Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 47
Filter
1.
Int J Mol Sci ; 23(12)2022 Jun 16.
Article in English | MEDLINE | ID: mdl-35743187

ABSTRACT

Transmissible spongiform encephalopathies (TSE), caused by abnormal prion protein (PrPSc), affect many species. The most classical scrapie isolates harbor mixtures of strains in different proportions. While the characterization of isolates has evolved from using wild-type mice to transgenic mice, no standardization is established yet. Here, we investigated the incubation period, lesion profile and PrPSc profile induced by well-defined sheep scrapie isolates, bovine spongiform encephalopathy (BSE) and ovine BSE after intracerebral inoculation into two lines of ovine PrP (both ARQ/ARQ) overexpressing transgenic mice (Tgshp IX and Tgshp XI). All isolates were transmitted to both mouse models with an attack rate of almost 100%, but genotype-dependent differences became obvious between the ARQ and VRQ isolates. Surprisingly, BSE induced a much longer incubation period in Tgshp XI compared to Tgshp IX. In contrast to the histopathological lesion profiles, the immunohistochemical PrPSc profiles revealed discriminating patterns in certain brain regions in both models with clear differentiation of both BSE isolates from scrapie. These data provide the basis for the use of Tgshp IX and XI mice in the characterization of TSE isolates. Furthermore, the results enable a deeper appreciation of TSE strain diversity using ovine PrP overexpressing transgenic mice as a biological prion strain typing approach.


Subject(s)
Encephalopathy, Bovine Spongiform , Prions , Scrapie , Animals , Brain/metabolism , Cattle , Encephalopathy, Bovine Spongiform/metabolism , Mice , Mice, Transgenic , PrPSc Proteins/genetics , PrPSc Proteins/metabolism , Prions/metabolism , Scrapie/metabolism , Sheep
2.
J Gen Virol ; 102(1)2021 01.
Article in English | MEDLINE | ID: mdl-32589123

ABSTRACT

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.


Subject(s)
Blood Transfusion/veterinary , Encephalopathy, Bovine Spongiform/transmission , Animals , Brain/metabolism , Cattle , Encephalopathy, Bovine Spongiform/blood , Encephalopathy, Bovine Spongiform/metabolism , Negative Results , PrPSc Proteins/chemistry , PrPSc Proteins/isolation & purification , Protein Folding
3.
Transfusion ; 61(4): 1266-1277, 2021 04.
Article in English | MEDLINE | ID: mdl-33605455

ABSTRACT

BACKGROUND: Hepatitis E virus (HEV) is the leading cause of acute hepatitis throughout the world. Increasing blood component transfusion-associated HEV infections highlight the need for reliable virus inactivation procedures for plasma derivatives from pooled plasma donations. STUDY DESIGN AND METHODS: An animal infection study was conducted to evaluate the efficiency of HEV inactivation by pasteurization during the manufacturing process of the von Willebrand Factor/Factor VIII (VWF/FVIII) concentrate Haemate P/Humate-P (CSL Behring, Marburg, Germany). For this purpose, groups of pigs were inoculated with stabilized VWF/FVIII intermediate spiked with HEV-positive liver homogenate and exposed to increasing incubation times of 0, 3, 6, and 10 h at 60°C. Animals were evaluated for virus replication over 27 days and in a subsequent trial over 92 days. RESULTS: Virus replication was detected in animals up to the 6-h pasteurization group. In contrast, pasteurization for 10 h did not reveal virus detection when the observation period was 27 days. In an additional experiment using the 10-h pasteurized material, two individuals started virus excretion and seroconverted when the observation period was extended to 92 days. Based on the total infection rate (2 of 12) of the animals inoculated with the sample pasteurized for 10 h, a virus reduction factor of at least 4.7 log10 is calculated. CONCLUSION: This study demonstrates that pasteurization at 60°C for 10 h of an HEV-positive plasma derivative leads to the effective reduction of infectivity, resulting in a VWF/FVIII product with an appropriate margin of safety for HEV.


Subject(s)
Blood Component Transfusion/adverse effects , Factor VIII/administration & dosage , Hepatitis E virus/genetics , Hepatitis E/etiology , Pasteurization/methods , von Willebrand Factor/administration & dosage , Acute Disease , Animals , Biological Assay/methods , Factor VIII/analysis , Female , Heating/adverse effects , Hepatitis/epidemiology , Hepatitis/virology , Hepatitis E/prevention & control , Male , Models, Animal , Safety , Swine , Time Factors , Virus Inactivation , Virus Replication/genetics , von Willebrand Factor/analysis
4.
Int J Mol Sci ; 22(19)2021 Sep 28.
Article in English | MEDLINE | ID: mdl-34638780

ABSTRACT

Portugal was among the first European countries to report cases of Atypical Scrapie (ASc), the dominant form of Transmissible Spongiform Encephalopathy (TSE) in Portuguese small ruminants. Although the diagnostic phenotypes observed in Portuguese ASc cases seem identical to those described for Nor98, unequivocal identification requires TSE strain-typing using murine bioassays. In this regard, we initiated characterization of ASc isolates from sheep either homozygous for the ARQ genotype or the classical scrapie-resistant ARR genotype. Isolates from such genotypes were transmitted to TgshpXI mice expressing ovine PrPARQ. Mean incubation periods were 414 ± 58 and 483 ± 107 days in mice inoculated with AL141RQ/AF141RQ and AL141RR/AL141RR sheep isolates, respectively. Both isolates produced lesion profiles similar to French ASc Nor98 'discordant cases', where vacuolation was observed in the hippocampus (G6), cerebral cortex at the thalamus (G8) level, cerebellar white matter (W1) and cerebral peduncles (W3). Immunohistochemical PrPSc deposition was observed in the hippocampus, cerebellar cortex, cerebellar white matter and cerebral peduncles in the form of aggregates and fine granules. These findings were consistent with previously reported cases of ASc Nor98 transmitted to transgenic TgshpXI mice, confirming that the ASc strain present in Portuguese sheep corresponds to ASc Nor98.


Subject(s)
Genotype , Prion Diseases , Prion Proteins , Scrapie , Animals , Mice , Mice, Transgenic , Prion Diseases/genetics , Prion Diseases/metabolism , Prion Proteins/genetics , Prion Proteins/metabolism , Scrapie/genetics , Scrapie/metabolism , Sheep
5.
Vet Res ; 50(1): 97, 2019 Nov 25.
Article in English | MEDLINE | ID: mdl-31767033

ABSTRACT

Scrapie in goats has been known since 1942, the archetype of prion diseases in which only prion protein (PrP) in misfolded state (PrPSc) acts as infectious agent with fatal consequence. Emergence of bovine spongiform encephalopathy (BSE) with its zoonotic behaviour and detection in goats enhanced fears that its source was located in small ruminants. However, in goats knowledge on prion strain typing is limited. A European-wide study is presented concerning the biochemical phenotypes of the protease resistant fraction of PrPSc (PrPres) in over thirty brain isolates from transmissible spongiform encephalopathy (TSE) affected goats collected in seven countries. Three different scrapie forms were found: classical scrapie (CS), Nor98/atypical scrapie and one case of CH1641 scrapie. In addition, CS was found in two variants-CS-1 and CS-2 (mainly Italy)-which differed in proteolytic resistance of the PrPres N-terminus. Suitable PrPres markers for discriminating CH1641 from BSE (C-type) appeared to be glycoprofile pattern, presence of two triplets instead of one, and structural (in)stability of its core amino acid region. None of the samples exhibited BSE like features. BSE and these four scrapie types, of which CS-2 is new, can be recognized in goats with combinations of a set of nine biochemical parameters.


Subject(s)
Blotting, Western/veterinary , Enzyme-Linked Immunosorbent Assay/veterinary , Goat Diseases/classification , Scrapie/classification , Animals , Blotting, Western/methods , Enzyme-Linked Immunosorbent Assay/methods , Europe , Goat Diseases/diagnosis , Goats , Scrapie/diagnosis
6.
BMC Vet Res ; 15(1): 399, 2019 Nov 06.
Article in English | MEDLINE | ID: mdl-31694666

ABSTRACT

BACKGROUND: TBE is an important tick-borne viral zoonosis in Europe and some parts of Asia. Humans can become infected by tick bite and in some cases also by consumption of nonpasteurized raw milk and raw milk products from ruminants. Serological investigations of milking flocks can help to assess the risk of TBEV infection for humans. 735 blood samples from 50 goat flocks from four federal states of Germany were tested by TBEV-VNT to assess a potential risk for TBEV infection. There are some gaps in the knowledge about immunity in animals, for example with regard to the longevity of TBEV immunity. Two goats and two sheep were immunized and TBEV antibody titers could be detected for up to 7 years. Furthermore, nothing is known about a possible long-lasting immunological memory that could quickly be reactivated by an additional contact to TBEV. Seven years after the first immunization two goats and two sheep as well as two naïve goats and two sheep were boostered and TBEV antibody titers followed. RESULTS: Only one sample in each of the three states was TBEV-antibody positive (VNT), albeit with low titers. However, in Baden-Württemberg seven samples were positive, among them four goats of the same flock. TBEV-antibody positive titers were detected in goats for up to 6 years and 10 months, in sheep for up to 4 years and 7 months. Seven years after immunization a clear immunological recall occurred in response to administration of one dose of vaccine in two goats and two sheep. CONCLUSION: It can be concluded that in the tested flocks the risk of an alimentary TBEV infection was low. However, in one single flock a considerably higher risk must be assumed. Antibody titers in goats and sheep can last very long after contact to TBEV, albeit at a low level. This should be taken into consideration in cases where the risk of an alimentary infection is assessed in a flock by serological investigations. The immunological recall gives rise to the suspicion that the immunological memory after a first contact to TBEV lasts for many years, probably lifelong.


Subject(s)
Antibodies, Viral/blood , Encephalitis Viruses, Tick-Borne/immunology , Encephalitis, Tick-Borne/veterinary , Goat Diseases/virology , Sheep Diseases/virology , Animals , Encephalitis Viruses, Tick-Borne/isolation & purification , Encephalitis, Tick-Borne/immunology , Female , Germany/epidemiology , Goat Diseases/immunology , Goats , Sheep , Sheep Diseases/immunology , Vaccines/pharmacology
7.
Emerg Infect Dis ; 24(12): 2334-2337, 2018 12.
Article in English | MEDLINE | ID: mdl-30457523

ABSTRACT

Eradication of small ruminant morbillivirus (PPRV) is targeted for 2030. PPRV lineage IV is found in much of Asia and Africa. We used PPRV lineage IV strain Kurdistan/2011 in transmission trials to investigate the role of pigs, wild boar, and small ruminants as PPRV reservoirs. Suids were a possible source of infection.


Subject(s)
Animal Diseases/virology , Host Specificity , Morbillivirus/physiology , Ruminants/virology , Viral Tropism , Animal Diseases/diagnosis , Animal Diseases/immunology , Animals , Antibodies, Neutralizing/immunology , Antibodies, Viral/immunology , Goats , Morbillivirus/isolation & purification , Neutralization Tests , Swine
8.
Vet Res ; 49(1): 122, 2018 Dec 20.
Article in English | MEDLINE | ID: mdl-30572960

ABSTRACT

BSE infectivity in mesentery fat is most likely associated with embedded nervous tissue. To prove this mesentery containing celiac ganglion was taken from oral BSE infected cattle in different stages of the disease and from one control animal. Fat was rendered according to standard tallow production methods and the prion infectivity therein analysed in transgenic mouse bioassay. Rendered fat of the clinical animal revealed low infectivity levels, whereas preclinical and control animals remained negative. This study, although not representative, provides a proof of principle, indicating the potential contamination of melted mesenteric fat by embedded nervous structures during standard tallow production.


Subject(s)
Abdominal Fat/physiopathology , Encephalopathy, Bovine Spongiform/transmission , Ganglia, Sympathetic/physiopathology , Animals , Cattle , Mesentery , Mice , Mice, Transgenic
9.
BMC Vet Res ; 14(1): 381, 2018 Dec 04.
Article in English | MEDLINE | ID: mdl-30514313

ABSTRACT

BACKGROUND: Hepatitis E virus (HEV) is one major cause of acute clinical hepatitis among humans throughout the world. In industrialized countries an increasing number of autochthonous HEV infections have been identified over the last years triggered by food borne as well as - to a much lower degree - by human to human transmission via blood transfusion. Pigs have been recognised as main reservoir for HEV genotype 3 (HEV-3), and zoonotic transmission to humans through undercooked/raw meat is reported repeatedly. The minimal infectious dose of HEV-3 for pigs is so far unknown. RESULTS: The minimum infectious dose of HEV-3 in a pig infection model was determined by intravenous inoculation of pigs with a dilution series of a liver homogenate of a HEV infected wild boar. Seroconversion, virus replication and shedding were determined by analysis of blood and faeces samples, collected over a maximum period of 91 days. A dose dependent incubation period was observed in faecal shedding of viruses employing a specific and sensitive PCR method. Faecal viral shedding and seroconversion was detected in animals inoculated with dilutions of up to 10- 7. This correlates with an intravenously (i.v.) administered infectious dose of only 6.5 copies in 2 ml (corresponding to 24 IU HEV RNA/ml). Furthermore the first detectable shedding of HEV RNA in faeces is clearly dose dependent. Unexpectedly one group infected with a 10- 4 dilution exhibited prolonged virus shedding for more than 60 days suggesting a persistent infection. CONCLUSION: The results indicate that pigs are highly susceptible to i.v. infection with HEV and that the swine model represents the most sensitive infectivity assay for HEV so far. Considering a minimum infectious dose of 24 IU RNA/ml our findings highlights the potential risk of HEV transmission via blood and blood products.


Subject(s)
Hepatitis E virus/physiology , Hepatitis E/transmission , Hepatitis E/virology , Sus scrofa , Administration, Intravenous , Animals , Feces/virology , Hepatitis E/blood , Virus Replication , Virus Shedding
10.
Emerg Infect Dis ; 23(3): 477-481, 2017 03.
Article in English | MEDLINE | ID: mdl-28221112

ABSTRACT

We screened squirrels in Germany and the Netherlands for the novel zoonotic variegated squirrel bornavirus 1 (VSBV-1). The detection of VSBV-1 in 11 squirrels indicates a considerable risk for transmission to humans handling those animals. Therefore, squirrels in contact with humans should routinely be tested for VSBV-1.


Subject(s)
Bornaviridae/classification , Bornaviridae/isolation & purification , Mononegavirales Infections/veterinary , Sciuridae/virology , Animals , Germany/epidemiology , Humans , Mononegavirales Infections/epidemiology , Mononegavirales Infections/virology , Netherlands/epidemiology , Risk Factors , Zoonoses
11.
Arch Virol ; 162(9): 2747-2754, 2017 Sep.
Article in English | MEDLINE | ID: mdl-28593419

ABSTRACT

The recently discovered variegated squirrel bornavirus 1 (VSBV-1) caused the death of three squirrel breeders in Germany. Subsequent first screening of squirrels with in vivo collected swab samples and a VSBV-1-specific RT-qPCR revealed not only variegated squirrel infections (Sciurus variegatoides), but also Prevost's squirrels (Callosciurus prevostii) as positive for VSBV-1 genome. In this study, 328 squirrels were tested using the established RT-qPCR assays. In 16 individual animals VSBV-1 RNA could be detected; 15 individuals were from small breedings and zoological gardens in Germany, with the remaining individual being from a zoological garden in Croatia. Positive animals belonged to the species C. prevostii, C. finlaysonii, and Tamiops swinhoei within the subfamily Callosciurinae and Sciurus granatensis within the subfamily Sciurinae. Repeated non-invasive oral swab sampling in one holding indicated positive animals months after a first negative result. Besides the oral swabs, VSBV-1 was also detected in fecal (pool) samples allowing the future monitoring of squirrel holdings based on RT-qPCR investigation of such samples. The detection in zoological gardens emphasizes the need for further investigations into the transmission route to humans in order to develop rational public health measures for prevention of transmission. Finally, the detection of several closely related VSBV-1 sequences in squirrels from different subfamilies raises questions as to the origin of the virus.


Subject(s)
Bornaviridae/classification , RNA, Viral/isolation & purification , Sciuridae/virology , Animals , Bornaviridae/genetics , Feces/virology , Phylogeny , RNA, Viral/genetics , Species Specificity , Zoonoses
12.
Vet Res ; 47(1): 99, 2016 10 06.
Article in English | MEDLINE | ID: mdl-27716411

ABSTRACT

Scrapie and bovine spongiform encephalopathy (BSE) are transmissible spongiform encephalopathies (TSE's) affecting sheep and goats. Susceptibility of goats to scrapie is influenced by polymorphisms of the prion protein gene (PRNP) of the host. Five polymorphisms are associated with reduced susceptibility to TSE's. In the study presented here caprine samples from a scrapie eradication program on Cyprus were genotyped and further characterized using BioRad TeSeE rapid test, histological, immunohistochemical and biochemical methods. In total 42 goats from 20 flocks were necropsied from which 25 goats showed a positive result in the rapid test, a spongiform encephalopathy and an accumulation of pathological prion protein (PrPSc) in the obex. PrPSc deposits were demonstrated in the placenta, peripheral nervous and lymphoreticular system. Two animals showed PrPSc-accumulations in peripheral tissues only. By discriminatory immunoblots a scrapie infection could be confirmed for all cases. Nevertheless, slight deviations in the glycosylation pattern might indicate the presence of different scrapie strains. Furthermore scrapie samples from goats in the current study demonstrated less long term resistance to proteinase K than ovine or caprine BSE control samples. Reduced scrapie susceptibility according to the PRNP genotype was demonstrated (Fishers Exact test, p < 0.05) for the goats with at least one polymorphism (p = 0.023) at the six codons examined and in particular for those with polymorphisms at codon 146 (p = 0.016). This work characterizes scrapie in goats having implications for breeding and surveillance strategies.


Subject(s)
Goat Diseases/genetics , Prion Diseases/veterinary , Animals , Cyprus/epidemiology , Female , Goat Diseases/epidemiology , Goat Diseases/pathology , Goats/genetics , Prion Diseases/epidemiology , Prion Diseases/genetics , Prion Diseases/pathology , Prion Proteins/metabolism
13.
J Infect Dis ; 212(4): 664-72, 2015 Aug 15.
Article in English | MEDLINE | ID: mdl-25722297

ABSTRACT

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.


Subject(s)
Encephalopathy, Bovine Spongiform/transmission , Goat Diseases/pathology , Goat Diseases/transmission , Prions/genetics , Animals , Cattle , Genetic Predisposition to Disease , Genotype , Goats , Mice , Mice, Transgenic , Species Specificity
14.
Vet Res ; 46: 87, 2015 Aug 17.
Article in English | MEDLINE | ID: mdl-26282836

ABSTRACT

As West Nile virus (WNV) can cause lethal diseases in raptors, a vaccination prophylaxis of free-living and captive populations is desirable. In the absence of vaccines approved for birds, equine vaccines have been used in falcons, but full protection against WNV infection was not achieved. Therefore, two DNA vaccines encoding the ectodomain of the envelope protein of WNV lineages 1 and 2, respectively, were evaluated in 28 large falcons. Four different vaccination protocols were used, including electroporation and booster-injections of recombinant WNV domain III protein, before challenge with the live WNV lineage 1 strain NY99. Drug safety, plasmid shedding and antibody production were monitored during the vaccination period. Serological, virological, histological, immunohistochemical and molecular biological investigations were performed during the challenge trials. Antibody response following vaccination was low overall and lasted for a maximum of three weeks. Plasmid shedding was not detected at any time. Viremia, mortality and levels, but not duration, of oral virus shedding were reduced in all of the groups during the challenge trial compared to the non-vaccinated control group. Likewise, clinical scoring, levels of cloacal virus shedding and viral load in organs were significantly reduced in three vaccination groups. Histopathological findings associated with WNV infections (meningo-encephalitis, myocarditis, and arteritis) were present in all groups, but immunohistochemical detection of the viral antigen was reduced. In conclusion, the vaccines can be used safely in falcons to reduce mortality and clinical signs and to lower the risk of virus transmission due to decreased levels of virus shedding and viremia, but full protection was not achieved in all groups.


Subject(s)
Bird Diseases/prevention & control , Falconiformes , Vaccines, DNA/pharmacology , Viral Envelope Proteins/genetics , West Nile Fever/veterinary , West Nile Virus Vaccines/pharmacology , West Nile virus/immunology , Animals , Antibodies, Viral/blood , Antibodies, Viral/metabolism , Bird Diseases/virology , Electroporation/veterinary , Injections, Intramuscular/veterinary , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Viral Envelope Proteins/metabolism , Viremia/prevention & control , Viremia/veterinary , Viremia/virology , Virus Shedding , West Nile Fever/prevention & control , West Nile Fever/virology
15.
Vet Res ; 45: 121, 2014 Nov 26.
Article in English | MEDLINE | ID: mdl-25421429

ABSTRACT

Hepatitis E virus (HEV) is the causative agent of acute hepatitis E in humans in developing countries, but sporadic and autochthonous cases do also occur in industrialised countries. In Europe, food-borne zoonotic transmission of genotype 3 (gt3) has been associated with domestic pig and wild boar. However, little is known about the course of HEV infection in European wild boar and their role in HEV transmission to domestic pigs. To investigate the transmissibility and pathogenesis of wild boar-derived HEVgt3, we inoculated four wild boar and four miniature pigs intravenously. Using quantitative real-time RT-PCR viral RNA was detected in serum, faeces and in liver, spleen and lymph nodes. The antibody response evolved after fourteen days post inoculation. Histopathological findings included mild to moderate lymphoplasmacytic hepatitis which was more prominent in wild boar than in miniature pigs. By immunohistochemical methods, viral antigens were detected mainly in Kupffer cells and liver sinusoidal endothelial cells, partially associated with hepatic lesions, but also in spleen and lymph nodes. While clinical symptoms were subtle and gross pathology was inconspicuous, increased liver enzyme levels in serum indicated hepatocellular injury. As the faecal-oral route is supposed to be the most likely transmission route, we included four contact animals to prove horizontal transmission. Interestingly, HEVgt3-infection was also detected in wild boar and miniature pigs kept in contact to intravenously inoculated wild boar. Given the high virus loads and long duration of viral shedding, wild boar has to be considered as an important HEV reservoir and transmission host in Europe.


Subject(s)
Hepatitis E virus/physiology , Hepatitis E/veterinary , Swine Diseases/transmission , Animals , Antibodies, Viral/blood , Antibody Formation , Genotype , Hepatitis E/transmission , Hepatitis E/virology , Hepatitis E virus/genetics , RNA, Viral/blood , RNA, Viral/metabolism , Real-Time Polymerase Chain Reaction/veterinary , Reverse Transcriptase Polymerase Chain Reaction/veterinary , Sus scrofa , Swine , Swine Diseases/virology , Swine, Miniature
16.
Vet Res ; 45: 41, 2014 Apr 07.
Article in English | MEDLINE | ID: mdl-24708385

ABSTRACT

West Nile virus (WNV) can lead to fatal diseases in raptor species. Unfortunately, there is no vaccine which has been designed specifically for use in breeding stocks of falcons. Therefore the immunogenicity and protective capacity of two commercially available WNV vaccines, both approved for use in horses, were evaluated in large falcons. One vaccine contained adjuvanted inactivated WNV lineage 1 immunogens, while the second represented a canarypox recombinant live virus vector vaccine. The efficacy of different vaccination regimes for these two vaccines was assessed serologically and by challenging the falcons with a WNV strain of homologous lineage 1. Our studies show that the recombinant vaccine conveys a slightly better protection than the inactivated vaccine, but moderate (recombinant vaccine) or weak (inactivated vaccine) side effects were observed at the injection sites. Using the recommended 2-dose regimen, both vaccines elicited only sub-optimal antibody responses and gave only partial protection following WNV challenge. Better results were obtained for both vaccines after a third dose, i.e. alleviation of clinical signs, absence of fatalities and reduction of virus shedding and viraemia. Therefore the consequences of WNV infections in falcons can be clearly alleviated by vaccination, especially if the amended triple administration scheme is used, although side effects at the vaccination site must be accepted.


Subject(s)
Bird Diseases/prevention & control , Falconiformes , West Nile Fever/veterinary , West Nile Virus Vaccines/adverse effects , West Nile virus/immunology , Animals , Bird Diseases/immunology , Immunohistochemistry/veterinary , Polymerase Chain Reaction/veterinary , Vaccines, Inactivated/immunology , Viremia/veterinary , Virus Shedding , West Nile Fever/immunology , West Nile Fever/prevention & control , West Nile Virus Vaccines/immunology
17.
Am J Pathol ; 181(2): 515-24, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22781833

ABSTRACT

An experimental oral bovine spongiform encephalopathy (BSE) challenge study was performed to elucidate the route of infectious prions from the gut to the central nervous system in preclinical and clinical infected animals. Tissue samples collected from the gut and the central and autonomic nervous system from animals sacrificed between 16 and 44 months post infection (mpi) were examined for the presence of the pathological prion protein (PrP(Sc)) by IHC. Moreover, parts of these samples were also bioassayed using bovine cellular prion protein (PrP(C)) overexpressing transgenic mice (Tgbov XV) that lack the species barrier for bovine prions. A distinct accumulation of PrP(Sc) was observed in the distal ileum, confined to follicles and/or the enteric nervous system, in almost all animals. BSE prions were found in the sympathetic nervous system starting at 16 mpi, and in the parasympathetic nervous system from 20 mpi. A clear dissociation between prion infectivity and detectable PrP(Sc) deposition became obvious. The earliest presence of infectivity in the brain stem was detected at 24 mpi, whereas PrP(Sc) accumulation was first detected after 28 mpi. In summary, our results decipher the centripetal spread of BSE prions along the autonomic nervous system to the central nervous system, starting already halfway in the incubation time.


Subject(s)
Brain/pathology , Encephalopathy, Bovine Spongiform/metabolism , Encephalopathy, Bovine Spongiform/pathology , Gastrointestinal Tract/pathology , Peripheral Nervous System/pathology , Prions/metabolism , Animals , Brain/metabolism , Cattle , Gastrointestinal Tract/metabolism , Ileum/metabolism , Ileum/pathology , Mice , Mice, Transgenic , Models, Biological , Peripheral Nervous System/metabolism , PrPSc Proteins/metabolism
18.
Vet Res ; 44: 123, 2013 Dec 21.
Article in English | MEDLINE | ID: mdl-24359408

ABSTRACT

Recently we have described the distribution of bovine spongiform encephalopathy (BSE) infectivity and/or PrPSc in Peyer's patches (PP) of the small intestine of orally BSE infected cattle. In this follow-up study additional jejunal and ileal PP's and ileocaecal-junction tissue samples from 1, 4, and 24 months post infection (mpi) were examined by mouse (Tgbov XV) bioassay. Infectivity was demonstrated in ileal PP's 4 mpi and the distribution/extent of infectivity at 24 mpi was comparable to those seen at earlier time points, revealing no indication for a decline/clearance. These data are relevant for the definition of Specified Risk Materials in the context of the TSE legislation worldwide.


Subject(s)
Cecum/pathology , Encephalopathy, Bovine Spongiform/pathology , Ileum/pathology , Jejunum/pathology , Peyer's Patches/pathology , PrPSc Proteins/analysis , Animals , Cattle , Encephalopathy, Bovine Spongiform/diagnosis , Encephalopathy, Bovine Spongiform/transmission , Female , Follow-Up Studies , Mice, Transgenic , Risk Assessment
19.
Pathogens ; 12(7)2023 Jul 20.
Article in English | MEDLINE | ID: mdl-37513806

ABSTRACT

Usutu virus (USUV) and West Nile virus (WNV) are closely related pathogens circulating between mosquitoes and birds, but also infecting mammals as dead-end hosts. Both viruses share the same susceptible hosts, vectors, and even distribution areas in Central Europe. The aim of the study was, therefore, to understand their amplification potential and interference upon a successive infection. Two-week old geese were initially infected with an USUV isolate from Germany and with a German WNV isolate17 days later. The geese were susceptible to the USUV and the WNV infections, as evidenced by specific flavivirus antibodies in all of the birds. Furthermore, in half of the USUV-inoculated geese, USUV genomes were detected in the blood and swab samples 2-4 days post-infection. Additionally, most of the examined organs contained USUV genomes and showed signs of encephalitis and ganglioneuritis. Interestingly, upon a sequential infection with WNV, the genome copy numbers in all of the examined samples were significantly lower and less frequent than after a WNV mono-infection. Similarly, the histopathological lesions were less severe. Therefore, it can be concluded that a previous USUV infection can protect birds from clinical disease in a subsequent WNV infection.

20.
Pathogens ; 12(2)2023 Feb 20.
Article in English | MEDLINE | ID: mdl-36839625

ABSTRACT

Bovine spongiform encephalopathy (BSE) belongs to the group of transmissible spongiform encephalopathies and is associated with the accumulation of a pathological isoform of the host-encoded glycoprotein, designated prion protein (PrPSc). Classical BSE (C-type) and two atypical BSE forms (L- and H-type) are known, and can be discriminated by biochemical characteristics. The goal of our study was to identify type-specific PrPSc profiles by using Immunohistochemistry. In our study, brain samples from 21 cattle, intracerebrally inoculated with C-, H-, and L-type BSE, were used. In addition, the corresponding samples from three orally C-type BSE infected animals were also included. From all animals, a lesion and PrPSc-profiles of six brain regions were determined. The lesion profile and the neuroanatomical distribution of PrPSc was highly consistent between the groups, but the immunohistochemical analysis revealed a distinct PrPSc profile for the different BSE-types, which included both the topographic and cellular pattern of PrPSc. This qualitative and quantitative analysis of PrPSc affected structures sheds new light into the pathogenesis of the different BSE types. Furthermore, immunohistochemical characterization is supported as an additional diagnostic tool in BSE surveillance programs, especially when only formalin-fixed tissue samples are available.

SELECTION OF CITATIONS
SEARCH DETAIL