Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 81
Filter
Add more filters

Publication year range
1.
Genome Res ; 34(6): 877-887, 2024 Jul 23.
Article in English | MEDLINE | ID: mdl-38977307

ABSTRACT

The zoonotic parasite Cryptosporidium parvum is a global cause of gastrointestinal disease in humans and ruminants. Sequence analysis of the highly polymorphic gp60 gene enabled the classification of C. parvum isolates into multiple groups (e.g., IIa, IIc, Id) and a large number of subtypes. In Europe, subtype IIaA15G2R1 is largely predominant and has been associated with many water- and food-borne outbreaks. In this study, we generated new whole-genome sequence (WGS) data from 123 human- and ruminant-derived isolates collected in 13 European countries and included other available WGS data from Europe, Egypt, China, and the United States (n = 72) in the largest comparative genomics study to date. We applied rigorous filters to exclude mixed infections and analyzed a data set from 141 isolates from the zoonotic groups IIa (n = 119) and IId (n = 22). Based on 28,047 high-quality, biallelic genomic SNPs, we identified three distinct and strongly supported populations: Isolates from China (IId) and Egypt (IIa and IId) formed population 1; a minority of European isolates (IIa and IId) formed population 2; and the majority of European (IIa, including all IIaA15G2R1 isolates) and all isolates from the United States (IIa) clustered in population 3. Based on analyses of the population structure, population genetics, and recombination, we show that population 3 has recently emerged and expanded throughout Europe to then, possibly from the United Kingdom, reach the United States, where it also expanded. The reason(s) for the successful spread of population 3 remain elusive, although genes under selective pressure uniquely in this population were identified.


Subject(s)
Cryptosporidiosis , Cryptosporidium parvum , Disease Outbreaks , Cryptosporidium parvum/genetics , United States/epidemiology , Europe/epidemiology , Humans , Cryptosporidiosis/parasitology , Cryptosporidiosis/epidemiology , Animals , Genomics/methods , Polymorphism, Single Nucleotide , Phylogeny , Whole Genome Sequencing/methods , Genome, Protozoan , China/epidemiology , Egypt/epidemiology
2.
Emerg Infect Dis ; 30(8): 1672-1676, 2024 Aug.
Article in English | MEDLINE | ID: mdl-39043445

ABSTRACT

Influenza D virus was isolated from pigs on a mixed pig and beef farm in France. Investigation suggested bull-to-pig transmission and spread among pigs. The swine influenza D virus recovered was a reassortant of D/660 and D/OK lineages. Reported mutations in the receptor binding site might be related to swine host adaptation.


Subject(s)
Farms , Orthomyxoviridae Infections , Phylogeny , Reassortant Viruses , Swine Diseases , Thogotovirus , Animals , Swine , Reassortant Viruses/genetics , France/epidemiology , Orthomyxoviridae Infections/virology , Orthomyxoviridae Infections/veterinary , Swine Diseases/virology , Cattle , Thogotovirus/genetics , Thogotovirus/classification , Thogotovirus/isolation & purification , Deltainfluenzavirus
3.
Vet Res ; 54(1): 66, 2023 Aug 22.
Article in English | MEDLINE | ID: mdl-37608341

ABSTRACT

Cryptosporidium spp. remain a major cause of waterborne diarrhea and illness in developing countries and represent a significant burden to farmers worldwide. Cryptosporidium parvum virus 1 (CSpV1), of the genus Cryspovirus, was first reported to be present in the cytoplasm of C. parvum in 1997. Full-length genome sequences have been obtained from C. parvum from Iowa (Iowa), Kansas (KSU) and China. We aimed at characterizing the genome of CSpV1 from France and used sequence analysis from Cryptosporidium isolates to explore whether CSpV1 genome diversity varies over time, with geographical sampling location, C. parvum genetic diversity, or ruminant host species. A total of 123 fecal samples of cattle, sheep and goats were collected from 17 different French departments (57 diseased animal fecal samples and 66 healthy animal fecal samples). Subtyping analysis of the C. parvum isolates revealed the presence of two zoonotic subtype families IIa and IId. Sequence analysis of CSpV1 revealed that all CSpV1 from France, regardless of the subtype of C. parvum (IIaA15G2R1, IIaA17G2R1 and IIdA18G1R1) are more closely related to CSpV1 from Turkey, and cluster on a distinct branch from CSpV1 collected from C. parvum subtype IIaA15G2R1 from Asia and North America. We also found that samples collected on a given year or successive years in a given location are more likely to host the same subtype of C. parvum and the same CSpV1 strain. Yet, there is no distinct clustering of CSpV1 per French department or ruminants, probably due to trade, and transmission of C. parvum among host species. Our results point towards (i) a close association between CSpV1 movement and C. parvum movement, (ii) recent migrations of C. parvum among distantly located departments and (iii) incidental transmission of C. parvum between ruminants. All together, these results provide insightful information regarding CSpV1 evolution and suggest the virus might be used as an epidemiological tracer for C. parvum. Future studies need to investigate CSpV1's role in C. parvum virulence and on subtype ability to infect different species.


Subject(s)
Cattle Diseases , Cryptosporidiosis , Cryptosporidium parvum , Cryptosporidium , Goat Diseases , Sheep Diseases , Sheep , Animals , Cattle , Goats , Cryptosporidium parvum/genetics , Cryptosporidiosis/epidemiology , France/epidemiology , Cattle Diseases/epidemiology , Goat Diseases/epidemiology , Sheep Diseases/epidemiology
4.
Avian Pathol ; 51(3): 291-294, 2022 Jun.
Article in English | MEDLINE | ID: mdl-35201915

ABSTRACT

Currently, turkey coronaviruses (TCoV) are isolated from homogenized intestines of experimentally infected embryos to ensure a maximum recovery of viral particles from all components of the intestines. However, the process of homogenization also ensures release of a significant amount of cellular RNAs into the sample that hinders downstream viral genome sequencing. This is especially the case for next generation sequencing (NGS) which sequences molecules at random. This characteristic means that the heavily abundant cellular RNA in the sample drowns out the minority viral RNA during the sequencing process and, consequently, very little to no viral genome data are obtained. To address this problem, a method was developed, in which 10 descendent isolates of the European strain of TCoV were recovered uniquely from the intestinal lumen without homogenization of the tissue. For nine out of 10 samples, NGS produced viral RNA reads with good coverage depth over the entire TCoV genomes. This is a much-needed new, simple and cost effective method of isolating TCoV that facilitates downstream NGS of viral RNA and should be considered as an alternative method for isolating other avian enteric coronaviruses in the interest of obtaining full-length genome sequences.


Subject(s)
Coronavirus, Turkey , Poultry Diseases , Animals , Coronavirus, Turkey/genetics , Genome, Viral , High-Throughput Nucleotide Sequencing/veterinary , Intestines , RNA, Viral/genetics , Turkeys
5.
J Fish Dis ; 45(5): 707-716, 2022 May.
Article in English | MEDLINE | ID: mdl-35172021

ABSTRACT

Gilthead seabream represents a species of importance in Mediterranean aquaculture. The larval stage is particularly sensitive and frequently impacted in suboptimal environmental or sanitary conditions. In the present study, investigations were carried out in a seabream hatchery following an unusual mortality reaching 70% among 50-day post-hatching. Anorexia, loss of appetite and abnormal swimming behaviour were observed in absence of parasites or pathogenic bacteria. Proliferation of rod-shaped bacteria in the gut lumen was associated with focal degeneration in the intestinal mucosa. Cytopathic effects on an EK-1 cell line after 21 days of culture at 14°C and 20°C in contact with homogenized affected larvae revealed the presence of a viral agent. Molecular characterization by high-throughput sequencing showed a typical picornavirus genome organization with a polyprotein precursor of 2276 amino acids sharing 46.3% identity with that of the Eel Picornavirus-1. A specific real-time PCR confirmed the presence of the viral genome in affected larval homogenate and corresponding cell culture supernatant. We propose the name Potamipivirus daurada for this novel species within the genus Potamipivirus. The etiological role of this virus remains uncertain at this time, and future studies will be necessary to investigate its prevalence in natural and aquaculture-reared populations as well as its ability to cause diseases in gilthead seabream.


Subject(s)
Fish Diseases , Sea Bream , Animals , DNA Viruses/genetics , Larva , Real-Time Polymerase Chain Reaction , Sea Bream/genetics
6.
Emerg Infect Dis ; 27(2): 508-516, 2021 02.
Article in English | MEDLINE | ID: mdl-33496244

ABSTRACT

We detected 3 genotypes of highly pathogenic avian influenza A(H5N8) virus in France during winter 2016-17. Genotype A viruses caused dramatic economic losses in the domestic duck farm industry in southwestern France. Our phylogenetic analysis suggests that genotype A viruses formed 5 distinct geographic clusters in southwestern France. In some clusters, local secondary transmission might have been started by a single introduction. The intensity of the viral spread seems to correspond to the density of duck holdings in each production area. To avoid the introduction of disease into an unaffected area, it is crucial that authorities limit the movements of potentially infected birds.


Subject(s)
Influenza A Virus, H5N8 Subtype , Influenza in Birds , Animals , Animals, Wild , Birds , Disease Outbreaks , France/epidemiology , Influenza A Virus, H5N8 Subtype/genetics , Influenza in Birds/epidemiology , Phylogeny
7.
Avian Pathol ; 50(6): 507-521, 2021 Dec.
Article in English | MEDLINE | ID: mdl-34545751

ABSTRACT

Guinea fowl fulminating enteritis has been reported in France since the 1970s. In 2014, a coronavirus was identified and appeared as a possible viral pathogen involved in the disease. In the present study, intestinal content from a guinea fowl involved in a new case of the disease in 2017 was analysed by deep sequencing, revealing the presence of a guinea fowl coronavirus (GfCoV) and a picornavirus (GfPic). Serial passage assays into the intra-amniotic cavity of 13-day-old specific pathogen-free chicken eggs and 20-day-old conventional guinea fowl eggs were attempted. In chicken eggs, isolation assays failed, but in guinea fowl eggs, both viruses were successfully obtained. Furthermore, two GfCoV and two GfPic isolates were obtained from the same bird but from different sections of its intestines. This shows that using eggs of the same species, in which the virus has been detected, can be the key for successful isolation. The consensus sequence of the full-length genomes of both GfCoV isolates was highly similar, and correlated to those previously described in terms of genome organization, ORF length and phylogenetic clustering. According to full-length genome analysis and the structure of the Internal Ribosome Entry Site, both GfPic isolates belong to the Anativirus genus and specifically the species Anativirus B. The availability of the first isolates of GfCoV and GfPic will now provide a means of assessing their pathogenicity in guinea fowl in controlled experimental conditions and to assess whether they are primary viral pathogens of the disease "guinea fowl fulminating enteritis".RESEARCH HIGHLIGHTSFirst isolation of guinea fowl coronaviruses and picornaviruses.Eggs homologous to the infected species are key for isolation.Isolates available to precisely evaluate the virus roles in fulminating enteritis.First full-length genome sequences of guinea fowl picornaviruses.


Subject(s)
Coronavirus/classification , Enteritis/virology , Galliformes/virology , Picornaviridae/classification , Animals , Coronavirus/isolation & purification , Coronavirus Infections/veterinary , Coronavirus Infections/virology , Enteritis/veterinary , Genome, Viral , Phylogeny , Picornaviridae/isolation & purification , Picornaviridae Infections/veterinary , Picornaviridae Infections/virology , Poultry Diseases/virology
8.
BMC Vet Res ; 17(1): 257, 2021 Jul 28.
Article in English | MEDLINE | ID: mdl-34321003

ABSTRACT

BACKGROUND: Rabbit haemorrhagic disease virus Lagovirus europaeus/GI.1d variant (GI.1d/RHDV) was identified in 1990 in France, and until the emergence of the new genotype GI.2, it was the main variant circulating in the country. The early stages of RHDV infection have been described in a few studies of rabbits experimentally infected with earlier strains, but no information was given on the minimum infective dose. We report the genomic and phenotypic characterisation of a GI.1d/RHDV strain collected in 2000 in France (GI.1d/00-21). RESULTS: We performed in vivo assays in rabbits to study virus replication kinetics in several tissues at the early stage of infection, and to estimate the minimum infective dose. Four tested doses, negligible (10- 1 viral genome copies), low (104), high (107) and very high (1011) were quantified using a method combining density gradient centrifugation of the viral particles and an RT-qPCR technique developed to quantify genomic RNA (gRNA). The GI.1d/00-21 genome showed the same genomic organisation as other lagoviruses; however, a substitution in the 5' untranslated region and a change in the potential p23/2C-like helicase cleavage site were observed. We showed that the liver of one of the two rabbits inoculated via the oral route was infected at 16 h post-infection and all tissues at 39 h post-infection. GI.1d/00-21 induced classical RHD signs (depression) and lesions (haemorrhage and splenomegaly). Although infective dose estimation should be interpreted with caution, the minimum infective dose that infected an inoculated rabbit was lower or equal to 104 gRNA copies, whereas between 104 and 107 gRNA copies were required to also induce mortality. CONCLUSIONS: These results provide a better understanding of GI.1d/RHDV infection in rabbits. The genome analysis showed a newly observed mutation in the 5' untranslated region of a lagovirus, whose role remains unknown. The phenotypic analysis showed that the pathogenicity of GI.1d/00-21 and the replication kinetics in infected organs were close to those reported for the original GI.1 strains, and could not alone explain the observed selective advantage of the GI.1d strains. Determining the minimum dose of viral particles required to cause mortality in rabbits is an important input for in vivo studies.


Subject(s)
Caliciviridae Infections/veterinary , Genetic Variation , Genome, Viral , Hemorrhagic Disease Virus, Rabbit/genetics , Rabbits/virology , Virus Replication , Animals , Caliciviridae Infections/epidemiology , Caliciviridae Infections/virology , France/epidemiology , Phylogeny , RNA, Viral/genetics
9.
Emerg Infect Dis ; 25(10): 1940-1943, 2019 10.
Article in English | MEDLINE | ID: mdl-31538914

ABSTRACT

In 2018, a veterinarian became sick shortly after swabbing sows exhibiting respiratory syndrome on a farm in France. Epidemiologic data and genetic analyses revealed consecutive human-to-swine and swine-to-human influenza A(H1N1)pdm09 virus transmission, which occurred despite some biosecurity measures. Providing pig industry workers the annual influenza vaccine might reduce transmission risk.


Subject(s)
Influenza A Virus, H1N1 Subtype , Influenza, Human/transmission , Orthomyxoviridae Infections/transmission , Swine Diseases/transmission , Zoonoses/transmission , Animals , Disease Outbreaks/statistics & numerical data , Disease Outbreaks/veterinary , Female , France/epidemiology , Humans , Influenza A Virus, H1N1 Subtype/genetics , Influenza, Human/epidemiology , Orthomyxoviridae Infections/epidemiology , Phylogeny , Swine , Swine Diseases/epidemiology , Zoonoses/epidemiology , Zoonoses/virology
10.
J Clin Microbiol ; 57(8)2019 08.
Article in English | MEDLINE | ID: mdl-31167846

ABSTRACT

Quality management and independent assessment of high-throughput sequencing-based virus diagnostics have not yet been established as a mandatory approach for ensuring comparable results. The sensitivity and specificity of viral high-throughput sequence data analysis are highly affected by bioinformatics processing using publicly available and custom tools and databases and thus differ widely between individuals and institutions. Here we present the results of the COMPARE [Collaborative Management Platform for Detection and Analyses of (Re-)emerging and Foodborne Outbreaks in Europe] in silico virus proficiency test. An artificial, simulated in silico data set of Illumina HiSeq sequences was provided to 13 different European institutes for bioinformatics analysis to identify viral pathogens in high-throughput sequence data. Comparison of the participants' analyses shows that the use of different tools, programs, and databases for bioinformatics analyses can impact the correct identification of viral sequences from a simple data set. The identification of slightly mutated and highly divergent virus genomes has been shown to be most challenging. Furthermore, the interpretation of the results, together with a fictitious case report, by the participants showed that in addition to the bioinformatics analysis, the virological evaluation of the results can be important in clinical settings. External quality assessment and proficiency testing should become an important part of validating high-throughput sequencing-based virus diagnostics and could improve the harmonization, comparability, and reproducibility of results. There is a need for the establishment of international proficiency testing, like that established for conventional laboratory tests such as PCR, for bioinformatics pipelines and the interpretation of such results.


Subject(s)
Computational Biology/methods , Computer Simulation , High-Throughput Nucleotide Sequencing/standards , Laboratory Proficiency Testing/statistics & numerical data , Sequence Analysis, DNA/standards , Viruses/genetics , Data Analysis , Europe , Genome, Viral , High-Throughput Nucleotide Sequencing/methods , Humans , Intersectoral Collaboration , Laboratory Proficiency Testing/organization & administration , Reproducibility of Results , Sequence Analysis, DNA/statistics & numerical data , Viruses/pathogenicity
11.
J Virol ; 92(24)2018 12 15.
Article in English | MEDLINE | ID: mdl-30258006

ABSTRACT

The H1N1 influenza virus responsible for the most recent pandemic in 2009 (H1N1pdm) has spread to swine populations worldwide while it replaced the previous seasonal H1N1 virus in humans. In France, surveillance of swine influenza A viruses in pig herds with respiratory outbreaks led to the detection of 44 H1N1pdm strains between 2009 and 2017, regardless of the season, and findings were not correlated with pig density. From these isolates, 17 whole-genome sequences were obtained, as were 6 additional hemagglutinin (HA)/neuraminidase (NA) sequences, in order to perform spatial and temporal analyses of genetic diversity and to compare evolutionary patterns of H1N1pdm in pigs to patterns for human strains. Following mutation accumulation and fixation over time, phylogenetic analyses revealed for the first time the divergence of a swine-specific genogroup within the H1N1pdm lineage. The divergence is thought to have occurred around 2011, although this was demonstrated only through strains isolated in 2015 to 2016 in the southern half of France. To date, these H1N1pdm swine strains have not been related to any increased virulence in swine herds and have not exhibited any antigenic drift compared to seasonal human strains. However, further monitoring is encouraged, as diverging evolutionary patterns in these two species, i.e., swine and humans, may lead to the emergence of viruses with a potentially higher risk to both animal and human health.IMPORTANCE Pigs are a "mixing vessel" for influenza A viruses (IAVs) because of their ability to be infected by avian and human IAVs and their propensity to facilitate viral genomic reassortment events. Also, as IAVs may evolve differently in swine and humans, pigs can become a reservoir for old human strains against which the human population has become immunologically naive. Thus, viruses from the novel swine-specific H1N1pdm genogroup may continue to diverge from seasonal H1N1pdm strains and/or from other H1N1pdm viruses infecting pigs and lead to the emergence of viruses that would not be covered by human vaccines and/or swine vaccines based on antigens closely related to the original H1N1pdm virus. This discovery confirms the importance of encouraging swine IAV monitoring because H1N1pdm swine viruses could carry an increased risk to both human and swine health in the future as a whole H1N1pdm virus or gene provider in subsequent reassortant viruses.


Subject(s)
Influenza A Virus, H1N1 Subtype/classification , Orthomyxoviridae Infections/epidemiology , Swine Diseases/virology , Whole Genome Sequencing/methods , Animals , Evolution, Molecular , France/epidemiology , Hemagglutinins/genetics , Influenza A Virus, H1N1 Subtype/genetics , Neuraminidase/genetics , Orthomyxoviridae Infections/virology , Pandemics , Phylogeny , Population Surveillance , Spatio-Temporal Analysis , Swine , Swine Diseases/epidemiology , Viral Proteins/genetics , Whole Genome Sequencing/veterinary
12.
Virol J ; 16(1): 140, 2019 11 21.
Article in English | MEDLINE | ID: mdl-31752912

ABSTRACT

BACKGROUND: Next generation sequencing (NGS) is becoming widely used among diagnostics and research laboratories, and nowadays it is applied to a variety of disciplines, including veterinary virology. The NGS workflow comprises several steps, namely sample processing, library preparation, sequencing and primary/secondary/tertiary bioinformatics (BI) analyses. The latter is constituted by a complex process extremely difficult to standardize, due to the variety of tools and metrics available. Thus, it is of the utmost importance to assess the comparability of results obtained through different methods and in different laboratories. To achieve this goal, we have organized a proficiency test focused on the bioinformatics components for the generation of complete genome sequences of salmonid rhabdoviruses. METHODS: Three partners, that performed virus sequencing using different commercial library preparation kits and NGS platforms, gathered together and shared with each other 75 raw datasets which were analyzed separately by the participants to produce a consensus sequence according to their own bioinformatics pipeline. Results were then compared to highlight discrepancies, and a subset of inconsistencies were investigated more in detail. RESULTS: In total, we observed 526 discrepancies, of which 39.5% were located at genome termini, 14.1% at intergenic regions and 46.4% at coding regions. Among these, 10 SNPs and 99 indels caused changes in the protein products. Overall reproducibility was 99.94%. Based on the analysis of a subset of inconsistencies investigated more in-depth, manual curation appeared the most critical step affecting sequence comparability, suggesting that the harmonization of this phase is crucial to obtain comparable results. The analysis of a calibrator sample allowed assessing BI accuracy, being 99.983%. CONCLUSIONS: We demonstrated the applicability and the usefulness of BI proficiency testing to assure the quality of NGS data, and recommend a wider implementation of such exercises to guarantee sequence data uniformity among different virology laboratories.


Subject(s)
Computational Biology/methods , Computational Biology/standards , High-Throughput Nucleotide Sequencing/standards , Infectious hematopoietic necrosis virus/genetics , Novirhabdovirus/genetics , Sequence Analysis, DNA/standards , Animals , Fishes , High-Throughput Nucleotide Sequencing/methods , Quality Control , Reproducibility of Results , Sequence Analysis, DNA/methods
13.
Vet Res ; 50(1): 77, 2019 Oct 07.
Article in English | MEDLINE | ID: mdl-31590684

ABSTRACT

This report describes the detection of a triple reassortant swine influenza A virus of H1avN2 subtype. It evolved from an avian-like swine H1avN1 that first acquired the N2 segment from a seasonal H3N2, then the M segment from a 2009 pandemic H1N1, in two reassortments estimated to have occurred 10 years apart. This study illustrates how recurrent influenza infections increase the co-infection risk and facilitate evolutionary jumps by successive gene exchanges. It recalls the importance of appropriate biosecurity measures inside holdings to limit virus persistence and interspecies transmissions, which both contribute to the emergence of new potentially zoonotic viruses.


Subject(s)
Influenza A Virus, H1N1 Subtype/physiology , Influenza A Virus, H1N2 Subtype/physiology , Influenza A Virus, H3N2 Subtype/physiology , Reassortant Viruses/physiology , Swine Diseases/virology , Animals , France , Influenza A Virus, H1N1 Subtype/genetics , Influenza A Virus, H1N2 Subtype/genetics , Influenza A Virus, H3N2 Subtype/genetics , Sus scrofa , Swine
14.
J Gen Virol ; 98(6): 1181-1184, 2017 Jun.
Article in English | MEDLINE | ID: mdl-28640747

ABSTRACT

Novirhabdoviruses like the Viral hemorrhagic septicemia virus (VHSV) are rhabdoviruses infecting fish. In the current study, RNA genomes of different VHSV field isolates classified as high, medium or low virulent phenotypes have been sequenced by next-generation sequencing and compared. Various amino acid changes, depending on the VHSV phenotype, have been identified in all the VHSV proteins. As a starting point, we focused our study on the non-virion (NV) non-structural protein in which an arginine residue (R116) is present in all the virulent isolates and replaced by a serine/asparagine residue S/N116 in the attenuated isolates. A recombinant virus derived from a virulent VHSV strain in which the NV R116 residue has been replaced by a serine, rVHSVNVR116S, was generated by reverse genetics and used to infect juvenile trout. We showed that rVHSVNVR116S was highly attenuated and that surviving fish were almost completely protected from a challenge with the wild-type VHSV.


Subject(s)
Amino Acid Substitution , Fish Diseases/pathology , Fish Diseases/virology , Novirhabdovirus/pathogenicity , Rhabdoviridae Infections/veterinary , Viral Proteins/genetics , Virulence Factors/genetics , Animals , Genome, Viral , Novirhabdovirus/genetics , Novirhabdovirus/isolation & purification , Phenotype , Reverse Genetics , Rhabdoviridae Infections/pathology , Rhabdoviridae Infections/virology , Sequence Analysis, DNA , Trout , Virulence
15.
Euro Surveill ; 22(9)2017 Mar 02.
Article in English | MEDLINE | ID: mdl-28277218

ABSTRACT

Several new highly pathogenic (HP) H5 avian influenza virus (AIV) have been detected in poultry farms from south-western France since November 2015, among which an HP H5N1. The zoonotic potential and origin of these AIVs immediately became matters of concern. One virus of each subtype H5N1 (150169a), H5N2 (150233) and H5N9 (150236) was characterised. All proved highly pathogenic for poultry as demonstrated molecularly by the presence of a polybasic cleavage site in their HA protein - with a sequence (HQRRKR/GLF) previously unknown among avian H5 HPAI viruses - or experimentally by the in vivo demonstration of an intravenous pathogenicity index of 2.9 for the H5N1 HP isolate. Phylogenetic analyses based on the full genomes obtained by NGS confirmed that the eight viral segments of the three isolates were all part of avian Eurasian phylogenetic lineage but differed from the Gs/Gd/1/96-like lineage. The study of the genetic characteristics at specific amino acid positions relevant for modulating the adaptation to and the virulence for mammals showed that presently, these viruses possess most molecular features characteristic of AIV and lack some major characteristics required for efficient respiratory transmission to or between humans. The three isolates are therefore predicted to have no significant pandemic potential.


Subject(s)
Influenza A Virus, H5N1 Subtype/genetics , Influenza A Virus, H5N1 Subtype/pathogenicity , Influenza A Virus, H5N2 Subtype/genetics , Influenza A Virus, H5N2 Subtype/pathogenicity , Influenza in Birds/virology , Animals , Birds , Chickens , Disease Outbreaks , Ducks , France/epidemiology , Genes, Viral/genetics , Hemagglutinin Glycoproteins, Influenza Virus/genetics , Humans , Influenza A Virus, H5N1 Subtype/classification , Influenza A Virus, H5N1 Subtype/isolation & purification , Influenza A Virus, H5N2 Subtype/classification , Influenza A Virus, H5N2 Subtype/isolation & purification , Influenza in Birds/epidemiology , Molecular Sequence Data , Phylogeny , Polymerase Chain Reaction , Poultry/virology , Poultry Diseases/virology , RNA, Viral/genetics , Sequence Analysis, DNA
16.
J Gen Virol ; 96(10): 3124-3130, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26296914

ABSTRACT

Porcine endogenous retroviruses (PERVs) are present in the genomes of pig cells. The PERV-A/C recombinant virus can infect human cells and is a major risk of zoonotic disease in the case of xenotransplantation of pig organs to humans. Raltegravir (RAL) is a viral integrase (IN) inhibitor used in highly active antiretroviral treatment. In the present study, we explored the potential use of RAL against PERV-A/C. We report (i) a three-dimensional model of the PERV-A/C intasome complexed with RAL, (ii) the sensitivity of PERV-A/C IN to RAL in vitro and (iii) the sensitivity of a PERV-A/C-IRES-GFP recombinant virus to RAL in cellulo. We demonstrated that RAL is a potent inhibitor against PERV-A/C IN and PERV-A/C replication with IC50s in the nanomolar range. To date, the use of retroviral inhibitors remains the only way to control the risk of zoonotic PERV infection during pig-to-human xenotransplantation.


Subject(s)
Antiviral Agents/pharmacology , Endogenous Retroviruses/enzymology , Endogenous Retroviruses/physiology , Integrases/analysis , Raltegravir Potassium/pharmacology , Virus Integration/drug effects , Animals , Antiviral Agents/chemistry , Crystallography, X-Ray , Endogenous Retroviruses/drug effects , Inhibitory Concentration 50 , Integrases/chemistry , Protein Binding , Protein Conformation , Raltegravir Potassium/chemistry , Swine
17.
Virol J ; 10: 279, 2013 Sep 10.
Article in English | MEDLINE | ID: mdl-24015922

ABSTRACT

BACKGROUND: Avian influenza A (AI) viruses of subtypes H5 can cause serious disease outbreaks in poultry including panzootic due to H5N1 highly pathogenic (HP) viruses. These viruses are a threat not only for animal health but also public health due to their zoonotic potential. The domestic duck plays a major role in the epidemiological cycle of influenza virus subtypes H5 but little is known concerning host/pathogen interactions during influenza infection in duck species. In this study, a subtracted library from duck trachea (a primary site of influenza virus infection) was constructed to analyse and compare the host response after a highly or low pathogenic (LP) H5N1-infection. RESULTS: Here, we show that more than 200 different genes were differentially expressed in infected duck trachea to a significant degree. In addition, significant differentially expressed genes between LPAI- and HPAI-infected tracheas were observed. Gene ontology annotation was used and specific signalling pathways were identified. These pathways were different for LPAI and HPAI-infected tracheas, except for the CXCR4 signalling pathway which is implicated in immune response. A different modulation of genes in the CXCR4 signalling pathway and TRIM33 was induced in duck tracheas infected with a HPAI- or a LPAI-H5N1. CONCLUSION: First, this study indicates that Suppressive Subtractive Hybridization (SSH) is an alternative approach to gain insights into the pathogenesis of influenza infection in ducks. Secondly, the results indicate that cellular gene expression in the duck trachea was differently modulated after infection with a LPAI-H5N1 or after infection with a HPAI-H5N1 virus. Such difference found in infected trachea, a primary infection site, could precede continuation of infection and could explain appearance of respiratory symptoms or not.


Subject(s)
Gene Expression Profiling , Host-Pathogen Interactions , Influenza A Virus, H5N1 Subtype/pathogenicity , Influenza in Birds/pathology , Influenza in Birds/virology , Trachea/pathology , Trachea/virology , Animals , Ducks , Nucleic Acid Hybridization/methods , Signal Transduction/genetics
18.
Viruses ; 15(12)2023 12 13.
Article in English | MEDLINE | ID: mdl-38140664

ABSTRACT

The European sea bass Dicentrarchus labrax is the main species reared in Mediterranean aquaculture. Its larval stage, which is very sensitive and highly affected by sanitary and environmental conditions, is particularly scrutinized in hatcheries. Recently, a Mediterranean sea bass farm had to deal with an abnormal increase in mortality, especially between 20 and 35 days post-hatching (dph). Biological investigations led to the observation of cytopathic effects on three different fish cell lines after almost 3 weeks of culture at 14 °C in contact with homogenized affected larvae, suggesting the presence of a viral agent. High-throughput sequencing revealed a 6818-nucleotide-long RNA genome with six putative ORFs, corresponding to the organization of viruses belonging to the Totiviridae family. This genome clustered with the newly described and suggested Pistolvirus genus, sharing 45.5% to 37.2% nucleotide identity with other piscine toti-like viruses such as Cyclopterus lumpus toti-like virus (CLuTLV) or piscine myocarditis virus (PMCV), respectively. Therefore, we propose to name this new viral agent sea bass toti-like virus (SBTLV). Specific real-time RT-PCR confirmed the presence of the viral genome in the affected larval homogenate from different production batches and the corresponding cell culture supernatant. Experimental infections performed on sea bass fingerlings did not induce mortality, although the virus could be detected in various organs and a specific immune response was developed. Additional studies are needed to understand the exact involvement of this virus in the mortality observed in hatcheries and the potential associated cofactors.


Subject(s)
Bass , Fish Diseases , Viruses , Animals , Bass/genetics , Genome , Aquaculture , Viruses/genetics , Nucleotides
19.
Trop Med Infect Dis ; 8(8)2023 Aug 16.
Article in English | MEDLINE | ID: mdl-37624355

ABSTRACT

The summer temperatures recorded in Poland in 2022 were among the highest in over 30 years and, combined with higher-than-expected rainfall, gave the impression of an almost tropical climate. Such climatic conditions were ideal for the transmission of vector-borne zoonotic diseases such as West Nile fever. In northeastern Poland, in the Mazowieckie region, the Polish event-based surveillance network reported increased fatalities of free-living hooded crows (Corvus corone cornix). West Nile virus (WNV) lineage 2 was identified for the first time as the etiological agent responsible for the death of the birds. WNV was detected in 17 out of the 99 (17.17%) free-living birds tested in this study. All the WNV-infected dead birds were collected in the same area and were diagnosed in September by the NVRI and confirmed by the EURL for equine diseases, ANSES, in October 2022. Unnaturally high temperatures recorded in Poland in 2022 likely favored the infection and spread of the virus in the avian population. A nationwide alert and awareness raising of blood transfusion centers and hospitals was carried out to prevent human infections by WNV.

20.
Microbiol Spectr ; 11(4): e0184423, 2023 08 17.
Article in English | MEDLINE | ID: mdl-37395665

ABSTRACT

Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) in infected patients mainly displays pulmonary and oronasal tropism; however, the presence of the virus has also been demonstrated in the stools of patients and consequently in wastewater treatment plant effluents, raising the question of the potential risk of environmental contamination (such as seawater contamination) through inadequately treated wastewater spillover into surface or coastal waters even if the environmental detection of viral RNA alone does not substantiate risk of infection. Therefore, here, we decided to experimentally evaluate the persistence of the porcine epidemic diarrhea virus (PEDv), considered as a coronavirus representative model, in the coastal environment of France. Coastal seawater was collected, sterile-filtered, and inoculated with PEDv before incubation for 0 to 4 weeks at four temperatures representative of those measured along the French coasts throughout the year (4, 8, 15, and 24°C). The decay rate of PEDv was determined using mathematical modeling and was used to determine the half-life of the virus along the French coast in accordance with temperatures from 2000 to 2021. We experimentally observed an inverse correlation between seawater temperature and the persistence of infectious viruses in seawater and confirm that the risk of transmission of infectious viruses from contaminated stool in wastewater to seawater during recreational practices is very limited. The present work represents a good model to assess the persistence of coronaviruses in coastal environments and contributes to risk evaluation, not only for SARS-CoV-2 persistence, but also for other coronaviruses, specifically enteric coronaviruses from livestock. IMPORTANCE The present work addresses the question of the persistence of coronavirus in marine environments because SARS-CoV-2 is regularly detected in wastewater treatment plants, and the coastal environment, subjected to increasing anthropogenic pressure and the final receiver of surface waters and sometimes insufficiently depurated wastewater, is particularly at risk. The problem also arises in the possibility of soil contamination by CoV from animals, especially livestock, during manure application, where, by soil impregnation and runoff, these viruses can end up in seawater. Our findings are of interest to researchers and authorities seeking to monitor coronaviruses in the environment, either in tourist areas or in regions of the world where centralized systems for wastewater treatment are not implemented, and more broadly, to the scientific community involved in "One Health" approaches.


Subject(s)
COVID-19 , Porcine epidemic diarrhea virus , Animals , Swine , COVID-19/epidemiology , Wastewater , SARS-CoV-2 , Soil
SELECTION OF CITATIONS
SEARCH DETAIL