Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 393
Filtrar
1.
Arch Virol ; 168(3): 91, 2023 Feb 14.
Artigo em Inglês | MEDLINE | ID: mdl-36786868

RESUMO

The first bovine parechovirus (Bo_ParV) was reported in 2021, and currently, only two nearly complete genome sequences of Bo_ParV are available. In this study, we detected Bo_ParVs in 10 out of 158 bovine fecal samples tested using real-time RT-PCR, and Bo_ParVs were isolated from three of these samples using MA104 cells. Analysis of the P1 region revealed that Bo_ParVs shared high pairwise amino acid sequence similarity (≥ 95.7% identity), suggesting antigenic similarity among Bo_ParVs, whereas nucleotide sequence identity values (≥ 84.8%) indicated more variability. A recombination breakpoint was identified in the 2B region, which may influence the evolution of this virus.


Assuntos
Bovinos , Parechovirus , Animais , Bovinos/virologia , Variação Genética , Genótipo , Parechovirus/genética , Filogenia , Prevalência
2.
J Virol ; 96(15): e0075122, 2022 08 10.
Artigo em Inglês | MEDLINE | ID: mdl-35867566

RESUMO

Lumpy skin disease virus (LSDV) is a poxvirus that causes severe systemic disease in cattle and is spread by mechanical arthropod-borne transmission. This study quantified the acquisition and retention of LSDV by four species of Diptera (Stomoxys calcitrans, Aedes aegypti, Culex quinquefasciatus, and Culicoides nubeculosus) from cutaneous lesions, normal skin, and blood from a clinically affected animal. The acquisition and retention of LSDV by Ae. aegypti from an artificial membrane feeding system was also examined. Mathematical models of the data were generated to identify the parameters which influence insect acquisition and retention of LSDV. For all four insect species, the probability of acquiring LSDV was substantially greater when feeding on a lesion compared with feeding on normal skin or blood from a clinically affected animal. After feeding on a skin lesion LSDV was retained on the proboscis for a similar length of time (around 9 days) for all four species and for a shorter time in the rest of the body, ranging from 2.2 to 6.4 days. Acquisition and retention of LSDV by Ae. aegypti after feeding on an artificial membrane feeding system that contained a high titer of LSDV was comparable to feeding on a skin lesion on a clinically affected animal, supporting the use of this laboratory model as a replacement for some animal studies. This work reveals that the cutaneous lesions of LSD provide the high-titer source required for acquisition of the virus by insects, thereby enabling the mechanical vector-borne transmission. IMPORTANCE Lumpy skin disease virus (LSDV) is a high consequence pathogen of cattle that is rapidly expanding its geographical boundaries into new regions such as Europe and Asia. This expansion is promoted by the mechanical transmission of the virus via hematogenous arthropods. This study quantifies the acquisition and retention of LSDV by four species of blood-feeding insects and reveals that the cutaneous lesions of LSD provide the high titer virus source necessary for virus acquisition by the insects. An artificial membrane feeding system containing a high titer of LSDV was shown to be comparable to a skin lesion on a clinically affected animal when used as a virus source. This promotes the use of these laboratory-based systems as replacements for some animal studies. Overall, this work advances our understanding of the mechanical vector-borne transmission of LSDV and provides evidence to support the design of more effective disease control programmes.


Assuntos
Sangue , Dípteros , Comportamento Alimentar , Insetos Vetores , Doença Nodular Cutânea , Vírus da Doença Nodular Cutânea , Aedes/anatomia & histologia , Aedes/virologia , Animais , Bovinos/virologia , Ceratopogonidae/anatomia & histologia , Ceratopogonidae/virologia , Culex/anatomia & histologia , Culex/virologia , Dípteros/anatomia & histologia , Dípteros/fisiologia , Dípteros/virologia , Insetos Vetores/anatomia & histologia , Insetos Vetores/fisiologia , Insetos Vetores/virologia , Doença Nodular Cutânea/virologia , Vírus da Doença Nodular Cutânea/isolamento & purificação , Vírus da Doença Nodular Cutânea/fisiologia , Membranas Artificiais , Muscidae/anatomia & histologia , Muscidae/virologia , Fatores de Tempo
3.
J Virol ; 96(15): e0055022, 2022 08 10.
Artigo em Inglês | MEDLINE | ID: mdl-35862708

RESUMO

The basis for rotavirus (RV) host range restriction (HRR) is not fully understood but is likely multigenic. RV genes encoding VP3, VP4, NSP1, NSP2, NSP3, and NSP4 have been associated with HRR in various studies. With the exception of NSP1, little is known about the relative contribution of the other RV genes to HRR. VP4 has been linked to HRR because it functions as the RV cell attachment protein, but its actual role in HRR has not been fully assessed. We generated a collection of recombinant RVs (rRVs) in an isogenic murine-like RV genetic background, harboring either heterologous or homologous VP4 genes from simian, bovine, porcine, human, and murine RV strains, and characterized these rRVs in vitro and in vivo. We found that a murine-like rRV encoding a simian VP4 was shed, spread to uninoculated littermates, and induced diarrhea comparably to rRV harboring a murine VP4. However, rRVs carrying VP4s from both bovine and porcine RVs had reduced diarrhea, but no change in fecal shedding was observed. Both diarrhea and shedding were reduced when VP4 originated from a human RV strain. rRVs harboring VP4s from human or bovine RVs did not transmit to uninoculated littermates. We also generated two rRVs harboring reciprocal chimeric murine or bovine VP4. Both chimeras replicated and caused disease as efficiently as the parental strain with a fully murine VP4. These data suggest that the genetic origin of VP4 partially modulates HRR in the suckling mouse and that both the VP8* and VP5* domains independently contribute to pathogenesis and transmission. IMPORTANCE Human group A rotaviruses (RVs) remain the most important cause of severe acute gastroenteritis among infants and young children worldwide despite the introduction of several safe and effective live attenuated vaccines. The lack of knowledge regarding fundamental aspects of RV biology, such as the genetic basis of host range restriction (HRR), has made it difficult to predictively and efficiently design improved, next-generation live attenuated rotavirus vaccines. Here, we engineered a collection of VP4 monoreassortant RVs to systematically explore the role of VP4 in replication, pathogenicity, and spread, as measures of HRR, in a suckling mouse model. The genetic and mechanistic bases of HRR have substantial clinical relevance given that this restriction forms the basis of attenuation for several replication-competent human RV vaccines. In addition, a better understanding of RV pathogenesis and the determinants of RV spread is likely to enhance our ability to improve antiviral drug and therapy development.


Assuntos
Proteínas do Capsídeo , Modelos Animais de Doenças , Especificidade de Hospedeiro , Infecções por Rotavirus , Rotavirus , Animais , Animais Lactentes , Proteínas do Capsídeo/metabolismo , Bovinos/virologia , Diarreia/veterinária , Diarreia/virologia , Haplorrinos/virologia , Humanos , Hibridização Genética , Camundongos/virologia , Rotavirus/classificação , Rotavirus/patogenicidade , Rotavirus/fisiologia , Infecções por Rotavirus/transmissão , Infecções por Rotavirus/veterinária , Infecções por Rotavirus/virologia , Suínos/virologia , Vacinas Atenuadas , Virulência , Replicação Viral/genética
4.
Virol Sin ; 37(2): 229-237, 2022 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-35527224

RESUMO

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


Assuntos
Alphavirus , Culicidae , Alphavirus/genética , Animais , Anticorpos Neutralizantes/sangue , Anticorpos Antivirais/sangue , Bovinos/virologia , China/epidemiologia , Culicidae/virologia , Cabras/virologia , Cavalos/virologia , Masculino , Filogenia , Análise de Sequência de DNA , Estudos Soroepidemiológicos , Ovinos/virologia , Suínos/virologia
5.
Viruses ; 14(2)2022 02 05.
Artigo em Inglês | MEDLINE | ID: mdl-35215918

RESUMO

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


Assuntos
Infecções por Alphavirus/veterinária , Infecções por Alphavirus/virologia , Alphavirus/fisiologia , Alphavirus/ultraestrutura , Montagem de Vírus , Alphavirus/classificação , Alphavirus/genética , Animais , Bovinos/virologia , Microscopia Crioeletrônica , Dimerização , Raposas/virologia , Cavalos/virologia , Humanos , Modelos Moleculares , Filogenia , Suínos/virologia , Proteínas do Envelope Viral/química , Proteínas do Envelope Viral/genética , Proteínas do Envelope Viral/metabolismo , Vírion/classificação , Vírion/genética , Vírion/fisiologia , Vírion/ultraestrutura
6.
Virus Res ; 308: 198632, 2022 01 15.
Artigo em Inglês | MEDLINE | ID: mdl-34793872

RESUMO

In 2020, to trace the prevalence and evolution of bovine coronavirus (BCoV) in China, a total of 1383 samples (1016 fecal samples and 367 nasal swab samples) were collected from 1016 cattle exhibiting diarrhea symptoms on dairy farms and beef cattle farms in Heilongjiang Province, Northeast China. All samples were subjected to reverse transcription-polymerase chain reaction (RT-PCR) detection of the BCoV N gene, followed by an analysis of its epidemiology and genetic evolution. The results indicated that of the 1016 diarrhea-affected cattle, 15.45% (157/1016) were positive for BCoV, in which positive rates of the fecal and nasal swab samples were 12.20% (124/1016) and 21.53% (79/367), respectively. Of the 367 cattle whose nasal swab samples were collected, the BCoV positive rate of the corresponding fecal samples was 15.26% (56/367). BCoV infection was significantly associated with age, farming pattern, cattle type, farm latitude, sample type, and clinical symptom (p < 0.05). Of the 203 BCoV-positive samples, 20 spike (S) genes were successfully sequenced. The 20 identified BCoV strains shared nucleotide homologies of 97.7-100.0%, and their N-terminal domain of S1 subunit (S1-NTD: residues 15-298) differed genetically from the reference strains of South Korea and Europe. The 20 identified BCoV strains were clustered in the Asia-North America group (GII group) in the global strain-based phylogenetic tree and formed three clades in the Chinese strain-based phylogenetic tree. The HLJ/HH-10/2020 strain was clustered into the Europe group (GI group) in the S1-NTD-based phylogenetic tree, exhibiting N146/I, D148/G, and L154/F mutations that affect the S protein structure. Of the identified BCoV strains, one potential recombination event occurred between the HLJ/HH-20/2020 and HLJ/HH-10/2020 strains, which led to the generation of the recombinant BCV-AKS-01 strain. A selective pressure analysis on the S protein revealed one positively selected site (Asn509) among the 20 identified BCoV strains located inside the putative receptor binding domain (residues 326-540). These data provide a greater understanding of the epidemiology and evolution of BCoV in China.


Assuntos
Doenças dos Bovinos , Infecções por Coronavirus , Coronavirus Bovino , Animais , Bovinos/virologia , Doenças dos Bovinos/epidemiologia , Doenças dos Bovinos/virologia , China/epidemiologia , Infecções por Coronavirus/epidemiologia , Infecções por Coronavirus/veterinária , Coronavirus Bovino/genética , Diarreia/epidemiologia , Diarreia/veterinária , Fezes , Variação Genética , Filogenia , Análise de Sequência de RNA
7.
J Gen Virol ; 102(12)2021 12.
Artigo em Inglês | MEDLINE | ID: mdl-34870577

RESUMO

Middle Point orbivirus (MPOV) is an Australian arbovirus, belongs to the Yunnan orbivirus species found in China. First detected and reported from Beatrice Hill, Northern Territory (NT), MPOV has to date, only been exclusively reported from the NT, Australia. Whilst genetic characterization of MPOV has been previously described, only restricted to sequence information for segments 2 and 3 coding core protein VP2 and outer capsid protein VP3, respectively. This study presents for the first time nearly full-length genome sequences of MPOV, which represent 24 isolates collected over a span of more than 20 years from 1997 to 2018. Whilst the majority of isolates were sampled at Beatrice Hill, NT where MPOV is most frequently isolated, this report also describes the first two isolations of MPOV from Queensland (QLD), Australia. One of which is the first non-bovine isolate obtained from the mosquito vector Aedes vittiger. We further compared these MPOV sequences with known sequences of the Yunnan orbivirus and other known orbivirus sequences of mosquito origin found in Australia. The phylogenetic analyses indicate the Australian MPOV sequences are more closely related to each other than other known sequences of Yunnan orbivirus. Furthermore, MPOV sequences are closely related to sequences from the Indonesian isolate JKT-8650. The clustering of Australian sequences in the phylogenetic tree suggests the monophyletic lineage of MPOV circulating in Australia. Further, ongoing surveillance is required to assess the existence and prevalence of this or other yet undetected lineages of MPOV and other orbiviruses in Australia.


Assuntos
Genoma Viral/genética , Orbivirus/genética , Filogenia , Aedes/virologia , Animais , Austrália , Bovinos/virologia , Mosquitos Vetores/virologia , Orbivirus/classificação , Orbivirus/isolamento & purificação , Infecções por Reoviridae/transmissão , Infecções por Reoviridae/veterinária , Infecções por Reoviridae/virologia , Especificidade da Espécie , Proteínas Virais/genética
8.
Emerg Microbes Infect ; 10(1): 2199-2201, 2021 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-34749583

RESUMO

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


Assuntos
COVID-19/veterinária , Especificidade de Hospedeiro , Gado/virologia , SARS-CoV-2/patogenicidade , Animais , Anticorpos Neutralizantes/sangue , Anticorpos Neutralizantes/imunologia , Anticorpos Antivirais/sangue , Anticorpos Antivirais/imunologia , COVID-19/imunologia , COVID-19/virologia , Camelídeos Americanos/virologia , Bovinos/virologia , Chlorocebus aethiops , Reservatórios de Doenças/virologia , Cabras/virologia , Cavalos/virologia , Especificidade de Hospedeiro/imunologia , Humanos , Cavidade Nasal/virologia , RNA Viral/análise , Coelhos/virologia , Reto/virologia , Sistema Respiratório/virologia , SARS-CoV-2/genética , SARS-CoV-2/isolamento & purificação , Ovinos/virologia , Especificidade da Espécie , Células Vero , Eliminação de Partículas Virais , Vísceras/virologia
9.
PLoS Negl Trop Dis ; 15(10): e0009837, 2021 10.
Artigo em Inglês | MEDLINE | ID: mdl-34695125

RESUMO

Rift Valley fever virus (RVFV) causes morbidity and mortality in humans and domestic ungulates in sub-Saharan Africa, Egypt, and the Arabian Peninsula. Mosquito vectors transmit RVFV between vertebrates by bite, and also vertically to produce infectious progeny. Arrival of RVFV into the United States by infected mosquitoes or humans could result in significant impacts on food security, human health, and wildlife health. Elucidation of the vectors involved in the post-introduction RVFV ecology is paramount to rapid implementation of vector control. We performed vector competence experiments in which field-collected mosquitoes were orally exposed to an epidemic strain of RVFV via infectious blood meals. We targeted floodwater Aedes species known to feed on cattle, and/or deer species (Aedes melanimon Dyar, Aedes increpitus Dyar, Aedes vexans [Meigen]). Two permanent-water-breeding species were targeted as well: Culiseta inornata (Williston) of unknown competence considering United States populations, and Culex tarsalis Coquillett as a control species for which transmission efficiency is known. We tested the potential for midgut infection, midgut escape (dissemination), ovarian infection (vertical transmission), and transmission by bite (infectious saliva). Tissues were assayed by plaque assay and RT-qPCR, to quantify infectious virus and confirm virus identity. Tissue infection data were analyzed using a within-host model under a Bayesian framework to determine the probabilities of infection outcomes (midgut-limited infection, disseminated infection, etc.) while estimating barriers to infection between tissues. Permanent-water-breeding mosquitoes (Cx. tarsalis and Cs. inornata) exhibited more efficient horizontal transmission, as well as potential for vertical transmission, which is contrary to the current assumptions of RVFV ecology. Barrier estimates trended higher for Aedes spp., suggesting systemic factors in the differences between these species and Cx. tarsalis and Cs. inornata. These data indicate higher potential for vertical transmission than previously appreciated, and support the consensus of RVFV transmission including a broad range of potential vectors.


Assuntos
Aedes/virologia , Culex/virologia , Mosquitos Vetores/virologia , Febre do Vale de Rift/transmissão , Vírus da Febre do Vale do Rift/fisiologia , Aedes/genética , Aedes/fisiologia , Animais , Bovinos/virologia , Colorado , Culex/fisiologia , Cervos/virologia , Mosquitos Vetores/classificação , Mosquitos Vetores/fisiologia , Febre do Vale de Rift/virologia , Vírus da Febre do Vale do Rift/genética , Vírus da Febre do Vale do Rift/isolamento & purificação , Saliva/virologia
10.
Viruses ; 13(10)2021 09 23.
Artigo em Inglês | MEDLINE | ID: mdl-34696337

RESUMO

Calf diarrhea is one of the common diseases involved in the process of calf feeding. In this study, a sample of calf diarrhea that tested positive for bovine coronavirus and bovine astrovirus was subjected to high-throughput sequencing. The reassembly revealed the complete genomes of bovine norovirus, bovine astrovirus, bovine kobuvirus, and the S gene of bovine coronavirus. Phylogenetic analysis showed that the ORF2 region of bovine astrovirus had the lowest similarity with other strains and gathered in the Mamastrovirus unclassified genogroup, suggesting a new serotype/genotype could appear. Compared with the most closely related strain, there are six amino acid mutation sites in the S gene of bovine coronavirus, most of which are located in the S1 subunit region. The bovine norovirus identified in our study was BNoV-GIII 2, based on the VP1 sequences. The bovine kobuvirus is distributed in the Aichi virus B genus; the P1 gene shows as highly variable, while the 3D gene is highly conserved. These findings enriched our knowledge of the viruses in the role of calf diarrhea, and help to develop an effective strategy for disease prevention and control.


Assuntos
Diarreia/etiologia , Genoma Viral/genética , Animais , Astroviridae/genética , Bovinos/virologia , Doenças dos Bovinos/virologia , Coronavirus/genética , Diarreia/virologia , Fezes/virologia , Variação Genética , Genótipo , Sequenciamento de Nucleotídeos em Larga Escala/métodos , Kobuvirus/genética , Norovirus/genética , Filogenia , Vírus/genética
11.
Viruses ; 13(10)2021 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-34696406

RESUMO

Coronaviruses (CoVs) are a group of enveloped positive-sense RNA viruses and can cause deadly diseases in animals and humans. Cell entry is the first and essential step of successful virus infection and can be divided into two ongoing steps: cell binding and membrane fusion. Over the past two decades, stimulated by the global outbreak of SARS-CoV and pandemic of SARS-CoV-2, numerous efforts have been made in the CoV research. As a result, significant progress has been achieved in our understanding of the cell entry process. Here, we review the current knowledge of this essential process, including the viral and host components involved in cell binding and membrane fusion, molecular mechanisms of their interactions, and the sites of virus entry. We highlight the recent findings of host restriction factors that inhibit CoVs entry. This knowledge not only enhances our understanding of the cell entry process, pathogenesis, tissue tropism, host range, and interspecies-transmission of CoVs but also provides a theoretical basis to design effective preventive and therapeutic strategies to control CoVs infection.


Assuntos
Infecções por Coronavirus/patologia , Coronavirus/metabolismo , Glicoproteína da Espícula de Coronavírus/metabolismo , Ligação Viral , Internalização do Vírus , Animais , Gatos/virologia , Bovinos/virologia , Galinhas/virologia , Coronavirus/genética , Cães/virologia , Gado/virologia , Fusão de Membrana/fisiologia , Receptores Virais/metabolismo , Glicoproteína da Espícula de Coronavírus/genética , Suínos/virologia , Tropismo Viral/fisiologia
12.
J Gen Virol ; 102(10)2021 10.
Artigo em Inglês | MEDLINE | ID: mdl-34714225

RESUMO

Calf diarrhoea has been a major cause of economic losses in the global dairy industry. Many factors, including multiple pathogen infections, can directly or indirectly cause calf diarrhoea. This study compared the faecal virome between 15 healthy calves and 15 calves with diarrhoea. Significantly lower diversity of viruses was found in samples from animals with diarrhoea than those in the healthy ones, and this feature may also be related to the age of the calves. Viruses belonging to the families Astroviridae and Caliciviridae that may cause diarrhoea in dairy calves have been characterized, which revealed that reads of caliciviruses and astroviruses in diarrhoea calves were much higher than those in healthy calves. Five complete genomic sequences closely related to Smacoviridae have been identified, which may participate in the regulation of the gut virus community ecology of healthy hosts together with bacteriophages. This research provides a theoretical basis for further understanding of known or potential enteric pathogens related to calf diarrhoea.


Assuntos
Doenças dos Bovinos/virologia , Bovinos/virologia , Diarreia/veterinária , Intestinos/virologia , Viroma , Animais , Caliciviridae/classificação , Caliciviridae/genética , Caliciviridae/isolamento & purificação , Vírus de DNA/classificação , Vírus de DNA/genética , Vírus de DNA/isolamento & purificação , Indústria de Laticínios , Diarreia/virologia , Fezes/virologia , Genoma Viral , Mamastrovirus/classificação , Mamastrovirus/genética , Mamastrovirus/isolamento & purificação , Metagenômica , Filogenia
13.
Viruses ; 13(8)2021 07 29.
Artigo em Inglês | MEDLINE | ID: mdl-34452357

RESUMO

Picobirnaviruses (PBVs) have been detected in several species of animals worldwide; however, data pertaining to their presence in Australian wild and domestic animals are limited. Although PBVs are mostly found in faecal samples, their detection in blood and respiratory tract samples raises questions concerning their tropism and pathogenicity. We report here PBV detection in wild deer and cattle from southeastern Australia. Through metagenomics, the presence of PBV genogroups I (GI) and II (GII) were detected in deer serum and plasma. Molecular epidemiology studies targeting the partial RNA-dependent RNA polymerase gene were performed in a wide range of specimens (serum, faeces, spleen, lung, nasal swabs, and trachea) collected from wild deer and cattle, with PCR amplification obtained in all specimen types except lung and spleen. Our results reveal the predominance of GI and concomitant detection of both genogroups in wild deer and cattle. In concordance with other studies, the detected GI sequences displayed high genetic diversity, however in contrast, GII sequences clustered into three distinct clades. Detection of both genogroups in the upper respiratory tract (trachea and nasal swab) of deer in the present study gives more evidence about the respiratory tract tropism of PBV. Although much remains unknown about the epidemiology and tropism of PBVs, our study suggests a wide distribution of these viruses in southeastern Australia.


Assuntos
Genótipo , Picobirnavirus/genética , Infecções por Vírus de RNA/epidemiologia , Infecções por Vírus de RNA/veterinária , Infecções Respiratórias/epidemiologia , Infecções Respiratórias/veterinária , Animais , Animais Selvagens/virologia , Austrália/epidemiologia , Bovinos/virologia , Cervos/virologia , Fezes/virologia , Variação Genética , Genoma Viral , Filogenia , Picobirnavirus/classificação , RNA Viral/genética , Infecções Respiratórias/virologia
14.
Prev Vet Med ; 195: 105446, 2021 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-34365125

RESUMO

A study was implemented to estimate the pestivirus seroprevalence in sheep and goats in Belgium, to identify circulating species and to check for a potential association between seropositivity of small ruminants and presence of cattle in the same farm. It was based on the testing of serum samples and bulk tank milk samples (BTM) collected in sheep and goat flocks in 2018-2019 all over the country. 7460 serum samples collected from 410 flocks were tested by a commercial ELISA able to detect antibodies (Ab) against Border Disease Virus (BDV), and Bovine Viral Diarrhea Virus (BVDV). BTM samples (n = 144) were collected from dairy flocks in November 2019 and tested with the same Ab ELISA. ELISA positive serum samples were also tested by virus neutralization test (VNT) for neutralizing antibodies against BDV, BVDV-type1 and BVDV-type2. Virological tests (RT-PCR) were performed on pools of serum samples from pestivirus-exposed flocks with at least two seropositive animals and on all Antibody-positive BTM samples. Information about serum and milk samples (identification, test results, farm of origin and location, presence of cattle) were gathered in animal-level and farm-level databases. Based on this study, the apparent animal seroprevalence for pestiviruses in small ruminant flocks in Belgium in 2018 was estimated to be 0.87 % (95 % C.I. [0.68 %-1.11 %]). The prevalence of flocks exposed to pestivirus (i.e. with at least one seropositive animal) was estimated to be 8.5 % (95 % C.I. [6.4 % - 11.6 %]). In exposed flocks, the average within-flock seroprevalence was 9.9 %. In dairy sheep and goats, the estimated proportion of exposed flocks in 2019, based on the detection of pestivirus antibodies in the bulk tank milk, was 9.7 % [5.9 %-15.7 %]. All PCR tests were negative, indicating the likely absence of active pestivirus circulation in these flocks. Although the observed pestivirus seroprevalence was found to be low in Belgian small ruminants, this study also showed, based on VNT results, that they are exposed to several pestivirus species: BDV, BVDV-1 and BVDV-2. 22.4 % of the farms included in the serological survey were holding both a small ruminant flock and a cattle herd, hence with a potential risk of contact between the two species. There was a significant positive association between pestivirus seropositivity in the sheep/goat flocks and the presence of a cattle herd in the same farm (OR = 2.42 (95 %C.I. [1.18-4.94]) but this association was not found for Ab-positive BTM in dairy flocks.


Assuntos
Doenças das Cabras , Infecções por Pestivirus , Doenças dos Ovinos , Animais , Anticorpos Antivirais , Bélgica/epidemiologia , Bovinos/virologia , Ensaio de Imunoadsorção Enzimática/veterinária , Doenças das Cabras/epidemiologia , Doenças das Cabras/virologia , Cabras/virologia , Infecções por Pestivirus/epidemiologia , Infecções por Pestivirus/veterinária , Estudos Soroepidemiológicos , Ovinos/virologia , Doenças dos Ovinos/epidemiologia , Doenças dos Ovinos/virologia
15.
Arch Virol ; 166(10): 2835-2839, 2021 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-34319454

RESUMO

The bovine adenovirus 7 (BAdV-7) isolate SD18-74 was recovered from lung tissue of calves in South Dakota. The 30,043-nucleotide (nt) genome has the typical organization of Atadenovirus genus members. The sequence shares over 99% nt sequence identity with two Japanese BAdV-7 sequences, followed by 74.9% nt sequence identity with the ovine adenovirus 7 strain OAV287, a member of the species Ovine atadenovirus D. SD18-74 was amplified in both bovine and ovine primary nasal turbinate cells, demonstrating greater fitness in bovine cells. The genomic and biological characteristics of BAdV-7 SD18-74 support the inclusion of the members of the BAdV-7 group in a new species in the genus Atadenovirus.


Assuntos
Infecções por Adenoviridae/veterinária , Atadenovirus/classificação , Atadenovirus/genética , Bovinos/virologia , Infecções por Adenoviridae/virologia , Animais , Atadenovirus/isolamento & purificação , Atadenovirus/fisiologia , Doenças dos Bovinos/virologia , Linhagem Celular , DNA Viral/genética , Genoma Viral/genética , Ovinos , Estados Unidos , Replicação Viral
16.
Viruses ; 13(7)2021 06 27.
Artigo em Inglês | MEDLINE | ID: mdl-34199054

RESUMO

Nairobi sheep disease orthonairovirus (NSDV) is a zoonotic tick-borne arbovirus, which causes severe gastroenteritis in small ruminants. To date, the virus is prevalent in East Africa and Asia. However, due to climate change, including the spread of transmitting tick vectors and increased animal movements, it is likely that the distribution range of NSDV is enlarging. In this project, sheep and cattle (hitherto classified as resistant to NSDV) were experimentally infected with NSDV for a comparative study of the species-specific pathogenesis. For this purpose, several new diagnostic assays (RT-qPCR, ELISA, iIFA, mVNT, PRNT) were developed, which will also be useful for future epidemiological investigations. All challenged sheep (three different doses groups) developed characteristic clinical signs, transient viremia and virus shedding-almost independent on the applied virus dose. Half of the sheep had to be euthanized due to severe clinical signs, including hemorrhagic diarrhea. In contrast, the course of infection in cattle was only subclinical. However, all ruminants showed seroconversion-implying that, indeed, both species are susceptible for NSDV. Hence, not only sheep but also cattle sera can be included in serological monitoring programs for the surveillance of NSDV occurrence and spread in the future.


Assuntos
Doenças dos Bovinos/diagnóstico , Doença dos Ovinos de Nairobi/diagnóstico , Doença dos Ovinos de Nairobi/patologia , Nairovirus/genética , Nairovirus/patogenicidade , Doenças dos Ovinos/diagnóstico , Animais , Bovinos/virologia , Doenças dos Bovinos/epidemiologia , Doenças dos Bovinos/imunologia , Feminino , Masculino , Técnicas de Diagnóstico Molecular/métodos , Doença dos Ovinos de Nairobi/epidemiologia , Doença dos Ovinos de Nairobi/imunologia , Nairovirus/imunologia , Soroconversão , Testes Sorológicos/métodos , Ovinos/virologia , Doenças dos Ovinos/epidemiologia , Doenças dos Ovinos/imunologia , Carrapatos/virologia
17.
Parasit Vectors ; 14(1): 342, 2021 Jun 29.
Artigo em Inglês | MEDLINE | ID: mdl-34187526

RESUMO

BACKGROUND: Crimean-Congo hemorrhagic fever virus (CCHFV) belongs to the genus Orthonairovirus (Nairovididae) and is a (re)emerging tick-borne pathogen. It is endemic in most parts of Africa, Asia and southern Europe, and can cause severe hemorrhagic symptoms in humans, with high fatality rates (5-30%). METHODS: Hyalomma ticks were collected from four different livestock herds (cattle and camels) in Mauritania in 2018. The tick species were determined morphologically and confirmed molecularly by using the cytochrome oxidase 1 gene marker. For the detection of CCHFV, ticks were tested individually by one-step multiplex real-time reverse-transcriptase quantitative polymerase chain reaction. The small segment of all positive samples was sequenced to determine the CCHFV genotype. RESULTS: In total, 39 of the 1523 ticks (2.56%) collected from 63 cattles and 28 camels tested positive for CCHFV. Three Hyalomma species were identified. Hyalomma rufipes had the largest proportion of positivity (5.67%; 16/282), followed by Hyalomma dromedarii (1.89%; 23/1214). No Hyalomma impeltatum tested positive (0%; 0/21). Positive ticks were found in only six out of 91 host animals. Viral sequence analysis revealed the presence of two different CCHFV lineages (Africa I and Africa III). CONCLUSIONS: In this study, 2.56% of Hyalomma ticks collected from camels and cattle in Mauritania tested positive for CCHFV. However, the true prevalence of CCHFV in unfed ticks may be lower, as a considerable number of ticks may have been passively infected during blood-feeding by co-feeding ticks or due to viremia of the host. The results indicate the need to track the actual area of circulation of this virus.


Assuntos
Sangue , Vírus da Febre Hemorrágica da Crimeia-Congo/genética , Gado/parasitologia , Carrapatos/virologia , Animais , Camelus/parasitologia , Camelus/virologia , Bovinos/parasitologia , Bovinos/virologia , Comportamento Alimentar , Feminino , Genótipo , Vírus da Febre Hemorrágica da Crimeia-Congo/isolamento & purificação , Febre Hemorrágica da Crimeia/virologia , Gado/virologia , Masculino , Mauritânia , Filogenia , RNA Viral/genética , Carrapatos/genética , Carrapatos/fisiologia
18.
Prev Vet Med ; 192: 105375, 2021 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-33989913

RESUMO

We present a new modelling framework to address the evaluation of national control/surveillance programs planned in line with the European Animal Health Law (AHL) for livestock diseases. Our modelling framework is applied to the cattle sector in Ireland where there is need for policy support to design an optimal programme to achieve bovine herpesvirus type 1 (BoHV-1) free status under the AHL. In this contribution, we show how our framework establishes a regional model that is able to mechanistically reproduce the demography, management practices and transport patterns of an entire cattle population without being dependent on continuous livestock registry data. An innovative feature of our model is the inclusion of herd typing, thereby extending these beyond the categories of dairy, beef and mixed herds that are frequently considered in other regional modelling studies. This detailed representation of herd type-specific management facilitates comparative assessment of BoHV-1 eradication strategies targeting different production types with individual strategy protocols. Finally, we apply our model to support current discussions regarding the structure and implementation of a potential national BoHV-1 eradication programme in Ireland.


Assuntos
Bovinos/virologia , Herpesvirus Bovino 1 , Rinotraqueíte Infecciosa Bovina , Animais , Tomada de Decisões , Rinotraqueíte Infecciosa Bovina/epidemiologia , Rinotraqueíte Infecciosa Bovina/prevenção & controle , Irlanda/epidemiologia , Modelos Teóricos
19.
J Gen Virol ; 102(4)2021 04.
Artigo em Inglês | MEDLINE | ID: mdl-33847554

RESUMO

Group A rotaviruses (RVAs) infect a wide variety of mammalian and avian species. Animals act as a potential reservoir to RVA human infections by direct virion transmission or by contributing genes to reassortants. Here, we report the molecular characterization of a rare human RVA strain Ni17-46 with a genotype G15P[14], isolated in Japan in 2017 during rotavirus surveillance in a paediatric outpatient clinic. The genome constellation of this strain was G15-P[14]-I2-R2-C2-M2-A13-N2-T9-E2-H3. This is the first report of an RVA with G15 genotype in humans, and sequencing and phylogenetic analysis results suggest that human infection with this strain has zoonotic origin from the bovine species. Given the fact that this strain was isolated from a patient with gastroenteritis and dehydration symptoms, we must take into account the virulence of this strain in humans.


Assuntos
Gastroenterite/virologia , Genoma Viral , Infecções por Rotavirus/virologia , Rotavirus , Zoonoses Virais/virologia , Adolescente , Animais , Bovinos/virologia , Feminino , Genótipo , Humanos , Japão , Rotavirus/genética , Rotavirus/isolamento & purificação
20.
Prev Vet Med ; 190: 105318, 2021 May.
Artigo em Inglês | MEDLINE | ID: mdl-33740596

RESUMO

The study investigated the important epidemiological parameters and farm-level economic costs of FMD incidence in cattle and buffaloes during 2013-14 to 2015-16 in various states of India. Multistage random sampling procedure was adopted for the primary survey and data was collected through face-to-face personal interview from 18,609 cattle and buffalo rearing farm households from 123 districts across twelve states and one Union Territory. Besides epidemiological parameters, different farm-level direct and indirect loss associated with FMD was assessed at disaggregated level (states) by employing deterministic mathematical models. Highest number of affected villages and disease incidence was observed in non- FMD control programme (FMD-CP) implemented Madhya Pradesh and Assam states, respectively whereas negligible incidence was in FMD-CP implemented Punjab state. The disease incidence was high during 2013-14 and declined during 2014-15 and 2015-16, respectively implied severe incidence scenario (2013-14) succeeded by moderate (2014-15) and mild (2015-16) scenarios. The crossbred and high productive animals were severely affected than local breeds whereas on sexwise and agewise comparison revealed higher incidence in females and adult animals. During severe incidence scenario, milk loss/animal ranged from USD 6.87-47.44, 18.42-125.88, 16.33-91.43, and 27.17-123.62; mortality loss/animal ranged from USD 32.61-804.27, 30.76-577.7, 65.36-502.2, and 188.04-413.7; distress sale loss/animal ranged from USD 3.22-188.63, 64.34-519.3, 214.47-341.8, and 209.11-450.3; and opportunity cost of labour/animal from USD 5.49-54.29, 5.49-67.78; 7.95-31.37 and 9.83-72.38 in indigenous cattle, crossbred cattle, local and improved buffalo, respectively. The estimated draught power loss/animal varied from USD 39.46-142.94 with least being in Madhya Pradesh and highest in Assam states whereas the median treatment cost/animal was USD 9.18 and USD 27.07 in indigenous cattle and upgraded buffaloes, respectively. The total farm-level economic loss projected due to FMD in cattle and buffaloes in India was USD 3159 million (INR 221,110 million), USD 270 million (INR 18,910 million) and USD 152 million (INR 10,610 million), respectively during the severe, moderate and mild incidence scenarios at 2015-16 constant prices. The loss varied across the states, and in severe incidence scenario, the country might lose USD 3.2 billion/year and hence, the bi-annual vaccination schedule need to be strictly implemented in all the states. Besides timely vaccination coverage, managing unabated animal movement, educating and motivating the farmers to vaccinate their animals might reduce the incidence and consequential losses to various stakeholders in endemic states like India.


Assuntos
Doenças dos Bovinos , Febre Aftosa , Animais , Búfalos/virologia , Bovinos/virologia , Doenças dos Bovinos/economia , Doenças dos Bovinos/epidemiologia , Doenças dos Bovinos/virologia , Surtos de Doenças , Fazendas/economia , Feminino , Febre Aftosa/economia , Febre Aftosa/epidemiologia , Incidência , Índia/epidemiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...