Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 66
Filtrar
Mais filtros








Base de dados
Intervalo de ano de publicação
1.
Emerg Microbes Infect ; 13(1): 2348526, 2024 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-38683015

RESUMO

The foot-and-mouth disease virus (FMDV) Leader proteinase Lpro inhibits host mRNA translation and blocks the interferon response which promotes viral survival. Lpro is not required for viral replication in vitro but serotype A FMDV lacking Lpro has been shown to be attenuated in cattle and pigs. However, it is not known, whether leaderless viruses can cause persistent infection in vivo after simulated natural infection and whether the attenuated phenotype is the same in other serotypes. We have generated an FMDV O/FRA/1/2001 variant lacking most of the Lpro coding region (ΔLb). Cattle were inoculated intranasopharyngeally and observed for 35 days to determine if O FRA/1/2001 ΔLb is attenuated during the acute phase of infection and whether it can maintain a persistent infection in the upper respiratory tract. We found that although this leaderless virus can replicate in vitro in different cell lines, it is unable to establish an acute infection with vesicular lesions and viral shedding nor is it able to persistently infect bovine pharyngeal tissues.


Assuntos
Doenças dos Bovinos , Vírus da Febre Aftosa , Febre Aftosa , Infecção Persistente , Sorogrupo , Replicação Viral , Animais , Bovinos , Vírus da Febre Aftosa/genética , Vírus da Febre Aftosa/fisiologia , Vírus da Febre Aftosa/classificação , Vírus da Febre Aftosa/patogenicidade , Vírus da Febre Aftosa/isolamento & purificação , Febre Aftosa/virologia , Doenças dos Bovinos/virologia , Infecção Persistente/virologia , Linhagem Celular , Endopeptidases/genética , Endopeptidases/metabolismo , Eliminação de Partículas Virais
2.
Virus Evol ; 9(2): vead062, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-38028148

RESUMO

Recently, we discovered two novel orthobornaviruses in colubrid and viperid snakes using an in silico data-mining approach. Here, we present the results of a screening of more than 100,000 nucleic acid sequence datasets of fish samples from the Sequence Read Archive (SRA) for potential bornaviral sequences. We discovered the potentially complete genomes of seven bornavirids in datasets from osteichthyans and chondrichthyans. Four of these are likely to represent novel species within the genus Cultervirus, and we propose that one genome represents a novel genus within the family of Bornaviridae. Specifically, we identified sequences of Wǔhàn sharpbelly bornavirus in sequence data from the widely used grass carp liver and kidney cell lines L8824 and CIK, respectively. A complete genome of Murray-Darling carp bornavirus was identified in sequence data from a goldfish (Carassius auratus). The newly discovered little skate bornavirus, identified in the little skate (Leucoraja erinacea) dataset, contained a novel and unusual genomic architecture (N-Vp1-Vp2-X-P-G-M-L), as compared to other bornavirids. Its genome is thought to encode two additional open reading frames (tentatively named Vp1 and Vp2), which appear to represent ancient duplications of the gene encoding the viral glycoprotein (G). The datasets also provided insights into the possible transcriptional gradients of these bornavirids and revealed previously unknown splicing mechanisms.

3.
Virus Evol ; 9(2): vead048, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37744713

RESUMO

Rustrela virus (RusV; species Rubivirus strelense, family Matonaviridae) was discovered in different zoo animal species affected by fatal encephalitis. Simultaneous RusV RNA detection in multiple yellow-necked field mice (Apodemus flavicollis) suggested this rodent as a reservoir of RusV. Here, we investigated 1,264 yellow-necked field mice and sympatric other small mammals from different regions in Germany for RusV RNA using an optimized reverse transcription-quantitative polymerase chain reaction (RT-qPCR) protocol and high-throughput sequencing. The investigation resulted in the detection of RusV RNA exclusively in 50 of 396 (12.6 per cent) yellow-necked field mice but absence in other sympatric species. RT-qPCR-determined tissue distribution of RusV RNA revealed the highest viral loads in the central nervous system, with other tissues being only very rarely affected. The histopathological evaluation did not reveal any hints of encephalitis in the brains of infected animals despite the detection of viral RNA in neurons by in situ hybridization (ISH). The positive association between the body mass of yellow-necked field mice and RusV RNA detection suggests a persistent infection. Phylogenetic analysis of partial E1 and full-genome sequences showed a high diversification with at least four RusV lineages (1A-1D) in northeastern Germany. Moreover, phylogenetic and isolation-by-distance analyses indicated evolutionary processes of RusV mostly in local reservoir populations. A comparison of complete genome sequences from all detected RusV lineages demonstrated a high level of amino acid and nucleotide sequence variability within a part of the p150 peptide of the non-structural polyprotein and its coding sequence, respectively. The location of this region within the RusV genome and its genetic properties were comparable to the hypervariable region of the rubella virus. The broad range of detected RusV spillover hosts in combination with its geographical distribution in northeastern Germany requires the assessment of its zoonotic potential and further analysis of encephalitis cases in mammals. Future studies have to prove a putative co-evolution scenario for RusV in the yellow-necked field mouse reservoir.

4.
Eur J Immunol ; 53(12): e2250332, 2023 12.
Artigo em Inglês | MEDLINE | ID: mdl-37609807

RESUMO

Knowledge about early immunity to SARS-CoV-2 variants of concern mainly comes from the analysis of human blood. Such data provide limited information about host responses at the site of infection and largely miss the initial events. To gain insights into compartmentalization and the early dynamics of host responses to different SARS-CoV-2 variants, we utilized human angiotensin converting enzyme 2 (hACE2) transgenic mice and tracked immune changes during the first days after infection by RNAseq, multiplex assays, and flow cytometry. Viral challenge infection led to divergent viral loads in the lungs, distinct inflammatory patterns, and innate immune cell accumulation in response to ancestral SARS-CoV-2, Beta (B.1.351) and Delta (B.1.617.2) variant of concern (VOC). Compared to other SARS-CoV-2 variants, infection with Beta (B.1.351) VOC spread promptly to the lungs, leading to increased inflammatory responses. SARS-CoV-2-specific antibodies and T cells developed within the first 7 days postinfection and were required to reduce viral spread and replication. Our studies show that VOCs differentially trigger transcriptional profiles and inflammation. This information contributes to the basic understanding of immune responses immediately postexposure to SARS-CoV-2 and is relevant for developing pan-VOC interventions including prophylactic vaccines.


Assuntos
COVID-19 , SARS-CoV-2 , Animais , Camundongos , Humanos , Enzima de Conversão de Angiotensina 2/genética , COVID-19/genética , Anticorpos Antivirais , Camundongos Transgênicos , Imunidade
5.
Arch Virol ; 168(9): 234, 2023 Aug 22.
Artigo em Inglês | MEDLINE | ID: mdl-37608200

RESUMO

A novel ephemerovirus was identified in a Holstein-Friesian cow in the Hefer Valley, Israel, that showed severe and fatal clinical signs resembling an arboviral infection. A sample taken during the acute phase tested negative for important endemic arboviral infectious cattle diseases. However, sequencing from blood revealed the full genome sequence of Hefer Valley virus, which is likely to represent a new species within the genus Ephemerovirus, family Rhabdoviridae. Archived samples from cattle with comparable clinical signs collected in Israel in 2021 and 2022 tested negative for the novel virus, and therefore, the actual distribution of the virus is unknown. As this is a recently identified new viral infection, the viral vector and the prevalence of the virus in the cattle population are still unknown but will be the subject of future investigations.


Assuntos
Ephemerovirus , Feminino , Bovinos , Animais , Israel/epidemiologia , Meio Ambiente
6.
Emerg Infect Dis ; 29(6): 1202-1205, 2023 06.
Artigo em Inglês | MEDLINE | ID: mdl-37209672

RESUMO

We detected a novel poxvirus from a gray seal (Halichoerus grypus) from the North Sea, Germany. The juvenile animal showed pox-like lesions and deteriorating overall health condition and was finally euthanized. Histology, electron microscopy, sequencing, and PCR confirmed a previously undescribed poxvirus of the Chordopoxvirinae subfamily, tentatively named Wadden Sea poxvirus.


Assuntos
Chordopoxvirinae , Poxviridae , Focas Verdadeiras , Animais , Poxviridae/genética , Mar do Norte , Alemanha/epidemiologia
7.
Emerg Infect Dis ; 29(5): 1042-1045, 2023 05.
Artigo em Inglês | MEDLINE | ID: mdl-37081716

RESUMO

Retrospective investigation of archived tissue samples from 3 lions displaying nonsuppurative meningoencephalitis and vasculitis led to the detection of rustrela virus (RusV). We confirmed RusV antigen and RNA in cortical neurons, axons, astrocytes and Purkinje cells by reverse transcription quantitative PCR, immunohistochemistry, and in situ hybridization.


Assuntos
Leões , Meningoencefalite , Vírus , Animais , Estudos Retrospectivos , Meningoencefalite/diagnóstico , Meningoencefalite/veterinária , Imuno-Histoquímica
8.
Nat Commun ; 14(1): 624, 2023 02 04.
Artigo em Inglês | MEDLINE | ID: mdl-36739288

RESUMO

'Staggering disease' is a neurological disease entity considered a threat to European domestic cats (Felis catus) for almost five decades. However, its aetiology has remained obscure. Rustrela virus (RusV), a relative of rubella virus, has recently been shown to be associated with encephalitis in a broad range of mammalian hosts. Here, we report the detection of RusV RNA and antigen by metagenomic sequencing, RT-qPCR, in-situ hybridization and immunohistochemistry in brain tissues of 27 out of 29 cats with non-suppurative meningoencephalomyelitis and clinical signs compatible with'staggering disease' from Sweden, Austria, and Germany, but not in non-affected control cats. Screening of possible reservoir hosts in Sweden revealed RusV infection in wood mice (Apodemus sylvaticus). Our work indicates that RusV is the long-sought cause of feline 'staggering disease'. Given its reported broad host spectrum and considerable geographic range, RusV may be the aetiological agent of neuropathologies in further mammals, possibly even including humans.


Assuntos
Encefalomielite , Humanos , Animais , Gatos , Camundongos , Causalidade , Suécia , Áustria , Alemanha , Mamíferos
9.
Microbiol Spectr ; : e0266422, 2023 Jan 26.
Artigo em Inglês | MEDLINE | ID: mdl-36700688

RESUMO

Surveillance of avian influenza viruses (AIV) in wild water bird populations is important for early warning to protect poultry from incursions of high-pathogenicity (HP) AIV. Access to individual water birds is difficult and restricted and limits sampling depth. Here, we focused on environmental samples such as surface water, sediments, and environmentally deposited fresh avian feces as matrices for AIV detection. Enrichment of viral particles by ultrafiltration of 10-L surface water samples using Rexeed-25-A devices was validated using a bacteriophage ϕ6 internal control system, and AIV detection was attempted using real-time RT-PCR and virus isolation. While validation runs suggested an average enrichment of about 60-fold, lower values of 10 to 15 were observed for field water samples. In total 25/36 (60%) of water samples and 18/36 (50%) of corresponding sediment samples tested AIV positive. Samples were obtained from shallow water bodies in habitats with large numbers of waterfowl during an HPAIV epizootic. Although AIV RNA was detected in a substantial percentage of samples virus isolation failed. Virus loads in samples often were too low to allow further sub- and pathotyping. Similar results were obtained with environmentally deposited avian feces. Moreover, the spectrum of viruses detected by these active surveillance methods did not fully mirror an ongoing HPAIV epizootic among waterfowl as detected by passive surveillance, which, in terms of sensitivity, remains unsurpassed. IMPORTANCE Avian influenza viruses (AIV) have a wide host range in the avian metapopulation and, occasionally, transmission to humans also occurs. Surface water plays a particularly important role in the epidemiology of AIV, as the natural virus reservoir is found in aquatic wild birds. Environmental matrices comprising surface water, sediments, and avian fecal matter deposited in the environment were examined for their usefulness in AIV surveillance. Despite virus enrichment efforts, environmental samples regularly revealed very low virus loads, which hampered further sub- and pathotyping. Passive surveillance based on oral and cloacal swabs of diseased and dead wild birds remained unsurpassed with respect to sensitivity.

10.
Front Vet Sci ; 10: 1339248, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-38239751

RESUMO

Bovine viral diarrhea virus (BVDV) is one of the most important cattle pathogens worldwide, causing major economic losses and animal welfare issues. Disease eradication programs have been implemented in several countries, including Germany where an obligatory nationwide control program is in force since 2011. As molecular epidemiology has become an essential tool to understand the transmission dynamics and evolution of BVDV, 5' untranslated region (UTR) sequences are generated from viruses present in persistently infected animals since the beginning of the BVDV control program. Here, we report the results of the sequence-based subtyping of BVDV strains found from 2018 through 2022 in calves born in Germany. In 2018, 2019 and 2020, BVDV-1d and-1b were the dominant subtypes and cases were spread throughout the area that was not yet officially declared BVDV-free at that time. In addition, BVDV-1a, -1e, -1f and -1h could rarely be detected. From 2021 onwards, subtype 1d clearly took over the dominance, while the other subtypes could be gradually nearly eliminated from the cattle population. The eradication success not only results in a drastic reduction of cases, but also in a marked reduction of strain diversity. Interestingly, before vaccination has been banned in regions and farms with a disease-free status, two live-vaccine virus strains were repeatedly detected in ear tissue samples of newborn calves (n = 14) whose mothers were immunized during gestation. The field-virus sequences are an important basis for molecular tracing and identification of potential relationships between the last outbreaks in the final phase of the German BVDV eradication program, thereby supporting classic epidemiological investigations. Furthermore, the monitoring of the composition of virus subtypes in the cattle population helps to maintain effective diagnostic methods and control measures and is an early warning system for the introduction of new pestiviruses in the naïve cattle population.

11.
Transbound Emerg Dis ; 69(6): 4016-4021, 2022 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-36135593

RESUMO

The rustrela virus (RusV) was recently described as a novel pathogen in a circumscribed area of northern Germany close to the Baltic Sea. Up to now, the virus has been detected in cases of fatal non-suppurative meningoencephalitis in zoo animals of different species and a single wild carnivore as well as in apparently healthy yellow-necked field mice (Apodemus flavicollis). Data regarding the background of this previously undiscovered pathogen, including clinical presentation of the disease, host range and distribution of the virus, are still limited. Here, three euthanized red-necked wallabies (Macropus rufogriseus) from zoos of different areas in northeastern Germany were submitted for necropsy after presenting with apathy and therapeutically unresponsive neurological signs. A moderate to severe, non-suppurative meningoencephalitis was diagnosed in all three cases. RusV was consistently detected via RT-qPCR and RNA in situ hybridization in the brains of all wallabies. Other commonly known neuropathogens could not be detected.


Assuntos
Doenças dos Roedores , Viroses , Camundongos , Animais , Macropodidae , Animais de Zoológico , Viroses/veterinária , Alemanha
12.
J Virol ; 96(17): e0099422, 2022 09 14.
Artigo em Inglês | MEDLINE | ID: mdl-35993736

RESUMO

Wild birds are the reservoir for all avian influenza viruses (AIV). In poultry, the transition from low pathogenic (LP) AIV of H5 and H7 subtypes to highly pathogenic (HP) AIV is accompanied mainly by changing the hemagglutinin (HA) monobasic cleavage site (CS) to a polybasic motif (pCS). Galliformes, including turkeys and chickens, succumb with high morbidity and mortality to HPAIV infections, although turkeys appear more vulnerable than chickens. Surprisingly, the genetic determinants for virulence and pathogenesis of HPAIV in turkeys are largely unknown. Here, we determined the genetic markers for virulence and transmission of HPAIV H7N1 in turkeys, and we explored the host responses in this species compared to those of chickens. We found that recombinant LPAIV H7N1 carrying pCS was avirulent in chickens but exhibited high virulence in turkeys, indicating that virulence determinants vary in these two galliform species. A transcriptome analysis indicated that turkeys mount a different host response than do chickens, particularly from genes involved in RNA metabolism and the immune response. Furthermore, we found that the HA glycosylation at residue 123, acquired by LP viruses shortly after transmission from wild birds and preceding the transition from LP to HP, had a role in virus fitness and virulence in chickens, though it was not a prerequisite for high virulence in turkeys. Together, these findings indicate variable virulence determinants and host responses in two closely related galliformes, turkeys and chickens, after infection with HPAIV H7N1. These results could explain the higher vulnerability to HPAIV of turkeys compared to chickens. IMPORTANCE Infection with HPAIV in chickens and turkeys, two closely related galliform species, results in severe disease and death. Although the presence of a polybasic cleavage site (pCS) in the hemagglutinin of AIV is a major virulence determinant for the transition of LPAIV to HPAIV, there are knowledge gaps on the genetic determinants (including pCS) and the host responses in turkeys compared to chickens. Here, we found that the pCS alone was sufficient for the transformation of a LP H7N1 into a HPAIV in turkeys but not in chickens. We also noticed that turkeys exhibited a different host response to an HPAIV infection, namely, a widespread downregulation of host gene expression associated with protein synthesis and the immune response. These results are important for a better understanding of the evolution of HPAIV from LPAIV and of the different outcomes and the pathomechanisms of HPAIV infections in chickens and turkeys.


Assuntos
Galinhas , Vírus da Influenza A Subtipo H7N1 , Influenza Aviária , Perus , Fatores de Virulência , Virulência , Animais , Galinhas/virologia , Glicoproteínas de Hemaglutininação de Vírus da Influenza/química , Glicoproteínas de Hemaglutininação de Vírus da Influenza/genética , Vírus da Influenza A Subtipo H7N1/genética , Vírus da Influenza A Subtipo H7N1/patogenicidade , Influenza Aviária/mortalidade , Influenza Aviária/virologia , Perus/virologia , Virulência/genética , Fatores de Virulência/química , Fatores de Virulência/genética
14.
Microbiol Spectr ; 10(2): e0010322, 2022 04 27.
Artigo em Inglês | MEDLINE | ID: mdl-35384712

RESUMO

Rustrela virus (RusV; species Rubivirus strelense) is a recently discovered relative of rubella virus (RuV) that has been detected in cases of encephalitis in diverse mammals. Here, we diagnosed two additional cases of fatal RusV-associated meningoencephalitis in a South American coati (Nasua nasua) and a Eurasian or European otter (Lutra lutra) that were detected in a zoological garden with history of prior RusV infections. Both animals showed abnormal movement or unusual behavior and their brains tested positive for RusV using specific reverse transcription quantitative PCR (RT-qPCR) and RNA in situ hybridization. As previous sequencing of the RusV genome proved to be very challenging, we employed a sophisticated target-specific capture enrichment with specifically designed RNA baits to generate complete RusV genome sequences from both detected encephalitic animals and apparently healthy wild yellow-necked field mice (Apodemus flavicollis). Furthermore, the technique was used to revise three previously published RusV genomes from two encephalitic animals and a wild yellow-necked field mouse. When comparing the newly generated RusV sequences to the previously published RusV genomes, we identified a previously undetected stretch of 309 nucleotides predicted to represent the intergenic region and the sequence encoding the N terminus of the capsid protein. This indicated that the original RusV sequence was likely incomplete due to misassembly of the genome at a region with an exceptionally high G+C content of >80 mol%. The new sequence data indicate that RusV has an overall genome length of 9,631 nucleotides with the longest intergenic region (290 nucleotides) and capsid protein-encoding sequence (331 codons) within the genus Rubivirus. IMPORTANCE The detection of rustrela virus (RusV)-associated encephalitis in two carnivoran mammal species further extends the knowledge on susceptible species. Furthermore, we provide clinical and pathological data for the two new RusV cases, which were until now limited to the initial description of this fatal encephalitis. Using a sophisticated enrichment method prior to sequencing of the viral genome, we markedly improved the virus-to-background sequence ratio compared to that of standard procedures. Consequently, we were able to resolve and update the intergenic region and the coding region for the N terminus of the capsid protein of the initial RusV genome sequence. The updated putative capsid protein now resembles those of rubella and ruhugu virus in size and harbors a predicted RNA-binding domain that had not been identified in the initial RusV genome version. The newly determined complete RusV genomes strongly improve our knowledge of the genome structure of this novel rubivirus.


Assuntos
Capsídeo , Encefalite , Animais , Proteínas do Capsídeo/genética , DNA Intergênico , Encefalite/veterinária , Mamíferos , Camundongos , Nucleotídeos , RNA , Rubivirus
15.
Microorganisms ; 10(3)2022 Feb 28.
Artigo em Inglês | MEDLINE | ID: mdl-35336101

RESUMO

Studies on tick-pathogen-host interactions are helping to identify candidates for vaccines against ticks and tick-borne diseases and to discover potent bioactive tick molecules. The tick midgut is the main tissue involved in blood feeding and, moreover, the first organ to have contact with pathogens ingested through the blood meal. As little is known about the molecular biology of feeding and tick defence mechanisms against microorganisms, but important for understanding vector-pathogen interactions, we explored the early transcriptional changes in the midgut of Ornithodoros moubata after feeding and in response to challenge with the relapsing-fever spirochete Borrelia duttonii using the Ion S5XL platform. Besides transcripts with metabolic function and immune-related transcripts we discovered numerous putative and uncharacterized protein sequences. Overall, our analyses support previous studies and provides a valuable reference database for further functional proteomic analysis of midgut proteins of O. moubata.

16.
J Gen Virol ; 103(12)2022 12.
Artigo em Inglês | MEDLINE | ID: mdl-36748520

RESUMO

The family Matonaviridae comprises enveloped viruses with positive-sense RNA genomes of 9.6-10 kb. The genus Rubivirus includes rubella virus (species Rubivirus rubellae) infecting humans, ruhugu virus (species Rubivirus ruteetense) infecting bats and rustrela virus (species Rubivirus strelense) infecting rodents and zoo animals. Rubella virus is spread via droplets. Postnatal infection leads to benign disease with rash and fever. Infection of seronegative women with rubella virus during the first trimester of pregnancy will often result in severe foetal malformations, known as congenital rubella syndrome. Vaccines are globally available. This is a summary of the International Committee on Taxonomy of Viruses (ICTV) Report on the family Matonaviridae, which is available at ictv.global/report/matonaviridae.


Assuntos
Vírus de RNA , Vírus , Animais , Feminino , Humanos , Vírus de RNA/genética , Vírus/genética , Vírus da Rubéola/genética , Genoma Viral
17.
PLoS Pathog ; 17(10): e1010006, 2021 10.
Artigo em Inglês | MEDLINE | ID: mdl-34673841

RESUMO

Marek's disease virus (MDV) is an alphaherpesvirus that causes immunosuppression and deadly lymphoma in chickens. Lymphoid organs play a central role in MDV infection in animals. B-cells in the bursa of Fabricius facilitate high levels of MDV replication and contribute to dissemination at early stages of infection. Several studies investigated host responses in bursal tissue of MDV-infected chickens; however, the cellular responses specifically in bursal B-cells has never been investigated. We took advantage of our recently established in vitro infection system to decipher the cellular responses of bursal B-cells to infection with a very virulent MDV strain. Here, we demonstrate that MDV infection extends the survival of bursal B-cells in culture. Microarray analyses revealed that most cytokine/cytokine-receptor-, cell cycle- and apoptosis-associated genes are significantly down-regulated in these cells. Further functional assays validated these strong effects of MDV infections on cell cycle progression and thus, B-cell proliferation. In addition, we confirmed that MDV infections protect B-cells from apoptosis and trigger an accumulation of the autophagy marker Lc3-II. Taken together, our data indicate that MDV-infected bursal B-cells show hallmarks of a senescence-like phenotype, leading to a prolonged B-cell survival. This study provides an in-depth analysis of bursal B-cell responses to MDV infection and important insights into how the virus extends the survival of these cells.


Assuntos
Linfócitos B/virologia , Doença de Marek , Animais , Senescência Celular/fisiologia , Galinhas , Mardivirus , Fenótipo
18.
Viruses ; 13(10)2021 09 28.
Artigo em Inglês | MEDLINE | ID: mdl-34696370

RESUMO

Camelpox virus (CMLV) is the causative agent of camelpox, which frequently occurs in the Old World camelids-rearing countries except for Australia. It has also been described in experimentally inoculated New World camelids. Camelpox outbreaks are often experienced shortly after the rainy season, which occurs twice a year on the Arabian Peninsula because of the increased density of the insect population, particularly mosquitos. A systemic form of camelpox outbreak in seven dromedary camels was diagnosed by histology, virus isolation, and PCR. A phylogenetic analysis using full length CMLV genomes of the isolated CMLV strains showed a single phylogenetic unit without any distinctive differences between them. The United Arab Emirates (UAE) isolate sequences showed phylogenetical relatedness with CMLV isolates from Israel with only minor sequence differences. Although the sequences of viruses from both countries were closely related, the disease manifestation was vastly different. Our study shows that the virulence is not only determined by genetic features of CMLV alone but may also depend on other factors such as unknown aspects of the host (e.g., age, overall fitness), management, and the environment.


Assuntos
Camelus/virologia , Surtos de Doenças/estatística & dados numéricos , Surtos de Doenças/veterinária , Orthopoxvirus/genética , Infecções por Poxviridae/epidemiologia , Infecções por Poxviridae/veterinária , Animais , Feminino , Sequenciamento de Nucleotídeos em Larga Escala , Masculino , Orthopoxvirus/classificação , Filogenia , Infecções por Poxviridae/mortalidade , Análise de Sequência de DNA , Emirados Árabes Unidos
19.
Viruses ; 13(10)2021 10 03.
Artigo em Inglês | MEDLINE | ID: mdl-34696419

RESUMO

Molecular details of field rabies virus (RABV) adaptation to cell culture replication are insufficiently understood. A better understanding of adaptation may not only reveal requirements for efficient RABV replication in cell lines, but may also provide novel insights into RABV biology and adaptation-related loss of virulence and pathogenicity. Using two recombinant field rabies virus clones (rRABV Dog and rRABV Fox), we performed virus passages in three different cell lines to identify cell culture adaptive mutations. Ten passages were sufficient for the acquisition of adaptive mutations in the glycoprotein G and in the C-terminus of phosphoprotein P. Apart from the insertion of a glycosylation sequon via the mutation D247N in either virus, both acquired additional and cell line-specific mutations after passages on BHK (K425N) and MDCK-II (R346S or R350G) cells. As determined by virus replication kinetics, complementation, and immunofluorescence analysis, the major bottleneck in cell culture replication was the intracellular accumulation of field virus G protein, which was overcome after the acquisition of the adaptive mutations. Our data indicate that limited release of extracellular infectious virus at the plasma membrane is a defined characteristic of highly virulent field rabies viruses and we hypothesize that the observed suboptimal release of infectious virions is due to the inverse correlation of virus release and virulence in vivo.


Assuntos
Antígenos Virais/genética , Vírus da Raiva/genética , Proteínas do Envelope Viral/genética , Liberação de Vírus/genética , Animais , Anticorpos Antivirais/sangue , Antígenos Virais/metabolismo , Técnicas de Cultura de Células , Linhagem Celular , Cães , Glicoproteínas/genética , Glicosilação , Mutação Puntual/genética , Raiva/virologia , Proteínas do Envelope Viral/metabolismo , Proteínas Virais/genética , Vírion/metabolismo , Virulência/genética , Replicação Viral/genética
20.
Sensors (Basel) ; 21(18)2021 Sep 21.
Artigo em Inglês | MEDLINE | ID: mdl-34577521

RESUMO

The SE(2) domain can be used to describe the position and orientation of objects in planar scenarios and is inherently nonlinear due to the periodicity of the angle. We present a novel filter that involves splitting up the joint density into a (marginalized) density for the periodic part and a conditional density for the linear part. We subdivide the state space along the periodic dimension and describe each part of the state space using the parameters of a Gaussian and a grid value, which is the function value of the marginalized density for the periodic part at the center of the respective area. By using the grid values as weighting factors for the Gaussians along the linear dimensions, we can approximate functions on the SE(2) domain with correlated position and orientation. Based on this representation, we interweave a grid filter with a Kalman filter to obtain a filter that can take different numbers of parameters and is in the same complexity class as a grid filter for circular domains. We thoroughly compared the filters with other state-of-the-art filters in a simulated tracking scenario. With only little run time, our filter outperformed an unscented Kalman filter for manifolds and a progressive filter based on dual quaternions. Our filter also yielded more accurate results than a particle filter using one million particles while being faster by over an order of magnitude.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA