Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
1.
J Gen Virol ; 102(1)2021 01.
Artigo em Inglês | MEDLINE | ID: mdl-32589123

RESUMO

While the presence of bovine spongiform encephalopathy (BSE) infectivity in the blood of clinically affected sheep has been proven by intraspecies blood-transfusion experiments, this question has remained open in the case of BSE-affected cattle. Although the absence of infectivity can be anticipated from the restriction of the agent to neuronal tissues in this species, evidence for this was still lacking. This particularly concerns the production and use of medicinal products and other applications containing bovine blood or preparations thereof. We therefore performed a blood-transfusion experiment from cattle in the clinical end stage of disease after experimental challenge with either classical (C-BSE) or atypical (H- and l-) BSE into calves at 4-6 months of age. The animals were kept in a free-ranging group for 10 years. Starting from 24 months post-transfusion, a thorough clinical examination was performed every 6 weeks in order to detect early symptoms of a BSE infection. Throughout the experiment, the clinical picture of all animals gave no indication of a BSE infection. Upon necropsy, the brainstem samples were analysed by BSE rapid test as well as by the highly sensitive Protein Misfolding Cyclic Amplification (PMCA), all with negative results. These results add resilient data to confirm the absence of BSE infectivity in the donor blood collected from C-, H- and l-BSE-affected cattle even in the final clinical phase of the disease. This finding has important implications for the risk assessment of bovine blood and blood products in the production of medicinal products and other preparations.


Assuntos
Transfusão de Sangue/veterinária , Encefalopatia Espongiforme Bovina/transmissão , Animais , Encéfalo/metabolismo , Bovinos , Encefalopatia Espongiforme Bovina/sangue , Encefalopatia Espongiforme Bovina/metabolismo , Resultados Negativos , Proteínas PrPSc/química , Proteínas PrPSc/isolamento & purificação , Dobramento de Proteína
2.
Int J Mol Sci ; 22(21)2021 Oct 20.
Artigo em Inglês | MEDLINE | ID: mdl-34768738

RESUMO

After oral exposure of cattle with classical bovine spongiform encephalopathy (C-BSE), the infectious agent ascends from the gut to the central nervous system (CNS) primarily via the autonomic nervous system. However, the timeline of this progression has thus far remained widely undetermined. Previous studies were focused on later time points after oral exposure of animals that were already 4 to 6 months old when challenged. In contrast, in this present study, we have orally inoculated 4 to 6 weeks old unweaned calves with high doses of BSE to identify any possible BSE infectivity and/or PrPBSE in peripheral nervous tissues during the first eight months post-inoculation (mpi). For the detection of BSE infectivity, we used a bovine PrP transgenic mouse bioassay, while PrPBSE depositions were analyzed by immunohistochemistry (IHC) and by protein misfolding cyclic amplification (PMCA). We were able to show that as early as 8 mpi the thoracic spinal cord as well as the parasympathetic nodal ganglion of these animals contained PrPBSE and BSE infectivity. This shows that the centripetal prion spread starts early after challenge at least in this age group, which represents an essential piece of information for the risk assessments for food, feed, and pharmaceutical products produced from young calves.


Assuntos
Encefalopatia Espongiforme Bovina/fisiopatologia , Encefalopatia Espongiforme Bovina/transmissão , Fatores Etários , Animais , Bovinos , Sistema Nervoso Central/metabolismo , Progressão da Doença , Encefalopatia Espongiforme Bovina/metabolismo , Feminino , Masculino , Camundongos , Camundongos Transgênicos , Nervos Periféricos/metabolismo , Proteínas PrPSc/metabolismo , Proteínas Priônicas/metabolismo , Príons/metabolismo , Príons/patogenicidade , Medula Espinal/metabolismo
3.
J Virol ; 90(1): 400-11, 2016 01 01.
Artigo em Inglês | MEDLINE | ID: mdl-26491158

RESUMO

UNLABELLED: In 1999, after circulation for a few months in poultry in Italy, low-pathogenic (LP) avian influenza (AI) H7N1 virus mutated into a highly pathogenic (HP) form by acquisition of a unique multibasic cleavage site (mCS), PEIPKGSRVRR*GLF (asterisk indicates the cleavage site), in the hemagglutinin (HA) and additional alterations with hitherto unknown biological function. To elucidate these virulence-determining alterations, recombinant H7N1 viruses carrying specific mutations in the HA of LPAI A/chicken/Italy/473/1999 virus (Lp) and HPAI A/chicken/Italy/445/1999 virus (Hp) were generated. Hp with a monobasic CS or carrying the HA of Lp induced only mild or no disease in chickens, thus resembling Lp. Conversely, Lp with the HA of Hp was as virulent and transmissible as Hp. While Lp with a multibasic cleavage site (Lp_CS445) was less virulent than Hp, full virulence was exhibited when HA2 was replaced by that of Hp. In HA2, three amino acid differences consistently detected between LP and HP H7N1 viruses were successively introduced into Lp_CS445. Q450L in the HA2 stem domain increased virulence and transmission but was detrimental to replication in cell culture, probably due to low-pH activation of HA. A436T and/or K536R restored viral replication in vitro and in vivo. Viruses possessing A436T and K536R were observed early in the HPAI outbreak but were later superseded by viruses carrying all three mutations. Together, besides the mCS, stepwise mutations in HA2 increased the fitness of the Italian H7N1 virus in vivo. The shift toward higher virulence in the field was most likely gradual with rapid optimization. IMPORTANCE: In 1999, after 9 months of circulation of low-pathogenic (LP) avian influenza virus (AIV), a devastating highly pathogenic (HP) H7N1 AIV emerged in poultry, marking the largest epidemic of AIV reported in a Western country. The HPAIV possessed a unique multibasic cleavage site (mCS) complying with the minimum motif for HPAIV. The main finding in this report is the identification of three mutations in the HA2 domain that are required for replication and stability, as well as for virulence, transmission, and tropism of H7N1 in chickens. In addition to the mCS, Q450L was required for full virulence and transmissibility of the virus. Nonetheless, it was detrimental to virus replication and required A436T and/or K536R to restore replication, systemic spread, and stability. These results are important for better understanding of the evolution of highly pathogenic avian influenza viruses from low-pathogenic precursors.


Assuntos
Hemaglutininas Virais/genética , Hemaglutininas Virais/metabolismo , Vírus da Influenza A Subtipo H7N1/crescimento & desenvolvimento , Vírus da Influenza A Subtipo H7N1/genética , Influenza Aviária/patologia , Influenza Aviária/virologia , Mutação de Sentido Incorreto , Animais , Galinhas , Itália , Proteínas Mutantes/genética , Proteínas Mutantes/metabolismo , Recombinação Genética , Genética Reversa , Virulência
4.
Vet Res ; 48(1): 88, 2017 12 19.
Artigo em Inglês | MEDLINE | ID: mdl-29258602

RESUMO

In classical bovine spongiform encephalopathy (C-BSE), an orally acquired prion disease of cattle, the ileal Peyer's patch (IPP) represents the main entry port for the BSE agent. In earlier C-BSE pathogenesis studies, cattle at 4-6 months of age were orally challenged, while there are strong indications that the risk of infection is highest in young animals. In the present study, unweaned calves aged 4-6 weeks were orally challenged to determine the earliest time point at which newly formed PrPBSE and BSE infectivity are detectable in the IPP. For this purpose, calves were culled 1 week as well as 2, 4, 6 and 8 months post-infection (mpi) and IPPs were examined for BSE infectivity using a bovine PrP transgenic mouse bioassay, and for PrPBSE by immunohistochemistry (IHC) and protein misfolding cyclic amplification (PMCA) assays. For the first time, BSE prions were detected in the IPP as early as 2 mpi by transgenic mouse bioassay and PMCA and 4 mpi by IHC in the follicular dendritic cells (FDCs) of the IPP follicles. These data indicate that BSE prions propagate in the IPP of unweaned calves within 2 months of oral uptake of the agent.


Assuntos
Encefalopatia Espongiforme Bovina/etiologia , Nódulos Linfáticos Agregados/metabolismo , Proteínas Priônicas/metabolismo , Animais , Bovinos , Feminino , Masculino , Camundongos , Camundongos Transgênicos
5.
Arch Virol ; 162(10): 3119-3129, 2017 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-28702933

RESUMO

In late 2011, Schmallenberg virus (SBV), a novel, arthropod-borne, teratogenic orthobunyavirus, emerged near the German/Dutch border and thereafter spread rapidly throughout the continent thereby causing great economic losses in European livestock. SBV mainly infects ruminants and closely related viruses such as Sabo virus (SABOV), Simbu virus (SIMBUV) and Sathuperi virus (SATV) have been isolated from their insect-vectors or putative ruminant hosts. However, information about their pathogenesis and in vivo studies with SABOV, SIMBUV, and SATV are scarce. As experimental infections of ruminants are comprehensive and time-consuming, an SBV small animal model was assessed regarding its suitability for studying Simbu viruses. Adult type I interferon deficient mice (IFNAR-/-) were subcutaneously infected with the Simbu serogroup members SABOV, SIMV and SATV, respectively, and compared to SBV-infected mice. All animals were clinically, virologically, serologically, and pathologically examined. The clinical signs were mainly characterised by the loss of body weight and by paralysis. In blood, and samples from the spleen and brain, high loads of viral genome were detected using newly developed real-time PCR assays. The most common histologic lesions included meningo-encephalomyelitis, perivascular cuffing of lymphocytes and macrophages, neuronal degeneration and gliosis. These lesions have also been described in foetuses after transplacental infection with SBV. In-situ hybridisation signals were widely distributed in multiple neurons of the brain and spinal cord in all examined, inoculated mice. In conclusion, IFNAR-/- mice are a suitable animal model for pathogenesis studies of a broad range of Simbu serogroup viruses since all the viruses examined displayed a common pattern of viral organ and tissue distribution in this mouse model.


Assuntos
Infecções por Bunyaviridae/imunologia , Receptor de Interferon alfa e beta/metabolismo , Vírus Simbu , Animais , Linhagem Celular , Modelos Animais de Doenças , Regulação da Expressão Gênica/imunologia , Camundongos , Camundongos Knockout , Reação em Cadeia da Polimerase em Tempo Real , Receptor de Interferon alfa e beta/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa
6.
J Gen Virol ; 97(12): 3193-3204, 2016 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-27902339

RESUMO

Highly pathogenic H5N1 avian influenza virus (A/H5N1) devastated the poultry industry and continues to pose a pandemic threat. Studying the progressive genetic changes in A/H5N1 after long-term circulation in poultry may help us to better understand A/H5N1 biology in birds. A/H5N1 clade 2.2.1.1 antigenic drift viruses have been isolated from vaccinated commercial poultry in Egypt. They exhibit a peculiar stepwise accumulation of glycosylation sites (GS) in the haemagglutinin (HA) with viruses carrying, beyond the conserved 5 GS, additional GS at amino acid residues 72, 154, 236 and 273 resulting in 6, 7, 8 or 9 GS in the HA. Available information about the impact of glycosylation on virus fitness and pathobiology is mostly derived from mammalian models. Here, we generated recombinant viruses imitating the progressive acquisition of GS in HA and investigated their biological relevance in vitro and in vivo. Our in vitro results indicated that the accumulation of GS correlated with increased glycosylation, increased virus replication, neuraminidase activity, cell-to-cell spread and thermostability, however, strikingly, without significant impact on virus escape from neutralizing antibodies. In vivo, glycosylation modulated virus virulence, tissue tropism, replication and chicken-to-chicken transmission. Predominance in the field was towards viruses with hyperglycosylated HA. Together, progressive glycosylation of the HA may foster persistence of A/H5N1 by increasing replication, stability and bird-to-bird transmission without significant impact on antigenic drift.


Assuntos
Variação Antigênica , Glicoproteínas de Hemaglutininação de Vírus da Influenza/genética , Glicoproteínas de Hemaglutininação de Vírus da Influenza/metabolismo , Virus da Influenza A Subtipo H5N1/fisiologia , Influenza Aviária/transmissão , Doenças das Aves Domésticas/virologia , Replicação Viral , Motivos de Aminoácidos , Animais , Galinhas , Egito , Glicosilação , Glicoproteínas de Hemaglutininação de Vírus da Influenza/química , Virus da Influenza A Subtipo H5N1/classificação , Virus da Influenza A Subtipo H5N1/genética , Virus da Influenza A Subtipo H5N1/patogenicidade , Influenza Aviária/virologia , Filogenia , Virulência
7.
J Infect Dis ; 212(4): 664-72, 2015 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-25722297

RESUMO

BACKGROUND: The prion protein-encoding gene (PRNP) is one of the major determinants for scrapie occurrence in sheep and goats. However, its effect on bovine spongiform encephalopathy (BSE) transmission to goats is not clear. METHODS: Goats harboring wild-type, R/Q211 or Q/K222 PRNP genotypes were orally inoculated with a goat-BSE isolate to assess their relative susceptibility to BSE infection. Goats were killed at different time points during the incubation period and after the onset of clinical signs, and their brains as well as several peripheral tissues were analyzed for the accumulation of pathological prion protein (PrP(Sc)) and prion infectivity by mouse bioassay. RESULTS: R/Q211 goats displayed delayed clinical signs compared with wild-type goats. Deposits of PrP(Sc) were detected only in brain, whereas infectivity was present in peripheral tissues too. In contrast, none of the Q/K222 goats showed any evidence of clinical prion disease. No PrP(Sc) accumulation was observed in their brains or peripheral tissues, but very low infectivity was detected in some tissues very long after inoculation (44-45 months). CONCLUSIONS: These results demonstrate that transmission of goat BSE is genotype dependent, and they highlight the pivotal protective effect of the K222 PRNP variant in the oral susceptibility of goats to BSE.


Assuntos
Encefalopatia Espongiforme Bovina/transmissão , Doenças das Cabras/patologia , Doenças das Cabras/transmissão , Príons/genética , Animais , Bovinos , Predisposição Genética para Doença , Genótipo , Cabras , Camundongos , Camundongos Transgênicos , Especificidade da Espécie
8.
J Virol ; 88(16): 9153-65, 2014 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-24899194

RESUMO

UNLABELLED: Human influenza cases caused by a novel avian H7N9 virus in China emphasize the zoonotic potential of that subtype. We compared the infectivity and pathogenicity of the novel H7N9 virus with those of a recent European avian H7N7 strain in chickens, pigeons, and ferrets. Neither virus induced signs of disease despite substantial replication in inoculated chickens and rapid transmission to contact chickens. Evidence of the replication of both viruses in pigeons, albeit at lower levels of RNA excretion, was also detected. No clear-cut differences between the two H7 isolates emerged regarding replication and antibody development in avian hosts. In ferrets, in contrast, greater replication of the avian H7N9 virus than of the H7N7 strain was observed with significant differences in viral presence, e.g., in nasal wash, lung, and cerebellum samples. Importantly, both viruses showed the potential to spread to the mammal brain. We conclude that efficient asymptomatic viral replication and shedding, as shown in chickens, facilitate the spread of H7 viruses that may harbor zoonotic potential. Biosafety measures are required for the handling of poultry infected with avian influenza viruses of the H7 subtype, independently of their pathogenicity for gallinaceous poultry. IMPORTANCE: This study is important to the field since it provides data about the behavior of the novel H7N9 avian influenza virus in chickens, pigeons, and ferrets in comparison with that of a recent low-pathogenicity H7N7 strain isolated from poultry. We clearly show that chickens, but not pigeons, are highly permissive hosts of both H7 viruses, allowing high-titer replication and virus shedding without any relevant clinical signs. In the ferret model, the potential of both viruses to infect mammals could be demonstrated, including infection of the brain. However, the replication efficiency of the H7N9 virus in ferrets was higher than that of the H7N7 strain. In conclusion, valuable data for the risk analysis of low-pathogenicity avian influenza viruses of the H7 subtype are provided that could also be used for the risk assessment of zoonotic potentials and necessary biosafety measures.


Assuntos
Galinhas/virologia , Columbidae/virologia , Furões/virologia , Vírus da Influenza A Subtipo H7N7/genética , Subtipo H7N9 do Vírus da Influenza A/genética , Virulência/genética , Adulto , Animais , Aves/virologia , China , Feminino , Humanos , Influenza Aviária/genética , Influenza Aviária/virologia , Influenza Humana/genética , Influenza Humana/virologia , Infecções por Orthomyxoviridae/genética , Infecções por Orthomyxoviridae/virologia , RNA Viral/genética , Replicação Viral/genética , Eliminação de Partículas Virais/genética
9.
Arch Virol ; 160(7): 1657-67, 2015 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-25916610

RESUMO

In 2007, African swine fever virus (ASFV) was introduced into the Transcaucasian countries and Russia. Since then, it has spread alarmingly and reached the European Union. ASFV strains are highly virulent and lead to almost 100% mortality under experimental conditions. However, the possibility of dose-dependent disease courses has been discussed. For this reason, a study was undertaken to assess the risk of chronic disease and the establishment of carriers upon low-dose oronasal infection of domestic pigs and European wild boar. It was demonstrated that very low doses of ASFV are sufficient to infect especially weak or runted animals by the oronasal route. Some of these animals did not show clinical signs indicative of ASF, and they developed almost no fever. However, no changes were observed in individual animal regarding the onset, course and outcome of infection as assessed by diagnostic tests. After amplification of ASFV by these animals, pen- and stablemates became infected and developed acute lethal disease with similar characteristics in all animals. Thus, we found no indication of prolonged or chronic individual courses upon low-dose infection in either species. The scattered onset of clinical signs and pathogen detection within and among groups confirms moderate contagiosity that is strongly linked with blood contact. In conclusion, the prolonged course at the "herd level" together with the exceptionally low dose that proved to be sufficient to infect a runted wild boar could be important for disease dynamics in wild-boar populations and in backyard settings.


Assuntos
Vírus da Febre Suína Africana/patogenicidade , Peste Suína Clássica/transmissão , Peste Suína Clássica/virologia , Vírus da Febre Suína Africana/classificação , Vírus da Febre Suína Africana/genética , Vírus da Febre Suína Africana/isolamento & purificação , Animais , Peste Suína Clássica/epidemiologia , Peste Suína Clássica/mortalidade , Europa (Continente)/epidemiologia , Federação Russa/epidemiologia , Sus scrofa/virologia , Suínos , Virulência
10.
Vet Ophthalmol ; 16 Suppl 1: 130-4, 2013 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-23217015

RESUMO

Two sibling snow leopards, a male and a female, with bilateral anterior segment dysgenesis (ASD), are reported. Both snow leopards also had colobomas of both upper eyelids. All eyes exhibited a central corneal opacity associated with a defect in posterior corneal stroma, endothelium and Descemet's membrane. Iris strands were present attached to the termination of Descemet's membrane and to the periphery of the posterior corneal defect. The iris was hypoplastic, and cataract was present in all four eyes. The left eye of the female was microphthalmic, with no trabecular meshwork and with persistent remnant of the hyaloid artery. The male had hydrocephalus and thus some of the features of Peters' plus syndrome (Peters' anomaly in addition to systemic malformations). The histological findings in the eyes of these snow leopard siblings are identical with those described in humans with Peters' anomaly.


Assuntos
Anormalidades do Olho/veterinária , Felidae , Animais , Animais de Zoológico , Olho/patologia , Anormalidades do Olho/patologia , Feminino , Masculino
11.
Transbound Emerg Dis ; 66(1): 454-462, 2019 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-30354028

RESUMO

Since its first appearance in 2011, Schmallenberg virus (SBV) has been repeatedly detected in aborted ruminant foetuses or severely malformed newborns whose mothers were naturally infected during pregnancy. However, especially the knowledge about dynamics of foetal infection in cattle is still scarce. Therefore, a total of 36 pregnant heifers were experimentally infected during two animal trials with SBV between days 60 and 150 of gestation. The foetuses were collected between 10 and 35 days after infection and virologically and pathologically investigated. Overall, 33 heifers yielded normally developed, macroscopically inconspicuous foetuses, but abundant virus replication was evident at the maternal/foetal interface and viral genome was detectable in at least one organ system of 18 out of 35 foetuses. One heifer was found to be not pregnant at autopsy. One of the animals aborted at day 4 after infection, viral RNA was detectable in the lymphatic tissue of the dam, in the maternal and foetal placenta, and in organs and lymphatic tissue of the foetus. In another foetus, SBV typical malformations like torticollis and arthrogryposis were observed. The corresponding dam was infected at day 90 of pregnancy and viral genome was detectable in the cerebellum of the unborn. Interestingly, no common patterns of infected foetal organs or maternal/foetal placentas could be identified, and both, sites of virus replication and genome loads, varied to a high degree in the individual foetuses. It is therefore concluded, that SBV infects in many cases also the bovine foetus of naïve pregnant cattle, however, the experimentally observed low abortion/malformation rate is in concordance to the reported low rates in the field during the first outbreak wave following the introduction of SBV. This observation speaks for a natural resistance of most bovine foetuses even during the vulnerable phase of early pregnancy, which has to be further studied in the future.


Assuntos
Infecções por Bunyaviridae/veterinária , Doenças dos Bovinos/transmissão , Transmissão Vertical de Doenças Infecciosas/veterinária , Orthobunyavirus/patogenicidade , Complicações Infecciosas na Gravidez/veterinária , Feto Abortado/virologia , Aborto Animal/virologia , Animais , Infecções por Bunyaviridae/transmissão , Infecções por Bunyaviridae/virologia , Bovinos , Doenças dos Bovinos/virologia , Surtos de Doenças , Feminino , Feto/virologia , Gravidez , Complicações Infecciosas na Gravidez/virologia , Prenhez , Ruminantes
12.
Viruses ; 11(1)2018 12 20.
Artigo em Inglês | MEDLINE | ID: mdl-30577433

RESUMO

Hepatitis E virus (HEV) is the causative agent of acute hepatitis E in humans in developing countries, but autochthonous cases of zoonotic genotype 3 (HEV-3) infection also occur in industrialized countries. In contrast to swine, rats, and rabbits, natural HEV infections in mice have not yet been demonstrated. The pig represents a well-established large animal model for HEV-3 infection, but a suitable small animal model mimicking natural HEV-3 infection is currently missing. Therefore, we experimentally inoculated C57BL/6 mice (wild-type, IFNAR-/-, CD4-/-, CD8-/-) and BALB/c nude (nu/nu) mice, Wistar rats, and European rabbits with a wild boar-derived HEV-3 strain and monitored virus replication and shedding, as well as humoral immune responses. HEV RNA and anti-HEV antibodies were detected in one and two out of eight of the rats and all rabbits inoculated, respectively, but not in any of the mouse strains tested. Remarkably, immunosuppressive dexamethasone treatment of rats did not enhance their susceptibility to HEV infection. In rabbits, immunization with recombinant HEV-3 and ratHEV capsid proteins induced protection against HEV-3 challenge. In conclusion, the rabbit model for HEV-3 infection may serve as a suitable alternative to the non-human primate and swine models, and as an appropriate basis for vaccine evaluation studies.


Assuntos
Modelos Animais de Doenças , Hepatite E/imunologia , Imunidade Humoral , Replicação Viral , Eliminação de Partículas Virais , Animais , Dexametasona/administração & dosagem , Fezes/virologia , Feminino , Genótipo , Anticorpos Anti-Hepatite/sangue , Hepatite E/prevenção & controle , Vírus da Hepatite E/genética , Vírus da Hepatite E/isolamento & purificação , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Knockout , RNA Viral , Coelhos , Ratos , Ratos Wistar , Vacinas Sintéticas/imunologia , Vacinas contra Hepatite Viral/imunologia
13.
Berl Munch Tierarztl Wochenschr ; 128(5-6): 169-76, 2015.
Artigo em Alemão | MEDLINE | ID: mdl-26054220

RESUMO

African swine fever (ASF), a disease notifiable to the World Organization of Animal Health (OIE), is characterized by severe, unspecific clinical signs and high mortality rates. Hosts for ASF virus (ASFV) are only members of the family Suidae and soft ticks of the genus Ornithodoros. Currently, no vaccine is available and therefore, the control is primarily based on strict sanitary measures. The most important part is the early detection of the disease within affected animal holdings and the fast and reliable confirmation by laboratory diagnosis. Infections of domestic pigs and European wild boar with recent Armenian, Sardinian, Lithuanian or Kenyan ASFV isolates lead to severe, acute disease courses with the predominant symptom of high fever (> 41 degrees C) accompanied by further unspecific clinical signs such as lethargy, loss of appetite, diarrhoea, respiratory symptoms, and an increased bleeding tendency. In experimental infection studies the mortality rate reached 100%. The most prominent pathomorphological findings included ebony-colored gastrohepatic lymph nodes, lung oedema, petechiae in the renal cortex, and oedema of the gallbladder wall. In the light of the current epidemiological situation with endemic ASFV infections on Sardinia, outbreaks in Russia and several Eastern EU Member States there is a risk for an introduction in further, previously unaffected EU countries including Germany. Hence, appropriate sample materials (serum, blood, spleen) of domestic pigs with unspecific clinical symptoms or pathomorphological findings should be examined for both ASFV and classical swine fever virus.


Assuntos
Vírus da Febre Suína Africana/classificação , Vírus da Febre Suína Africana/isolamento & purificação , Febre Suína Africana/virologia , Febre Suína Africana/epidemiologia , Febre Suína Africana/fisiopatologia , Animais , Surtos de Doenças/veterinária , Europa (Continente)/epidemiologia , Suínos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA