Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 33
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
J Virol ; 97(3): e0009923, 2023 03 30.
Artículo en Inglés | MEDLINE | ID: mdl-36856426

RESUMEN

The emergence of several bat coronavirus-related disease outbreaks in human and domestic animals has fueled surveillance of coronaviruses in bats worldwide. However, little is known about how these viruses interact with their natural hosts. We demonstrate a Betacoronavirus (subgenus Merbecovirus), PN-ßCoV, in the intestine of its natural host, Nathusius's Pipistrelle Bat (Pipistrellus nathusii), by combining molecular and microscopy techniques. Eighty-eight P. nathusii bat carcasses were tested for PN-ßCoV RNA by RT-qPCR, of which 25 bats (28%) tested positive. PN-ßCoV RNA was more often detected in samples of the intestinal tract than in other sample types. In addition, viral RNA loads were higher in intestinal samples compared to other sample types, both on average and in each individual bat. In one bat, we demonstrated Merbecovirus antigen and PN-ßCoV RNA expression in intestinal epithelium and the underlying connective tissue using immunohistochemistry and in situ hybridization, respectively. These results indicate that PN-ßCoV has a tropism for the intestinal epithelium of its natural host, Nathusius's Pipistrelle Bat, and imply that the fecal-oral route is a possible route of transmission. IMPORTANCE Virtually all mammal species circulate coronaviruses. Most of these viruses will infect one host species; however, coronaviruses are known to include species that can infect multiple hosts, for example the well-known virus that caused a pandemic, SARS-CoV-2. Chiroptera (bats) include over 1,400 different species, which are expected to harbor a great variety of coronaviruses. However, we know very little about how any of these coronaviruses interact with their bat hosts; for example, we do not know their modes of transmissions, or which cells they infect. Thus, we have a limited understanding of coronavirus infections in this important host group. The significance of our study is that we learned that a bat coronavirus that occurs in a common bat species in Europe has a tropism for the intestines. This implies the fecal-oral route is a likely transmission route.


Asunto(s)
COVID-19 , Quirópteros , Coronaviridae , Coronavirus del Síndrome Respiratorio de Oriente Medio , Animales , Humanos , Filogenia , SARS-CoV-2 , Intestinos , Tropismo , ARN
2.
Emerg Infect Dis ; 29(4): 835-838, 2023 04.
Artículo en Inglés | MEDLINE | ID: mdl-36958025

RESUMEN

In August 2021, a large-scale mortality event affected harbor porpoises (Phocoena phocoena) in the Netherlands. Pathology and ancillary testing of 22 animals indicated that the most likely cause of death was Erysipelothrix rhusiopathiae infection. This zoonotic agent poses a health hazard for cetaceans and possibly for persons handling cetacean carcasses.


Asunto(s)
Erysipelothrix , Phocoena , Animales , Países Bajos/epidemiología
3.
J Zoo Wildl Med ; 53(1): 41-49, 2022 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-35339148

RESUMEN

Since the emergence of the Goose/Guangdong H5 lineage in 1996 and spillover of highly pathogenic avian influenza (HPAI) from poultry to wild birds, outbreaks have become increasingly frequent in wild birds. The latest outbreak in the Netherlands occurred in the fall-winter of 2020-2021 and was linked to incursions of HPAI H5N8 virus. During the larger national outbreak, wild birds in rehabilitation center "Vogelklas Karel Schot (VKS)" in Rotterdam presented with clinical signs compatible with HPAI, including head shaking, torticollis, and abnormal gait. During an epidemiologic investigation at VKS, water samples from the pools in the enclosures and oropharyngeal and cloacal swabs from 128 birds of different species were analyzed for the presence of H5N8 virus. Forty-five birds and the pool water tested positive for the virus. The outbreak at VKS was likely introduced by one or more infected geese (Anser anser, Anser anser domesticus, Branta leucopsis), after which the virus spread via pool water and with the relocation of infected birds within the center. In principle, such outbreaks are preventable. Recent updates about HPAI to provide guidance to help avoid future incursions of HPAI into wildlife rescue centers are reported.


Asunto(s)
Subtipo H5N8 del Virus de la Influenza A , Gripe Aviar , Animales , Animales Salvajes , Brotes de Enfermedades/veterinaria , Gripe Aviar/epidemiología , Países Bajos/epidemiología
4.
J Virol ; 93(16)2019 08 15.
Artículo en Inglés | MEDLINE | ID: mdl-31167913

RESUMEN

Middle East respiratory syndrome coronavirus (MERS-CoV) uses the S1B domain of its spike protein to bind to dipeptidyl peptidase 4 (DPP4), its functional receptor, and its S1A domain to bind to sialic acids. The tissue localization of DPP4 in humans, bats, camelids, pigs, and rabbits generally correlates with MERS-CoV tropism, highlighting the role of DPP4 in virus pathogenesis and transmission. However, MERS-CoV S1A does not indiscriminately bind to all α2,3-sialic acids, and the species-specific binding and tissue distribution of these sialic acids in different MERS-CoV-susceptible species have not been investigated. We established a novel method to detect these sialic acids on tissue sections of various organs of different susceptible species by using nanoparticles displaying multivalent MERS-CoV S1A We found that the nanoparticles specifically bound to the nasal epithelial cells of dromedary camels, type II pneumocytes in human lungs, and the intestinal epithelial cells of common pipistrelle bats. Desialylation by neuraminidase abolished nanoparticle binding and significantly reduced MERS-CoV infection in primary susceptible cells. In contrast, S1A nanoparticles did not bind to the intestinal epithelium of serotine bats and frugivorous bat species, nor did they bind to the nasal epithelium of pigs and rabbits. Both pigs and rabbits have been shown to shed less infectious virus than dromedary camels and do not transmit the virus via either contact or airborne routes. Our results depict species-specific colocalization of MERS-CoV entry and attachment receptors, which may be relevant in the transmission and pathogenesis of MERS-CoV.IMPORTANCE MERS-CoV uses the S1B domain of its spike protein to attach to its host receptor, dipeptidyl peptidase 4 (DPP4). The tissue localization of DPP4 has been mapped in different susceptible species. On the other hand, the S1A domain, the N-terminal domain of this spike protein, preferentially binds to several glycotopes of α2,3-sialic acids, the attachment factor of MERS-CoV. Here we show, using a novel method, that the S1A domain specifically binds to the nasal epithelium of dromedary camels, alveolar epithelium of humans, and intestinal epithelium of common pipistrelle bats. In contrast, it does not bind to the nasal epithelium of pigs or rabbits, nor does it bind to the intestinal epithelium of serotine bats and frugivorous bat species. This finding supports the importance of the S1A domain in MERS-CoV infection and tropism, suggests its role in transmission, and highlights its potential use as a component of novel vaccine candidates.


Asunto(s)
Infecciones por Coronavirus/metabolismo , Infecciones por Coronavirus/virología , Coronavirus del Síndrome Respiratorio de Oriente Medio/fisiología , Receptores Virales/metabolismo , Internalización del Virus , Animales , Camelus , Línea Celular , Quirópteros , Células Epiteliales/metabolismo , Células Epiteliales/virología , Especificidad del Huésped , Humanos , Membrana Mucosa/metabolismo , Membrana Mucosa/patología , Membrana Mucosa/virología , Unión Proteica , Dominios y Motivos de Interacción de Proteínas , Conejos , Glicoproteína de la Espiga del Coronavirus/química , Glicoproteína de la Espiga del Coronavirus/metabolismo , Porcinos
5.
Vet Res ; 51(1): 117, 2020 Sep 14.
Artículo en Inglés | MEDLINE | ID: mdl-32928280

RESUMEN

In 2016/2017, H5N8 highly pathogenic avian influenza (HPAI) virus of the Goose/Guangdong lineage spread from Asia to Europe, causing the biggest and most widespread HPAI epidemic on record in wild and domestic birds in Europe. We hypothesized that the wide dissemination of the 2016 H5N8 virus resulted at least partly from a change in tissue tropism from the respiratory tract, as in older HPAIV viruses, to the intestinal tract, as in low pathogenic avian influenza (LPAI) viruses, allowing more efficient faecal-oral transmission. Therefore, we determined the tissue tropism and associated lesions in wild birds found dead during the 2016 H5N8 epidemic, as well as the pattern of attachment of 2016 H5N8 virus to respiratory and intestinal tissues of four key wild duck species. We found that, out of 39 H5N8-infected wild birds of 12 species, four species expressed virus antigen in both respiratory and intestinal epithelium, one species only in respiratory epithelium, and one species only in intestinal epithelium. Virus antigen expression was association with inflammation and necrosis in multiple tissues. The level of attachment to wild duck intestinal epithelia of 2016 H5N8 virus was comparable to that of LPAI H4N5 virus, and higher than that of 2005 H5N1 virus for two of the four duck species and chicken tested. Overall, these results indicate that 2016 H5N8 may have acquired a similar enterotropism to LPAI viruses, without having lost the respirotropism of older HPAI viruses of the Goose/Guangdong lineage. The increased enterotropism of 2016 H5N8 implies that this virus had an increased chance to persist long term in the wild waterbird reservoir.


Asunto(s)
Animales Salvajes , Patos , Subtipo H5N8 del Virus de la Influenza A/fisiología , Gripe Aviar/virología , Tropismo Viral , Animales
7.
J Infect Dis ; 217(8): 1237-1246, 2018 03 28.
Artículo en Inglés | MEDLINE | ID: mdl-29329410

RESUMEN

Background: The 1918 Spanish H1N1 influenza pandemic was the most severe recorded influenza pandemic with an estimated 20-50 million deaths worldwide. Even though it is known that influenza viruses can cause extrarespiratory tract complications-which are often severe or even fatal-the potential contribution of extrarespiratory tissues to the pathogenesis of 1918 H1N1 virus infection has not been studied comprehensively. Methods: Here, we performed a time-course study in ferrets inoculated intranasally with 1918 H1N1 influenza virus, with special emphasis on the involvement of extrarespiratory tissues. Respiratory and extrarespiratory tissues were collected after inoculation for virological, histological, and immunological analysis. Results: Infectious virus was detected at high titers in respiratory tissues and, at lower titers in most extrarespiratory tissues. Evidence for active virus replication, as indicated by the detection of nucleoprotein by immunohistochemistry, was observed in the respiratory tract, peripheral and central nervous system, and liver. Proinflammatory cytokines were up-regulated in respiratory tissues, olfactory bulb, spinal cord, liver, heart, and pancreas. Conclusions: 1918 H1N1 virus spread to and induced cytokine responses in tissues outside the respiratory tract, which likely contributed to the severity of infection. Moreover, our data support the suggested link between 1918 H1N1 infection and central nervous system disease.


Asunto(s)
Citocinas/metabolismo , Subtipo H1N1 del Virus de la Influenza A/inmunología , Infecciones por Orthomyxoviridae/virología , Replicación Viral/fisiología , Animales , Citocinas/genética , Hurones , Regulación de la Expresión Génica , Humanos , Inflamación/metabolismo , Pulmón/patología , Infecciones por Orthomyxoviridae/patología , Enfermedades Respiratorias/virología , Distribución Tisular , Pérdida de Peso
8.
Euro Surveill ; 23(4)2018 01.
Artículo en Inglés | MEDLINE | ID: mdl-29382414

RESUMEN

IntroductionHighly pathogenic avian influenza (HPAI) viruses of subtype H5N8 were re-introduced into the Netherlands by late 2016, after detections in south-east Asia and Russia. This second H5N8 wave resulted in a large number of outbreaks in poultry farms and the deaths of large numbers of wild birds in multiple European countries. Methods: Here we report on the detection of HPAI H5N8 virus in 57 wild birds of 12 species sampled during active (32/5,167) and passive (25/36) surveillance activities, i.e. in healthy and dead animals respectively, in the Netherlands between 8 November 2016 and 31 March 2017. Moreover, we further investigate the experimental approach of wild bird serology as a contributing tool in HPAI outbreak investigations. Results: In contrast to the first H5N8 wave, local virus amplification with associated wild bird mortality has occurred in the Netherlands in 2016/17, with evidence for occasional gene exchange with low pathogenic avian influenza (LPAI) viruses. Discussion: These apparent differences between outbreaks and the continuing detections of HPAI viruses in Europe are a cause of concern. With the current circulation of zoonotic HPAI and LPAI virus strains in Asia, increased understanding of the drivers responsible for the global spread of Asian poultry viruses via wild birds is needed.


Asunto(s)
Animales Salvajes/virología , Aves/virología , Brotes de Enfermedades/veterinaria , Subtipo H5N8 del Virus de la Influenza A/aislamiento & purificación , Subtipo H5N8 del Virus de la Influenza A/patogenicidad , Gripe Aviar/mortalidad , Animales , Subtipo H5N8 del Virus de la Influenza A/clasificación , Subtipo H5N8 del Virus de la Influenza A/genética , Gripe Aviar/patología , Gripe Aviar/virología , Países Bajos/epidemiología , ARN Viral/genética , Vigilancia de Guardia , Análisis de Secuencia de ADN
9.
Arch Virol ; 162(7): 2035-2040, 2017 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-28283815

RESUMEN

Harbour porpoises (Phocoena phocoena) are the most prevalent cetaceans in the North Sea. The fecal viral flora of 21 harbour porpoises stranded along the Dutch coastline was analyzed by a metagenomics approach. Sequences of a novel cetacean mastadenovirus, designated harbour porpoise adenovirus 1 (HpAdV-1), were detected. The sequence of a 23-kbp genomic region, spanning the conserved late region, was determined using primer walking. Phylogenetic analysis indicated that HpAdV-1 is most closely related to bottlenose dolphin adenovirus and clusters with Cetartiodactyla adenoviruses. The prevalence of HpAdV-1 was low (2.6%) based on targeted PCR-screening of the intestinal contents of 151 harbour porpoises stranded between 2010 and 2013.


Asunto(s)
Infecciones por Adenoviridae/veterinaria , Adenoviridae/genética , Adenoviridae/aislamiento & purificación , Enfermedades de los Animales/virología , Phocoena/virología , Secuencia de Aminoácidos , Animales , Heces/virología , Metagenómica , Mar del Norte , Filogenia
10.
Dis Aquat Organ ; 127(1): 49-56, 2017 Dec 19.
Artículo en Inglés | MEDLINE | ID: mdl-29256427

RESUMEN

Harbour porpoises are often found to be infected by endoparasites in several organs including the lungs and stomach as well as the heart, liver and ears. Nevertheless there is still little knowledge about the impact, ecology, transmission, and virulence of these parasitic infections. Here, we profile the presence of parasites in 4 frequently infected organs (lungs, stomach, liver and ears) in relation to biological parameters of harbour porpoises stranded along the Dutch coastline between December 2008 and December 2013. We found that parasites were common, with prevalence of 68% in lungs, 74.4% in ears, 26% in stomach and 23.5% in liver. We used generalised linear models to further quantify parasite presence in relation to biological data gathered during necropsy (sex, body length and nutritive condition). Body length (used as a proxy for age) was significant in explaining parasite presence for all organs with increasing probability of having the parasite with increasing body length. For the parasitic infections in the ears and stomach the nutritive condition was an additional significant factor, with a higher probability of parasite presence in porpoises in a poorer nutritive condition. The results of this study can be used as a baseline for assessing parasite presence in harbour porpoises and are a first step towards linking parasite infections to basic biological data gathered during necropsy.


Asunto(s)
Enfermedades Parasitarias en Animales/parasitología , Phocoena/parasitología , Animales , Enfermedades del Oído/parasitología , Enfermedades del Oído/veterinaria , Hepatopatías/parasitología , Hepatopatías/veterinaria , Enfermedades Pulmonares Parasitarias/veterinaria , Países Bajos , Enfermedades Parasitarias en Animales/patología , Gastropatías/parasitología , Gastropatías/veterinaria
11.
Proc Biol Sci ; 282(1798): 20142429, 2015 Jan 07.
Artículo en Inglés | MEDLINE | ID: mdl-25429021

RESUMEN

Harbour porpoises (Phocoena phocoena) stranding in large numbers around the southern North Sea with fatal, sharp-edged mutilations have spurred controversy among scientists, the fishing industry and conservationists, whose views about the likely cause differ. The recent detection of grey seal (Halichoerus grypus) DNA in bite marks on three mutilated harbour porpoises, as well as direct observations of grey seal attacks on porpoises, have identified this seal species as a probable cause. Bite mark characteristics were assessed in a retrospective analysis of photographs of dead harbour porpoises that stranded between 2003 and 2013 (n = 1081) on the Dutch coastline. There were 271 animals that were sufficiently fresh to allow macroscopic assessment of grey seal-associated wounds with certainty. In 25% of these, bite and claw marks were identified that were consistent with the marks found on animals that had tested positive for grey seal DNA. Affected animals were mostly healthy juveniles that had a thick blubber layer and had recently fed. We conclude that the majority of the mutilated harbour porpoises were victims of grey seal attacks and that predation by this species is one of the main causes of death in harbour porpoises in The Netherlands. We provide a decision tree that will help in the identification of future cases of grey seal predation on porpoises.


Asunto(s)
Phocoena/fisiología , Conducta Predatoria , Phocidae/fisiología , Animales , Femenino , Cadena Alimentaria , Masculino , Países Bajos , Mar del Norte , Estudios Retrospectivos
12.
Dis Aquat Organ ; 112(3): 257-63, 2015 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-25590777

RESUMEN

Cetaceans are well known definitive hosts of parasitic nematodes of the genus Anisakis (Nematoda: Anisakidae). Anisakid nematodes are also a health hazard for humans, potentially causing gastrointestinal infections or allergic reactions following the consumption of infected fish. In marine mammals, the nematodes develop from third-stage larvae to adults in the stomachs. In the first (or fore-) stomach, these parasites are typically associated with mucosal ulceration; parasites have not been identified in other organs. Two small cetaceans, a bottlenose dolphin Tursiops truncatus and a harbour porpoise Phocoena phocoena, presented marked gastric A. simplex infection, as well as chronic granulomatous and ulcerative dermatitis with intralesional nematodes, bordered by epithelial hyperplasia. Nematodes in the skin of the bottlenose dolphin were morphologically similar to Anisakis spp. Morphology of the parasitic remnants in the skin lesion of the harbour porpoise was indistinct, but molecular identification confirmed the presence of A. simplex. This is the first report of Anisakis spp. infection in the skin of marine mammals.


Asunto(s)
Anisakiasis/veterinaria , Anisakis/aislamiento & purificación , Delfín Mular , Dermatitis/veterinaria , Phocoena , Animales , Anisakiasis/diagnóstico , Anisakiasis/parasitología , Anisakis/genética , Dermatitis/diagnóstico , Dermatitis/parasitología , Diagnóstico Diferencial , Femenino , Masculino , Países Bajos
13.
J Comp Pathol ; 208: 54-60, 2024 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-38211467

RESUMEN

The aim of this study was to describe the pathology in seals from which Listeria monocytogenes was isolated and investigate if the lesions' nature and severity were related to the phylogeny of isolates. L. monocytogenes was isolated from 13 of 50 (26%) dead grey seal (Halichoerus grypus) pups, six (12%) in systemic distribution, on the Isle of May, Scotland. Similar fatal L. monocytogenes-associated infections were found in a grey seal pup from Carnoustie, Scotland, and a juvenile harbour seal (Phoca vitulina) in the Netherlands. Whole genome sequencing of 15 of the L. monocytogenes isolates identified 13 multilocus sequence types belonging to the L. monocytogenes lineages I and II, but with scant phenotypic and genotypic antimicrobial resistance and limited variation in virulence factors. The phylogenetic diversity present suggests there are multiple sources of L. monocytogenes, even for seal pups born in the same colony and breeding season. This is the first description of L. monocytogenes isolated from, and detected in lesions in, pinnipeds and indicates that infection can be systemic and fatal. Therefore, listeriosis may be an emerging or overlooked disease in seals with infection originating from contamination of the marine environment.


Asunto(s)
Caniformia , Listeria monocytogenes , Phoca , Phocidae , Animales , Listeria monocytogenes/genética , Filogenia , Genotipo
14.
Front Microbiol ; 14: 1151524, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37056741

RESUMEN

Zoonotic viral infections that cause severe disease or even death in some people may be asymptomatic or mild in reservoir hosts. Comparison of the pathogenesis of these two host categories may potentially explain the difference in disease. However, infections in reservoir hosts are often neglected. Therefore, we compared the pathogenesis of rabies virus, macacine alphaherpesvirus, West Nile virus, Puumala orthohantavirus, monkeypox virus, Lassa mammarenavirus, H5N1 highly pathogenic avian influenza, Marburg virus, Nipah virus, Middle East respiratory syndrome, and simian/human immunodeficiency viruses in both humans and reservoir hosts. We showed that most aspects of the pathogeneses were remarkably similar. The remaining differences lead to the identification of tipping points in the pathogeneses that are important for explaining the disease outcome in severe human cases. Further elucidating these tipping points by studying zoonotic viral infections in their reservoir hosts may teach us how to reduce the severity of zoonotic viral diseases in humans.

15.
PLoS One ; 18(11): e0293649, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-38019737

RESUMEN

Changes in land use can modify habitat and roosting behaviour of bats, and therefore the transmission dynamics of viruses. Within bat roosts the density and contact rate among individuals increase and may facilitate the transmission of bat coronaviruses (CoVs). Landscape components supporting larger bat populations may thus lead to higher CoVs prevalence, as the number of roosts and/or roost size are likely to be higher. Hence, relationships between landscape composition and the presence of CoVs are expected to exist. To increase our understanding of the spread and shedding of coronaviruses in bat populations we studied the relationships between landscape composition and CoVs prevalence in the species Pipistrellus pipistrellus and Pipistrellus nathusii. Faecal samples were collected across The Netherlands, and were screened to detect the presence of CoV RNA. Coordinates were recorded for all faecal samples, so that landscape attributes could be quantified. Using a backward selection procedure on the basis of AIC, the landscape variables that best explained the presence of CoVs were selected in the final model. Results suggested that relationships between landscape composition and CoVs were likely associated with optimal foraging opportunities in both species, e.g. nearby water in P. nathusii or in areas with more grassland situated far away from forests for P. pipistrellus. Surprisingly, we found no positive association between built-up cover (where roosts are frequently found) and the presence of bat-CoVs for both species. We also show that samples collected from large bat roosts, such as maternity colonies, substantially increased the probability of finding CoVs in P. pipistrellus. Interestingly, while maternity colonies of P. nathusii are rarely present in The Netherlands, CoVs prevalence was similar in both species, suggesting that other mechanisms besides roost size, participate in the transmission of bat-CoVs. We encourage further studies to quantify bat roosts and colony networks over the different landscape compositions to better understand the ecological mechanisms involved in the transmission of bat-CoVs.


Asunto(s)
Quirópteros , Infecciones por Coronavirus , Coronavirus , Humanos , Embarazo , Animales , Femenino , Coronavirus/genética , Infecciones por Coronavirus/epidemiología , Infecciones por Coronavirus/veterinaria , Ecosistema , Bosques
16.
mSphere ; 8(4): e0020023, 2023 08 24.
Artículo en Inglés | MEDLINE | ID: mdl-37428085

RESUMEN

Continued circulation of A/H5N1 influenza viruses of the A/goose/Guangdong/1/96 lineage in poultry has resulted in the diversification in multiple genetic and antigenic clades. Since 2009, clade 2.3.4.4 hemagglutinin (HA) containing viruses harboring the internal and neuraminidase (NA) genes of other avian influenza A viruses have been detected. As a result, various HA-NA combinations, such as A/H5N1, A/H5N2, A/H5N3, A/H5N5, A/H5N6, and A/H5N8 have been identified. As of January 2023, 83 humans have been infected with A/H5N6 viruses, thereby posing an apparent risk for public health. Here, as part of a risk assessment, the in vitro and in vivo characterization of A/H5N6 A/black-headed gull/Netherlands/29/2017 is described. This A/H5N6 virus was not transmitted between ferrets via the air but was of unexpectedly high pathogenicity compared to other described A/H5N6 viruses. The virus replicated and caused severe lesions not only in respiratory tissues but also in multiple extra-respiratory tissues, including brain, liver, pancreas, spleen, lymph nodes, and adrenal gland. Sequence analyses demonstrated that the well-known mammalian adaptation substitution D701N was positively selected in almost all ferrets. In the in vitro experiments, no other known viral phenotypic properties associated with mammalian adaptation or increased pathogenicity were identified. The lack of transmission via the air and the absence of mammalian adaptation markers suggest that the public health risk of this virus is low. The high pathogenicity of this virus in ferrets could not be explained by the known mammalian pathogenicity factors and should be further studied. IMPORTANCE Avian influenza A/H5 viruses can cross the species barrier and infect humans. These infections can have a fatal outcome, but fortunately these influenza A/H5 viruses do not spread between humans. However, the extensive circulation and reassortment of A/H5N6 viruses in poultry and wild birds warrant risk assessments of circulating strains. Here an in-depth characterization of the properties of an avian A/H5N6 influenza virus isolated from a black-headed gull in the Netherlands was performed in vitro and in vivo, in ferrets. The virus was not transmissible via the air but caused severe disease and spread to extra-respiratory organs. Apart from the detection in ferrets of a mutation that increased virus replication, no other mammalian adaptation phenotypes were identified. Our results suggest that the risk of this avian A/H5N6 virus for public health is low. The underlying reasons for the high pathogenicity of this virus are unexplained and should be further studied.


Asunto(s)
Subtipo H5N1 del Virus de la Influenza A , Subtipo H5N2 del Virus de la Influenza A , Virus de la Influenza A , Gripe Aviar , Humanos , Animales , Hurones , Subtipo H5N1 del Virus de la Influenza A/genética , Subtipo H5N2 del Virus de la Influenza A/genética , Virus de la Influenza A/genética , Aves de Corral
17.
Acta Neuropathol Commun ; 10(1): 124, 2022 09 05.
Artículo en Inglés | MEDLINE | ID: mdl-36058935

RESUMEN

Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection is associated with various neurological complications. Although the mechanism is not fully understood, several studies have shown that neuroinflammation occurs in the acute and post-acute phase. As these studies have predominantly been performed with isolates from 2020, it is unknown if there are differences among SARS-CoV-2 variants in their ability to cause neuroinflammation. Here, we compared the neuroinvasiveness, neurotropism and neurovirulence of the SARS-CoV-2 ancestral strain D614G, the Delta (B.1.617.2) and Omicron BA.1 (B.1.1.529) variants using in vitro and in vivo models. The Omicron BA.1 variant showed reduced neurotropism and neurovirulence compared to Delta and D614G in human induced pluripotent stem cell (hiPSC)-derived cortical neurons co-cultured with astrocytes. Similar differences were obtained in Syrian hamsters inoculated with D614G, Delta and the Omicron BA.1 variant 5 days post infection. Replication in the olfactory mucosa was observed in all hamsters, but most prominently in D614G inoculated hamsters. Furthermore, neuroinvasion into the CNS via the olfactory nerve was observed in D614G, but not Delta or Omicron BA.1 inoculated hamsters. Furthermore, neuroinvasion was associated with neuroinflammation in the olfactory bulb of hamsters inoculated with D614G. Altogether, our findings suggest differences in the neuroinvasive, neurotropic and neurovirulent potential between SARS-CoV-2 variants using in vitro hiPSC-derived neural cultures and in vivo in hamsters during the acute phase of the infection.


Asunto(s)
COVID-19 , Células Madre Pluripotentes Inducidas , Animales , Cricetinae , Humanos , Mesocricetus , SARS-CoV-2
18.
Transbound Emerg Dis ; 69(6): 3339-3349, 2022 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-35988158

RESUMEN

In the Netherlands, 69 of the 126 (55%) mink farms in total became infected with SARS-CoV-2 in 2020. Despite strict biosecurity measures and extensive epidemiological investigations, the main transmission route remained unclear. A better understanding of SARS-CoV-2 transmission between mink farms is of relevance for countries where mink farming is still common practice and can be used as a case study to improve future emerging disease preparedness. We assessed whether SARS-CoV-2 spilled over from mink to free-ranging animals, and whether free-ranging animals may have played a role in farm-to-farm transmission in the Netherlands. The study encompassed farm visits, farm questionnaires, expert workshops and SARS-CoV-2 RNA and antibody testing of samples from target animal species (bats, birds and free-ranging carnivores). In this study, we show that the open housing system of mink allowed access to birds, bats and most free-ranging carnivores, and that direct and indirect contact with mink was likely after entry, especially for free-ranging carnivores and birds. This allowed SARS-CoV-2 exposure to animals entering the mink farm, and subsequent infection or mechanical carriage by the target animal species. Moreover, mink can escape farms in some cases, and two SARS-CoV-2-positive mink were found outside farm premises. No other SARS-CoV-2-RNA-positive free-ranging animals were detected, suggesting there was no abundant circulation in the species tested during the study period. To investigate previous SARS-CoV-2 infections, SARS-CoV-2 antibody detection using lung extracts of carcasses was set up and validated. One tested beech marten did have SARS-CoV-2 antibodies, but the closest SARS-CoV-2-infected mink farm was outside of its home range, making infection at a mink farm unlikely. Knowing that virus exchange between different species and the formation of animal reservoirs affects SARS-CoV-2 evolution, continued vigilance and monitoring of mink farms and surrounding wildlife remains vital.


Asunto(s)
COVID-19 , Quirópteros , Mustelidae , Animales , Visón , SARS-CoV-2 , COVID-19/epidemiología , COVID-19/veterinaria , Países Bajos/epidemiología , ARN Viral , Granjas
19.
Front Immunol ; 12: 622516, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33679766

RESUMEN

Rabies virus (RABV) is able to reach the central nervous system (CNS) without triggering a strong immune response, using multiple mechanisms to evade and suppress the host immune system. After infection via a bite or scratch from a rabid animal, RABV comes into contact with macrophages, which are the first antigen-presenting cells (APCs) that are recruited to the area and play an essential role in the onset of a specific immune response. It is poorly understood how RABV affects macrophages, and if the interaction contributes to the observed immune suppression. This study was undertaken to characterize the interactions between RABV and human monocyte-derived macrophages (MDMs). We showed that street RABV does not replicate in human MDMs. Using a recombinant trimeric RABV glycoprotein (rRABV-tG) we showed binding to the nicotinic acetylcholine receptor alpha 7 (nAChr α7) on MDMs, and confirmed the specificity using the nAChr α7 antagonist alpha-bungarotoxin (α-BTX). We found that this binding induced the cholinergic anti-inflammatory pathway (CAP), characterized by a significant decrease in tumor necrosis factor α (TNF-α) upon LPS challenge. Using confocal microscopy we found that induction of the CAP is associated with significant cytoplasmic retention of nuclear factor κB (NF-κB). Co-cultures of human MDMs exposed to street RABV and autologous T cells further revealed that the observed suppression of MDMs might affect their function as T cell activators as well, as we found a significant decrease in proliferation of CD8+ T cells and an increased production of the anti-inflammatory cytokine IL-10. Lastly, using flow cytometric analysis we observed a significant increase in expression of the M2-c surface marker CD163, hinting that street RABV might be able to affect macrophage polarization. Taken together, these results show that street RABV is capable of inducing an anti-inflammatory state in human macrophages, possibly affecting T cell functioning.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Macrófagos/inmunología , Virus de la Rabia/fisiología , Rabia/inmunología , Receptor Nicotínico de Acetilcolina alfa 7/metabolismo , Antiinflamatorios , Antígenos CD/metabolismo , Antígenos de Diferenciación Mielomonocítica/metabolismo , Diferenciación Celular , Células Cultivadas , Colinérgicos , Técnicas de Cocultivo , Humanos , Interleucina-10/metabolismo , Activación de Linfocitos , FN-kappa B/metabolismo , Neuroinmunomodulación , Unión Proteica , Receptores de Superficie Celular/metabolismo , Transducción de Señal , Células Th2/inmunología
20.
Viruses ; 13(12)2021 12 03.
Artículo en Inglés | MEDLINE | ID: mdl-34960690

RESUMEN

Many people worldwide suffer from hepatitis C virus (HCV) infection, which is frequently persistent. The lack of efficient vaccines against HCV and the unavailability of or limited compliance with existing antiviral therapies is problematic for health care systems worldwide. Improved small animal models would support further hepacivirus research, including development of vaccines and novel antivirals. The recent discovery of several mammalian hepaciviruses may facilitate such research. In this study, we demonstrated that bank voles (Clethrionomys glareolus) were susceptible to bank vole-associated Hepacivirus F and Hepacivirus J strains, based on the detection of hepaciviral RNA in 52 of 55 experimentally inoculated voles. In contrast, interferon α/ß receptor deficient C57/Bl6 mice were resistant to infection with both bank vole hepaciviruses (BvHVs). The highest viral genome loads in infected voles were detected in the liver, and viral RNA was visualized by in situ hybridization in hepatocytes, confirming a marked hepatotropism. Furthermore, liver lesions in infected voles resembled those of HCV infection in humans. In conclusion, infection with both BvHVs in their natural hosts shares striking similarities to HCV infection in humans and may represent promising small animal models for this important human disease.


Asunto(s)
Arvicolinae , Modelos Animales de Enfermedad , Hepacivirus/fisiología , Hepatitis C , Animales , Femenino , Hepacivirus/aislamiento & purificación , Hepacivirus/patogenicidad , Hepatitis C/patología , Hepatitis C/transmisión , Hepatitis C/veterinaria , Hepatitis C/virología , Interacciones Microbiota-Huesped , Humanos , Hígado/patología , Hígado/virología , Masculino , Ratones , Ratones Endogámicos C57BL , Carga Viral/fisiología , Tropismo Viral
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA