Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 33
Filter
1.
Viruses ; 13(12)2021 12 06.
Article in English | MEDLINE | ID: mdl-34960715

ABSTRACT

Newcastle disease virus (NDV) can infect over 250 bird species with variable pathogenicity; it can also infect humans in rare cases. The present study investigated an outbreak in feral pigeons in São Paulo city, Brazil, in 2019. Affected birds displayed neurological signs, and hemorrhages were observed in different tissues. Histopathology changes with infiltration of mononuclear inflammatory cells were also found in the brain, kidney, proventriculus, heart, and spleen. NDV staining was detected by immunohistochemistry. Twenty-seven out of thirty-four tested samples (swabs and tissues) were positive for Newcastle disease virus by RT-qPCR test, targeting the M gene. One isolate, obtained from a pool of positive swab samples, was characterized by the intracerebral pathogenicity index (ICPI) and the hemagglutination inhibition (HI) tests. This isolate had an ICPI of 0.99, confirming a virulent NDV strain. The monoclonal antibody 617/161, which recognizes a distinct epitope in pigeon NDV strains, inhibited the isolate with an HI titer of 512. A complete genome of NDV was obtained using next-generation sequencing. Phylogenetic analysis based on the complete CDS F gene grouped the detected isolate with other viruses from subgenotype VI.2.1.2, class II, including one previously reported in Southern Brazil in 2014. This study reports a comprehensive characterization of the subgenotype VI.2.1.2, which seems to have been circulating in Brazilian urban areas since 2014. Due to the zoonotic risk of NDV, virus surveillance in feral pigeons should also be systematically performed in urban areas.


Subject(s)
Columbidae , Disease Outbreaks/veterinary , Newcastle Disease/epidemiology , Newcastle disease virus/genetics , Animals , Brazil/epidemiology , Genome, Viral , Genotype , High-Throughput Nucleotide Sequencing , Newcastle Disease/pathology , Newcastle Disease/virology , Newcastle disease virus/classification , Newcastle disease virus/isolation & purification , Newcastle disease virus/pathogenicity , Phylogeny , Virulence , Whole Genome Sequencing
2.
J Virol ; 95(18): e0095521, 2021 08 25.
Article in English | MEDLINE | ID: mdl-34232725

ABSTRACT

Highly pathogenic avian influenza (HPAI) viruses from the H5Nx Goose/Guangdong/96 lineage continue to cause outbreaks in domestic and wild bird populations. Two distinct genetic groups of H5N8 HPAI viruses, hemagglutinin (HA) clades 2.3.4.4A and 2.3.4.4B, caused intercontinental outbreaks in 2014 to 2015 and 2016 to 2017, respectively. Experimental infections using viruses from these outbreaks demonstrated a marked difference in virulence in mallards, with the H5N8 virus from 2014 causing mild clinical disease and the 2016 H5N8 virus causing high mortality. To assess which gene segments are associated with enhanced virulence of H5N8 HPAI viruses in mallards, we generated reassortant viruses with 2014 and 2016 viruses. For single-segment reassortants in the genetic backbone of the 2016 virus, pathogenesis experiments in mallards revealed that morbidity and mortality were reduced for all eight single-segment reassortants compared to the parental 2016 virus, with significant reductions in mortality observed with the polymerase basic protein 2 (PB2), nucleoprotein (NP), and matrix (M) reassortants. No differences in morbidity and mortality were observed with reassortants that either have the polymerase complex segments or the HA and neuraminidase (NA) segments of the 2016 virus in the genetic backbone of the 2014 virus. In vitro assays showed that the NP and polymerase acidic (PA) segments of the 2014 virus lowered polymerase activity when combined with the polymerase complex segments of the 2016 virus. Furthermore, the M segment of the 2016 H5N8 virus was linked to filamentous virion morphology. Phylogenetic analyses demonstrated that gene segments related to the more virulent 2016 H5N8 virus have persisted in the contemporary H5Nx HPAI gene pool until 2020. IMPORTANCE Outbreaks of H5Nx HPAI viruses from the goose/Guangdong/96 lineage continue to occur in many countries and have resulted in substantial impact on wild birds and poultry. Epidemiological evidence has shown that wild waterfowl play a major role in the spread of these viruses. While HPAI virus infection in gallinaceous species causes high mortality, a wide range of disease outcomes has been observed in waterfowl species. In this study, we examined which gene segments contribute to severe disease in mallards infected with H5N8 HPAI viruses. No virus gene was solely responsible for attenuating the high virulence of a 2016 H5N8 virus, but the PB2, NP, and M segments significantly reduced mortality. The findings herein advance our knowledge on the pathobiology of avian influenza viruses in waterfowl and have potential implications on the ecology and epidemiology of H5Nx HPAI in wild bird populations.


Subject(s)
Ducks/virology , Influenza A Virus, H5N8 Subtype/classification , Influenza A Virus, H5N8 Subtype/pathogenicity , Influenza in Birds/transmission , Influenza in Birds/virology , Poultry Diseases/virology , Viral Proteins/genetics , Animals , Influenza A Virus, H5N8 Subtype/genetics , Phylogeny , Poultry Diseases/genetics , Virulence
3.
Vaccines (Basel) ; 9(2)2021 Feb 21.
Article in English | MEDLINE | ID: mdl-33669907

ABSTRACT

The efficacy of an adenovirus-vectored Newcastle disease virus (NDV) vaccine expressing the fusion (F) NDV protein (adeno-F) was evaluated against challenges with virulent heterologous and homologous NDV strains to the F protein. In a preliminary study, two different doses (low and high) of adeno-F were tested against a virulent NDV strain containing the homologous NDV F protein, CA02. In a second study, at three weeks post-vaccination, the efficacy of the high dose of adeno-F was compared to a live attenuated NDV vaccine strain (LaSota) against three antigenically distinct virulent NDV challenge strains, one homologous (CA02) and two heterologous (TZ12, EG14) to F in the vectored vaccine. In both experiments, clinical signs, mortality, virus shedding, and humoral response were evaluated. In the first experiment, the survival rates from birds vaccinated with adeno-F at a high and low dose were 100% and 25%, respectively. In the second experiment, birds vaccinated with the high dose of adeno-F had a survival rate of 80%, 75%, and 65% after challenge with the CA02, TZ12, and EG14 viruses, respectively. All of the LaSota-vaccinated birds survived post-challenge no matter the NDV challenge strain. High antibody titers were detected after vaccination with LaSota by HI and ELISA tests. The majority of adeno-F-vaccinated birds had detectable antibodies using the ELISA test, but not using the HI test, before the challenge. The data show that both the similarity of the F protein of the adeno-F vaccine to the challenge virus and the adeno-F vaccination dose affect the efficacy of an adenovirus-vectored NDV vaccine against a virulent NDV challenge.

4.
Viruses ; 13(1)2021 Jan 13.
Article in English | MEDLINE | ID: mdl-33451125

ABSTRACT

Kenyan poultry consists of ~80% free-range indigenous chickens kept in small flocks (~30 birds) on backyard poultry farms (BPFs) and they are traded via live bird markets (LBMs). Newcastle disease virus (NDV) was detected in samples collected from chickens, wild farm birds, and other domestic poultry species during a 2017-2018 survey conducted at 66 BPFs and 21 LBMs in nine Kenyan counties. NDV nucleic acids were detected by rRT-PCR L-test in 39.5% (641/1621) of 1621 analyzed samples, of which 9.67% (62/641) were NDV-positive by both the L-test and a fusion-test designed to identify the virulent virus, with a majority being at LBMs (64.5%; 40/62) compared to BPFs (25.5%; 22/62). Virus isolation and next-generation sequencing (NGS) on a subset of samples resulted in 32 complete NDV genome sequences with 95.8-100% nucleotide identities amongst themselves and 95.7-98.2% identity with other east African isolates from 2010-2016. These isolates were classified as a new sub-genotype, V.3, and shared 86.5-88.9% and 88.5-91.8% nucleotide identities with subgenotypes V.1 and V.2 viruses, respectively. The putative fusion protein cleavage site (113R-Q-K-R↓F 117) in all 32 isolates, and a 1.86 ICPI score of an isolate from a BPF chicken that had clinical signs consistent with Newcastle disease, confirmed the high virulence of the NDVs. Compared to genotypes V and VI viruses, the attachment (HN) protein of 18 of the 32 vNDVs had amino acid substitutions in the antigenic sites. A time-scaled phylogeographic analysis suggests a west-to-east dispersal of the NDVs via the live chicken trade, but the virus origins remain unconfirmed due to scarcity of continuous and systematic surveillance data. This study reveals the widespread prevalence of vNDVs in Kenyan backyard poultry, the central role of LBMs in the dispersal and possibly generation of new virus variants, and the need for robust molecular epidemiological surveillance in poultry and non-poultry avian species.


Subject(s)
Chickens/virology , Genotype , Newcastle Disease/epidemiology , Newcastle Disease/virology , Newcastle disease virus/classification , Newcastle disease virus/genetics , Poultry Diseases/epidemiology , Poultry Diseases/virology , Animals , Farms , Genome, Viral , Genomics/methods , Kenya/epidemiology , Molecular Epidemiology , Newcastle disease virus/isolation & purification , Newcastle disease virus/pathogenicity , Phylogeny , Phylogeography , Public Health Surveillance , RNA, Viral , Spatio-Temporal Analysis , Virulence
5.
Vaccine ; 38(34): 5507-5515, 2020 07 22.
Article in English | MEDLINE | ID: mdl-32591288

ABSTRACT

Vaccines against virulent Newcastle disease virus (NDV) are widely available and can be protective, but improved vaccination protocols are needed to prevent clinical disease and reduce virus circulation. The present study evaluated the efficacy of two commercial vaccines alone or in combination: a live attenuated NDV vaccine (LV) and a recombinant herpesvirus of turkeys vector expressing the fusion protein of NDV and the virus protein 2 of infectious bursal disease virus (rHVT-ND-IBD). Chickens were vaccinated with one of four vaccination protocols: live vaccine (LV) at 1 and 11 days of age (DOA), rHVT ND-IBD and LV at 1 DOA, rHVT ND-IBD at 1 DOA boosted with an LV at 11 DOA, and rHVT ND-IBD at 1 DOA. The vaccinated birds were challenged at different time points (3 or 4 weeks of age) with the California 2018 virus. The mortality, clinical signs, mean death time (MDT), humoral response before and after vaccination, and virus shedding after challenge were evaluated. All vaccination protocols were able to prevent mortality, reduce virus shedding, and induce antibody levels before the challenge at 3 and 4 weeks-old. Overall, the antibody levels before the challenge at 4 weeks were significantly higher in all groups vaccinated with the rHVT ND-IBD when compared to levels in 3 week old birds. The combination of recombinant rHVT ND-IBD with a live vaccine at one-day-old seems to be a better combination, due to the absence of clinical signs, higher antibody levels pre and post-challenge, and reduced virus shedding at any time point after the challenge at 3 or 4 weeks of age with the California 2018 virus.


Subject(s)
Newcastle Disease , Poultry Diseases , Viral Vaccines , Animals , Antibodies, Viral , California , Chickens , Newcastle Disease/prevention & control , Newcastle disease virus/genetics , Poultry Diseases/prevention & control , Vaccination , Vaccines, Attenuated , Vaccines, Synthetic/genetics , Viral Vaccines/genetics
7.
Infect Genet Evol ; 78: 104074, 2020 03.
Article in English | MEDLINE | ID: mdl-31634645

ABSTRACT

Poultry production plays an important role in the economy and livelihoods of rural households in Kenya. As part of a surveillance program, avian influenza virus (AIV)-specific real-time RT-PCR (RRT-PCR) was used to screen 282 oropharyngeal swabs collected from chickens at six live bird markets (LBMs) and 33 backyard poultry farms in Kenya and 8 positive samples were detected. Virus was isolated in eggs from five samples, sequenced, and identified as H9N2 low pathogenic AIV (LPAIV) G1 lineage, with highest nucleotide sequence identity (98.6-99.9%) to a 2017 Ugandan H9N2 isolate. The H9N2 contained molecular markers for mammalian receptor specificity, implying their zoonotic potential. Virus pathogenesis and transmissibility was assessed by inoculating low and medium virus doses of a representative Kenyan H9N2 LPAIV isolate into experimental chickens and exposing them to naïve uninfected chickens at 2 -days post inoculation (dpi). Virus shedding was determined at 2/4/7 dpi and 2/5 days post placement (dpp), and seroconversion determined at 14 dpi/12 dpp. None of the directly-inoculated or contact birds exhibited any mortality or clinical disease signs. All directly-inoculated birds in the low dose group shed virus during the experiment, while only one contact bird shed virus at 2 dpp. Only two directly-inoculated birds that shed high virus titers seroconverted in that group. All birds in the medium dose group shed virus at 4/7 dpi and at 5 dpp, and they all seroconverted at 12/14 dpp. This is the first reported detection of H9N2 LPAIV from Kenya and it was shown to be infectious and transmissible in chickens by direct contact and represents a new disease threat to poultry and potentially to people.


Subject(s)
Eggs/virology , Influenza A Virus, H9N2 Subtype/pathogenicity , Influenza in Birds/diagnosis , Oropharynx/virology , Reassortant Viruses/pathogenicity , Animals , Chickens , Influenza A Virus, H9N2 Subtype/classification , Influenza A Virus, H9N2 Subtype/genetics , Influenza in Birds/virology , Kenya , Phylogeny , Population Surveillance , Reassortant Viruses/classification , Reassortant Viruses/genetics , Sequence Analysis, RNA , Virus Shedding , Whole Genome Sequencing
8.
Arch Virol ; 165(1): 261, 2020 01.
Article in English | MEDLINE | ID: mdl-31784908

ABSTRACT

The Editor-in-Chief has retracted this article [1]. Figures 1A, 1D and 2B (bottom right) are identical with Figures 1A, 1H and 1B respectively in another article [2] which reports a study in a different species. In addition, Table 1 contains data presented in a third article [3], which also reports a study in a different species. The Editor-in-Chief therefore no longer has confidence in the validity of the data and the conclusions drawn. Tereza C. Cardoso disagrees with this retraction. Helena L. Ferreira agrees with this retraction. Sergio E. L. da Silva, Andrea F. Garcia, Felipe E. S. Silva, Roberto Gameiro, Carolina U. F. Fabri and Dielson S. Vieira have not responded to any correspondence about this retraction.

9.
PLoS One ; 14(9): e0222457, 2019.
Article in English | MEDLINE | ID: mdl-31513638

ABSTRACT

Highly pathogenic avian influenza (HPAI) virus subtype H7N3 has been circulating in poultry in Mexico since 2012 and vaccination has been used to control the disease. In this study, eight Mexican H7N3 HPAI viruses from 2015-2017 were isolated and fully sequenced. No evidence of reassortment was detected with other avian influenza (AI) viruses, but phylogenetic analyses show divergence of all eight gene segments into three genetic clusters by 2015, with 94.94 to 98.78 percent nucleotide homology of the HA genes when compared to the index virus from 2012. The HA protein of viruses from each cluster showed a different number of basic amino acids (n = 5-7) in the cleavage site, and six different patterns at the predicted N-glycosylation sites. Comparison of the sequences of the Mexican lineage H7N3 HPAI viruses and American ancestral wild bird AI viruses to characterize the virus evolutionary dynamics showed that the nucleotide substitution rates in PB2, PB1, PA, HA, NP, and NS genes greatly increased once the virus was introduced into poultry. The global nonsynonymous and synonymous ratios imply strong purifying selection driving the evolution of the virus. Forty-nine positively selected sites out of 171 nonsynonymous mutations were identified in the Mexican H7N3 HPAI viruses, including 7 amino acid changes observed in higher proportion in North American poultry origin AI viruses isolates than in wild bird-origin viruses. Continuous monitoring and molecular characterization of the H7N3 HPAI virus is important for better understanding of the virus evolutionary dynamics and further improving control measures including vaccination.


Subject(s)
Influenza A Virus, H7N3 Subtype/genetics , Influenza in Birds/epidemiology , Poultry Diseases/genetics , Animals , Birds/genetics , Chickens/genetics , Chickens/virology , Disease Outbreaks , Evolution, Molecular , Influenza A Virus, H7N3 Subtype/pathogenicity , Influenza in Birds/virology , Mexico/epidemiology , Phylogeny , Poultry/genetics , Poultry Diseases/virology
11.
Vet Microbiol ; 235: 25-34, 2019 Aug.
Article in English | MEDLINE | ID: mdl-31282376

ABSTRACT

Five, class II, virulent Newcastle disease virus (vNDV) isolates of different genotypes from different host species were evaluated for their ability to infect, cause disease, and transmit to naïve chickens. Groups of five birds received a low, medium, or high dose, by the oculonasal route, of one of the following vNDV: three chicken-origin, one cormorant-origin, and one pigeon-origin. Three naïve birds were added to each group at two days post-inoculation (DPI) to evaluate transmission. Virus shedding was quantified from swabs (2/4/7 DPI), and seroconversion was evaluated at 14 DPI. All inoculated and contact birds in the chicken-origin vNDV groups succumbed to infection, displaying clinical signs typical of Newcastle disease and shed virus titers above 6 log10 EID50/ml. Birds receiving a high and medium dose of the cormorant virus showed primarily neurological clinical signs with 80% and 60% mortality, respectively. The chickens showing clinical disease shed virus at titers below 4 log10 EID50/ml, and the remaining bird in the high dose group seroconverted with a high HI titer. For the pigeon-origin virus, no clinical signs were observed in any of the birds, but all 5 chickens in the high challenge dose and one bird in the medium challenge group shed virus at mean titers of 3.1 and 2.2 log10 EID50/ml, respectively. Overall, the chicken-origin viruses infected chickens and efficiently transmitted to naïve birds, while the cormorant- and pigeon-origin viruses infected chickens only at the higher doses and did not transmit to other birds.


Subject(s)
Chickens/virology , Columbidae/virology , Newcastle Disease/transmission , Newcastle disease virus/pathogenicity , Poultry Diseases/transmission , Animals , Animals, Wild/virology , Vaccination , Virulence , Virus Shedding
12.
Infect Genet Evol ; 74: 103917, 2019 10.
Article in English | MEDLINE | ID: mdl-31200111

ABSTRACT

Several Avian paramyxoviruses 1 (synonymous with Newcastle disease virus or NDV, used hereafter) classification systems have been proposed for strain identification and differentiation. These systems pioneered classification efforts; however, they were based on different approaches and lacked objective criteria for the differentiation of isolates. These differences have created discrepancies among systems, rendering discussions and comparisons across studies difficult. Although a system that used objective classification criteria was proposed by Diel and co-workers in 2012, the ample worldwide circulation and constant evolution of NDV, and utilization of only some of the criteria, led to identical naming and/or incorrect assigning of new sub/genotypes. To address these issues, an international consortium of experts was convened to undertake in-depth analyses of NDV genetic diversity. This consortium generated curated, up-to-date, complete fusion gene class I and class II datasets of all known NDV for public use, performed comprehensive phylogenetic neighbor-Joining, maximum-likelihood, Bayesian and nucleotide distance analyses, and compared these inference methods. An updated NDV classification and nomenclature system that incorporates phylogenetic topology, genetic distances, branch support, and epidemiological independence was developed. This new consensus system maintains two NDV classes and existing genotypes, identifies three new class II genotypes, and reduces the number of sub-genotypes. In order to track the ancestry of viruses, a dichotomous naming system for designating sub-genotypes was introduced. In addition, a pilot dataset and sub-trees rooting guidelines for rapid preliminary genotype identification of new isolates are provided. Guidelines for sequence dataset curation and phylogenetic inference, and a detailed comparison between the updated and previous systems are included. To increase the speed of phylogenetic inference and ensure consistency between laboratories, detailed guidelines for the use of a supercomputer are also provided. The proposed unified classification system will facilitate future studies of NDV evolution and epidemiology, and comparison of results obtained across the world.


Subject(s)
Newcastle disease virus/classification , RNA, Viral/genetics , Sequence Analysis, RNA/methods , Bayes Theorem , Consensus , Data Curation , Databases, Genetic , Genotype , Guidelines as Topic , International Cooperation , Likelihood Functions , Newcastle disease virus/genetics , Phylogeny
13.
Virology ; 531: 203-218, 2019 05.
Article in English | MEDLINE | ID: mdl-30928699

ABSTRACT

In May of 2018, virulent Newcastle disease virus was detected in sick, backyard, exhibition chickens in southern California. Since, the virus has affected 401 backyard and four commercial flocks, and one live bird market in California, and one backyard flock in Utah. The pathogenesis and transmission potential of this virus, along with two genetically related and widely studied viruses, chicken/California/2002 and chicken/Belize/2008, were evaluated in both 3-week- and 62-week-old chickens given a low, medium, or high challenge dose. All three viruses were highly virulent causing clinical signs, killing all the chickens in the medium and high dose groups, and efficiently transmitting to contacts. The three viruses also replicated in the reproductive tract of the adult hens. Virus shedding for all viruses was detected 24 hours after challenge, peaking with high titers at day 4 post challenge. Although not genetically identical, the studied isolates were shown to be phenotypically very similar, which allows the utilization of the available literature in the control of the current outbreak.


Subject(s)
Newcastle Disease/transmission , Newcastle disease virus/physiology , Newcastle disease virus/pathogenicity , Poultry Diseases/transmission , Age Factors , Animals , California/epidemiology , Chickens , Disease Outbreaks , Female , Male , Newcastle Disease/epidemiology , Newcastle Disease/virology , Newcastle disease virus/genetics , Newcastle disease virus/isolation & purification , Poultry Diseases/epidemiology , Poultry Diseases/virology , Virulence , Virus Shedding
14.
Pesqui. vet. bras ; 39(3): 209-213, Mar. 2019. tab, ilus
Article in English | VETINDEX, LILACS | ID: biblio-1002804

ABSTRACT

The present study investigated the circulation of avian metapneumovirus (aMPV) in wild birds in Brazil. To do so, 131 samples from 366 oropharyngeal or cloacal swabs collected from 18 species of birds were tested individually or in pools by RT-PCR. Samples detected by RT-PCR were selected for DNA sequencing. Thirteen (9.9%) samples were detected by the RT-PCR targeting the N gene and four out of 13 samples were sequenced. Sequencing results showed a high identity with the aMPV subtype A. Our results confirm the circulation of the aMPV subtype A in wild birds in Brazil even five years after its last detection.(AU)


O presente estudo investigou a circulação de metapneumovírus aviário em aves silvestres no Brasil. Para tanto, 131 amostras de 366 suabes orofaringeanos ou cloacais coletados de 18 espécies de aves foram testadas individualmente ou na forma de pools por RT-PCR. As amostras detectadas por RT-PCR foram selecionadas para sequenciamento. Treze (9,9%) das amostras foram detectadas por RT-PCR tendo o gene N como alvo; destas, quatro foram sequenciadas com sucesso. Resultados do sequenciamento mostraram alta identidade com o aMPV de subtipo A. Nossos resultados confirmam a circulação de aMPV subtipo A em aves silvestres no Brasil mesmo cinco anos após sua última detecção.(AU)


Subject(s)
Animals , Psittaciformes/virology , Paramyxoviridae Infections/veterinary , Paramyxoviridae Infections/epidemiology , Strigiformes/virology , Metapneumovirus/isolation & purification , Anseriformes/virology , Columbiformes/virology , Falconiformes/virology , Birds/virology
15.
Vet Microbiol ; 229: 153-158, 2019 02.
Article in English | MEDLINE | ID: mdl-30642592

ABSTRACT

This article has been retracted: please see Elsevier Policy on Article Withdrawal (https://www.elsevier.com/about/our-business/policies/article-withdrawal). This article has been retracted at the request of the Editors-in-Chief and Authors. Fig 1A is a duplicate of a figure that has already been published in da Silva SEL et al. Archives of Virology 2018;163:1043-1049; 10.1007/s00705-018-3704-2. These two papers report studies performed with cells from two different animal species (bovine cells for the Veterinary Microbiology paper and chicken cells for the Archives of Virology paper). The reuse of the same figure in the Veterinary Microbiology paper to describe cells that were supposed to be from a different species is thus inappropriate and also puts into question the reliability of the other results presented in this paper. In addition, the Editors-in-Chief have remaining concerns about the strong similarities of other data presented in the two papers. Even if these concerns were addressed, the re-use of any data has to be clearly indicated and appropriately cited. As such this article represents a misuse of the scientific publishing system. The scientific community takes a very strong view on this matter and apologies are offered to readers of the journal that this was not detected during the submission process.


Subject(s)
Herpesvirus 5, Bovine , Macrophages/virology , Mitochondria/pathology , Virus Replication/physiology , Animals , Cattle , Cell Proliferation , Cell Survival , Cells, Cultured , Macrophages/metabolism , Membrane Potential, Mitochondrial , Nitric Oxide
16.
Article in English | MEDLINE | ID: mdl-30574579

ABSTRACT

Newcastle disease virus (NDV) was detected by reverse transcriptase PCR (RT-PCR) from total RNA isolated from a chicken spleen of a backyard flock in Jordan. The complete coding genome sequence of NDV/chicken/Jordan/J11-spleen/2018 was obtained with MiSeq (Illumina) sequencing. Phylogenetic analysis of the concatenated coding sequences classified the virus as class II subgenotype VIIi.

17.
Pesqui. vet. bras ; 38(6): 1058-1063, jun. 2018. graf
Article in English | LILACS, VETINDEX | ID: biblio-955451

ABSTRACT

This study aimed to verify the occurrence of Leishmania spp. and Leishmania (Leishmania) infantum in horses from a visceral leishmaniasis endemic area in Brazil. DNA samples from blood and conjunctival swab (CS) were tested by PCR and Indirect Immunofluorescence Antibody Test (IFAT). Although none of the horses was clinically sick, animals infected by Leishmania spp. were found and some could be characterized as infected by L. (L.) infantum. From 40 horses, 100% of the animals were positive by blood PCR, 90% (36/40) by CS PCR, and 2.5% (01/40) in serodiagnosis, by IFAT. Six from these 40 horses were L. (L.) infantum positive by blood PCR. Direct sequencing and analysis of amplicons resulted in a sequence to evolutionary analysis. Results indicate the presence of Leishmania spp. and L. (L.) infantum infecting healthy horses in Brazil. The presence of Leishmania spp. and L. (L.) infantum DNA in asymptomatic horses suggests that they can be important reservoirs of these parasites, a highly relevant finding for the epidemiological surveillance of the diseases they cause.(AU)


O estudo objetivou verificar a ocorrência de Leishmania spp. e Leishmania (Leishmania) infantum em cavalos de uma região endêmica para leishmaniose visceral do Brasil. Amostras de DNA de sangue e suabe conjuntival (SC) foram testadas pela PCR e pela Reação de Imunofluorescência Indireta (RIFI). Embora nenhum cavalo estivesse clinicamente doente, animais infectados por Leishmania spp. e L. (L.) infantum foram encontrados em Ilha Solteira/SP. Dos 40 cavalos, 100% (40/40) foram positivos pela PCR de sangue, 90% (36/40) pela PCR de SC, e 2,5% (01/40) no sorodiagnóstico, pela RIFI. Seis desses 40 cavalos foram positivos para L. (L.) infantum pela PCR de sangue. O sequenciamento direto e a análise dos amplicons resultaram em uma sequência para análise evolutiva. Os resultados indicam a presença de Leishmania spp. e L. (L.) infantum infectando cavalos saudáveis no Brasil. A presença de DNA de Leishmania spp. and L. (L.) infantum em cavalos saudáveis sugere que eles podem ser importantes reservatórios desses parasitas, um achado altamente relevante para a vigilância epidemiológica das doenças que causam.(AU)


Subject(s)
Animals , Serologic Tests/veterinary , Leishmania infantum/immunology , Leishmania/classification
18.
Exp Parasitol ; 188: 42-49, 2018 May.
Article in English | MEDLINE | ID: mdl-29522766

ABSTRACT

In a previous study in Brazil, six isolates of Sarcocystis spp. recovered from budgerigars fed sporocysts excreted by opossums of the genus Didelphis were characterized by means of sequencing fragments of gene coding cytochrome B (CYTB), internal transcribed spacer 1 (ITS1), and surface antigen genes (SAG2, SAG3 and SAG4). The isolates shared identical ITS1 and CYTB sequences, but differed at SAG2, SAG3 and SAG4: three allele variants of SAG2, 3 allele variants of SAG3 and 2 allele variants of SAG4 were encountered in three multilocus genotypes (MLGs) (MLG1, MLG2, and MLG3). At ITS1 and CYTB, all the isolates from budgerigars were identical to the Sarcocystis falcatula-like isolate 59-2016-RS-BR that was detected in a barefaced ibis (Phimosus infuscatus) causing necrotizing meningoencephalitis in Brazil. At ITS1 locus, all the above isolates were clearly distinct from Sarcocystis neurona, Sarcocystis falcatula, Sarcocystis lindsayi, and Sarcocystis speeri, the four known species of Sarcocystis that use opossums of the genus Didelphis as definitive hosts. Here, we replicated the experiment above to identify additional MLGs or other species of Sarcocystis. Fifteen budgerigars were experimentally infected with sporocysts of Sarcocystis spp. from 12 opossums of the genus Didelphis. All the birds died 9-19 days after infection and tissue samples containing merozoites and schizonts of Sarcocystis spp. were recovered. Fractions of sequences coding for 18S ribosomal RNA gene (18S), CYTB, ITS1, SAG2, SAG3 and SAG4 were PCR amplified and sequenced from the infected lungs. In addition, fractions of 18S, SAG2, SAG3 and SAG4 were sequenced from the isolate 59-2016-RS-BR and fractions of 18S were sequenced from the six isolates from budgerigars described above. From the results, all the isolates shared identical 18S, ITS1 and CYTB sequences. Among the 15 new isolates from budgerigars, three allele variants of SAG2, 3 allele variants of SAG3 and 2 allele variants of SAG4 were encountered in five MLGs, of which four were novel (MLG1, MLG4, MLG5, MLG6 and MLG7). Isolate 59-2016-RS-BR was assigned to an eighth MLG (MLG8). Molecular data pointed that Sarcocystis assigned to MLGs 1 to 8 are variants of the same species, but the SAG-based trees of the isolates conflicted, which supports genetic admixture among them. The sarcocystinae studied have high diversity of SAG alleles per locus and the correlation of such an abundant variety of SAG alleles to host specificity and pathogenicity needs to be assessed. Remains to be elucidated if the parasites studied here and S. falcatula are variants of the same species that have diverged to the point of possessing differences at ITS1 level, but that are still capable of exchanging genes.


Subject(s)
Alleles , Antigens, Protozoan/genetics , Bird Diseases/parasitology , Opossums/parasitology , Sarcocystis/genetics , Sarcocystosis/veterinary , Animals , Antigens, Surface/genetics , Biological Evolution , Birds , Brain/parasitology , Brazil , DNA, Protozoan/chemistry , DNA, Protozoan/isolation & purification , Genetic Variation/genetics , Lung/parasitology , Melopsittacus , Meningoencephalitis/parasitology , Meningoencephalitis/veterinary , Multilocus Sequence Typing , Polymerase Chain Reaction/veterinary , Raccoons/parasitology , Sarcocystis/classification , Sarcocystis/immunology , Sarcocystis/isolation & purification , Sarcocystosis/parasitology , Sequence Analysis, DNA/veterinary
19.
Avian Pathol ; 47(3): 286-293, 2018 Jun.
Article in English | MEDLINE | ID: mdl-29517348

ABSTRACT

The detection of avian coronaviruses (AvCoV) in wild birds and the emergence of new AvCoV have increased in the past few years. In the present study, the pathogenicity of three AvCoV isolates was investigated in day-old chicks. One AvCoV isolated from a pigeon, which clustered with the Massachusetts vaccine serotype, and two AvCoV isolated from chickens, which grouped with a Brazilian genotype lineage, were used. Clinical signs, gross lesions, histopathological changes, ciliary activity, viral RNA detection, and serology were evaluated during 42 days post infection. All AvCoV isolates induced clinical signs, gross lesions in the trachea, moderate histopathological changes in the respiratory tract, and mild changes in other tissues. AvCoV isolated from the pigeon sample caused complete tracheal ciliostasis over a longer time span. Specific viral RNA was detected in all tissues, but the highest RNA loads were detected in the digestive tract (cloacal swabs and ileum). The highest antibody levels were also detected in the group infected with an isolate from the pigeon. These results confirm the pathogenicity of Brazilian variants, which can cause disease and induce gross lesions and histopathological changes in chickens. Our results suggest that non-Galliformes birds can also play a role in the ecology of AvCoV.


Subject(s)
Antibodies, Viral/blood , Chickens/virology , Columbidae/virology , Coronavirus Infections/veterinary , Gammacoronavirus/pathogenicity , Poultry Diseases/virology , Tracheal Diseases/veterinary , Animals , Coronavirus Infections/virology , Gammacoronavirus/genetics , Gammacoronavirus/immunology , Gammacoronavirus/isolation & purification , Genotype , Infectious bronchitis virus/genetics , Infectious bronchitis virus/immunology , Infectious bronchitis virus/isolation & purification , Infectious bronchitis virus/pathogenicity , Trachea/virology , Tracheal Diseases/virology
20.
Arch Virol ; 163(4): 1043-1049, 2018 Apr.
Article in English | MEDLINE | ID: mdl-29302792

ABSTRACT

To establish an association between mitochondrial dysfunction and apoptosis following infectious bronchitis virus (IBV) infection, HD11 avian macrophage cells were infected with the Massachusetts 41 (M41) strain. Our results show that the M41 strain of IBV induced cytopathic effects followed by the release of new viral particles. Elevated numbers of apoptotic cells were observed at 24, 48 and 72 h post-infection (p.i.). Viral infection was associated with mitochondrial membrane depolarization and reactive oxygen species (ROS) production at all of the examined timepoints p.i. In summary, IBV M41 replication in infected HD11 macrophages seems to induce mitochondrial bioenergy failure, acting as a respiratory chain uncoupler, without compromising viral replication.


Subject(s)
Host-Pathogen Interactions , Infectious bronchitis virus/pathogenicity , Macrophages/virology , Mitochondria/virology , Virion/pathogenicity , Animals , Apoptosis , Cell Line , Cell Proliferation , Chickens , Infectious bronchitis virus/growth & development , Macrophages/metabolism , Membrane Potential, Mitochondrial , Mitochondria/metabolism , Reactive Oxygen Species/metabolism , Virion/growth & development , Virus Replication
SELECTION OF CITATIONS
SEARCH DETAIL
...