Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
1.
Virology ; 592: 109996, 2024 04.
Artigo em Inglês | MEDLINE | ID: mdl-38301448

RESUMO

While vaccination and therapeutics for prevention/treatment of influenza are available, new strategies are needed to combat influenza disease in susceptible populations, particularly young children and newborns. Host associated microbiota play an important role in modulating the virulence of numerous pathogens, including the influenza A virus. In this study, we examined microbiome-influenza interactions in a neonatal piglet model system. The nasal microbiome of newborn piglets was longitudinally sampled before and after intranasal infection with recombinant viruses expressing hemagglutinins (HAs) derived from distinct zoonotic H1 subtypes. We found that viruses expressing different parental HAs manifested unique patterns of pathogenicity, and varied impacts on microbial community diversity. Despite these virus specific differences, a consistent microbial signature of viral infection was detected. Our results indicate that influenza A virus infection associates with the restructuring of nasal microbiome and such shifts in microbial diversity may contribute to outcomes of viral infection in neonatal piglets.


Assuntos
Vírus da Influenza A , Influenza Humana , Microbiota , Infecções por Orthomyxoviridae , Recém-Nascido , Criança , Animais , Humanos , Suínos , Pré-Escolar , Vírus da Influenza A/genética , Hemaglutininas
2.
mSphere ; 8(4): e0006723, 2023 08 24.
Artigo em Inglês | MEDLINE | ID: mdl-37409816

RESUMO

The global pandemic of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) poses a significant threat to public health. Besides humans, SARS-CoV-2 can infect several animal species. Highly sensitive and specific diagnostic reagents and assays are urgently needed for rapid detection and implementation of strategies for prevention and control of the infection in animals. In this study, we initially developed a panel of monoclonal antibodies (mAbs) against SARS-CoV-2 nucleocapsid protein. To detect SARS-CoV-2 antibodies in a broad spectrum of animal species, an mAb-based blocking enzyme-linked immunosorbent assay (bELISA) was developed. Test validation using a set of animal serum samples with known infection status obtained an optimal percentage of inhibition cut-off value of 17.6% with diagnostic sensitivity of 97.8% and diagnostic specificity of 98.9%. The assay demonstrates high repeatability as determined by a low coefficient of variation (7.23%, 4.89%, and 3.16%) between-runs, within-run, and within-plate, respectively. Testing of samples collected over time from experimentally infected cats showed that the bELISA was able to detect seroconversion as early as 7 days post-infection. Subsequently, the bELISA was applied for testing pet animals with coronavirus disease 2019 (COVID-19)-like symptoms and specific antibody responses were detected in two dogs. The panel of mAbs generated in this study provides a valuable tool for SARS-CoV-2 diagnostics and research. The mAb-based bELISA provides a serological test in aid of COVID-19 surveillance in animals. IMPORTANCE Antibody tests are commonly used as a diagnostic tool for detecting host immune response following infection. Serology (antibody) tests complement nucleic acid assays by providing a history of virus exposure, no matter symptoms developed from infection or the infection was asymptomatic. Serology tests for COVID-19 are in high demand, especially when the vaccines become available. They are important to determine the prevalence of the viral infection in a population and identify individuals who have been infected or vaccinated. ELISA is a simple and practically reliable serological test, which allows high-throughput implementation in surveillance studies. Several COVID-19 ELISA kits are available. However, they are mostly designed for human samples and species-specific secondary antibody is required for indirect ELISA format. This paper describes the development of an all species applicable monoclonal antibody (mAb)-based blocking ELISA to facilitate the detection and surveillance of COVID-19 in animals.


Assuntos
COVID-19 , SARS-CoV-2 , Humanos , Animais , Cães , COVID-19/diagnóstico , Anticorpos Monoclonais , Sensibilidade e Especificidade , Ensaio de Imunoadsorção Enzimática
3.
bioRxiv ; 2023 Mar 13.
Artigo em Inglês | MEDLINE | ID: mdl-36993307

RESUMO

The global pandemic of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) poses a significant threat to public health. Besides humans, SARS-CoV-2 can infect several animal species. Highly sensitive and specific diagnostic reagents and assays are urgently needed for rapid detection and implementation of strategies for prevention and control of the infection in animals. In this study, we initially developed a panel of monoclonal antibodies (mAbs) against SARS-CoV-2 nucleocapsid (N) protein. To detect SARS-CoV-2 antibodies in a broad spectrum of animal species, a mAb-based bELISA was developed. Test validation using a set of animal serum samples with known infection status obtained an optimal percentage of inhibition (PI) cut-off value of 17.6% with diagnostic sensitivity of 97.8% and diagnostic specificity of 98.9%. The assay demonstrates high repeatability as determined by a low coefficient of variation (7.23%, 6.95%, and 5.15%) between-runs, within-run, and within-plate, respectively. Testing of samples collected over time from experimentally infected cats showed that the bELISA was able to detect seroconversion as early as 7 days post-infection. Subsequently, the bELISA was applied for testing pet animals with COVID-19-like symptoms and specific antibody responses were detected in two dogs. The panel of mAbs generated in this study provides a valuable tool for SARS-CoV-2 diagnostics and research. The mAb-based bELISA provides a serological test in aid of COVID-19 surveillance in animals. IMPORTANCE: Antibody tests are commonly used as a diagnostic tool for detecting host immune response following infection. Serology (antibody) tests complement nucleic acid assays by providing a history of virus exposure, no matter symptoms developed from infection or the infection was asymptomatic. Serology tests for COVID-19 are in high demand, especially when the vaccines become available. They are important to determine the prevalence of the viral infection in a population and identify individuals who have been infected or vaccinated. ELISA is a simple and practically reliable serological test, which allows high-throughput implementation in surveillance studies. Several COVID-19 ELISA kits are available. However, they are mostly designed for human samples and species-specific secondary antibody is required for indirect ELISA format. This paper describes the development of an all species applicable monoclonal antibody (mAb)-based blocking ELISA to facilitate the detection and surveillance of COVID-19 in animals.

4.
Vaccines (Basel) ; 11(3)2023 Mar 14.
Artigo em Inglês | MEDLINE | ID: mdl-36992230

RESUMO

While it is well appreciated that maternal immunity can provide neonatal protection, the contribution of maternal vaccination toward generating such immunity is not well characterized. In our previous work, we created a candidate influenza vaccine using our chimeric hemagglutinin (HA) construct, HA-129. The HA-129 was expressed as part of a whole-virus vaccine that was built on the A/swine/Texas/4199-2/98-H3N2 backbone to generate the recombinant virus TX98-129. The TX98-129 candidate vaccine has the ability to induce broadly protective immune responses against genetically diversified influenza viruses in both mice and nursery pigs. In the current study, we established a pregnant sow-neonate model to evaluate the maternal immunity induced by this candidate vaccine to protect pregnant sows and their neonatal piglets against influenza virus infection. In pregnant sows, the results consistently show that TX98-129 induced a robust immune response against the TX98-129 virus and the parental viruses that were used to construct HA-129. After challenge with a field strain of influenza A virus, a significant increase in antibody titers was observed in vaccinated sows at both 5 and 22 days post challenge (dpc). The challenge virus was detected at a low level in the nasal swab of only one vaccinated sow at 5 dpc. Evaluation of cytokine responses in blood and lung tissue showed that levels of IFN-α and IL-1ß were increased in the lung of vaccinated sows at 5 dpc, when compared to unvaccinated pigs. Further analysis of the T-cell subpopulation in PBMCs showed a higher ratio of IFN-γ-secreting CD4+CD8+ and CD8+ cytotoxic T cells in vaccinated sows at 22 dpc after stimulation with either challenge virus or vaccine virus. Finally, we used a neonatal challenge model to demonstrate that vaccine-induced maternal immunity can be passively transferred to newborn piglets. This was observed in the form of both increased antibody titers and deceased viral loads in neonates born from immunized sows. In summary, this study provides a swine model system to evaluate the impact of vaccination on maternal immunity and fetal/neonatal development.

5.
J Virol ; 96(17): e0096122, 2022 09 14.
Artigo em Inglês | MEDLINE | ID: mdl-36000850

RESUMO

Omicron (B.1.1.529) is the most recent SARS-CoV-2 variant of concern, which emerged in late 2021 and rapidly achieved global predominance by early 2022. In this study, we compared the infection dynamics, tissue tropism, and pathogenesis and pathogenicity of SARS-CoV-2 D614G (B.1), Delta (B.1.617.2), and Omicron BA.1.1 (B.1.1.529) variants in a highly susceptible feline model of infection. Although D614G- and Delta-inoculated cats became lethargic and showed increased body temperatures between days 1 and 3 postinfection (pi), Omicron-inoculated cats remained subclinical and, similar to control animals, gained weight throughout the 14-day experimental period. Intranasal inoculation of cats with D614G- and the Delta variants resulted in high infectious virus shedding in nasal secretions (up to 6.3 log10 TCID50.Ml-1), whereas strikingly lower level of viruses shedding (<3.1 log10 TCID50.Ml-1) was observed in Omicron-inoculated animals. In addition, tissue distribution of the Omicron variant was markedly reduced in comparison to the D614G and Delta variants, as evidenced by lower in situ viral RNA detection, in situ viral immunofluorescence staining, and viral loads in tissues on days 3, 5, and 14 pi. Nasal turbinate, trachea, and lung were the main-but not the only-sites of replication for all three viral variants. However, only scarce virus staining and lower viral titers suggest lower levels of viral replication in tissues from Omicron-infected animals. Notably, while D614G- and Delta-inoculated cats presented pneumonia, histologic examination of the lungs from Omicron-infected cats revealed mild to modest inflammation. Together, these results demonstrate that the Omicron variant BA.1.1 is less pathogenic than D614G and Delta variants in a highly susceptible feline model. IMPORTANCE The SARS-CoV-2 Omicron (B.1.1.529) variant of concern emerged in South Africa late in 2021 and rapidly spread across the world causing a significant increase in the number of infections. Importantly, this variant was also associated with an increased risk of reinfections. However, the number of hospitalizations and deaths due to COVID-19 did not follow the same trends. These early observations suggested effective protection conferred by immunizations and/or overall lower virulence of the highly mutated variant virus. In this study we present novel evidence demonstrating that the Omicron BA.1.1 variant of concern presents a lower pathogenicity when compared to D614G- or Delta variants in cats. Clinical, virological, and pathological evaluations revealed lower disease severity, viral replication, and lung pathology in Omicron-infected cats when compared with D614G and Delta variant inoculated animals, confirming that Omicron BA.1.1 is less pathogenic in a highly susceptible feline model of infection.


Assuntos
COVID-19/virologia , SARS-CoV-2 , Animais , Gatos , Modelos Animais de Doenças , Humanos , SARS-CoV-2/patogenicidade , Virulência , Replicação Viral
6.
bioRxiv ; 2022 Jun 16.
Artigo em Inglês | MEDLINE | ID: mdl-35734088

RESUMO

Omicron (B.1.1.529) is the most recent SARS-CoV-2 variant of concern (VOC), which emerged in late 2021 and rapidly achieved global predominance in early 2022. In this study, we compared the infection dynamics, tissue tropism and pathogenesis and pathogenicity of SARS-CoV-2 D614G (B.1), Delta (B.1.617.2) and Omicron BA.1.1 sublineage (B.1.1.529) variants in a highly susceptible feline model of infection. While D614G- and Delta-inoculated cats became lethargic, and showed increased body temperatures between days 1 and 3 post-infection (pi), Omicron-inoculated cats remained subclinical and, similar to control animals, gained weight throughout the 14-day experimental period. Intranasal inoculation of cats with D614G- and the Delta variants resulted in high infectious virus shedding in nasal secretions (up to 6.3 log10 TCID 50 .ml -1 ), whereas strikingly lower level of viruses shedding (<3.1 log10 TCID 50 .ml -1 ) was observed in Omicron-inoculated animals. In addition, tissue distribution of the Omicron variant was markedly reduced in comparison to the D614G and Delta variants, as evidenced by in situ viral RNA detection, in situ immunofluorescence, and quantification of viral loads in tissues on days 3, 5, and 14 pi. Nasal turbinate, trachea, and lung were the main - but not the only - sites of replication for all three viral variants. However, only scarce virus staining and lower viral titers suggest lower levels of viral replication in tissues from Omicron-infected animals. Notably, while D614G- and Delta-inoculated cats had severe pneumonia, histologic examination of the lungs from Omicron-infected cats revealed mild to modest inflammation. Together, these results demonstrate that the Omicron variant BA.1.1 is less pathogenic than D614G and Delta variants in a highly susceptible feline model. Author Summary: The SARS-CoV-2 Omicron (B.1.1.529) variant of concern (VOC) emerged in South Africa late in 2021 and rapidly spread across the world causing a significant increase in the number of infections. Importantly, this variant was also associated with an increased risk of reinfections. However, the number of hospitalizations and deaths due to COVID-19 did not follow the same trends. These early observations, suggested effective protection conferred by immunizations and/or overall lower virulence of the highly mutated variant virus. In this study we present novel evidence demonstrating that the Omicron BA.1.1 variant of concern (VOC) presents a lower pathogenicity when compared to D614G- or Delta variants in cats. Clinical, virological and pathological evaluations revealed lower disease severity, viral replication and lung pathology in Omicron-infected cats when compared to D614G and Delta variant inoculated animals, confirming that Omicron BA.1.1 is less pathogenic in a highly susceptible feline model of infection.

7.
Viruses ; 14(5)2022 05 18.
Artigo em Inglês | MEDLINE | ID: mdl-35632822

RESUMO

Porcine reproductive and respiratory syndrome virus (PRRSV) is one of the most economically important pathogens affecting the global swine industry. Vaccination is still a main strategy for PRRSV control; however, host factors associated with vaccine efficacy remain poorly understood. Growing evidence suggests that mucosa-associated microbiomes may play a role in the responses to vaccination. In this study, we investigated the effects of a killed virus vaccine on the gut microbiome diversity in pigs. Fecal microbial communities were longitudinally assessed in three groups of pigs (vaccinated/challenged with PRRSV, unvaccinated/challenged with PRRSV, and unvaccinated/unchallenged) before and after vaccination and after viral challenge. We observed significant interaction effects between viral challenge and vaccination on both taxonomic richness and community diversity of the gut microbiota. While some specific taxonomic alterations appear to be enhanced in vaccinated/challenged pigs, others appeared to be more consistent with the levels in control animals (unvaccinated/unchallenged), indicating that vaccination incompletely protects against viral impacts on the microbiome. The abundances of several microbial taxa were further determined to be correlated with the level of viral load and the amount of PRRSV reactive CD4+ and CD8+ T-cells. This study highlights the potential roles of gut microbiota in the response of pigs to vaccination, which may pave the road for the development of novel strategies to enhance vaccine efficacy.


Assuntos
Microbioma Gastrointestinal , Síndrome Respiratória e Reprodutiva Suína , Vírus da Síndrome Respiratória e Reprodutiva Suína , Vacinas Virais , Animais , Linfócitos T CD8-Positivos , Síndrome Respiratória e Reprodutiva Suína/prevenção & controle , Suínos , Vacinas Atenuadas , Vacinas de Produtos Inativados
8.
Virology ; 570: 107-116, 2022 05.
Artigo em Inglês | MEDLINE | ID: mdl-35398774

RESUMO

Porcine respirovirus 1 (PRV1) is widely spread in many countries. In this study, we isolated an emgerging PRV1 strain (KS17-258) from a US swine farm. A full-length genome sequence of the virus was obtained, and the mRNA editing mechanism utilized for the expression of V/W proteins by P gene was confirmed. The virus shares 91.3-98% nucleotide sequence identity with the other PRV1 genomes reported previously. Phylogenetic analysis showed that KS17-258 forms a clade with the other US isolates. Infectious clone of the KS17-258 isolate was constructed, which was further explored as a viral vector to express enhanced green fluorescent protein (EGFP). The expression cassette of EGFP in the recombinant virus remained stable for 10 passages in cell culture. The availability of PRV1 infectious clone provides an important tool for study the basic PRV1 replication mechanisms. It also provides a novel platform for potential development of vectored vaccines against swine diseases.


Assuntos
Respirovirus , Doenças dos Suínos , Animais , DNA Complementar/genética , Vetores Genéticos/genética , Genoma Viral , Filogenia , Respirovirus/genética , Suínos
9.
Transbound Emerg Dis ; 69(2): 753-763, 2022 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-33621429

RESUMO

Atypical porcine pestivirus (APPV), a highly divergent pestivirus, has a wide geographical distribution around the world. APPV is known to cause type A-II congenital tremors in newborn piglets. The main objective of this study is to access APPV prevalence in the US swine herds utilizing a newly developed quantitative real-time RT-PCR assay. Retrospective analysis of 1,785 samples revealed a 19.0% prevalence in Midwest swine herds over a period of three years (2016-2018). Among all clinical and field samples that were APPV positive, 82 samples (24.19%) were also positive for one or more swine viral pathogens. Two APPV US strains identified in this study demonstrated significant sequence diversity (~12% in full genome) compared to the first reported APPV strain from the United States in 2014. Of the two strains identified in this study, USA/023005/2016 is closer to two strains identified in Germany, and USA/047310/2017 shares more similarities with two US strains including Minnesota-1 and ISDVDL2014016573. Partial NS5B sequences (9127-9836 nt of the polyprotein gene) obtained from 54 APPV-positive samples revealed considerable sequence diversity, ranging from 85.8% to 100% nucleotide identity, within the US strains in samples from different geographic regions. Analysis of all US samples indicates high prevalence of APPV in Minnesota (37.35%), followed by Illinois (32.86%), Iowa (30.60%) and Kansas (21.89%). APPV was detected in 15.48% of samples assayed from 2017, slightly higher than that in 2016 (13.08%), but much lower than 2018 (28.77%). Among the various sample types tested, oral fluid samples had the highest prevalence and lowest average Ct value suggesting their suitability as a reliable diagnostic specimen for APPV detection. Overall, sequence variation among APPV strains and prevalence of the pathogen within the United States provides a basis for understanding the genetic diversity and molecular epidemiology of APPV in the US swine herds.


Assuntos
Infecções por Pestivirus , Pestivirus , Doenças dos Suínos , Animais , Variação Genética , Pestivirus/genética , Infecções por Pestivirus/veterinária , Filogenia , Prevalência , Estudos Retrospectivos , Suínos
10.
Pathogens ; 10(6)2021 Jun 17.
Artigo em Inglês | MEDLINE | ID: mdl-34204199

RESUMO

The incursion of African swine fever virus (ASFV) into Eurasia presents a threat to the world's swine industry. Highly sensitive and specific diagnostic assays are urgently needed for rapid detection during an outbreak, post-outbreak investigation, and disease surveillance. In this study, a highly specific and repeatable blocking ELISA (bELISA) was developed using a recombinant p30 protein as the antigen combined with biotinylated mAb against p30 as the detection antibody. Initial test validation included sera from 810 uninfected animals and 106 animals experimentally inoculated with ASFV or recombinant alphavirus/adenovirus expressing p30. Receiver operating characteristic (ROC) analysis of the data calculated an optimal percentage of inhibition (PI) cutoff value of 45.92%, giving a diagnostic sensitivity of 98.11% and diagnostic specificity of 99.42%. The coefficient of variation of an internal quality control serum was 6.81% for between runs, 6.71% for within run, and 6.14% for within plate. A time course study of infected pigs showed that bELISA was able to detect seroconversion as early as 7 days post-inoculation. Taken together, these results demonstrate that bELISA can be used as an alternative serological test for detecting ASFV infection.

11.
J Virol Methods ; 296: 114220, 2021 10.
Artigo em Inglês | MEDLINE | ID: mdl-34182037

RESUMO

Atypical Porcine Pestivirus (APPV) is reported as the etiologic agent for type AII congenital tremors in newborn piglets. Initial PCR-based diagnostic tests to detect APPV were designed based on the limited sequence information and are not capable of detecting the majority of APPV strains. A sensitive and reliable PCR-based diagnostic test is critical for accurate detection of APPV. In this study, a quantitative reverse transcription PCR (RT-qPCR) assay was developed for reliable detection of all currently known APPV strains. The assay design also included swine 18S rRNA gene as an internal control to monitor RNA extraction efficiency. Two APPV gene fragments, one each from NS5b and NS3, were cloned and used to determine the dynamic range of detection, linearity and analytical sensitivity/limit of detection (LOD). Both individual and multiplex assays (duplex and triplex) had correlation coefficients of >0.99 and PCR amplification efficiencies of >90 %. Comparison of detection limit and analytical sensitivity between individual, and multiplex assays indicated no inhibition of PCR sensitivity upon multiplexing. The detection limit for APPV target, based on analytical sensitivity, is 7.75 copies (NS5b) and 5.2 copies (NS3) per reaction. Assay specificity was verified by testing nucleic acids of other closely related pestiviruses and clinical samples that are positive for other common swine pathogens. Assay sensitivity was also assessed on synthesized gene fragments of the most divergent China strains. Testing 339 known APPV-positive and 202 negative clinical samples demonstrated a good diagnostic sensitivity and specificity. Data from six independent runs, including 5 replicates of three clinical samples with three Ct ranges, were utilized to assess inter-assay repeatability and intra-assay reproducibility. This analysis demonstrated intra-assay/inter-assay coefficients of variation of 0.71 % and 0.01 %, respectively, with a PCR efficiency of 92.71 % for the triplex assay. Testing of 1785 clinical samples revealed ∼19 % prevalence of APPV in the US swine herds and oral fluids demonstrates to be a reliable specimen for viral detection. This multiplex RT-qPCR assay offers a rapid and reliable detection of APPV in swine herds and serves as useful tool in APPV surveillance and epidemiological investigations.


Assuntos
Infecções por Pestivirus , Pestivirus , Doenças dos Suínos , Animais , Pestivirus/genética , Infecções por Pestivirus/diagnóstico , Infecções por Pestivirus/veterinária , Filogenia , Reprodutibilidade dos Testes , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Suínos , Doenças dos Suínos/diagnóstico , Tremor/diagnóstico , Tremor/veterinária
12.
Infect Genet Evol ; 92: 104866, 2021 08.
Artigo em Inglês | MEDLINE | ID: mdl-33872785

RESUMO

Atypical porcine pestivirus (APPV) is an emerging virus discovered in 2014 and it can cause congenital tremors in pigs. Molecular epidemiology serves as an essential tool in monitoring and controlling the disease. Virus epidemiology mainly relies on genome sequencing and phylogenetic characterization. Previous molecular epidemiology studies have been using different genes/regions for phylogeny, namely whole genome, Npro, and E2 coding sequences. However, with increasing number of APPV sequences available in GenBank, no systemic studies have been performed for detailed classification of APPV strains around the globe. The goal of this study is to propose a classification strategy or taxonomy of APPV strains at genotype, subgenotype, and isolate levels. A total of 76 whole genomes and 16 partial polyprotein coding sequences were analyzed for genetic variability and suitability of all individual genes for viral phylogenies. Our results revealed that, among all the viral genes, NS5a coding sequences were proved to be the most suitable alternative for tracing APPV strains supported by its capability of reproducing the same phylogenetic and evolutionary information as the whole viral genome did. Also, a reliable cutoff to accurately classify APPV at different levels is established. We propose a genotyping scheme with three well-defined genotypes (1-3) and 7 subgenotypes for genotype 1 (1.1-1.7). For whole genome analysis, a threshold value of 84%-91% pairwise identity allows separation of all APPV subgenotypes, whereas 80% identity clearly segregate the three major APPV genotypes. For NS5a gene analysis, 82%-91% identity allows subgenotype separation and 76% identity segregate APPV genotypes. Additionally, genetic distance of whole genome exhibits ≤8% in isolate level, 9%-14% in subgenotype level, and 17%-22% in genotype level, while for NS5a encoding sequences the genetic distance displays ≤9% in isolate level, 9.9%-19.1% in subgenotype level, and 21.6%-29.7% in genotype level. These allow a clear segregation among APPV genotypes, subgenotypes, and isolates. Therefore, the proposed strategy in this study provides a solid and improved basis for molecular phylogenetics to understand APPV genetic diversity, trace the origins and control the spread of new disease outbreaks.


Assuntos
Genótipo , Pestivirus/genética , RNA Polimerase Dependente de RNA/genética , Doenças dos Suínos/virologia , Proteínas não Estruturais Virais/genética , Animais , Sus scrofa , Suínos
13.
Transbound Emerg Dis ; 68(6): 3288-3304, 2021 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-33207070

RESUMO

Since first identified in December of 2019, COVID-19 has been quickly spreading to the world in few months and COVID-19 cases are still undergoing rapid surge in most countries worldwide. The causative agent, severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), adapts and evolves rapidly in nature. With the availability of 16,092 SARS-CoV-2 full genomes in GISAID as of 13 May, we removed the poor-quality genomes and performed mutational profiling analysis for the remaining 11,183 viral genomes. Global analysis of all sequences identified all single nucleotide polymorphisms (SNPs) across the whole genome and critical SNPs with high mutation frequency that contributes to five-clade classification of global strains. A total of 119 SNPs were found with 74 non-synonymous mutations, 43 synonymous mutations and two mutations in intergenic regions. Analysis of geographic pattern of mutational profiling for the whole genome reveals differences between each continent. A transition mutation from C to T represents the most mutation types across the genome, suggesting rapid evolution and adaptation of the virus in host. Amino acid (AA) deletions and insertions found across the genome results in changes in viral protein length and potential function alteration. Mutational profiling for each gene was analysed, and results show that nucleocapsid gene demonstrates the highest mutational frequency, followed by Nsp2, Nsp3 and Spike gene. We further focused on non-synonymous mutational distributions on four key viral proteins, spike with 75 mutations, RNA-dependent-RNA-polymerase with 41 mutations, 3C-like protease with 22 mutations and Papain-like protease with 10 mutations. Results show that non-synonymous mutations on critical sites of these four proteins pose great challenge for development of anti-viral drugs and other countering measures. Overall, this study provides more understanding of genetic diversity/variability of SARS-CoV-2 and insights for development of anti-viral therapeutics.


Assuntos
Genoma Viral , SARS-CoV-2 , Glicoproteína da Espícula de Coronavírus , COVID-19/virologia , Variação Genética , Humanos , Mutação , Filogenia , Polimorfismo de Nucleotídeo Único , SARS-CoV-2/genética , Glicoproteína da Espícula de Coronavírus/genética
14.
mBio ; 11(5)2020 10 13.
Artigo em Inglês | MEDLINE | ID: mdl-33051368

RESUMO

Despite numerous barriers to transmission, zoonoses are the major cause of emerging infectious diseases in humans. Among these, severe acute respiratory syndrome (SARS), Middle East respiratory syndrome (MERS), and ebolaviruses have killed thousands; the human immunodeficiency virus (HIV) has killed millions. Zoonoses and human-to-animal cross-species transmission are driven by human actions and have important management, conservation, and public health implications. The current SARS-CoV-2 pandemic, which presumably originated from an animal reservoir, has killed more than half a million people around the world and cases continue to rise. In March 2020, New York City was a global epicenter for SARS-CoV-2 infections. During this time, four tigers and three lions at the Bronx Zoo, NY, developed mild, abnormal respiratory signs. We detected SARS-CoV-2 RNA in respiratory secretions and/or feces from all seven animals, live virus in three, and colocalized viral RNA with cellular damage in one. We produced nine whole SARS-CoV-2 genomes from the animals and keepers and identified different SARS-CoV-2 genotypes in the tigers and lions. Epidemiologic and genomic data indicated human-to-tiger transmission. These were the first confirmed cases of natural SARS-CoV-2 animal infections in the United States and the first in nondomestic species in the world. We highlight disease transmission at a nontraditional interface and provide information that contributes to understanding SARS-CoV-2 transmission across species.IMPORTANCE The human-animal-environment interface of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is an important aspect of the coronavirus disease 2019 (COVID-19) pandemic that requires robust One Health-based investigations. Despite this, few reports describe natural infections in animals or directly link them to human infections using genomic data. In the present study, we describe the first cases of natural SARS-CoV-2 infection in tigers and lions in the United States and provide epidemiological and genetic evidence for human-to-animal transmission of the virus. Our data show that tigers and lions were infected with different genotypes of SARS-CoV-2, indicating two independent transmission events to the animals. Importantly, infected animals shed infectious virus in respiratory secretions and feces. A better understanding of the susceptibility of animal species to SARS-CoV-2 may help to elucidate transmission mechanisms and identify potential reservoirs and sources of infection that are important in both animal and human health.


Assuntos
Animais de Zoológico/virologia , Betacoronavirus/fisiologia , Infecções por Coronavirus/transmissão , Infecções por Coronavirus/veterinária , Pandemias/veterinária , Panthera/virologia , Pneumonia Viral/transmissão , Pneumonia Viral/veterinária , Animais , Betacoronavirus/classificação , Betacoronavirus/genética , Betacoronavirus/isolamento & purificação , COVID-19 , Infecções por Coronavirus/diagnóstico , Infecções por Coronavirus/virologia , Genoma Viral/genética , Haplótipos , Humanos , Cidade de Nova Iorque/epidemiologia , Saúde Única , Filogenia , Pneumonia Viral/diagnóstico , Pneumonia Viral/virologia , SARS-CoV-2 , Zoonoses/epidemiologia , Zoonoses/transmissão , Zoonoses/virologia
15.
Microbiol Resour Announc ; 9(22)2020 May 28.
Artigo em Inglês | MEDLINE | ID: mdl-32467283

RESUMO

This report describes the identification and characterization of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) in a Malayan tiger in a U.S. zoo.

16.
Virology ; 543: 63-75, 2020 04.
Artigo em Inglês | MEDLINE | ID: mdl-32174300

RESUMO

Viruses exploit phosphorylation of both viral and host proteins to support viral replication. In this study, we demonstrate that porcine reproductive and respiratory syndrome virus replicase nsp2, and two nsp2-related -2/-1 frameshifting products, nsp2TF and nsp2N, are hyper-phosphorylated. By mapping phosphorylation sites, we subdivide an extended, previously uncharacterized region, located between the papain-like protease-2 (PLP2) domain and frameshifting site, into three distinct domains. These domains include two large hypervariable regions (HVR) with putative intrinsically disordered structures, separated by a conserved and partly structured interval domain that we defined as the inter-HVR conserved domain (IHCD). Abolishing phosphorylation of the inter-species conserved residue serine918, which is located within the IHCD region, abrogates accumulation of viral genomic and subgenomic RNAs and recombinant virus production. Our study reveals the biological significance of phosphorylation events in nsp2-related proteins, emphasizes pleiotropic functions of nsp2-related proteins in the viral life cycle, and presents potential links to pathogenesis.


Assuntos
Vírus da Síndrome Respiratória e Reprodutiva Suína/metabolismo , Proteínas não Estruturais Virais/metabolismo , Animais , Linhagem Celular , Genoma Viral , Interações entre Hospedeiro e Microrganismos , Espectrometria de Massas , Mutação , Fosforilação , Vírus da Síndrome Respiratória e Reprodutiva Suína/química , Vírus da Síndrome Respiratória e Reprodutiva Suína/genética , Vírus da Síndrome Respiratória e Reprodutiva Suína/crescimento & desenvolvimento , Domínios Proteicos , Suínos , Proteínas não Estruturais Virais/química , Proteínas não Estruturais Virais/genética , Replicação Viral/genética , Replicação Viral/fisiologia
17.
Virus Res ; 269: 197632, 2019 08.
Artigo em Inglês | MEDLINE | ID: mdl-31129172

RESUMO

Among the structural proteins that compose the virion of African swine fever virus (ASFV), p30 is one of the most immunogenic proteins and is produced during early stage of ASFV infection. These two characteristics make p30 a good target for diagnostic assays to detect ASFV infection. In this study, we describe a panel of newly generated p30-specific monoclonal antibodies (mAbs). The reactivity of these mAbs was confirmed by immunoprecipitation and Western blot analysis in Vero cells infected with alphavirus replicon particles that express p30 (RP-p30). Furthermore, this panel of mAbs recognized ASFV strains BA71 V (Genotype I) and Georgia/2007 (Genotype II) in immunofluorescence assays on virus-infected Vero cells and swine macrophages, respectively. These mAbs also detected p30 expression by immunohistochemistry in tissue samples from ASFV-infected pigs. Epitope mapping revealed that a selected mAb from the panel recognized a linear epitope within the 32-amino acid region, 61-93. In contrast, two of the mAbs recognize the C-terminal region of the protein, which is highly hydrophilic, enriched in glutamic acid residues, and predicted to contain an intrinsically disordered protein region (IDPR). This panel of mAbs and mAb-based diagnostic assays potentially represent valuable tools for ASFV detection, surveillance and disease control.


Assuntos
Vírus da Febre Suína Africana/química , Febre Suína Africana/diagnóstico , Anticorpos Monoclonais/imunologia , Anticorpos Antivirais/imunologia , Fosfoproteínas/imunologia , Proteínas Virais/imunologia , Febre Suína Africana/imunologia , Vírus da Febre Suína Africana/genética , Alphavirus/genética , Alphavirus/imunologia , Animais , Anticorpos Monoclonais/isolamento & purificação , Antígenos Virais/imunologia , Chlorocebus aethiops , Epitopos/química , Epitopos/imunologia , Macrófagos/imunologia , Macrófagos/virologia , Suínos , Células Vero
18.
Diagn Microbiol Infect Dis ; 95(1): 59-66, 2019 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-31130238

RESUMO

Influenza is a common and contagious respiratory disease caused by influenza A, B, C, and D viruses (IAV, IBV, ICV, and IDV). A multiplex real-time RT-PCR assay was developed for simultaneous detection of IAV, IBV, ICV, and IDV. The assay was designed to target unique sequences in the matrix gene of IBV and ICV, the RNA polymerase subunit PB1 of IDV, and combined with USDA and CDC IAV assays, both target the matrix gene. The host 18S rRNA gene was included as an internal control. In silico analyses indicated high strain coverages: 97.9% for IBV, 99.5% for ICV, and 100% for IDV. Transcribed RNA, viral isolates and clinical samples were used for validation. The assay specifically detected target viruses without cross-reactivity, nor detection of other common pathogens. The limit of detection was approximately 30 copies for each viral RNA template, which was equivalent to a threshold cycle value of ~37.


Assuntos
Doenças dos Bovinos/diagnóstico , Infecções por Orthomyxoviridae/veterinária , Orthomyxoviridae/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa/normas , Doenças dos Suínos/diagnóstico , Animais , Bovinos , Doenças dos Bovinos/virologia , Diagnóstico Diferencial , Genes Virais/genética , Orthomyxoviridae/classificação , Infecções por Orthomyxoviridae/virologia , RNA Viral/genética , Sensibilidade e Especificidade , Suínos , Doenças dos Suínos/virologia
19.
J Virol ; 91(1)2017 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-27795441

RESUMO

Porcine circovirus-associated disease (PCVAD) is clinically manifested by postweaning multisystemic wasting syndrome (PMWS), respiratory and enteric disease, reproductive failure, and porcine dermatitis and nephropathy syndrome (PDNS). Porcine circovirus 2 (PCV2) is an essential component of PCVAD, although an etiologic role in PDNS is not well established. Here, a novel circovirus, designated porcine circovirus 3 (PCV3), was identified in sows that died acutely with PDNS-like clinical signs. The capsid and replicase proteins of PCV3 are only 37% and 55% identical to PCV2 and bat circoviruses, respectively. Aborted fetuses from sows with PDNS contained high levels of PCV3 (7.57 × 107 genome copies/ml), and no other viruses were detected by PCR and metagenomic sequencing. Immunohistochemistry (IHC) analysis of sow tissue samples identified PCV3 antigen in skin, kidney, lung, and lymph node samples localized in typical PDNS lesions, including necrotizing vasculitis, glomerulonephritis, granulomatous lymphadenitis, and bronchointerstitial pneumonia. Further study of archived PDNS tissue samples that were negative for PCV2 by IHC analysis identified 45 of 48 that were PCV3 positive by quantitative PCR (qPCR), with 60% of a subset also testing positive for PCV3 by IHC analysis. Analysis by qPCR of 271 porcine respiratory disease diagnostic submission samples identified 34 PCV3-positive cases (12.5%), and enzyme-linked immunosorbent assay detection of anti-PCV3 capsid antibodies in serum samples found that 46 (55%) of 83 samples tested were positive. These results suggest that PCV3 commonly circulates within U.S. swine and may play an etiologic role in reproductive failure and PDNS. Because of the high economic impact of PCV2, this novel circovirus warrants further studies to elucidate its significance and role in PCVAD. IMPORTANCE: While porcine circovirus 2 (PCV2) was first identified in sporadic cases of postweaning multisystemic wasting syndrome in Canada in the early 1990s, an epidemic of severe systemic disease due to PCV2 spread worldwide in the ensuing decade. Despite being effectively controlled by commercial vaccines, PCV2 remains one of the most economically significant viruses of swine. Here, a novel porcine circovirus (PCV3) that is distantly related to known circoviruses was identified in sows with porcine dermatitis and nephropathy syndrome (PDNS) and reproductive failure. PCV2, which has previously been associated with these clinical presentations, was not identified. High levels of PCV3 nucleic acid were observed in aborted fetuses by quantitative PCR, and PCV3 antigen was localized in histologic lesions typical of PDNS in sows by immunohistochemistry (IHC) analysis. PCV3 was also identified in archival PDNS diagnostic samples that previously tested negative for PCV2 by IHC analysis. The emergence of PCV3 warrants further investigation.


Assuntos
Aborto Espontâneo/epidemiologia , Circovirus/genética , Dermatite/epidemiologia , Genoma Viral , Filogenia , Síndrome Definhante Multissistêmico de Suínos Desmamados/epidemiologia , Doenças dos Suínos/epidemiologia , Aborto Espontâneo/mortalidade , Aborto Espontâneo/patologia , Aborto Espontâneo/virologia , Doença Aguda , Animais , Antígenos Virais/genética , Antígenos Virais/imunologia , Canadá/epidemiologia , Capsídeo/química , Capsídeo/imunologia , Proteínas do Capsídeo/genética , Proteínas do Capsídeo/imunologia , Circovirus/classificação , Circovirus/imunologia , Circovirus/isolamento & purificação , Dermatite/mortalidade , Dermatite/patologia , Dermatite/virologia , Feminino , Feto , Vigilância Imunológica , Rim/patologia , Rim/virologia , Pulmão/patologia , Pulmão/virologia , Linfonodos/patologia , Linfonodos/virologia , North Carolina/epidemiologia , Síndrome Definhante Multissistêmico de Suínos Desmamados/mortalidade , Síndrome Definhante Multissistêmico de Suínos Desmamados/patologia , Síndrome Definhante Multissistêmico de Suínos Desmamados/virologia , RNA Polimerase Dependente de RNA/genética , RNA Polimerase Dependente de RNA/imunologia , Pele/patologia , Pele/virologia , Análise de Sobrevida , Suínos , Doenças dos Suínos/mortalidade , Doenças dos Suínos/patologia , Doenças dos Suínos/virologia
20.
Virology ; 497: 111-124, 2016 10.
Artigo em Inglês | MEDLINE | ID: mdl-27459668

RESUMO

A full-length cDNA infectious clone, pKS15-01-Clone, was constructed from an emerging Senecavirus A (SVA; strain KS15-01). To explore the potential use as a viral backbone for expressing marker genes, the enhanced green fluorescent protein (EGFP)-tagged reporter virus (vKS15-01-EGFP) was generated using reverse genetics. Compared to the parental virus, the pKS15-01-Clone derived virus (vKS15-01-Clone) replicated efficiently in vitro and in vivo, and induced similar levels of neutralizing antibody and cytokine responses in infected animals. In contrast, the vKS15-01-EGFP virus showed impaired growth ability and induced lower level of immune response in infected animals. Lesions on the dorsal snout and coronary bands were observed in all pigs infected by parental virus KS15-01, but not in pigs infected with vKS15-01-Clone or vKS15-01-EGFP viruses. These results demonstrated that the infectious clone and EGFP reporter virus could be used as important tools in further elucidating the SVA pathogenesis and development of control measures.


Assuntos
DNA Complementar , Genoma Viral , Picornaviridae/genética , RNA Viral , Imunidade Adaptativa , Animais , Anticorpos Antivirais/imunologia , Sequência de Bases , Linhagem Celular , Clonagem Molecular , Citocinas/metabolismo , DNA Complementar/química , DNA Complementar/genética , Expressão Gênica , Genes Reporter , Vetores Genéticos/genética , Imunidade Inata , Mutação , Fenótipo , Picornaviridae/imunologia , Infecções por Picornaviridae/veterinária , Recombinação Genética , Suínos , Doenças dos Suínos/diagnóstico , Doenças dos Suínos/imunologia , Doenças dos Suínos/metabolismo , Doenças dos Suínos/virologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA