Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 561
Filtrar
1.
Viruses ; 14(2)2022 01 18.
Artigo em Inglês | MEDLINE | ID: mdl-35215769

RESUMO

Bats have been recognized as an exceptional viral reservoir, especially for coronaviruses. At least three bat zoonotic coronaviruses (SARS-CoV, MERS-CoV and SARS-CoV-2) have been shown to cause severe diseases in humans and it is expected more will emerge. One of the major features of CoVs is that they are all highly prone to recombination. An extreme example is the insertion of the P10 gene from reoviruses in the bat CoV GCCDC1, first discovered in Rousettus leschenaultii bats in China. Here, we report the detection of GCCDC1 in four different bat species (Eonycteris spelaea, Cynopterus sphinx, Rhinolophus shameli and Rousettus sp.) in Cambodia. This finding demonstrates a much broader geographic and bat species range for this virus and indicates common cross-species transmission. Interestingly, one of the bat samples showed a co-infection with an Alpha CoV most closely related to RsYN14, a virus recently discovered in the same genus (Rhinolophus) of bat in Yunnan, China, 2020. Taken together, our latest findings highlight the need to conduct active surveillance in bats to assess the risk of emerging CoVs, especially in Southeast Asia.


Assuntos
Quirópteros/virologia , Infecções por Coronaviridae/veterinária , Coronaviridae/classificação , Coronaviridae/genética , Reservatórios de Doenças/veterinária , Reservatórios de Doenças/virologia , Filogeografia , Recombinação Genética , Animais , Camboja/epidemiologia , China/epidemiologia , Quirópteros/classificação , Coronaviridae/isolamento & purificação , Infecções por Coronaviridae/epidemiologia , Infecções por Coronaviridae/transmissão , Evolução Molecular , Genoma Viral , Filogenia
2.
PLoS One ; 16(11): e0260038, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34813610

RESUMO

At present, global immunity to SARS-CoV-2 resides within a heterogeneous combination of susceptible, naturally infected and vaccinated individuals. The extent to which viral shedding and transmission occurs on re-exposure to SARS-CoV-2 is an important determinant of the rate at which COVID-19 achieves endemic stability. We used Sialodacryoadenitis Virus (SDAV) in rats to model the extent to which immune protection afforded by prior natural infection via high risk (inoculation; direct contact) or low risk (fomite) exposure, or by vaccination, influenced viral shedding and transmission on re-exposure. On initial infection, we confirmed that amount, duration and consistency of viral shedding, and seroconversion rates were correlated with exposure risk. Animals were reinfected after 3.7-5.5 months using the same exposure paradigm. 59% of seropositive animals shed virus, although at lower amounts. Previously exposed seropositive reinfected animals were able to transmit virus to 25% of naive recipient rats after 24-hour exposure by direct contact. Rats vaccinated intranasally with a related virus (Parker's Rat Coronavirus) were able to transmit SDAV to only 4.7% of naive animals after a 7-day direct contact exposure, despite comparable viral shedding. Cycle threshold values associated with transmission in both groups ranged from 29-36 cycles. Observed shedding was not a prerequisite for transmission. Results indicate that low-level shedding in both naturally infected and vaccinated seropositive animals can propagate infection in susceptible individuals. Extrapolated to COVID-19, our results suggest that continued propagation of SARS-CoV-2 by seropositive previously infected or vaccinated individuals is possible.


Assuntos
COVID-19/transmissão , Infecções por Coronaviridae/veterinária , Coronavirus do Rato/fisiologia , Modelos Biológicos , Modelos Estatísticos , Doenças dos Roedores/transmissão , Eliminação de Partículas Virais , Animais , COVID-19/virologia , Infecções por Coronaviridae/transmissão , Feminino , Masculino , Ratos , Ratos Sprague-Dawley , SARS-CoV-2/fisiologia , Soroconversão
3.
Avian Pathol ; 50(4): 295-310, 2021 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-34126817

RESUMO

Infectious bronchitis virus (IBV) was first isolated in Australia in 1962. Ongoing surveillance and characterization of Australian IBVs have shown that they have evolved separately from strains found throughout the rest of the world, resulting in the evolution of a range of unique strains and changes in the dominant wild-type strains, affecting tissue tropism, pathogenicity, antigenicity, and gene arrangement. Between 1961 and 1976 highly nephropathogenic genotype GI-5 and GI-6 strains, causing mortalities of 40% to 100%, predominated, while strains causing mainly respiratory disease, with lower mortality rates, have predominated since then. Since 1988, viruses belonging to two distinct and novel genotypes, GIII and GV, have been detected. The genome organization of the GIII strains has not been seen in any other gammacoronavirus. Mutations that emerged soon after the introduction of vaccination, incursion of strains with a novel lineage from unknown sources, recombination between IBVs from different genetic lineages, and gene translocations and deletions have contributed to an increasingly complex IBV population. These processes and the consequences of this variation for the biology of these viruses provide an insight into the evolution of endemic coronaviruses during their control by vaccination and may provide a better understanding of the potential for evolution of other coronaviruses, including SARS-CoV-2. Furthermore, the continuing capacity of attenuated IBV vaccines developed over 40 years ago to provide protection against viruses in the same genetic lineage provides some assurance that coronavirus vaccines developed to control other coronaviruses may continue to be effective for an extended period.


Assuntos
Evolução Biológica , Galinhas , Infecções por Coronaviridae/veterinária , Vírus da Bronquite Infecciosa/fisiologia , Doenças das Aves Domésticas/virologia , Animais , Variação Antigênica , Austrália/epidemiologia , Infecções por Coronaviridae/epidemiologia , Infecções por Coronaviridae/prevenção & controle , Infecções por Coronaviridae/virologia , Evolução Molecular , Variação Genética , Vírus da Bronquite Infecciosa/classificação , Vírus da Bronquite Infecciosa/genética , Vírus da Bronquite Infecciosa/imunologia , Fenótipo , Filogenia , Doenças das Aves Domésticas/epidemiologia , Doenças das Aves Domésticas/prevenção & controle , Vacinas Virais
4.
J Vet Diagn Invest ; 33(3): 457-468, 2021 May.
Artigo em Inglês | MEDLINE | ID: mdl-33739188

RESUMO

Every day, thousands of samples from diverse populations of animals are submitted to veterinary diagnostic laboratories (VDLs) for testing. Each VDL has its own laboratory information management system (LIMS), with processes and procedures to capture submission information, perform laboratory tests, define the boundaries of test results (i.e., positive or negative), and report results, in addition to internal business and accounting applications. Enormous quantities of data are accumulated and stored within VDL LIMSs. There is a need for platforms that allow VDLs to exchange and share portions of laboratory data using standardized, reliable, and sustainable information technology processes. Here we report concepts and applications for standardization and aggregation of data from swine submissions to multiple VDLs to detect and monitor porcine enteric coronaviruses by RT-PCR. Oral fluids, feces, and fecal swabs were the specimens submitted most frequently for enteric coronavirus testing. Statistical algorithms were used successfully to scan and monitor the overall and state-specific percentage of positive submissions. Major findings revealed a consistently recurrent seasonal pattern, with the highest percentage of positive submissions detected during December-February for porcine epidemic diarrhea virus, porcine deltacoronavirus, and transmissible gastroenteritis virus (TGEV). After 2014, very few submissions tested positive for TGEV. Monitoring VDL data proactively has the potential to signal and alert stakeholders early of significant changes from expected detection. We demonstrate the importance of, and applications for, data organized and aggregated by using LOINC and SNOMED CTs, as well as the use of customized messaging to allow inter-VDL exchange of information.


Assuntos
Infecções por Coronaviridae/veterinária , Coronaviridae/isolamento & purificação , Laboratórios/normas , Doenças dos Suínos/virologia , Animais , Teste para COVID-19/veterinária , Infecções por Coronaviridae/diagnóstico , Infecções por Coronaviridae/virologia , Surtos de Doenças , Fezes/virologia , Padrões de Referência , Estações do Ano , Suínos , Doenças dos Suínos/diagnóstico
5.
Transbound Emerg Dis ; 68(3): 1097-1110, 2021 May.
Artigo em Inglês | MEDLINE | ID: mdl-32799433

RESUMO

COVID-19 is a highly contagious disease caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). It has rapidly spread to 216 countries and territories since first outbreak in December of 2019, posing a substantial economic losses and extraordinary threats to the public health worldwide. Although bats have been suggested as the natural host of SARS-CoV-2, transmission chains of this virus, role of animals during cross-species transmission, and future concerns remain unclear. Diverse animal coronaviruses have extensively been studied since the discovery of avian coronavirus in 1930s. The current article comprehensively reviews and discusses the current understanding about animal coronaviruses and SARS-CoV-2 for their emergence, transmission, zoonotic potential, alteration of tissue/host tropism, evolution, status of vaccines and surveillance. This study aims at providing guidance for control of COVID-19 and preventative strategies for possible future outbreaks of zoonotic coronavirus via cross-species transmission.


Assuntos
COVID-19/virologia , Infecções por Coronaviridae/veterinária , Coronavirus/classificação , SARS-CoV-2/genética , Animais , Infecções por Coronaviridae/virologia , Humanos
6.
Med Sci (Paris) ; 36(8-9): 783-796, 2020.
Artigo em Francês | MEDLINE | ID: mdl-32773024

RESUMO

SARS-CoV-2 is a new human coronavirus (CoV), which emerged in People's Republic of China at the end of 2019 and is responsible for the global Covid-19 pandemic that caused more than 540 000 deaths in six months. Understanding the origin of this virus is an important issue and it is necessary to determine the mechanisms of its dissemination in order to be able to contain new epidemics. Based on phylogenetic inferences, sequence analysis and structure-function relationships of coronavirus proteins, informed by the knowledge currently available, we discuss the different scenarios evoked to account for the origin - natural or synthetic - of the virus. On the basis of currently available data, it is impossible to determine whether SARS-CoV-2 is the result of a natural zoonotic emergence or an accidental escape from experimental strains. Regardless of its origin, the study of the evolution of the molecular mechanisms involved in the emergence of this pandemic virus is essential to develop therapeutic and vaccine strategies.


TITLE: Retrouver les origines du SARS-CoV-2 dans les phylogénies de coronavirus. ABSTRACT: Le SARS-CoV-2 est un nouveau coronavirus (CoV) humain. Il a émergé en Chine fin 2019 et est responsable de la pandémie mondiale de Covid-19 qui a causé plus de 540 000 décès en six mois. La compréhension de l'origine de ce virus est une question importante et il est nécessaire de déterminer les mécanismes de sa dissémination afin de pouvoir se prémunir de nouvelles épidémies. En nous fondant sur des inférences phylogénétiques, l'analyse des séquences et les relations structure-fonction des protéines de coronavirus, éclairées par les connaissances actuellement disponibles, nous discutons les différents scénarios évoqués pour rendre compte de l'origine - naturelle ou synthétique - du virus.


Assuntos
Betacoronavirus/genética , Doenças Transmissíveis Emergentes/virologia , Infecções por Coronavirus/virologia , Coronavirus/classificação , Evolução Molecular , Pandemias , Filogenia , Pneumonia Viral/virologia , RNA Viral/genética , Sequência de Aminoácidos , Animais , Betacoronavirus/classificação , Betacoronavirus/isolamento & purificação , Derramamento de Material Biológico , COVID-19 , China/epidemiologia , Infecções por Coronaviridae/transmissão , Infecções por Coronaviridae/veterinária , Infecções por Coronaviridae/virologia , Infecções por Coronavirus/epidemiologia , Infecções por Coronavirus/transmissão , Reservatórios de Doenças , Mutação com Ganho de Função , Genoma Viral , HIV/genética , Especificidade de Hospedeiro , Humanos , Mamíferos/virologia , Pneumonia Viral/epidemiologia , Pneumonia Viral/transmissão , Vírus Reordenados/genética , SARS-CoV-2 , Alinhamento de Sequência , Homologia de Sequência de Aminoácidos , Glicoproteína da Espícula de Coronavírus/química , Glicoproteína da Espícula de Coronavírus/genética , Glicoproteína da Espícula de Coronavírus/fisiologia , Zoonoses
7.
Virulence ; 11(1): 707-718, 2020 01 01.
Artigo em Inglês | MEDLINE | ID: mdl-32490723

RESUMO

With the outbreak of the recent severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) in 2019, coronaviruses have become a global research hotspot in the field of virology. Coronaviruses mainly cause respiratory and digestive tract diseases, several coronaviruses are responsible for porcine diarrhea, such as porcine epidemic diarrhea virus (PEDV), porcine deltacoronavirus (PDCoV), and emerging swine acute diarrhea syndrome coronavirus (SADS-CoV). Those viruses have caused huge economic losses and are considered as potential public health threats. Porcine torovirus (PToV) and coronaviruses, sharing similar genomic structure and replication strategy, belong to the same order Nidovirales. Here, we developed a multiplex TaqMan-probe-based real-time PCR for the simultaneous detection of PEDV, PDCoV, PToV, and SADS-CoV for the first time. Specific primers and TaqMan fluorescent probes were designed targeting the ORF1a region of PDEV, PToV, and SADS-CoV and the ORF1b region of PDCoV. The method showed high sensitivity and specificity, with a detection limit of 1 × 102 copies/µL for each pathogen. A total of 101 clinical swine samples with signs of diarrhea were analyzed using this method, and the result showed good consistency with conventional reverse transcription PCR (RT-PCR). This method improves the efficiency for surveillance of these emerging and reemerging swine enteric viruses and can help reduce economic losses to the pig industry, which also benefits animal and public health.


Assuntos
Doenças Transmissíveis Emergentes/veterinária , Infecções por Coronaviridae/veterinária , Coronaviridae/isolamento & purificação , Reação em Cadeia da Polimerase , Doenças dos Suínos/diagnóstico , Animais , Coinfecção/diagnóstico , Coinfecção/veterinária , Doenças Transmissíveis Emergentes/diagnóstico , Coronaviridae/genética , Infecções por Coronaviridae/diagnóstico , Diarreia/diagnóstico , Diarreia/veterinária , Fases de Leitura Aberta/genética , Reação em Cadeia da Polimerase/normas , RNA Viral/genética , Reprodutibilidade dos Testes , Sensibilidade e Especificidade , Suínos
8.
Vet Microbiol ; 237: 108398, 2019 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-31585653

RESUMO

Feline infectious peritonitis (FIP) is a highly fatal disease caused by a virulent feline coronavirus in domestic and wild cats. We have previously reported the synthesis of potent coronavirus 3C-like protease (3CLpro) inhibitors and the efficacy of a protease inhibitor, GC376, in client-owned cats with FIP. In this study, we studied the effect of the amino acid changes in 3CLpro of feline coronavirus from a feline patient who received antiviral treatment for prolonged duration. We generated recombinant 3CLpro containing the identified amino acid changes (N25S, A252S or K260 N) and determined their susceptibility to protease inhibitors in the fluorescence resonance energy transfer assay. The assay showed that N25S in 3CLpro confers a small change (up to 1.68-fold increase in the 50% inhibitory concentration) in susceptibility to GC376, but other amino acid changes do not affect susceptibility. Modelling of 3CLpro carrying the amino acid changes was conducted to probe the structural basis for these findings. The results of this study may explain the observed absence of clinical resistance to the long-term antiviral treatment in the patients.


Assuntos
Doenças do Gato/virologia , Infecções por Coronaviridae/veterinária , Coronavirus Felino/enzimologia , Peritonite Infecciosa Felina/complicações , Inibidores de Proteases/uso terapêutico , Pirrolidinas/uso terapêutico , Sequência de Aminoácidos , Animais , Sequência de Bases , Sítios de Ligação , Gatos , Infecções por Coronaviridae/tratamento farmacológico , Infecções por Coronaviridae/virologia , Masculino , Modelos Moleculares , Inibidores de Proteases/farmacologia , Conformação Proteica , Pirrolidinas/farmacologia , RNA Viral , Alinhamento de Sequência , Ácidos Sulfônicos , Proteínas Virais/química , Proteínas Virais/metabolismo
9.
J Microbiol Biotechnol ; 29(11): 1817-1829, 2019 Nov 28.
Artigo em Inglês | MEDLINE | ID: mdl-31546302

RESUMO

Porcine deltacoronavirus (PDCoV) is an emerging swine enteric coronavirus that causes diarrhea in neonatal piglets. Like other coronaviruses, PDCoV encodes at least three accessory or species-specific proteins; however, the biological roles of these proteins in PDCoV replication remain undetermined. As a first step toward understanding the biology of the PDCoV accessory proteins, we established a stable porcine cell line constitutively expressing the PDCoV NS7 protein in order to investigate the functional characteristics of NS7 for viral replication. Confocal microscopy and subcellular fractionation revealed that the NS7 protein was extensively distributed in the mitochondria. Proteomic analysis was then conducted to assess the expression dynamics of the host proteins in the PDCoV NS7-expressing cells. Highresolution two-dimensional gel electrophoresis initially identified 48 protein spots which were differentially expressed in the presence of NS7. Seven of these spots, including two upregulated and five down-regulated protein spots, showed statistically significant alterations, and were selected for subsequent protein identification. The affected cellular proteins identified in this study were classified into functional groups involved in various cellular processes such as cytoskeleton networks and cell communication, metabolism, and protein biosynthesis. A substantial down-regulation of α-actinin-4 was confirmed in NS7-expressing and PDCoV-infected cells. These proteomic data will provide insights into the understanding of specific cellular responses to the accessory protein during PDCoV infection.


Assuntos
Infecções por Coronaviridae/veterinária , Coronaviridae/fisiologia , Doenças dos Suínos/virologia , Proteínas Virais Reguladoras e Acessórias/metabolismo , Actinina/metabolismo , Animais , Linhagem Celular , Coronaviridae/genética , Coronaviridae/metabolismo , Infecções por Coronaviridae/virologia , Interações Hospedeiro-Patógeno , Mitocôndrias/metabolismo , Proteômica , Suínos , Proteínas Virais Reguladoras e Acessórias/genética
10.
J Virol ; 92(15)2018 08 01.
Artigo em Inglês | MEDLINE | ID: mdl-29769348

RESUMO

The emergence of Middle East respiratory syndrome showed once again that coronaviruses (CoVs) in animals are potential source for epidemics in humans. To explore the diversity of deltacoronaviruses in animals in the Middle East, we tested fecal samples from 1,356 mammals and birds in Dubai, The United Arab Emirates. Four novel deltacoronaviruses were detected from eight birds of four species by reverse transcription-PCR (RT-PCR): FalCoV UAE-HKU27 from a falcon, HouCoV UAE-HKU28 from a houbara bustard, PiCoV UAE-HKU29 from a pigeon, and QuaCoV UAE-HKU30 from five quails. Complete genome sequencing showed that FalCoV UAE-HKU27, HouCoV UAE-HKU28, and PiCoV UAE-HKU29 belong to the same CoV species, suggesting recent interspecies transmission between falcons and their prey, houbara bustards and pigeons, possibly along the food chain. Western blotting detected specific anti-FalCoV UAE-HKU27 antibodies in 33 (75%) of 44 falcon serum samples, supporting genuine infection in falcons after virus acquisition. QuaCoV UAE-HKU30 belongs to the same CoV species as porcine coronavirus HKU15 (PorCoV HKU15) and sparrow coronavirus HKU17 (SpCoV HKU17), discovered previously from swine and tree sparrows, respectively, supporting avian-to-swine transmission. Recombination involving the spike protein is common among deltacoronaviruses, which may facilitate cross-species transmission. FalCoV UAE-HKU27, HouCoV UAE-HKU28, and PiCoV UAE-HKU29 originated from recombination between white-eye coronavirus HKU16 (WECoV HKU16) and magpie robin coronavirus HKU18 (MRCoV HKU18), QuaCoV UAE-HKU30 from recombination between PorCoV HKU15/SpCoV HKU17 and munia coronavirus HKU13 (MunCoV HKU13), and PorCoV HKU15 from recombination between SpCoV HKU17 and bulbul coronavirus HKU11 (BuCoV HKU11). Birds in the Middle East are hosts for diverse deltacoronaviruses with potential for interspecies transmission.IMPORTANCE During an attempt to explore the diversity of deltacoronaviruses among mammals and birds in Dubai, four novel deltacoronaviruses were detected in fecal samples from eight birds of four different species: FalCoV UAE-HKU27 from a falcon, HouCoV UAE-HKU28 from a houbara bustard, PiCoV UAE-HKU29 from a pigeon, and QuaCoV UAE-HKU30 from five quails. Genome analysis revealed evidence of recent interspecies transmission between falcons and their prey, houbara bustards and pigeons, possibly along the food chain, as well as avian-to-swine transmission. Recombination, which is known to occur frequently in some coronaviruses, was also common among these deltacoronaviruses and occurred predominantly at the spike region. Such recombination, involving the receptor binding protein, may contribute to the emergence of new viruses capable of infecting new hosts. Birds in the Middle East are hosts for diverse deltacoronaviruses with potential for interspecies transmission.


Assuntos
Doenças das Aves , Aves/virologia , Infecções por Coronaviridae , Coronavirus , Sequenciamento de Nucleotídeos em Larga Escala , Suínos/virologia , Animais , Doenças das Aves/genética , Doenças das Aves/transmissão , Infecções por Coronaviridae/genética , Infecções por Coronaviridae/transmissão , Infecções por Coronaviridae/veterinária , Coronavirus/classificação , Coronavirus/genética , Coronavirus/isolamento & purificação , Coronavirus/patogenicidade , Arábia Saudita
11.
Virology ; 502: 33-38, 2017 02.
Artigo em Inglês | MEDLINE | ID: mdl-27984784

RESUMO

Porcine deltacoronavirus (PDCoV) causes acute enteric disease and mortality in seronegative neonatal piglets. Previously we have demonstrated that PDCoV infection suppresses the production of interferon-beta (IFN-ß), while the detailed mechanisms are poorly understood. Here, we demonstrate that nonstructural protein 5 (nsp5) of PDCoV, the 3C-like protease, significantly inhibits Sendai virus (SEV)-induced IFN-ß production by targeting the NF-κB essential modulator (NEMO), confirmed by the diminished function of NEMO cleaved by PDCoV. The PDCoV nsp5 cleavage site in the NEMO protein was identified as glutamine 231, and was identical to the porcine epidemic diarrhea virus nsp5 cleavage site, revealing the likelihood of a common target in NEMO for coronaviruses. Furthermore, this cleavage impaired the ability of NEMO to activate the IFN response and downstream signaling. Taken together, our findings reveal PDCoV nsp5 to be a newly identified IFN antagonist and enhance the understanding of immune evasion by deltacoronaviruses.


Assuntos
Infecções por Coronaviridae/veterinária , Coronaviridae/enzimologia , Cisteína Endopeptidases/metabolismo , Quinase I-kappa B/metabolismo , Interferon beta/metabolismo , Doenças dos Suínos/enzimologia , Proteínas não Estruturais Virais/metabolismo , Animais , Coronaviridae/genética , Infecções por Coronaviridae/enzimologia , Infecções por Coronaviridae/metabolismo , Infecções por Coronaviridae/virologia , Proteases 3C de Coronavírus , Cisteína Endopeptidases/genética , Interações Hospedeiro-Patógeno , Quinase I-kappa B/genética , Interferon beta/genética , Processamento de Proteína Pós-Traducional , Suínos , Doenças dos Suínos/genética , Doenças dos Suínos/metabolismo , Doenças dos Suínos/virologia , Proteínas não Estruturais Virais/genética
12.
Prev Vet Med ; 135: 87-94, 2016 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-27931933

RESUMO

Porcine epidemic diarrhea virus (PEDV) has caused economic losses in the Americas, Asia and Europe in recent years. Reliable serological assays are essential for epidemiological studies and vaccine evaluation. The objective of this study was to compare the ability of five enzyme-linked immunosorbent assays (ELISAs) to detect antibodies against different PEDV strains in pig serum. A total of 732 serum samples from North American or European pigs were tested. Samples included experimental samples from pigs infected with classical (G1a PEDV) or variant genogroup 1 PEDV (G1b PEDV), pandemic genogroup 2 PEDV (G2b PEDV) or non-infected controls. Field samples from herds with confirmed or unknown PEDV exposure were also used. Three indirect ELISAs based on G2b antigens (ELISAs 1, 2 and 3), a competitive ELISA based on the G2b antigen (ELISA 4) and a competitive ELISA based on the G1a antigen (ELISA 5) were compared. Overall, the tests had a moderate agreement (κ=0.61). G1a PEDV infected pigs were earliest detected by ELISA 3, G1b PEDV infected pigs were earliest detected by ELISAs 4 and 5 and the performance of all tests was similar for the G2b PEDV group. ELISA 1 showed the overall lowest detection on experimentally and field derived samples. Diagnostic sensitivity and specificity with a 95% probability interval were estimated to be 68.2% (62.1-74.4%) and 97.5% (95.2-99.0%) for ELISA 1, 73.7% (71.5-79.6%) and 98.4% (96.6-99.5%) for ELISA 2, 86.2% (81.1-90.6%) and 91.6% (87.7-94.8%) for ELISA 3, 78.3% (72.8-83.5%) and 99.7% (98.2-100%) for ELISA 4, and 93.5% (90.3-96.0%) and 91.2% (83.8-97.9%) for ELISA 5. Differences in detection among assays seem to be more related to intrinsic factors of an assay than to the PEDV antigen used.


Assuntos
Anticorpos Antivirais/sangue , Infecções por Coronaviridae/veterinária , Técnicas e Procedimentos Diagnósticos/veterinária , Ensaio de Imunoadsorção Enzimática/veterinária , Vírus da Diarreia Epidêmica Suína/isolamento & purificação , Doenças dos Suínos/diagnóstico , Animais , Infecções por Coronaviridae/diagnóstico , Infecções por Coronaviridae/virologia , Dinamarca , Itália , Sensibilidade e Especificidade , Suínos , Doenças dos Suínos/virologia , Estados Unidos
13.
Vet Microbiol ; 193: 28-35, 2016 Sep 25.
Artigo em Inglês | MEDLINE | ID: mdl-27599927

RESUMO

Population of wild boar is increasing in the whole Europe, the animals migrate close to human habitats which greatly increases the possibility of natural transmission between domestic animals or humans and wild boars. The aim of the study was to estimate in population of free-living wild boar in the Czech Republic the prevalence of enteric viral pathogens, namely rotavirus groups A and C (RVA and RVC), porcine reproductive and respiratory syndrome virus (PRRSV), and members of family Coronaviridae (transmissible gastroenteritis virus - TGEV, porcine epidemic diarrhea virus - PEDV, porcine respiratory coronavirus - PRCV, and porcine hemagglutination encephalomyelitis virus - PHEV) and Picornaviridae,(teschovirus A - PTV, sapelovirus A - PSV, and enterovirus G - EV-G). In our study, stool samples from 203 wild boars culled during hunting season 2014-2015 (from October to January) were examined by RT-PCR. RVA was detected in 2.5% of tested samples. Nucleotide analysis of VP7, VP4, and VP6 genes revealed that four RVA strains belong to G4P[25]I1, G4P[6]I5, G11P[13]I5, and G5P[13]I5 genotypes and phylogenetic analysis suggested close relation to porcine and human RVAs. The prevalence of RVC in wild boar population reached 12.8%, PTV was detected in 20.2%, PSV in 8.9%, and EV-G in 2.5% of samples. During our study no PRRSV or coronaviruses were detected. Our study provides the first evidence of RVC prevalence in wild boars and indicates that wild boars might contribute to the genetic variability of RVA and also serve as an important reservoir of other enteric viruses.


Assuntos
Infecções por Coronaviridae/veterinária , Infecções por Picornaviridae/veterinária , Infecções por Rotavirus/veterinária , Rotavirus/isolamento & purificação , Doenças dos Suínos/virologia , Animais , Antígenos Virais/genética , Proteínas do Capsídeo/genética , Coronaviridae/genética , Coronaviridae/isolamento & purificação , Infecções por Coronaviridae/epidemiologia , Infecções por Coronaviridae/virologia , República Tcheca/epidemiologia , Reservatórios de Doenças , Fezes/virologia , Feminino , Genótipo , Humanos , Masculino , Filogenia , Picornaviridae/genética , Picornaviridae/isolamento & purificação , Infecções por Picornaviridae/epidemiologia , Infecções por Picornaviridae/virologia , Rotavirus/genética , Infecções por Rotavirus/epidemiologia , Infecções por Rotavirus/virologia , Sus scrofa , Suínos , Doenças dos Suínos/epidemiologia
14.
Virol J ; 13: 136, 2016 08 05.
Artigo em Inglês | MEDLINE | ID: mdl-27496131

RESUMO

BACKGROUND: Following the initial isolation of porcine deltacoronavirus (PDCoV) from pigs with diarrheal disease in the United States in 2014, the virus has been detected on swine farms in some provinces of China. To date, little is known about the molecular epidemiology of PDCoV in southern China where major swine production is operated. RESULTS: To investigate the prevalence of PDCoV in this region and compare its activity to other enteric disease of swine caused by porcine epidemic diarrhea virus (PEDV), transmissible gastroenteritis coronavirus (TGEV), and porcine rotavirus group C (Rota C), 390 fecal samples were collected from swine of various ages from 15 swine farms with reported diarrhea. Fecal samples were tested by reverse transcription-PCR (RT-PCR) that targeted PDCoV, PEDV, TGEV, and Rota C, respectively. PDCoV was detected exclusively from nursing piglets with an overall prevalence of approximate 1.28 % (5/390), not in suckling and fattening piglets. Interestingly, all of PDCoV-positive samples were from 2015 rather than 2012-2014. Despite a low detection rate, PDCoV emerged in each province/region of southern China. In addition, compared to TGEV (1.54 %, 5/390) or Rota C (1.28 %, 6/390), there were highly detection rates of PEDV (22.6 %, 88/390) in those samples. Notably, all five PDCoV-positive piglets were co-infected by PEDV. Furthermore, phylogenetic analysis of spike (S) and nucleocapsid (N) gene sequences of PDCoVs revealed that currently circulating PDCoVs in southern China were more closely related to other Chinese strains of PDCoVs than to those reported in United States, South Korea and Thailand. CONCLUSIONS: This study demonstrated that PDCoV was present in southern China despite the low prevalence, and supported an evolutionary theory of geographical clustering of PDCoVs.


Assuntos
Infecções por Coronaviridae/veterinária , Coronaviridae/isolamento & purificação , Fezes/virologia , Doenças dos Suínos/virologia , Animais , China/epidemiologia , Coronaviridae/classificação , Coronaviridae/genética , Infecções por Coronaviridae/epidemiologia , Infecções por Coronaviridae/virologia , Filogenia , Análise de Sequência de DNA , Suínos , Proteínas Virais/genética
15.
Virus Res ; 226: 142-151, 2016 12 02.
Artigo em Inglês | MEDLINE | ID: mdl-27397100

RESUMO

Porcine enteric coronaviruses (CoVs) cause severe disease in the porcine herds worldwide, leading to important economic losses. Despite the knowledge of these viruses since the 1970s, vaccination strategies have not been implemented, leading to continuous re-emergence of novel virulent strains. Live attenuated vaccines historically have been the most efficient. We consider that the new trend is the development of recombinant vaccines by using reverse genetics systems to engineer attenuated viruses, which could be used as effective and safe modified live vaccine candidates. To this end, host cell signaling pathways influencing porcine CoV virulence should be identified. Similarly, the identity of viral proteins involved in the modulation of host cell pathways influencing CoV pathogenesis should be analyzed. With this information, and using reverse genetics systems, it is possible to design viruses with modifications in the viral proteins acting as virulence factors, which may lead to attenuated viruses and, therefore, vaccine candidates. In addition, novel antiviral drugs may be developed once the host cell pathways and the molecular mechanism affecting porcine CoV replication and virulence are known. This review is focused in the host cell responses to enteric porcine CoV infection and the viral proteins involved in pathogenesis.


Assuntos
Infecções por Coronaviridae/veterinária , Coronavirus/imunologia , Coronavirus/patogenicidade , Doenças dos Suínos/imunologia , Doenças dos Suínos/virologia , Vacinas Virais/imunologia , Fatores de Virulência/imunologia , Animais , Antígenos Virais/genética , Antígenos Virais/imunologia , Coronavirus/genética , Interações Hospedeiro-Patógeno/imunologia , Imunidade Inata , Suínos , Doenças dos Suínos/metabolismo , Doenças dos Suínos/prevenção & controle , Proteínas Virais/genética , Proteínas Virais/imunologia , Vacinas Virais/genética , Fatores de Virulência/genética
16.
Avian Dis ; 60(2): 411-2, 2016 06.
Artigo em Inglês | MEDLINE | ID: mdl-27309279

RESUMO

Viruses within the Coronaviridae family show variations within their genome sequences, especially within the major structural protein, the Spike (S) glycoprotein gene. Therefore, many different antigenic forms, serotypes, or variant strains of avian coronaviruses (AvCoV) exist worldwide. Only a few of them, the so called protectotypes, cross protect against different serotypes. New serotypes arise by recombination or spontaneous mutations. From time to time, antigenic virus variants appear which differ significantly from known serotypes. The result of this variability is an inconsistent nomenclature and classification of virus strains. Furthermore, there are currently no standard classification methods defined. Within the framework of the COST Action FA1207 "Towards control of avian coronaviruses: strategies for diagnosis, surveillance, and vaccination" (working groups "Molecular virology" and "Epidemiology"), we aimed at defining and developing a unified and internationally standardized nomenclature and classification of AvCoVs. We recommend the use of "CoV Genus/AvCov/host/country/specimen id/year" to refer to AvCoV strains.


Assuntos
Doenças das Aves/classificação , Infecções por Coronaviridae/veterinária , Coronaviridae/classificação , Animais , Doenças das Aves/virologia , Infecções por Coronaviridae/classificação , Infecções por Coronaviridae/virologia , Terminologia como Assunto
17.
BMC Vet Res ; 12: 95, 2016 Jun 08.
Artigo em Inglês | MEDLINE | ID: mdl-27277214

RESUMO

BACKGROUND: A novel porcine deltacoronavirus (PDCoV), also known as porcine coronavirus HKU15, was reported in China in 2012 and identified in the U.S. in early 2014. Since then, PDCoV has been identified in a number of U.S. states and linked with clinical disease including acute diarrhea and vomiting in the absence of other identifiable pathogens. Since PDCoV was just recently linked with clinical disease, few specific antibody-based reagents were available to assist in diagnosis of PDCoV and limited serological capabilities were available to detect an antibody response to this virus. Therefore, the overall objective of this project was to develop and validate selected diagnostic reagents and assays for PDCoV antigen and antibody detection. RESULTS: The nucleoprotein of PDCoV was expressed as a recombinant protein and purified for use as an antigen to immunize mice for polyclonal, hyperimmune sera and monoclonal antibody (mAb) production. The resulting mAbs were evaluated for use in fluorescent antibody staining methods to detect PDCoV infected cells following virus isolation attempts and for immunohistochemistry staining of intestinal tissues of infected pigs. The same antigen was used to develop serological tests to detect the antibody response to PDCoV in pigs following infection. Serum samples from swine herds with recent documentation of PDCoV infection and samples from expected naïve herds were used for initial assay optimization. The tests were optimized in a checkerboard fashion to reduce signal to noise ratios using samples of known status. Statistical analysis was performed to establish assay cutoff values and assess diagnostic sensitivities and specificities. At least 629 known negative serum samples and 311 known positive samples were evaluated for each assay. The enzyme linked immunosorbent assay (ELISA) showed diagnostic sensitivity (DSe) of 96.1% and diagnostic specificity (DSp) of 96.2%. The fluorescent microsphere immunoassay (FMIA) showed a DSe of 95.8% and DSp of 98.1%. Both ELISA and FMIA detected seroconversion of challenged pigs between 8-14 days post-infection (DPI). An indirect fluorescent antibody (IFA) test was also developed using cell culture adapted PDCoV for comparative purposes. CONCLUSION: These new, specific reagents and serological assays will allow for improved diagnosis of PDCoV. Since many aspects of PDCoV infection and transmission are still not fully understood, the reagents and assays developed in this project should provide valuable tools to help understand this disease and to aid in the control and surveillance of porcine deltacoronavirus outbreaks.


Assuntos
Anticorpos Monoclonais/imunologia , Anticorpos Antivirais/imunologia , Infecções por Coronaviridae/veterinária , Coronaviridae/imunologia , Ensaio de Imunoadsorção Enzimática/veterinária , Doenças dos Suínos/diagnóstico , Animais , Antígenos Virais/imunologia , Células Cultivadas , Infecções por Coronaviridae/diagnóstico , Infecções por Coronaviridae/imunologia , Reações Cruzadas , Ensaio de Imunoadsorção Enzimática/métodos , Técnica Indireta de Fluorescência para Anticorpo/métodos , Microesferas , Nucleoproteínas/imunologia , Dobramento de Proteína , Testes Sorológicos/métodos , Testes Sorológicos/veterinária , Suínos , Doenças dos Suínos/imunologia , Doenças dos Suínos/virologia
18.
Transbound Emerg Dis ; 63(3): 248-52, 2016 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-26968326

RESUMO

This study applied molecular-based method to investigate the presence of porcine deltacoronavirus (PDCoV) in 59 commercial pig farms in South Korea. The results of RT-PCR screening on a relatively large collection of faeces samples (n = 681) from January 2013 to March 2015 did not reveal the presence of PDCoV until the end of 2014. However, on March 2015, PDCoV-positive samples (SL2, SL5) were detected from SL swine farm in Gyeongbuk province. The phylogenetic trees based on the complete spike- and nucleocapsid protein-coding genes showed that SL2 and SL5 closely related to the US PDCoV strains rather than those in China. Thought Korean strains of PDCoV isolated in 2014 (KNU14.04) and in 2015 (SL2 and SL5) grouped within US PDCoV cluster, the reconstruction of ancestral amino acid changes suggested that they are different.


Assuntos
Infecções por Coronaviridae/veterinária , Coronaviridae/isolamento & purificação , Diarreia/veterinária , Doenças dos Suínos/epidemiologia , Animais , Coronaviridae/genética , Infecções por Coronaviridae/epidemiologia , Infecções por Coronaviridae/virologia , Diarreia/epidemiologia , Diarreia/virologia , Fezes/virologia , Feminino , Proteínas do Nucleocapsídeo/genética , Filogenia , Reação em Cadeia da Polimerase/veterinária , República da Coreia/epidemiologia , Análise de Sequência de DNA/veterinária , Glicoproteína da Espícula de Coronavírus/genética , Suínos , Doenças dos Suínos/virologia
19.
Proc Natl Acad Sci U S A ; 113(11): 3048-53, 2016 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-26976607

RESUMO

Outbreaks from zoonotic sources represent a threat to both human disease as well as the global economy. Despite a wealth of metagenomics studies, methods to leverage these datasets to identify future threats are underdeveloped. In this study, we describe an approach that combines existing metagenomics data with reverse genetics to engineer reagents to evaluate emergence and pathogenic potential of circulating zoonotic viruses. Focusing on the severe acute respiratory syndrome (SARS)-like viruses, the results indicate that the WIV1-coronavirus (CoV) cluster has the ability to directly infect and may undergo limited transmission in human populations. However, in vivo attenuation suggests additional adaptation is required for epidemic disease. Importantly, available SARS monoclonal antibodies offered success in limiting viral infection absent from available vaccine approaches. Together, the data highlight the utility of a platform to identify and prioritize prepandemic strains harbored in animal reservoirs and document the threat posed by WIV1-CoV for emergence in human populations.


Assuntos
Quirópteros/virologia , Doenças Transmissíveis Emergentes/virologia , Infecções por Coronaviridae/virologia , Coronaviridae/patogenicidade , Enzima de Conversão de Angiotensina 2 , Animais , Anticorpos Monoclonais/imunologia , Anticorpos Neutralizantes/imunologia , Anticorpos Antivirais/imunologia , Células Cultivadas , Chlorocebus aethiops , Coronaviridae/genética , Coronaviridae/imunologia , Coronaviridae/isolamento & purificação , Coronaviridae/fisiologia , Infecções por Coronaviridae/prevenção & controle , Infecções por Coronaviridae/transmissão , Infecções por Coronaviridae/veterinária , Reações Cruzadas , Encefalite Viral/virologia , Células Epiteliais/virologia , Especificidade de Hospedeiro , Humanos , Pulmão/citologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Transgênicos , Modelos Moleculares , Peptidil Dipeptidase A/genética , Peptidil Dipeptidase A/fisiologia , Mutação Puntual , Conformação Proteica , Receptores Virais/genética , Receptores Virais/fisiologia , Proteínas Recombinantes de Fusão/metabolismo , Coronavírus Relacionado à Síndrome Respiratória Aguda Grave/imunologia , Especificidade da Espécie , Glicoproteína da Espícula de Coronavírus/genética , Glicoproteína da Espícula de Coronavírus/fisiologia , Células Vero , Replicação Viral , Zoonoses
20.
PLoS One ; 11(1): e0146975, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-26771312

RESUMO

To trace the evolution of canine coronavirus (CCoV), 201 stool samples from diarrheic dogs in northeast China were subjected to reverse transcription-polymerase chain reactions (RT-PCRs) targeting the partial M and S genes of CCoV, followed by an epidemiological analysis. M gene RT-PCRs showed that 28.36% (57/201) of the samples were positive for CCoV; of the 57 positive samples, CCoV-I and CCoV-II accounted for 15.79% (9/57) and 84.21% (48/57), respectively. A sequence comparison of the partial M gene revealed nucleotide homologies of 88.4%-100% among the 57 CCoV strains, and 88.7%-96.2% identity between the 57 CCoV strains and the Chinese reference strain HF3. The CCoV-I and CCoV-II strains exhibited genetic diversity when compared with reference strains from China and other countries. The 57 CCoV strains exhibited high co-infection rates with canine kobuvirus (CaKV) (33.33%) and canine parvovirus-2 (CPV-2) (31.58%). The CCoV prevalence in diarrheic dogs differed significantly with immunization status, regions, seasons, and ages. Moreover, 28 S genes were amplified from the 57 CCoV-positive samples, including 26 CCoV-IIa strains, one CCoV-IIb strain, and one CCoV-I strain. A sequence comparison of the partial S gene revealed 86.3%-100% nucleotide identity among the 26 CCoV-IIa strains, and 89.6%-92.2% identity between the 26 CCoV-IIa strains and the Chinese reference strain V1. The 26 CCoV-IIa strains showed genetic diversity when compared with reference strains from China and other countries. Our data provide evidence that CCoV-I, CCoV-IIa, and CCoV-IIb strains co-circulate in the diarrhoetic dogs in northeast China, high co-infection rates with CaKV and CPV-2 were observed, and the CCoV-II strains exhibited high prevalence and genetic diversity.


Assuntos
Infecções por Coronaviridae/veterinária , Coronavirus Canino/genética , Doenças do Cão/epidemiologia , Doenças do Cão/virologia , Variação Genética/genética , Animais , China , Infecções por Coronaviridae/epidemiologia , Infecções por Coronaviridae/virologia , Cães
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA