Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 535
Filter
1.
Viruses ; 16(8)2024 Jul 26.
Article in English | MEDLINE | ID: mdl-39205178

ABSTRACT

Equine rhinitis A (ERAV) and B (ERBV) viruses are respiratory pathogens with worldwide distribution. The current study aimed to determine the frequency of infection of ERAV and ERBV among horses and foals at Polish national studs, and to determine genetic variability within the viruses obtained. Virus-specific quantitative RT-PCR assays targeting a 5' untranslated region were used to screen nasal swabs collected from 621 horses at 16 national horse studs from throughout Poland, including 553 healthy horses and 68 horses with respiratory disease. A partial DNA polymerase gene was amplified and sequenced from the qRT-PCR-positive samples. The obtained sequences were analysed using phylogeny and genetic network analysis. None of the nasal swabs were positive for ERAV, whereas ERBV was found in 11/621 (1.78%) samples collected from 10 healthy horses and one foal affected by respiratory disease. Partial DNA polymerase gene sequence variability was correlated with individual horses and studs from which samples were collected when only Polish sequences were analysed, but there was no correlation between country of origin and ERBV sequence when Polish and international sequences were included in the network. The report presents the first detection of ERBV in Poland.


Subject(s)
Horse Diseases , Phylogeny , Picornaviridae Infections , Horses/virology , Animals , Poland/epidemiology , Horse Diseases/virology , Horse Diseases/epidemiology , Picornaviridae Infections/veterinary , Picornaviridae Infections/virology , Picornaviridae Infections/epidemiology , Prevalence , Genetic Variation , Erbovirus/genetics , Erbovirus/isolation & purification , Rhinovirus/genetics , Rhinovirus/isolation & purification , Rhinovirus/classification , Sequence Analysis, DNA
2.
Viruses ; 16(7)2024 Jul 07.
Article in English | MEDLINE | ID: mdl-39066254

ABSTRACT

BACKGROUND: Equid alphaherpesvirus 1 (EHV-1) is a highly contagious respiratory tract pathogen of horses, and infection may be followed by myeloencephalopathy or abortion. Surveillance and early detection have focused on PCR assays using less tolerated nasal swabs. Here, we assess non-invasive non-contact sampling techniques as surveillance tools in naturally equid gammaherpesvirus 2-shedding horses as surrogates for EHV-1. METHODS: Horses were individually housed for 10 h periods on 2 consecutive days. Sampling included nasal swabs, nostril wipes, environmental swabs, droplet-catching devices, and air sampling. The latter was completed via two strategies: a combined air sample collected while going from horse to horse and a collective air sample collected at a stationary central point for 6 h. Samples were screened through quantitative PCR and digital PCR. RESULTS: Nine horses on day 1 and 11 horses on day 2 were positive for EHV-1; overall, 90.9% of the nostril wipes, 81.8% of the environmental surfaces, and 90.9% of the droplet-catching devices were found to be positive. Quantitative analysis showed that the mean DNA copies detection per cm2 of nostril wipe sampled concentration (4.3 × 105 per day) was significantly (p < 0.05) comparable to that of nasal swabs (3.6 × 105 per day) followed by environmental swabs (4.3 × 105 per day) and droplet catchers (3.5 × 103 per day), respectively. Overall, 100% of the air samples collected were positive on both qPCR and dPCR. In individual air samples, a mean concentration of 1.0 × 104 copies of DNA were detected in per m3 air sampled per day, while in the collective air samples, the mean concentration was 1.1 × 103. CONCLUSIONS: Environmental samples look promising in replacing direct contact sampling. Environmental and air sampling could become efficient surveillance tools at equestrian events; however, it needs threshold calculations for minimum detection levels.


Subject(s)
Herpesviridae Infections , Herpesvirus 1, Equid , Horse Diseases , Animals , Horses/virology , Horse Diseases/virology , Horse Diseases/diagnosis , Herpesviridae Infections/veterinary , Herpesviridae Infections/virology , Herpesviridae Infections/diagnosis , Herpesvirus 1, Equid/isolation & purification , Herpesvirus 1, Equid/genetics , Specimen Handling/methods , Female , Virus Shedding
3.
Virol J ; 21(1): 163, 2024 Jul 23.
Article in English | MEDLINE | ID: mdl-39044231

ABSTRACT

Usutu virus (USUV), an arbovirus from the Flaviviridae family, genus Flavivirus, has recently gained increasing attention because of its potential for emergence. After his discovery in South Africa, USUV spread to other African countries, then emerged in Europe where it was responsible for epizootics. The virus has recently been found in Asia. USUV infection in humans is considered to be most often asymptomatic or to cause mild clinical signs. However, a few cases of neurological complications such as encephalitis or meningo-encephalitis have been reported in both immunocompromised and immunocompetent patients. USUV natural life cycle involves Culex mosquitoes as its main vector, and multiple bird species as natural viral reservoirs or amplifying hosts, humans and horses can be incidental hosts. Phylogenetic studies carried out showed eight lineages, showing an increasing genetic diversity for USUV. This work describes the development and validation of a novel whole-genome amplicon-based sequencing approach to Usutu virus. This study was carried out on different strains from Senegal and Italy. The new approach showed good coverage using samples derived from several vertebrate hosts and may be valuable for Usutu virus genomic surveillance to better understand the dynamics of evolution and transmission of the virus.


Subject(s)
Flavivirus Infections , Flavivirus , Genome, Viral , Phylogeny , Flavivirus/genetics , Flavivirus/classification , Flavivirus/isolation & purification , Animals , Flavivirus Infections/virology , Flavivirus Infections/veterinary , Humans , Senegal , Italy , Birds/virology , RNA, Viral/genetics , Genetic Variation , Culex/virology , Whole Genome Sequencing , Horses/virology
4.
Virus Genes ; 60(5): 559-562, 2024 Oct.
Article in English | MEDLINE | ID: mdl-39028407

ABSTRACT

The Equid alphaherpesvirus type 1 (EHV-1) infection can have devastating economic consequences in the horse industry due to large-scale outbreaks of abortions, perinatal foal mortality, and myeloencephalopathy. The present study analyzed the genome of two isolates obtained from aborted fetuses in Argentina, E/745/99 and E/1297/07. The E745/99 genome shares 98.2% sequence identity with Ab4, a reference EHV-1 strain. The E/1297/07 genome shares 99.8% identity with NY03, a recombinant strain containing part of ORF64 and part of the intergenic region from Equid alphaherpesvirus-4 (EHV-4). The E/1297/07 genome has the same breakpoints as other United States and Japanese recombinants, including NY03. The recombinant regions have varying numbers of tandem repeat sequences and different minor parental sequences (EHV-4), suggesting distinct origins of the recombinant events. These are the first complete genomes of EHV-1 from Argentina and South America available in the Databases.


Subject(s)
Genome, Viral , Herpesviridae Infections , Herpesvirus 1, Equid , Phylogeny , Argentina , Herpesvirus 1, Equid/genetics , Herpesvirus 1, Equid/isolation & purification , Herpesvirus 1, Equid/classification , Animals , Genome, Viral/genetics , Herpesviridae Infections/veterinary , Herpesviridae Infections/virology , Horses/virology , Recombination, Genetic , Horse Diseases/virology , Open Reading Frames/genetics , Whole Genome Sequencing , DNA, Viral/genetics
5.
Infect Dis (Lond) ; 56(9): 743-758, 2024 Sep.
Article in English | MEDLINE | ID: mdl-38836293

ABSTRACT

BACKGROUND: West Nile Virus (WNV) is a zoonotic arbovirus worldwide spread. Seasonal WNV outbreaks occur in the Mediterranean basin since the late 1990's with ever-increasing incidence. In Southern Spain WNV is endemic, as disease foci - caused by WNV lineage 1 (WNV-L1) strains - occur every year. On the contrary, WNV-L2 is the dominant lineage in Europe, so most European WNV sequences available belong to this lineage, WNV-L1 sequences being still scarce. METHODS: To fill this gap, this study reports the genetic characterisation of 27 newly described WNV-L1 strains, involved in outbreaks affecting wild birds and horses during the last decade in South-Western Spain. RESULTS: All strains except one belong to the Western Mediterranean-1 sub-cluster (WMed-1), related phylogenetically to Italian, French, Portuguese, Moroccan and, remarkably, Senegalese strains. This sub-cluster persisted, spread and evolved into three distinguishable WMed-1 phylogenetic groups that co-circulated, notably, in the same province (Cádiz). They displayed different behaviours: from long-term persistence and rapid spread to neighbouring regions within Spain, to long-distance spread to different countries, including transcontinental spread to Africa. Among the different introductions of WNV in Spain revealed in this study, some of them succeeded to get established, some extinguished from the territory shortly afterwards. Furthermore, Spain's southernmost province, Cádiz, constitutes a hotspot for virus incursion. CONCLUSION: Southern Spain seems a likely scenario for emergence of exotic pathogens of African origin. Therefore, circulation of diverse WNV-L1 variants in Spain prompts for an extensive surveillance under a One Health approach.


Subject(s)
Birds , Phylogeny , West Nile Fever , West Nile virus , West Nile virus/genetics , West Nile virus/classification , West Nile virus/isolation & purification , West Nile Fever/epidemiology , West Nile Fever/virology , West Nile Fever/transmission , Animals , Spain/epidemiology , Birds/virology , Bird Diseases/virology , Bird Diseases/epidemiology , Horses/virology , Europe/epidemiology , Disease Outbreaks , Africa/epidemiology , Horse Diseases/virology , Horse Diseases/epidemiology , Humans , Animals, Wild/virology
6.
Microb Pathog ; 193: 106755, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38897362

ABSTRACT

Equid herpesviruses (EHVs) are a group of highly impactful viral pathogens that affect horses, presenting a substantial risk to the global equine industry. Among these, equid herpesvirus-1 (EHV-1) primarily causes respiratory infections. However, its ability to spread to distant organs can lead to severe consequences such as abortion and neurological diseases. These viruses can enter a dormant phase, with minimal activity, and later reactivate to trigger active infections at any time. Recently, there has been a notable rise in the prevalence of a particularly devastating strains of EHV-1 known as equid herpesviral myeloencephalopathy (EHM). In the light of dynamic nature of EHV-1, this review provides a thorough overview of EHV-1 and explores how advances in viral biology affect the pathophysiology of viral infection. The information presented here is crucial for understanding the dynamics of EHV-1 infections and creating practical plans to stop the virus's global spread among equid populations.


Subject(s)
Herpesviridae Infections , Herpesvirus 1, Equid , Horse Diseases , Animals , Horses/virology , Herpesviridae Infections/veterinary , Herpesviridae Infections/virology , Horse Diseases/virology
7.
J Virol Methods ; 328: 114952, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38754768

ABSTRACT

Primary cell cultures derived from human embryo lung play a crucial role in virology by aiding virus propagation and vaccine development. These cultures exhibit a notable ability to undergo multiple subcultures, often reaching up to 70 passages. However, finding alternative primary cell cultures with similar longevity and usefulness is challenging. In this study, we introduce a novel primary culture cells derived from equine embryo brain (FEB), which cells exhibited remarkable long-term cultivation potential. The FEB was established and maintained using Sumitomo Nerve-Cell Culture System Comparison studies were conducted with fetal equine kidney cell line (FEK-Tc13) to assess growth rates and subculture longevity. Immunological characterization was performed using neuronal markers to confirm the neural nature of FEB cells. Viral growth assessments were conducted using equine herpesviruses (EHV-1 and EHV-4) to evaluate infectivity and cytopathic effects in FEB cells. PCR analysis and real-time PCR assays were employed to detect viral genomic DNA and transcription activity of EHVs in infected FEB cells. FEB cells demonstrated faster growth rates compared to fetal equine kidney cell line (FEK-Tc13 cells) and exhibited sustained subculture capability exceeding 50 passages. Immunostaining confirmed the glial identity of FEB cells. Both equine herpesviruses 1 and 4 EHV-1 and EHV-4 viruses efficiently replicated in FEB cells, resulting in clear cytopathic effects. PCR analysis detected genomic DNA of EHVs in infected FEB cells, indicating successful viral infection. The establishment of FEB cells with extended subculture capability highlights their potential utility as a model system for studying neural cell biology and viral infections.


Subject(s)
Brain , Animals , Horses/virology , Brain/virology , Brain/embryology , Brain/cytology , Primary Cell Culture/methods , Herpesvirus 1, Equid/growth & development , Herpesvirus 1, Equid/physiology , Cell Line , Neurons/virology , Virus Cultivation/methods , Cell Culture Techniques/methods , Cell Culture Techniques/veterinary , Cells, Cultured , Virus Replication
8.
Viruses ; 16(4)2024 04 16.
Article in English | MEDLINE | ID: mdl-38675957

ABSTRACT

Equine hepacivirus (EqHV, Flaviviridae, hepacivirus) is a small, enveloped RNA virus generally causing sub-clinical hepatitis with occasional fatalities. EqHV is reported in equids worldwide, but for Italy data are limited. To address this, a survey study was set up to estimate prevalence at a national level and among different production categories (equestrian; competition; work and meat; reproduction) and national macro-regions (North, Central, South, and Islands). Data obtained testing 1801 horse serum samples by Real-Time RT PCR were compared within the categories and regions. The NS3 fragment of the PCR-positive samples was sequenced by Sanger protocol for phylogenetic and mutational analysis. The tertiary structure of the NS3 protein was also assessed. The estimated national prevalence was 4.27% [1.97-6.59, 95% CI] and no statistical differences were detected among production categories and macro-regions. The phylogenesis confirmed the distribution in Italy of the three known EqHV subtypes, also suggesting a possible fourth sub-type that, however, requires further confirmation. Mutational profiles that could also affect the NS3 binding affinity to the viral RNA were detected. The present paper demonstrates that EqHV should be included in diagnostic protocols when investigating causes of hepatitis, and in quality control protocols for blood derived products due to its parental transmission.


Subject(s)
Hepacivirus , Hepatitis C , Horse Diseases , Phylogeny , Animals , Italy/epidemiology , Horses/virology , Horse Diseases/virology , Horse Diseases/epidemiology , Prevalence , Hepacivirus/genetics , Hepacivirus/classification , Hepacivirus/isolation & purification , Hepatitis C/epidemiology , Hepatitis C/virology , Hepatitis C/veterinary , Viral Nonstructural Proteins/genetics , Genotype , RNA, Viral/genetics
9.
J Virol ; 98(4): e0194123, 2024 Apr 16.
Article in English | MEDLINE | ID: mdl-38470143

ABSTRACT

Influenza A viruses (IAVs) can overcome species barriers by adaptation of the receptor-binding site of the hemagglutinin (HA). To initiate infection, HAs bind to glycan receptors with terminal sialic acids, which are either N-acetylneuraminic acid (NeuAc) or N-glycolylneuraminic acid (NeuGc); the latter is mainly found in horses and pigs but not in birds and humans. We investigated the influence of previously identified equine NeuGc-adapting mutations (S128T, I130V, A135E, T189A, and K193R) in avian H7 IAVs in vitro and in vivo. We observed that these mutations negatively affected viral replication in chicken cells but not in duck cells and positively affected replication in horse cells. In vivo, the mutations reduced virus virulence and mortality in chickens. Ducks excreted high viral loads longer than chickens, although they appeared clinically healthy. To elucidate why these viruses infected chickens and ducks despite the absence of NeuGc, we re-evaluated the receptor binding of H7 HAs using glycan microarray and flow cytometry studies. This re-evaluation demonstrated that mutated avian H7 HAs also bound to α2,3-linked NeuAc and sialyl-LewisX, which have an additional fucose moiety in their terminal epitope, explaining why infection of ducks and chickens was possible. Interestingly, the α2,3-linked NeuAc and sialyl-LewisX epitopes were only bound when presented on tri-antennary N-glycans, emphasizing the importance of investigating the fine receptor specificities of IAVs. In conclusion, the binding of NeuGc-adapted H7 IAV to tri-antennary N-glycans enables viral replication and shedding by chickens and ducks, potentially facilitating interspecies transmission of equine-adapted H7 IAVs.IMPORTANCEInfluenza A viruses (IAVs) cause millions of deaths and illnesses in birds and mammals each year. The viral surface protein hemagglutinin initiates infection by binding to host cell terminal sialic acids. Hemagglutinin adaptations affect the binding affinity to these sialic acids and the potential host species targeted. While avian and human IAVs tend to bind to N-acetylneuraminic acid (sialic acid), equine H7 viruses prefer binding to N-glycolylneuraminic acid (NeuGc). To better understand the function of NeuGc-specific adaptations in hemagglutinin and to elucidate interspecies transmission potential NeuGc-adapted viruses, we evaluated the effects of NeuGc-specific mutations in avian H7 viruses in chickens and ducks, important economic hosts and reservoir birds, respectively. We also examined the impact on viral replication and found a binding affinity to tri-antennary N-glycans containing different terminal epitopes. These findings are significant as they contribute to the understanding of the role of receptor binding in avian influenza infection.


Subject(s)
Chickens , Ducks , Horses , Influenza A virus , Influenza in Birds , Neuraminic Acids , Animals , Humans , Chickens/genetics , Chickens/metabolism , Chickens/virology , Ducks/genetics , Ducks/metabolism , Ducks/virology , Epitopes/chemistry , Epitopes/metabolism , Hemagglutinin Glycoproteins, Influenza Virus/chemistry , Hemagglutinin Glycoproteins, Influenza Virus/genetics , Hemagglutinin Glycoproteins, Influenza Virus/metabolism , Horses/genetics , Horses/metabolism , Horses/virology , Influenza A virus/chemistry , Influenza A virus/classification , Influenza A virus/metabolism , Influenza in Birds/genetics , Influenza in Birds/transmission , Influenza in Birds/virology , Mutation , N-Acetylneuraminic Acid/chemistry , N-Acetylneuraminic Acid/metabolism , Neuraminic Acids/chemistry , Neuraminic Acids/metabolism , Receptors, Virus/chemistry , Receptors, Virus/genetics , Receptors, Virus/metabolism , Swine/virology , Viral Zoonoses/metabolism , Viral Zoonoses/transmission , Viral Zoonoses/virology
10.
Microbiol Spectr ; 11(6): e0267623, 2023 Dec 12.
Article in English | MEDLINE | ID: mdl-37943512

ABSTRACT

IMPORTANCE: Spike-receptor interaction is a critical determinant for the host range of coronaviruses. In this study, we investigated the SARS-CoV-2 WHU01 strain and five WHO-designated SARS-CoV-2 variants of concern (VOCs), including Alpha, Beta, Gamma, Delta, and the early Omicron variant, for their Spike interactions with ACE2 proteins of 18 animal species. First, the receptor-binding domains (RBDs) of Alpha, Beta, Gamma, and Omicron were found to display progressive gain of affinity to mouse ACE2. More interestingly, these RBDs were also found with progressive loss of affinities to multiple ACE2 orthologs. The Omicron RBD showed decreased or complete loss of affinity to eight tested animal ACE2 orthologs, including that of some livestock animals (horse, donkey, and pig), pet animals (dog and cat), and wild animals (pangolin, American pika, and Rhinolophus sinicus bat). These findings shed light on potential host range shift of SARS-CoV-2 VOCs, especially that of the Omicron variant.


Subject(s)
Angiotensin-Converting Enzyme 2 , COVID-19 , Cat Diseases , Chiroptera , Dog Diseases , SARS-CoV-2 , Spike Glycoprotein, Coronavirus , Animals , Cats , Dogs , Mice , Angiotensin-Converting Enzyme 2/metabolism , Animals, Wild/virology , Cat Diseases/virology , Chiroptera/virology , COVID-19/metabolism , Dog Diseases/virology , Horses/virology , Mutation , Protein Binding , SARS-CoV-2/genetics , SARS-CoV-2/metabolism , Swine/virology , Spike Glycoprotein, Coronavirus/genetics
11.
J Virol ; 97(4): e0193222, 2023 04 27.
Article in English | MEDLINE | ID: mdl-37022231

ABSTRACT

High-throughput sequences were generated from DNA and cDNA from four Southern white rhinoceros (Ceratotherium simum simum) located in the Taronga Western Plain Zoo in Australia. Virome analysis identified reads that were similar to Mus caroli endogenous gammaretrovirus (McERV). Previous analysis of perissodactyl genomes did not recover gammaretroviruses. Our analysis, including the screening of the updated white rhinoceros (Ceratotherium simum) and black rhinoceros (Diceros bicornis) draft genomes identified high-copy orthologous gammaretroviral ERVs. Screening of Asian rhinoceros, extinct rhinoceros, domestic horse, and tapir genomes did not identify related gammaretroviral sequences in these species. The newly identified proviral sequences were designated SimumERV and DicerosERV for the white and black rhinoceros retroviruses, respectively. Two long terminal repeat (LTR) variants (LTR-A and LTR-B) were identified in the black rhinoceros, with different copy numbers associated with each (n = 101 and 373, respectively). Only the LTR-A lineage (n = 467) was found in the white rhinoceros. The African and Asian rhinoceros lineages diverged approximately 16 million years ago. Divergence age estimation of the identified proviruses suggests that the exogenous retroviral ancestor of the African rhinoceros ERVs colonized their genomes within the last 8 million years, a result consistent with the absence of these gammaretroviruses from Asian rhinoceros and other perissodactyls. The black rhinoceros germ line was colonized by two lineages of closely related retroviruses and white rhinoceros by one. Phylogenetic analysis indicates a close evolutionary relationship with ERVs of rodents including sympatric African rats, suggesting a possible African origin of the identified rhinoceros gammaretroviruses. IMPORTANCE Rhinoceros genomes were thought to be devoid of gammaretroviruses, as has been determined for other perissodactyls (horses, tapirs, and rhinoceros). While this may be true of most rhinoceros, the African white and black rhinoceros genomes have been colonized by evolutionarily young gammaretroviruses (SimumERV and DicerosERV for the white and black rhinoceros, respectively). These high-copy endogenous retroviruses (ERVs) may have expanded in multiple waves. The closest relative of SimumERV and DicerosERV is found in rodents, including African endemic species. Restriction of the ERVs to African rhinoceros suggests an African origin for the rhinoceros gammaretroviruses.


Subject(s)
Biological Evolution , Endogenous Retroviruses , Gammaretrovirus , Perissodactyla , Animals , Mice , Rats , Endogenous Retroviruses/classification , Endogenous Retroviruses/genetics , Gammaretrovirus/classification , Gammaretrovirus/genetics , Horses/genetics , Horses/virology , Perissodactyla/genetics , Perissodactyla/virology , Phylogeny , Proviruses/genetics
12.
Viruses ; 15(3)2023 03 08.
Article in English | MEDLINE | ID: mdl-36992416

ABSTRACT

INTRODUCTION: Eastern equine encephalitis virus (EEEV) and Venezuelan equine encephalitis virus (VEEV) viruses are zoonotic pathogens affecting humans, particularly equines. These neuroarboviruses compromise the central nervous system and can be fatal in different hosts. Both have significantly influenced Colombia; however, few studies analyse its behaviour, and none develop maps using geographic information systems to characterise it. OBJECTIVE: To describe the temporal-spatial distribution of those viruses in Colombia between 2008 and 2019. METHODS: Retrospective cross-sectional descriptive study, based on weekly reports by municipalities of the ICA, of the surveillance of both arboviruses in equines, in Colombia, from 2008 to 2019. The data were converted into databases in Microsoft Access 365®, and multiple epidemiological maps were generated with the Kosmo RC1®3.0 software coupled to shape files of all municipalities in the country. RESULTS: In the study period, 96 cases of EEE and 70 of VEE were reported, with 58% of EEE cases occurring in 2016 and 20% of EEV cases in 2013. The most affected municipalities for EEE corresponded to the department of Casanare: Yopal (20), Aguazul (16), and Tauramena (10). In total, 40 municipalities in the country reported ≥1 case of EEE. CONCLUSIONS: The maps allow a quick appreciation of groups of neighbouring municipalities in different departments (1° political division) and regions of the country affected by those viruses, which helps consider the expansion of the disease associated with mobility and transport of equines between other municipalities, also including international borders, such as is the case with Venezuela. In that country, especially for EEV, municipalities in the department of Cesar are bordering and at risk for that arboviral infection. there is a high risk of equine encephalitis outbreaks, especially for VEE. This poses a risk also, for municipalities in the department of Cesar, bordering with Venezuela.


Subject(s)
Encephalitis Virus, Venezuelan Equine , Encephalomyelitis, Venezuelan Equine , Horses , Animals , Colombia/epidemiology , Cross-Sectional Studies , Encephalomyelitis, Venezuelan Equine/epidemiology , Geographic Information Systems , Horses/virology , Retrospective Studies
13.
Nature ; 613(7943): 340-344, 2023 01.
Article in English | MEDLINE | ID: mdl-36384167

ABSTRACT

During recent decades, pathogens that originated in bats have become an increasing public health concern. A major challenge is to identify how those pathogens spill over into human populations to generate a pandemic threat1. Many correlational studies associate spillover with changes in land use or other anthropogenic stressors2,3, although the mechanisms underlying the observed correlations have not been identified4. One limitation is the lack of spatially and temporally explicit data on multiple spillovers, and on the connections among spillovers, reservoir host ecology and behaviour and viral dynamics. We present 25 years of data on land-use change, bat behaviour and spillover of Hendra virus from Pteropodid bats to horses in subtropical Australia. These data show that bats are responding to environmental change by persistently adopting behaviours that were previously transient responses to nutritional stress. Interactions between land-use change and climate now lead to persistent bat residency in agricultural areas, where periodic food shortages drive clusters of spillovers. Pulses of winter flowering of trees in remnant forests appeared to prevent spillover. We developed integrative Bayesian network models based on these phenomena that accurately predicted the presence or absence of clusters of spillovers in each of the 25 years. Our long-term study identifies the mechanistic connections between habitat loss, climate and increased spillover risk. It provides a framework for examining causes of bat virus spillover and for developing ecological countermeasures to prevent pandemics.


Subject(s)
Chiroptera , Ecology , Ecosystem , Hendra Virus , Horses , Animals , Humans , Australia , Bayes Theorem , Chiroptera/virology , Climate , Horses/virology , Public Health , Hendra Virus/isolation & purification , Natural Resources , Agriculture , Forests , Food Supply , Pandemics/prevention & control , Pandemics/veterinary
14.
Viruses ; 14(11)2022 11 11.
Article in English | MEDLINE | ID: mdl-36423106

ABSTRACT

While some companion animals have been shown to be susceptible to SARS-CoV-2, their role in the COVID-19 pandemic has remained poorly investigated. Equids are susceptible to SARS-CoV-2 based on the similarity of the human ACE-2 receptor and reports of infection. Clinical disease and prevalence factors associated with SARS-CoV-2 infection in equids have not yet been investigated. The aim of this study was to determine the seroprevalence of SARS-CoV-2 and selected prevalence factors in 1186 equids presented for various conditions to a Veterinary Medical Teaching Hospital over a two-year period. Blood samples were tested for SARS-CoV-2 antibodies using an ELISA targeting the receptor binding domain (RBD) of the SARS-CoV-2 spike protein. Further, selected prevalence factors (season, age, breed, sex, presenting complaint) were retrieved from the medical records. No information was available on whether the horses had come into contact with COVID-19-positive individuals. Among the study animals, 42/1186 (3.5%) horses had detectable SARS-CoV-2 antibodies. Amongst the prevalence factors investigated, only seasonality (spring) was associated with a greater frequency of seropositivity to SARS-CoV-2. Horses with medical and surgical complaints were more likely to test seropositive to SARS-CoV-2 compared to horses presented for routine health care procedures, suggesting more frequent and/or longer interactions with individuals with COVID-19. While horses can become infected with SARS-CoV-2 via the occasional spillover from COVID-19 individuals, clinical disease expression remains subclinical, making horses an unlikely contributor to the spread of SARS-CoV-2.


Subject(s)
COVID-19 , SARS-CoV-2 , Animals , Antibodies, Viral , California , COVID-19/epidemiology , COVID-19/veterinary , Horses/virology , Hospitals, Teaching , Pandemics , Seroepidemiologic Studies , Hospitals, Animal
15.
Euro Surveill ; 27(25)2022 06.
Article in English | MEDLINE | ID: mdl-35748300

ABSTRACT

BackgroundWest Nile virus (WNV) and Usutu virus (USUV), two closely related flaviviruses, mainly follow an enzootic cycle involving mosquitoes and birds, but also infect humans and other mammals. Since 2010, their epidemiological situation may have shifted from irregular epidemics to endemicity in several European regions; this requires confirmation, as it could have implications for risk assessment and surveillance strategies.AimTo explore the seroprevalence in animals and humans and potential endemicity of WNV and USUV in Southern France, given a long history of WNV outbreaks and the only severe human USUV case in France in this region.MethodsWe evaluated the prevalence of WNV and USUV in a repeated cross-sectional study by serological and molecular analyses of human, dog, horse, bird and mosquito samples in the Camargue area, including the city of Montpellier, between 2016 and 2020.ResultsWe observed the active transmission of both viruses and higher USUV prevalence in humans, dogs, birds and mosquitoes, while WNV prevalence was higher in horses. In 500 human samples, 15 were positive for USUV and 6 for WNV. Genetic data showed that the same lineages, WNV lineage 1a and USUV lineage Africa 3, were found in mosquitoes in 2015, 2018 and 2020.ConclusionThese findings support existing literature suggesting endemisation in the study region and contribute to a better understanding of USUV and WNV circulation in Southern France. Our study underlines the importance of a One Health approach for the surveillance of these viruses.


Subject(s)
Culicidae , Flavivirus Infections , One Health , West Nile Fever , Animals , Birds/virology , Cross-Sectional Studies , Culicidae/virology , Dogs/virology , Flavivirus/genetics , Flavivirus Infections/epidemiology , Flavivirus Infections/veterinary , France/epidemiology , Horses/virology , Humans , Seroepidemiologic Studies , West Nile Fever/epidemiology , West Nile Fever/veterinary , West Nile virus/genetics
16.
Virol Sin ; 37(2): 229-237, 2022 Apr.
Article in English | MEDLINE | ID: mdl-35527224

ABSTRACT

The Getah virus (GETV), a mosquito-borne RNA virus, is widely distributed in Oceania and Asia. GETV is not the only pathogenic to horses, pigs, cattle, foxes and boars, but it can also cause fever in humans. Since its first reported case in Chinese mainland in 2017, the number of GETV-affected provinces has increased to seventeen till now. Therefore, we performed an epidemiologic investigation of GETV in the Xinjiang region, located in northwestern China, during the period of 2017-2020. ELISA was used to analyze 3299 serum samples collected from thoroughbred horse, local horse, sheep, goat, cattle, and pigs, with thoroughbred horse (74.8%), local horse (67.3%), goat (11.7%), sheep (10.0%), cattle (25.1%) and pigs (51.1%) being positive for anti-GETV antibodies. Interestingly, the neutralizing antibody titer in horses was much higher than in other species. Four samples from horses and pigs were positive for GETV according to RT-PCR. Furthermore, from the serum of a local horse, we isolated GETV which was designated as strain XJ-2019-07, and determined its complete genome sequence. From the phylogenetic relationships, it belongs to the Group III lineage. This is the first evidence of GETV associated to domestic animals in Xinjiang. Overall, GETV is prevalent in Xinjiang and probably has been for several years. Since no vaccine against GETV is available in China, detection and monitoring strategies should be improved in horses and pigs, especially imported and farmed, in order to prevent economic losses.


Subject(s)
Alphavirus , Culicidae , Alphavirus/genetics , Animals , Antibodies, Neutralizing/blood , Antibodies, Viral/blood , Cattle/virology , China/epidemiology , Culicidae/virology , Goats/virology , Horses/virology , Male , Phylogeny , Sequence Analysis, DNA , Seroepidemiologic Studies , Sheep/virology , Swine/virology
17.
Viruses ; 14(2)2022 02 05.
Article in English | MEDLINE | ID: mdl-35215918

ABSTRACT

Getah virus (GETV) is a member of the alphavirus genus, and it infects a variety of animal species, including horses, pigs, cattle, and foxes. Human infection with this virus has also been reported. The structure of GETV has not yet been determined. In this study, we report the cryo-EM structure of GETV at a resolution of 3.5 Å. This structure reveals conformational polymorphism of the envelope glycoproteins E1 and E2 at icosahedral 3-fold and quasi-3-fold axes, which is believed to be a necessary organization in forming a curvature surface of virions. In our density map, three extra densities are identified, one of which is believed a "pocket factor"; the other two are located by domain D of E2, and they may maintain the stability of E1/E2 heterodimers. We also identify three N-glycosylations at E1 N141, E2 N200, and E2 N262, which might be associated with receptor binding and membrane fusion. The resolving of the structure of GETV provides new insights into the structure and assembly of alphaviruses and lays a basis for studying the differences of biology and pathogenicity between arthritogenic and encephalitic alphaviruses.


Subject(s)
Alphavirus Infections/veterinary , Alphavirus Infections/virology , Alphavirus/physiology , Alphavirus/ultrastructure , Virus Assembly , Alphavirus/classification , Alphavirus/genetics , Animals , Cattle/virology , Cryoelectron Microscopy , Dimerization , Foxes/virology , Horses/virology , Humans , Models, Molecular , Phylogeny , Swine/virology , Viral Envelope Proteins/chemistry , Viral Envelope Proteins/genetics , Viral Envelope Proteins/metabolism , Virion/classification , Virion/genetics , Virion/physiology , Virion/ultrastructure
18.
Int J Mol Sci ; 23(4)2022 Feb 10.
Article in English | MEDLINE | ID: mdl-35216085

ABSTRACT

The equine sarcoid is one of the most common neoplasias in the Equidae family. Despite the association of this tumor with the presence of bovine papillomavirus (BPV), the molecular mechanism of this lesion has not been fully understood. The transgenization of equine adult cutaneous fibroblast cells (ACFCs) was accomplished by nucleofection, followed by detection of molecular modifications using high-throughput NGS transcriptome sequencing. The results of the present study confirm that BPV-E4- and BPV-E1^E4-mediated nucleofection strategy significantly affected the transcriptomic alterations, leading to sarcoid-like neoplastic transformation of equine ACFCs. Furthermore, the results of the current investigation might contribute to the creation of in vitro biomedical models suitable for estimating the fates of molecular dedifferentiability and the epigenomic reprogrammability of BPV-E4 and BPV-E4^E1 transgenic equine ACFC-derived sarcoid-like cell nuclei in equine somatic cell-cloned embryos. Additionally, these in vitro models seem to be reliable for thoroughly recognizing molecular mechanisms that underlie not only oncogenic alterations in transcriptomic signatures, but also the etiopathogenesis of epidermal and dermal sarcoid-dependent neoplastic transformations in horses and other equids. For those reasons, the aforementioned transgenic models might be useful for devising clinical treatments in horses afflicted with sarcoid-related neoplasia of cutaneous and subcutaneous tissues.


Subject(s)
Fibroblasts/virology , Horse Diseases/virology , Horses/virology , Neoplasms/virology , Papillomaviridae/genetics , Sarcoidosis/virology , Skin Diseases/virology , Animals , Animals, Genetically Modified/virology , Equidae/virology , Papillomavirus Infections/virology , Skin/virology , Transcriptome/genetics
19.
Arq. Inst. Biol. (Online) ; 89: e00462020, 2022. tab, mapas
Article in English | VETINDEX, LILACS | ID: biblio-1383683

ABSTRACT

The present objective was to investigate the presence of anti-equine viral encephalomyelitis (EVE) antibodies and the possible risk factors for its dissemination in horses raised in the East and West Potiguar mesoregions of the state of Rio Grande do Norte, Brazil. Serological diagnosis for neutralizing antibodies against Eastern (EEEV), Western (WEEV) and Venezuelan (VEEV). Equine viral encephalomyelitis was performed using a seroneutralization technique on 811 blood samples from horses from ninety properties and sixteen municipalities between July 2018 and February 2019. Factors associated with EVE were evaluated using an investigative epidemiological questionnaire, and the data were statistically analyzed using the Epi Info 3.5.2 software with a confidence level of 95%. The seroprevalence of anti-EVE antibodies was 14.2% (115), with 10.36% (84) for EEEV, 6.9% (56) for WEEV, and null for EVE. When analyzing risk factors, it can be concluded that horses raised in properties that do not clean installations and/or rent out their pasture are more likely to have anti-EVE antibodies. These results show evidence that horses raised in the East and West Potiguar mesoregions were exposed to EEEV and WEEV, thus reinforcing the importance of vaccination and serological survey of nonvaccinated horses as a means of monitoring the disease.


Subject(s)
Animals , Encephalomyelitis, Equine/epidemiology , Horse Diseases/diagnosis , Horses/virology , Brazil , Seroepidemiologic Studies , Encephalitis, Viral/veterinary
20.
Emerg Microbes Infect ; 10(1): 2199-2201, 2021 Dec.
Article in English | MEDLINE | ID: mdl-34749583

ABSTRACT

We report pilot studies to evaluate the susceptibility of common domestic livestock (cattle, sheep, goat, alpaca, rabbit, and horse) to intranasal infection with SARS-CoV-2. None of the infected animals shed infectious virus via nasal, oral, or faecal routes, although viral RNA was detected in several animals. Further, neutralizing antibody titres were low or non-existent one month following infection. These results suggest that domestic livestock are unlikely to contribute to SARS-CoV-2 epidemiology.


Subject(s)
COVID-19/veterinary , Host Specificity , Livestock/virology , SARS-CoV-2/pathogenicity , Animals , Antibodies, Neutralizing/blood , Antibodies, Neutralizing/immunology , Antibodies, Viral/blood , Antibodies, Viral/immunology , COVID-19/immunology , COVID-19/virology , Camelids, New World/virology , Cattle/virology , Chlorocebus aethiops , Disease Reservoirs/virology , Goats/virology , Horses/virology , Host Specificity/immunology , Humans , Nasal Cavity/virology , RNA, Viral/analysis , Rabbits/virology , Rectum/virology , Respiratory System/virology , SARS-CoV-2/genetics , SARS-CoV-2/isolation & purification , Sheep/virology , Species Specificity , Vero Cells , Virus Shedding , Viscera/virology
SELECTION OF CITATIONS
SEARCH DETAIL