Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 33
Filtrar
Más filtros

Intervalo de año de publicación
1.
J Virol ; 95(18): e0095521, 2021 08 25.
Artículo en Inglés | MEDLINE | ID: mdl-34232725

RESUMEN

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.


Asunto(s)
Patos/virología , Subtipo H5N8 del Virus de la Influenza A/clasificación , Subtipo H5N8 del Virus de la Influenza A/patogenicidad , Gripe Aviar/transmisión , Gripe Aviar/virología , Enfermedades de las Aves de Corral/virología , Proteínas Virales/genética , Animales , Subtipo H5N8 del Virus de la Influenza A/genética , Filogenia , Enfermedades de las Aves de Corral/genética , Virulencia
3.
Arch Virol ; 165(1): 261, 2020 01.
Artículo en Inglés | MEDLINE | ID: mdl-31784908

RESUMEN

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.

4.
Arch Virol ; 163(4): 1043-1049, 2018 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-29302792

RESUMEN

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.


Asunto(s)
Interacciones Huésped-Patógeno , Virus de la Bronquitis Infecciosa/patogenicidad , Macrófagos/virología , Mitocondrias/virología , Virión/patogenicidad , Animales , Apoptosis , Línea Celular , Proliferación Celular , Pollos , Virus de la Bronquitis Infecciosa/crecimiento & desarrollo , Macrófagos/metabolismo , Potencial de la Membrana Mitocondrial , Mitocondrias/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Virión/crecimiento & desarrollo , Replicación Viral
5.
Avian Pathol ; 47(3): 286-293, 2018 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-29517348

RESUMEN

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.


Asunto(s)
Anticuerpos Antivirales/sangre , Pollos/virología , Columbidae/virología , Infecciones por Coronavirus/veterinaria , Gammacoronavirus/patogenicidad , Enfermedades de las Aves de Corral/virología , Enfermedades de la Tráquea/veterinaria , Animales , Infecciones por Coronavirus/virología , Gammacoronavirus/genética , Gammacoronavirus/inmunología , Gammacoronavirus/aislamiento & purificación , Genotipo , Virus de la Bronquitis Infecciosa/genética , Virus de la Bronquitis Infecciosa/inmunología , Virus de la Bronquitis Infecciosa/aislamiento & purificación , Virus de la Bronquitis Infecciosa/patogenicidad , Tráquea/virología , Enfermedades de la Tráquea/virología
6.
Exp Parasitol ; 188: 42-49, 2018 May.
Artículo en Inglés | MEDLINE | ID: mdl-29522766

RESUMEN

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.


Asunto(s)
Alelos , Antígenos de Protozoos/genética , Enfermedades de las Aves/parasitología , Zarigüeyas/parasitología , Sarcocystis/genética , Sarcocistosis/veterinaria , Animales , Antígenos de Superficie/genética , Evolución Biológica , Aves , Encéfalo/parasitología , Brasil , ADN Protozoario/química , ADN Protozoario/aislamiento & purificación , Variación Genética/genética , Pulmón/parasitología , Melopsittacus , Meningoencefalitis/parasitología , Meningoencefalitis/veterinaria , Tipificación de Secuencias Multilocus , Reacción en Cadena de la Polimerasa/veterinaria , Mapaches/parasitología , Sarcocystis/clasificación , Sarcocystis/inmunología , Sarcocystis/aislamiento & purificación , Sarcocistosis/parasitología , Análisis de Secuencia de ADN/veterinaria
7.
Cell Tissue Res ; 367(2): 243-256, 2017 02.
Artículo en Inglés | MEDLINE | ID: mdl-27677269

RESUMEN

The possibility of isolating bovine mesenchymal multipotent stromal cells (MSCs) from fetal adnexa is an interesting prospect due to the potential use of these cells in biotechnological applications. However, little is known about the properties of these progenitor cells in bovine species. Wharton's jelly (WJ) MSC cells were obtained from the umbilical cord of bovine fetuses at three different stages of pregnancy and divided into groups 1, 2 and 3 according to gestational trimester. Cell morphology, from the three stages of pregnancy, typically appeared fibroblast-like spindle-shaped, presenting the same viability and number. Moreover, the proliferative ability of T-cells in response to a mitogenic stimulus was suppressed when WJMSC cells were added to the culture. Multilineage properties were confirmed by their ability to undergo adipogenic, osteogenic/chondrogenic and neurogenic differentiation. Mesenchymal phenotyping, CD105+, CD29+, CD73+ and CD90+ cell markers were detected in all three cell groups, yet these markers were considered more expressed in MSCs of group 2 (p < 0.005). Expression of cytokines IL2, IL6RR, INFAC, INFB1, IFNG, TNF and LTBR were downregulated, whereas IL1F10 expression was upregulated in all tested WJMSCs. The present study demonstrated that WJMSCs harvested from the bovine umbilical cord at different gestational stages showed proliferative capacity, immune privilege and stemness potential.


Asunto(s)
Separación Celular/métodos , Inmunomodulación/genética , Células Madre Mesenquimatosas/citología , Células Madre Multipotentes/citología , Trimestres del Embarazo/genética , Transcripción Genética , Gelatina de Wharton/citología , Animales , Biomarcadores/metabolismo , Bovinos , Diferenciación Celular , Linaje de la Célula , Proliferación Celular , Forma de la Célula , Supervivencia Celular , Femenino , Citometría de Flujo , Perfilación de la Expresión Génica , Células Madre Mesenquimatosas/metabolismo , Células Madre Multipotentes/metabolismo , Fenotipo , Embarazo , Telomerasa/metabolismo , Cordón Umbilical/citología
8.
J Zoo Wildl Med ; 48(2): 529-531, 2017 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-28749292

RESUMEN

Upper respiratory tract disease is a complex infectious disease process with multiple pathogens involved. Identification of infectious agents in wild animals is of great importance for wildlife conservation. The aim of this study was to evaluate the molecular detection of feline herpesvirus type 1, feline calicivirus (FCV), Bordetella bronchiseptica , Chlamydophila felis , and Mycoplasma felis using ocular and nasal swabs in three species of captive nondomestic felids. Mycoplasma felis was detected in two ocular samples of Puma concolor and in one nasal sample of one Panthera onca . FCV was detected in association with M. felis in one P. concolor . The other pathogens tested were not detected. To the authors' knowledge, this is the first report of M. felis in nondomestic felids from Brazil.


Asunto(s)
Bacterias/clasificación , Infecciones Bacterianas/veterinaria , Infecciones por Caliciviridae/veterinaria , Calicivirus Felino/aislamiento & purificación , Felidae , Herpesviridae/clasificación , Infecciones del Sistema Respiratorio/veterinaria , Animales , Bacterias/aislamiento & purificación , Infecciones Bacterianas/diagnóstico , Infecciones Bacterianas/microbiología , Infecciones por Caliciviridae/virología , Herpesviridae/aislamiento & purificación , Infecciones del Sistema Respiratorio/microbiología
9.
J Neurovirol ; 22(6): 725-735, 2016 12.
Artículo en Inglés | MEDLINE | ID: mdl-27311457

RESUMEN

Oncolytic viruses have the ability to infect tumor cells and leave healthy cells intact. In this study, bovine herpesvirus 1 (BHV1; Los Angeles, Cooper, and SV56/90 strains) and bovine herpesvirus 5 (BHV5; SV507/99 and GU9457818 strains) were used to infect two neuronal tumor cell lineages: neuro2a (mouse neuroblastoma cells) and C6 (rat glial cells). BHV1 and BHV5 strains infected both cell lines and positively correlated with viral antigen detection (p < 0.005). When neuro2a cells were infected by Los Angeles, SV507/99, and GU9457818 strains, 40 % of infected cells were under early apoptosis and necroptosis pathways. Infected C6 cells were >40 % in necroptosis phase when infected by BHV5 (GU9457818 strain). Blocking caspase activation did not interfere with cell death. However, when necroptosis was blocked, 60-80 % of both infected cells with either virus switched to early apoptosis pathway with no interference with virus replication. Moreover, reactive oxygen species production and mitochondrial membrane dysfunction were detected at high levels in both infected cell lines. In spite of apoptosis and necroptosis blockage, tumor necrosis factor alpha (TNFA) and virus transcription were positively correlated for all viral strains studied. Thus, these results contribute to the characterization of BHV1 and BHV5 as potential oncolytic viruses for non-human cells. Nonetheless, the mechanisms underlying their oncolytic activity in human cells are still to be determined.


Asunto(s)
Apoptosis/genética , Herpesvirus Bovino 1/crecimiento & desarrollo , Herpesvirus Bovino 5/crecimiento & desarrollo , Necrosis/virología , Neuroglía/virología , Neuronas/virología , Animales , Antígenos Virales/genética , Bovinos , Línea Celular Tumoral , Expresión Génica , Herpesvirus Bovino 1/genética , Herpesvirus Bovino 5/genética , Interacciones Huésped-Patógeno , Humanos , Ratones , Mitocondrias/metabolismo , Mitocondrias/virología , Necrosis/genética , Necrosis/patología , Neuroglía/metabolismo , Neuroglía/patología , Neuronas/metabolismo , Neuronas/patología , Virus Oncolíticos/genética , Virus Oncolíticos/crecimiento & desarrollo , Especificidad de Órganos , Estrés Oxidativo , Ratas , Especies Reactivas de Oxígeno/metabolismo , Factor de Necrosis Tumoral alfa/genética , Factor de Necrosis Tumoral alfa/metabolismo , Replicación Viral
10.
Exp Parasitol ; 164: 71-8, 2016 May.
Artículo en Inglés | MEDLINE | ID: mdl-26905780

RESUMEN

Although few species of Sarcocystis are known to use marsupials of the genus Didelphis as definitive host, an extensive diversity of alleles of surface antigen genes (sag2, sag3, and sag4) has been described in samples of didelphid opossums in Brazil. In this work, we studied 25 samples of Sarcocystis derived from gastrointestinal tract of opossums of the genus Didelphis by accessing the variability of sag2, sag3, sag4, gene encoding cytochrome b (cytB) and first internal transcribed spacer (ITS1). Reference samples of Sarcocystis neurona (SN138) and Sarcocystis falcatula (SF1) maintained in cell culture were also analyzed. We found four allele variants of cytB, seven allele variants of ITS1, 10 allele variants of sag2, 13 allele variants of sag3, and 6 allele variants of sag4. None of the sporocyst-derived sequences obtained from Brazilian opossums revealed 100% identity to SN138 at cytB gene, nor to SN138 or SF1 at ITS1 locus. In addition, none of the sag alleles were found identical to either SF1 or SN138 homologous sequences, and a high number of new sag allele types were found other than those previously described in Brazil. Out of ten sag2 alleles, four are novel, while eight out of 13 sag3 alleles are novel and one out of six sag4 alleles is novel. Further studies are needed to clarify if such a vast repertoire of allele variants of Sarcocystis is the consequence of re-assortments driven by sexual exchange, in order to form individuals with highly diverse characteristics, such as pathogenicity, host spectrum, among others or if it only represents allele variants of different species with different biological traits.


Asunto(s)
ADN Protozoario/química , Didelphis/parasitología , Variación Genética , Filogenia , Sarcocystis/clasificación , Alelos , Animales , Antígenos de Protozoos/genética , Antígenos de Superficie/genética , Brasil , Citocromos b/genética , ADN Intergénico/genética , ADN Protozoario/genética , Tracto Gastrointestinal/parasitología , Genotipo , Melopsittacus , Reacción en Cadena de la Polimerasa , Proteínas Protozoarias/genética , Mapaches , Sarcocystis/genética , Sarcocistosis/parasitología , Sarcocistosis/veterinaria , Análisis de Secuencia de ADN
11.
J Mol Evol ; 81(1-2): 21-3, 2015 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-26250156

RESUMEN

This study showed that the most of the coronaviruses (CoVs) detected in Brazilian wild birds clustered with the mouse hepatitis virus A59 strain, belonging to the BetaCoV group. Furthermore, CoV detected in two different bird species, Amazona vinacea and Brotogeris tirica, clustered with a CoV isolated from Sparrow (SpaCoV HKU17) belonging to a monophyletic group related with the CoVs isolated from swines (PorCoV HKU15), both belonging to the DeltaCoV genus, previously unreported in South America. Considering the risk of inter-species host switching and further adaptation to new hosts, detection in bird species of CoVs closely related to mammal CoVs should warn for the potential emergence of new threatening viruses.


Asunto(s)
Evolución Biológica , Enfermedades de las Aves/virología , Infecciones por Coronavirus/veterinaria , Coronavirus/genética , Animales , Brasil , Infecciones por Coronavirus/virología , Genoma Viral , Especificidad del Huésped , Mamíferos/virología , Filogenia
12.
Arch Virol ; 160(11): 2683-91, 2015 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-26239341

RESUMEN

Members of the subfamily Alphaherpesvirinae use the epithelium of the upper respiratory and/or genital tract as preferential sites for primary replication. However, bovine herpesvirus 5 (BoHV5) is neurotropic and neuroinvasive and responsible for meningoencephalitis in cattle and in animal models. A related virus, BoHV1 has also been occasionally implicated in natural cases of neurological infection and disease in cattle. The aim of the present study was to assess the in vitro effects of BoHV1 and BoHV5 replication in neuron-like cells. Overall, cytopathic effects, consisting of floating rounded cells, giant cells and monolayer lysis, induced by both viruses at 48 h postinfection (p.i.) resulted in a loss of cell viability and high virus titres (r = 0.978). The BoHV1 Cooper strain produced the lowest titres in neuron-like cells, although viral DNA was detected in infected cells during all experiments. Virus replication in infected cells was demonstrated by immunocytochemistry, flow cytometry and qPCR assays. BoHV antigens were better visualized at 48 h p.i. and flow cytometry analysis showed that SV56/90 and Los Angeles antigens were present at higher levels. In spite of the fact that BoHV titres dropped at 48 h p.i, viral DNA remained detectable until 120 h p.i. Sensitive TUNEL (terminal deoxynucleotidyltransferase-mediated dUTP-biotin nick end labeling) and annexin V assays were used to identify apoptosis. BoHV5 induced death in approximately 50% of cells within 24 h p.i., similar to what has been observed for BoHV1 Los Angeles. Infection with the BoHV1 Cooper strain resulted in 26.37% of cells being in the early stages of apoptosis; 63.69% of infected cells were considered viable. Modulation of mitochondrial function, as measured by mitochondrial membrane depolarization, was synchronous with the virus replication cycle, cell viability and virus titres at 48 h p.i. Our results indicate that apoptosis plays an important role in preventing neuronal death and provides a bovine-derived in vitro system to study herpesvirus-neuron interactions.


Asunto(s)
Enfermedades de los Bovinos/virología , Infecciones por Herpesviridae/veterinaria , Herpesvirus Bovino 1/fisiología , Herpesvirus Bovino 5/fisiología , Neuronas/virología , Replicación Viral , Animales , Apoptosis , Bovinos , Enfermedades de los Bovinos/fisiopatología , Células Cultivadas , Infecciones por Herpesviridae/fisiopatología , Infecciones por Herpesviridae/virología , Herpesvirus Bovino 1/genética , Herpesvirus Bovino 5/genética
13.
Arch Virol ; 160(7): 1785-90, 2015 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-25951972

RESUMEN

The ability of avian coronaviruses to replicate in mice was investigated to investigate interspecies transmission. Two inbred mouse strains (BALB/c and A/J) with different genetic backgrounds were inoculated with the avian coronavirus strains Mass and BR-I and monitored for at least 10 days. Analysis of viral RNA, histopathological examinations, immunohistochemistry and serology were performed. After virus inoculation, neither clinical signs nor evident gross lesions were observed. Viral RNA, histopathological changes, and viral nucleoprotein were observed in the lung, trachea and sinus of all inoculated mice. Our study demonstrates the importance of elucidating the epidemiology of coronaviruses, including in rodents that are pests in poultry production.


Asunto(s)
Infecciones por Coronavirus/veterinaria , Virus de la Bronquitis Infecciosa/fisiología , Animales , Enfermedades de las Aves/genética , Enfermedades de las Aves/patología , Enfermedades de las Aves/virología , Infecciones por Coronavirus/genética , Infecciones por Coronavirus/patología , Infecciones por Coronavirus/virología , Modelos Animales de Enfermedad , Virus de la Bronquitis Infecciosa/genética , Virus de la Bronquitis Infecciosa/patogenicidad , Pulmón/patología , Pulmón/virología , Ratones , Ratones Endogámicos BALB C , Tráquea/patología , Tráquea/virología
14.
Vaccines (Basel) ; 9(2)2021 Feb 21.
Artículo en Inglés | MEDLINE | ID: mdl-33669907

RESUMEN

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.

15.
Viruses ; 13(1)2021 Jan 13.
Artículo en Inglés | MEDLINE | ID: mdl-33451125

RESUMEN

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.


Asunto(s)
Pollos/virología , Genotipo , Enfermedad de Newcastle/epidemiología , Enfermedad de Newcastle/virología , Virus de la Enfermedad de Newcastle/clasificación , Virus de la Enfermedad de Newcastle/genética , Enfermedades de las Aves de Corral/epidemiología , Enfermedades de las Aves de Corral/virología , Animales , Granjas , Genoma Viral , Genómica/métodos , Kenia/epidemiología , Epidemiología Molecular , Virus de la Enfermedad de Newcastle/aislamiento & purificación , Virus de la Enfermedad de Newcastle/patogenicidad , Filogenia , Filogeografía , Vigilancia en Salud Pública , ARN Viral , Análisis Espacio-Temporal , Virulencia
16.
Viruses ; 13(12)2021 12 06.
Artículo en Inglés | MEDLINE | ID: mdl-34960715

RESUMEN

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.


Asunto(s)
Columbidae , Brotes de Enfermedades/veterinaria , Enfermedad de Newcastle/epidemiología , Virus de la Enfermedad de Newcastle/genética , Animales , Brasil/epidemiología , Genoma Viral , Genotipo , Secuenciación de Nucleótidos de Alto Rendimiento , Enfermedad de Newcastle/patología , Enfermedad de Newcastle/virología , Virus de la Enfermedad de Newcastle/clasificación , Virus de la Enfermedad de Newcastle/aislamiento & purificación , Virus de la Enfermedad de Newcastle/patogenicidad , Filogenia , Virulencia , Secuenciación Completa del Genoma
17.
Vaccine ; 38(34): 5507-5515, 2020 07 22.
Artículo en Inglés | MEDLINE | ID: mdl-32591288

RESUMEN

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.


Asunto(s)
Enfermedad de Newcastle , Enfermedades de las Aves de Corral , Vacunas Virales , Animales , Anticuerpos Antivirales , California , Pollos , Enfermedad de Newcastle/prevención & control , Virus de la Enfermedad de Newcastle/genética , Enfermedades de las Aves de Corral/prevención & control , Vacunación , Vacunas Atenuadas , Vacunas Sintéticas/genética , Vacunas Virales/genética
18.
Infect Genet Evol ; 78: 104074, 2020 03.
Artículo en Inglés | MEDLINE | ID: mdl-31634645

RESUMEN

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.


Asunto(s)
Huevos/virología , Subtipo H9N2 del Virus de la Influenza A/patogenicidad , Gripe Aviar/diagnóstico , Orofaringe/virología , Virus Reordenados/patogenicidad , Animales , Pollos , Subtipo H9N2 del Virus de la Influenza A/clasificación , Subtipo H9N2 del Virus de la Influenza A/genética , Gripe Aviar/virología , Kenia , Filogenia , Vigilancia de la Población , Virus Reordenados/clasificación , Virus Reordenados/genética , Análisis de Secuencia de ARN , Esparcimiento de Virus , Secuenciación Completa del Genoma
19.
Vet Microbiol ; 235: 25-34, 2019 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-31282376

RESUMEN

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.


Asunto(s)
Pollos/virología , Columbidae/virología , Enfermedad de Newcastle/transmisión , Virus de la Enfermedad de Newcastle/patogenicidad , Enfermedades de las Aves de Corral/transmisión , Animales , Animales Salvajes/virología , Vacunación , Virulencia , Esparcimiento de Virus
20.
PLoS One ; 14(9): e0222457, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31513638

RESUMEN

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.


Asunto(s)
Subtipo H7N3 del Virus de la Influenza A/genética , Gripe Aviar/epidemiología , Enfermedades de las Aves de Corral/genética , Animales , Aves/genética , Pollos/genética , Pollos/virología , Brotes de Enfermedades , Evolución Molecular , Subtipo H7N3 del Virus de la Influenza A/patogenicidad , Gripe Aviar/virología , México/epidemiología , Filogenia , Aves de Corral/genética , Enfermedades de las Aves de Corral/virología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA