais | MEDLINE | ID: mdl-39039404

RÉSUMÉ

Several techniques have been developed to diagnose Rift Valley fever infection. Viral isolation is one of the most difficult techniques to apply but offers great opportunities for further research. It is useful, for example, for the development of an accurate diagnostic test suitable for screening for Rift Valley fever virus infection, specific treatments by testing known antiviral molecules that act on the replication cycle to assess their therapeutic or even prophylactic potential, therapeutic applications, and vaccine candidates. Understanding how the virus replicates and interacts with the host cell and organism and identifying biomarkers of infection or new targets for the development of treatments are made possible through field virus isolates. Biosafety level 3 conditions are a pre-requisite for viral isolation by a trained staff member. Here, we describe the procedure to isolate Rift Valley fever virus from field samples by cell culture.


Sujet(s)
Techniques de culture cellulaire , Fièvre de la Vallée du Rift , Virus de la fièvre de la vallée du Rift , Virus de la fièvre de la vallée du Rift/isolement et purification , Fièvre de la Vallée du Rift/virologie , Fièvre de la Vallée du Rift/diagnostic , Animaux , Techniques de culture cellulaire/méthodes , Humains , Cellules Vero , Chlorocebus aethiops
3.
Methods Mol Biol ; 2824: 67-80, 2024.
Article de Anglais | MEDLINE | ID: mdl-39039406

RÉSUMÉ

RT-qPCR allows the detection of viruses and the monitoring of viral replication. This technique was extensively employed during the SARS-CoV-2 pandemic, where it demonstrated its efficiency and robustness. Here we describe the analysis of Rift Valley fever and Toscana virus infections over time, achieved through the RT-qPCR quantification of the viral genome. We further elaborate on the method to discriminate between genomic and antigenomic viral RNAs by using primers specific for each strand during the reverse transcription step.


Sujet(s)
ARN viral , Fièvre de la Vallée du Rift , Virus de la fièvre de la vallée du Rift , Virus de la fièvre de la vallée du Rift/génétique , ARN viral/génétique , Fièvre de la Vallée du Rift/virologie , Fièvre de la Vallée du Rift/diagnostic , Humains , Génome viral , Réaction de polymérisation en chaine en temps réel/méthodes , Réplication virale/génétique , Animaux
4.
Methods Mol Biol ; 2824: 35-65, 2024.
Article de Anglais | MEDLINE | ID: mdl-39039405

RÉSUMÉ

Rift Valley fever virus (RVFV) is a globally important mosquito-borne virus that can also be directly transmitted via aerosolization of body fluids from infected animals. RVFV outbreaks cause mass mortality of young livestock and abortions in animals. In most severe human cases, the disease can progress to hemorrhagic fever and encephalitis, leading to death. RVF has a significant economic impact due to the loss of livestock that is a great challenge for people who depend on animals for income and food. Several vaccines are available for animal use, but none are yet licensed for use in human populations. This situation emphasizes the need to have robust and efficient diagnostic methods that can be used for early case confirmation, assessment of seroprevalence, and virus surveillance as well as vaccine efficacy evaluation. Despite the existence of different diagnostic methods for RVFV, we still have untimely reporting or underreporting of cases, probably due to lack of appropriate surveillance systems or diagnostic tools in some endemic countries. Here, we describe different methods available for detection and diagnosis of RVFV.


Sujet(s)
Fièvre de la Vallée du Rift , Virus de la fièvre de la vallée du Rift , Virus de la fièvre de la vallée du Rift/isolement et purification , Virus de la fièvre de la vallée du Rift/immunologie , Fièvre de la Vallée du Rift/diagnostic , Fièvre de la Vallée du Rift/épidémiologie , Animaux , Humains , Anticorps antiviraux/immunologie , Anticorps antiviraux/sang , Test ELISA/méthodes
5.
Methods Mol Biol ; 2824: 15-25, 2024.
Article de Anglais | MEDLINE | ID: mdl-39039403

RÉSUMÉ

Vector competence assays allow to measure, in the laboratory, the ability of a mosquito to get infected and then retransmit an arbovirus while mimicking natural vector infection route. Aedes aegypti is a major vector of arboviruses worldwide and thus a reference species used in vector competence assays. Rift Valley fever virus (RVFV) is a major public health threat, mostly in Africa, that infects humans and animals through the bite of mosquito vectors. Here, we describe vector competence assay of Aedes aegypti mosquitoes for RVFV, from mosquito exposure to the virus through an infectious artificial blood meal to the measurement of virus prevalence in the mosquito's body, head, and saliva.


Sujet(s)
Aedes , Vecteurs moustiques , Fièvre de la Vallée du Rift , Virus de la fièvre de la vallée du Rift , Animaux , Aedes/virologie , Virus de la fièvre de la vallée du Rift/physiologie , Virus de la fièvre de la vallée du Rift/isolement et purification , Vecteurs moustiques/virologie , Fièvre de la Vallée du Rift/transmission , Fièvre de la Vallée du Rift/virologie , Salive/virologie , Humains
6.
Methods Mol Biol ; 2824: 81-89, 2024.
Article de Anglais | MEDLINE | ID: mdl-39039407

RÉSUMÉ

The Rift Valley fever virus (RVFV) is an arthropod-borne, zoonotic, hemorrhagic fever virus that can cause severe diseases both in livestock and humans. The spread of RVFV in areas previously considered as non-endemic together with the absence of licensed vaccines for use in humans and animals poses a major health and economic threat worldwide. It is therefore crucial to make major progresses in our understanding and management of this virus and its zoonosis. RVFV is considered a bioterrorism pathogen, and, thus, only a few institutes, facilities, and personnel are legally authorized to detain it and handle it. Moreover, this virus must be manipulated in a biosafety level 3 (BSL3) laboratory following strict biosafety protocols to ensure that biosecurity's highest standards are met. Only certain attenuated strains such as the MP12 strain can be handled in BSL2 laboratories, depending on the country considered. To assist researchers in working with RVFV in the safest possible conditions, this chapter presents validated methods for effective RVFV decontamination and inactivation.


Sujet(s)
Décontamination , Virus de la fièvre de la vallée du Rift , Inactivation virale , Animaux , Décontamination/méthodes , Humains , Fièvre de la Vallée du Rift/prévention et contrôle , Fièvre de la Vallée du Rift/transmission , Fièvre de la Vallée du Rift/virologie , Confinement de risques biologiques/méthodes , Cellules Vero , Chlorocebus aethiops
7.
Methods Mol Biol ; 2824: 91-104, 2024.
Article de Anglais | MEDLINE | ID: mdl-39039408

RÉSUMÉ

Rift Valley fever virus (RVFV) is an arthropod-borne virus (arbovirus) responsible for a severe zoonotic disease affecting a wide range of domestic and wild ruminants as well as humans. RVFV is endemic in many African countries and has also caused outbreaks in Madagascar and Arabian Peninsula. With regard to its wide geographical distribution, its potential to emerge in a new area, and its capability to trigger major health and economic crisis, it is essential to study and better understand several aspects of its life cycle and, in particular, its interactions with mammalian hosts and arthropod vectors. To do so, it is key for researchers to be able to amplify in vitro viral strains isolated from the field and determine accurately the viral titers of RVFV stocks. In this chapter, we present protocols that can be easily implemented to produce and titrate RVFV stocks in your laboratory.


Sujet(s)
Fièvre de la Vallée du Rift , Virus de la fièvre de la vallée du Rift , Virus de la fièvre de la vallée du Rift/isolement et purification , Animaux , Fièvre de la Vallée du Rift/virologie , Humains , Charge virale , Chlorocebus aethiops , Cellules Vero , Culture virale/méthodes
8.
Methods Mol Biol ; 2824: 147-164, 2024.
Article de Anglais | MEDLINE | ID: mdl-39039412

RÉSUMÉ

Single-domain antibodies, referred to as VHH (variable heavy chains of heavy chain-only antibodies) or in their commercial name as nanobodies, are potent tools for the detection of target proteins in biological samples. They have the advantage of being highly stable, specific, and sensitive, with affinities reaching the nanomolar range. We utilized this tool to develop a rapid detection method that discriminates cells infected with Rift Valley fever virus (RVFV), based on the intracellular detection of the viral nonstructural NSm protein localized on the outer membrane of mitochondria. Here we describe how NSm-specific VHHs have been produced, cloned, and characterized, highlighting their value in RVFV research and diagnosis. This work may also raise interest in other potential applications such as antiviral therapy.


Sujet(s)
Fièvre de la Vallée du Rift , Virus de la fièvre de la vallée du Rift , Anticorps à domaine unique , Protéines virales non structurales , Virus de la fièvre de la vallée du Rift/immunologie , Anticorps à domaine unique/immunologie , Humains , Fièvre de la Vallée du Rift/immunologie , Fièvre de la Vallée du Rift/diagnostic , Fièvre de la Vallée du Rift/virologie , Protéines virales non structurales/immunologie , Animaux , Anticorps antiviraux/immunologie
9.
Methods Mol Biol ; 2824: 165-188, 2024.
Article de Anglais | MEDLINE | ID: mdl-39039413

RÉSUMÉ

Rift Valley fever virus (RVFV) is a mosquito-borne pathogen that represents a significant threat to both human and veterinary public health. Since its discovery in the Great Rift Valley of Kenya in the 1930s, the virus has spread across Africa and beyond, now posing a risk of introduction into Southern Europe and Asia. Despite recent progresses, early RVFV-host cell interactions remain largely uncharacterized. In this method chapter, we delineate the procedure for labeling RVFV particles with fluorescent organic dyes. This approach makes it feasible to visualize single viral particles in both fixed and living cells and study RVFV entry into host cells. We provide additional examples with two viruses closely related to RVFV, namely, Toscana virus and Uukuniemi virus. Furthermore, we illustrate how to utilize fluorescent viral particles to examine and quantify each step of the cell entry program of RVFV, which includes state-of-the-art fluorescence-based detection techniques such as fluorescence microscopy, flow cytometry, and fluorimetry.


Sujet(s)
Colorants fluorescents , Microscopie de fluorescence , Virus de la fièvre de la vallée du Rift , Virion , Virus de la fièvre de la vallée du Rift/isolement et purification , Humains , Virion/isolement et purification , Animaux , Colorants fluorescents/composition chimique , Microscopie de fluorescence/méthodes , Cytométrie en flux/méthodes , Pénétration virale , Fièvre de la Vallée du Rift/virologie , Fièvre de la Vallée du Rift/diagnostic , Coloration et marquage/méthodes , Lignée cellulaire
10.
Methods Mol Biol ; 2824: 189-202, 2024.
Article de Anglais | MEDLINE | ID: mdl-39039414

RÉSUMÉ

Affinity enrichment coupled with liquid chromatography-tandem mass spectrometry (AE-LC-MS/MS) enables a comprehensive study of virus-host protein-protein interactions in cells and tissues infected with Rift Valley fever virus (RVFV) or ectopically expressing RVFV proteins. Depending on the research question, different experimental setups with carefully chosen controls are needed. Here, we describe the detailed workflow of sample preparation, processing, and cleanup, while also outlining critical points to consider when designing and performing AE-LC-MS/MS experiments.


Sujet(s)
Interactions hôte-pathogène , Protéomique , Virus de la fièvre de la vallée du Rift , Spectrométrie de masse en tandem , Protéomique/méthodes , Spectrométrie de masse en tandem/méthodes , Chromatographie en phase liquide/méthodes , Humains , Protéines virales/métabolisme , Fièvre de la Vallée du Rift/virologie , Fièvre de la Vallée du Rift/métabolisme , Animaux
11.
Methods Mol Biol ; 2824: 281-318, 2024.
Article de Anglais | MEDLINE | ID: mdl-39039419

RÉSUMÉ

Rift Valley fever virus (RVFV; genus Phlebovirus, family Phenuiviridae, order Bunyavirales) is a mosquito-borne zoonotic pathogen endemic in Africa. Its negative-stranded genomic RNA (vRNA) is divided into three segments termed L, M, and S. Both vRNAs and antigenomic cRNAs are encapsidated by viral nucleoprotein (N) to form nucleocapsids, which constitute the template for genome transcription and replication. Based on a number of electron microscopy and structural studies, the viral RNAs of negative-strand RNA viruses, including phleboviruses, are commonly considered to be entirely and uniformly covered by N protein. However, high resolution data supporting this notion was missing to date.Here, we describe a method how to globally map all N-RNA interactions of RVFV by using iCLIP (individual-nucleotide resolution UV cross-linking and immunoprecipitation). The protocol is based on covalent cross-linking of direct protein-RNA interactions by UV irradiation. Following sample lysis, a selective isolation of N in complex with its RNA targets is achieved by immunoprecipitation. Then, N-RNA complexes are separated by SDS-PAGE, and after membrane transfer, RNA is isolated and subjected to library preparation and high-throughput sequencing. We explain how the standard iCLIP protocol can be adapted to RVFV N-RNA interaction studies. The protocol describes mapping of all N interactions with the vRNAs and cRNAs derived either from RVFV particles or from infected cells.


Sujet(s)
Génome viral , Nucléoprotéines , ARN viral , Virus de la fièvre de la vallée du Rift , Virus de la fièvre de la vallée du Rift/génétique , ARN viral/génétique , ARN viral/métabolisme , Nucléoprotéines/métabolisme , Nucléoprotéines/génétique , Cartographie nucléotidique/méthodes , Immunoprécipitation/méthodes , Humains , Fièvre de la Vallée du Rift/virologie , Fièvre de la Vallée du Rift/métabolisme , Animaux
12.
Methods Mol Biol ; 2824: 385-395, 2024.
Article de Anglais | MEDLINE | ID: mdl-39039425

RÉSUMÉ

Rift Valley fever (RVF) caused by Rift Valley fever virus (RVFV) is a major health concern for both domesticated animals and humans in certain endemic areas of Africa. With changing environmental conditions and identification of vectors capable of transmitting the virus, there is high risk of RVFV spreading into other parts of the world. Furthermore, unavailability of effective vaccines in the event of an outbreak can be a major challenge as witnessed recently in case of SARS-CoV2 pandemic. Hence, identifying potential vaccines and testing their protective efficacy in preclinical models before clinical testing is the absolute need of the hour. Here, we describe methods used to quantify virus-specific T cell responses in mice that were immunized with RVFV strains or antigens.


Sujet(s)
Virus de la fièvre de la vallée du Rift , Lymphocytes T , Vaccins antiviraux , Animaux , Souris , Lymphocytes T/immunologie , Virus de la fièvre de la vallée du Rift/immunologie , Vaccins antiviraux/immunologie , Fièvre de la Vallée du Rift/immunologie , Fièvre de la Vallée du Rift/prévention et contrôle , Immunisation/méthodes , Vaccination/méthodes , Antigènes viraux/immunologie
13.
Methods Mol Biol ; 2824: 373-383, 2024.
Article de Anglais | MEDLINE | ID: mdl-39039424

RÉSUMÉ

RNAseq is a valuable tool that can aid researchers in uncovering the transcriptional changes that occur when a viral pathogen infects a host cell. Viral infection will invariably cause differential expression of many genes, from transcription of mRNA to alternative splicing and degradation. This change in gene expression can be a result of immune activation or a direct activity of the virus to alter the host cell's environment to make it more favorable for viral replication. Studying the innate immune response to a pathogen can reveal which cellular pathways are active, indicating the steps that the host takes to halt viral infection, and detecting virus-mediated mRNA expression changes can help with identifying the pathways which may be exploited by the virus. Gene expression changes-both cell-caused and virus-caused-can be studied through RNAseq, helping to provide a clearer picture of the cellular changes that occur during viral infection. In this protocol, we outline methods to carry out mRNA sequencing in Rift Valley fever virus-infected cell cultures, from infection to library prep and analysis.


Sujet(s)
Fièvre de la Vallée du Rift , Virus de la fièvre de la vallée du Rift , Virus de la fièvre de la vallée du Rift/génétique , Virus de la fièvre de la vallée du Rift/physiologie , Humains , Fièvre de la Vallée du Rift/virologie , Fièvre de la Vallée du Rift/génétique , Interactions hôte-pathogène/génétique , Analyse de séquence d'ARN/méthodes , ARN messager/génétique , ARN messager/métabolisme , Animaux , Réplication virale/génétique , Épissage alternatif , Épissage des ARN , Transcription génétique , Lignée cellulaire
14.
Methods Mol Biol ; 2824: 447-459, 2024.
Article de Anglais | MEDLINE | ID: mdl-39039429

RÉSUMÉ

Rift Valley fever virus is able to infect multiple organs and cell types, and the course of infection varies between viral strains and between individuals in particular according to age, genetic background, and physiological status. Studies on viral and host factors involve detecting and quantifying viral load at multiple time points and in multiple tissues. While this is classically performed by genome quantification or viral titration, in vivo imaging techniques using recombinant viruses expressing a bioluminescent or fluorescent protein allow noninvasive longitudinal studies on the same group of mice over the entire course of disease and the detection of unsuspected sites of infection. Here, we describe the protocol to monitor and characterize mouse infection with Rift Valley fever virus by in vivo imaging using recombinant viruses expressing light-emitting reporter genes.


Sujet(s)
Gènes rapporteurs , Mesures de luminescence , Virus de la fièvre de la vallée du Rift , Animaux , Souris , Mesures de luminescence/méthodes , Virus de la fièvre de la vallée du Rift/génétique , Fièvre de la Vallée du Rift/virologie , Fièvre de la Vallée du Rift/diagnostic , Charge virale/méthodes , Modèles animaux de maladie humaine , Humains , Protéines luminescentes/génétique , Protéines luminescentes/métabolisme
15.
Methods Mol Biol ; 2824: 425-445, 2024.
Article de Anglais | MEDLINE | ID: mdl-39039428

RÉSUMÉ

Rift Valley fever virus (RVFV) is an arboviral pathogen of clinical and agricultural relevance. The ongoing development of targeted RVFV prophylactics and therapeutics is overwhelmingly dependent on animal models due to both natural, that is, sporadic outbreaks, and structural, for example, underresourcing of endemic regions, limitations in accessing human patient samples and cohorts. Elucidating mechanisms of viral pathogenesis and testing therapeutics is further complicated by the diverse manifestations of RVFV disease and the heterogeneity of the host response to infection. In this chapter, we describe major clinical manifestations of RVFV infection and discuss the laboratory animal models used to study each.


Sujet(s)
Modèles animaux de maladie humaine , Fièvre de la Vallée du Rift , Virus de la fièvre de la vallée du Rift , Fièvre de la Vallée du Rift/virologie , Animaux , Virus de la fièvre de la vallée du Rift/pathogénicité , Humains , Souris , Animaux de laboratoire/virologie
16.
Emerg Microbes Infect ; 13(1): 2373313, 2024 Dec.
Article de Anglais | MEDLINE | ID: mdl-38946528

RÉSUMÉ

Rift Valley fever (RVF) is a mosquito-borne zoonotic disease caused by RVF virus (RVFV). RVFV infections in humans are usually asymptomatic or associated with mild febrile illness, although more severe cases of haemorrhagic disease and encephalitis with high mortality also occur. Currently, there are no licensed human vaccines available. The safety and efficacy of a genetically engineered four-segmented RVFV variant (hRVFV-4s) as a potential live-attenuated human vaccine has been tested successfully in mice, ruminants, and marmosets though the correlates of protection of this vaccine are still largely unknown. In the present study, we have assessed hRVFV-4s-induced humoral and cellular immunity in a mouse model of RVFV infection. Our results confirm that a single dose of hRVFV-4s is highly efficient in protecting naïve mice from developing severe disease following intraperitoneal challenge with a highly virulent RVFV strain and data show that virus neutralizing (VN) serum antibody titres in a prime-boost regimen are significantly higher compared to the single dose. Subsequently, VN antibodies from prime-boost-vaccinated recipients were shown to be protective when transferred to naïve mice. In addition, hRVFV-4s vaccination induced a significant virus-specific T cell response as shown by IFN-γ ELISpot assay, though these T cells did not provide significant protection upon passive transfer to naïve recipient mice. Collectively, this study highlights hRVFV-4s-induced VN antibodies as a major correlate of protection against lethal RVFV infection.


Sujet(s)
Anticorps neutralisants , Anticorps antiviraux , Fièvre de la Vallée du Rift , Virus de la fièvre de la vallée du Rift , Vaccins atténués , Vaccins antiviraux , Animaux , Virus de la fièvre de la vallée du Rift/immunologie , Virus de la fièvre de la vallée du Rift/génétique , Fièvre de la Vallée du Rift/prévention et contrôle , Fièvre de la Vallée du Rift/immunologie , Vaccins antiviraux/immunologie , Vaccins antiviraux/administration et posologie , Souris , Anticorps antiviraux/sang , Anticorps antiviraux/immunologie , Anticorps neutralisants/sang , Anticorps neutralisants/immunologie , Femelle , Vaccins atténués/immunologie , Vaccins atténués/administration et posologie , Modèles animaux de maladie humaine , Immunité cellulaire , Lymphocytes T/immunologie , Immunité humorale , Souris de lignée BALB C , Interféron gamma/immunologie , Vaccination
17.
Viruses ; 16(7)2024 Jun 21.
Article de Anglais | MEDLINE | ID: mdl-39066162

RÉSUMÉ

Rift Valley fever (RVF) is a mosquito-borne zoonotic viral disease endemic to Africa and the Middle East. Live-attenuated RVF vaccines have been studied for both veterinary and human use due to their strong immunogenicity and cost-effective manufacturing. The live-attenuated MP-12 vaccine has been conditionally approved for veterinary use in the U.S.A., and next-generation live-attenuated RVF vaccine candidates are being actively researched. Assessing the virulence phenotype of vaccine seeds or lots is crucial for managing vaccine safety. Previously, preweaning 19-day-old outbred CD1 mice have been used to evaluate the MP-12 strain. This study aimed to characterize the relative virulence of three live-attenuated RVF vaccine strains in 19-day-old inbred C57BL/6 mice: the recombinant MP-12 (rMP-12), the RVax-1, and the ∆NSs-∆NSm-rZH501 strains. Although this mouse model did not show dose-dependent pathogenesis, mice that succumbed to the infection exhibited distinct brain pathology. Mice infected with ∆NSs-∆NSm-rZH501 showed an infiltration of inflammatory cells associated with infected neurons, and focal lesions formed around virus-infected cells. In contrast, mice infected with rMP-12 or RVax-1 showed a minimal association of inflammatory cells in the brain, yet the virus spread diffusely. The preweaning model is likely useful for evaluating host responses to attenuated RVFV strains, although further refinement may be necessary to quantitate the virulence among different RVFV strains or vaccine lots.


Sujet(s)
Modèles animaux de maladie humaine , Souris de lignée C57BL , Fièvre de la Vallée du Rift , Virus de la fièvre de la vallée du Rift , Vaccins atténués , Vaccins antiviraux , Animaux , Virus de la fièvre de la vallée du Rift/pathogénicité , Virus de la fièvre de la vallée du Rift/immunologie , Virus de la fièvre de la vallée du Rift/génétique , Fièvre de la Vallée du Rift/virologie , Fièvre de la Vallée du Rift/anatomopathologie , Fièvre de la Vallée du Rift/prévention et contrôle , Fièvre de la Vallée du Rift/immunologie , Souris , Vaccins atténués/immunologie , Vaccins atténués/administration et posologie , Vaccins antiviraux/immunologie , Vaccins antiviraux/administration et posologie , Virulence , Femelle
18.
Viruses ; 16(7)2024 Jun 25.
Article de Anglais | MEDLINE | ID: mdl-39066182

RÉSUMÉ

Rift Valley fever (RVF) is a re-emerging vector-borne zoonosis with a high public health and veterinary impact. In West Africa, many lineages were previously detected, but since 2020, lineage H from South Africa has been the main cause of the outbreaks. In this study, clinical samples collected through national surveillance were screened for RVF virus (RVFV) acute infection by RT-PCR and IgM ELISA tests. Sequencing, genome mapping and in vitro phenotypic characterization in mammal cells were performed on RT-PCR positive samples in comparison with other epidemic lineages (G and C). Four RVFV human cases were detected in Senegal and the sequence analyses revealed that the strains belonged to lineage H. The in vitro kinetics and genome mapping showed different replication efficiency profiles for the tested RVFV lineages and non-conservative mutations, which were more common to lineage G or specific to lineage H. Our findings showed the re-emergence of lineage H in Senegal in 2022, its high viral replication efficiency in vitro and support the findings that genetic diversity affects viral replication. This study gives new insights into the biological properties of lineage H and calls for deeper studies to better assess its potential to cause a future threat in Senegal.


Sujet(s)
Génome viral , Phylogenèse , Fièvre de la Vallée du Rift , Virus de la fièvre de la vallée du Rift , Réplication virale , Virus de la fièvre de la vallée du Rift/génétique , Virus de la fièvre de la vallée du Rift/isolement et purification , Virus de la fièvre de la vallée du Rift/classification , Virus de la fièvre de la vallée du Rift/physiologie , Fièvre de la Vallée du Rift/virologie , Fièvre de la Vallée du Rift/épidémiologie , Fièvre de la Vallée du Rift/transmission , Sénégal/épidémiologie , Humains , Animaux , Maladies transmissibles émergentes/virologie , Maladies transmissibles émergentes/épidémiologie , Maladies transmissibles émergentes/médecine vétérinaire , Épidémies de maladies , Afrique de l'Ouest/épidémiologie , Variation génétique , Mutation
19.
Viruses ; 16(7)2024 Jul 17.
Article de Anglais | MEDLINE | ID: mdl-39066310

RÉSUMÉ

Rift Valley fever (RVF), a mosquito-borne transboundary zoonosis, was first confirmed in Rwanda's livestock in 2012 and since then sporadic cases have been reported almost every year. In 2018, the country experienced its first large outbreak, which was followed by a second one in 2022. To determine the circulating virus lineages and their ancestral origin, two genome sequences from the 2018 outbreak, and thirty-six, forty-one, and thirty-eight sequences of small (S), medium (M), and large (L) genome segments, respectively, from the 2022 outbreak were generated. All of the samples from the 2022 outbreak were collected from slaughterhouses. Both maximum likelihood and Bayesian-based phylogenetic analyses were performed. The findings showed that RVF viruses belonging to a single lineage, C, were circulating during the two outbreaks, and shared a recent common ancestor with RVF viruses isolated in Uganda between 2016 and 2019, and were also linked to the 2006/2007 largest East Africa RVF outbreak reported in Kenya, Tanzania, and Somalia. Alongside the wild-type viruses, genetic evidence of the RVFV Clone 13 vaccine strain was found in slaughterhouse animals, demonstrating a possible occupational risk of exposure with unknown outcome for people working in meat-related industry. These results provide additional evidence of the ongoing wide spread of RVFV lineage C in Africa and emphasize the need for an effective national and international One Health-based collaborative approach in responding to RVF emergencies.


Sujet(s)
Épidémies de maladies , Génome viral , Bétail , Phylogenèse , Fièvre de la Vallée du Rift , Virus de la fièvre de la vallée du Rift , Animaux , Rwanda/épidémiologie , Fièvre de la Vallée du Rift/épidémiologie , Fièvre de la Vallée du Rift/virologie , Fièvre de la Vallée du Rift/transmission , Virus de la fièvre de la vallée du Rift/génétique , Virus de la fièvre de la vallée du Rift/classification , Virus de la fièvre de la vallée du Rift/isolement et purification , Bétail/virologie , Bovins , Abattoirs , Génomique/méthodes
20.
Onderstepoort J Vet Res ; 91(1): e1-e7, 2024 Jun 28.
Article de Anglais | MEDLINE | ID: mdl-38949427

RÉSUMÉ

Wild animals, sharing pathogens with domestic animals, play a crucial role in the epidemiology of infectious diseases. Sampling from wild animals poses significant challenges, yet it is vital for inclusion in disease surveillance and monitoring programmes. Often, mass surveillance involves serological screenings using enzyme-linked immunosorbent assay (ELISA) tests, typically validated only for domestic animals. This study assessed the diagnostic specificity of commercially available ELISA tests on 342 wild ruminant serum samples and 100 from wild boars. We evaluated three tests for foot-and-mouth disease: two for Peste des petits ruminants, two for Rift Valley fever and one for Capripox virus. Diagnostic specificity was calculated using the formula True Negative/(False Positive + True Negative). Cohen's kappa coefficient measured agreement between tests. Results showed high specificity and agreement across all tests. Specificity for foot-and-mouth disease (FMD) ranged from 93.89% for Prionics to 100% for IDEXX, with IDvet showing 99.6%. The highest agreement was between FMD IDvet and IDEXX at 97.1%. Rift Valley fever (RVF) tests, Ingezim and IDvet, achieved specificities of 100% and 98.83%, respectively. The optimal specificity was attained by retesting single reactors and inactivating the complement.Contribution: Commercially available ELISA kits are specific for foot-and-mouth disease and similar transboundary animal diseases and can be used for highly specific wild animal testing.


Sujet(s)
Animaux sauvages , Test ELISA , Sensibilité et spécificité , Animaux , Test ELISA/médecine vétérinaire , Fièvre aphteuse/diagnostic , Fièvre de la Vallée du Rift/diagnostic , Fièvre de la Vallée du Rift/sang , Sus scrofa , Ruminants , Anticorps antiviraux/sang
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE