Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 26
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Viruses ; 15(10)2023 10 17.
Artículo en Inglés | MEDLINE | ID: mdl-37896883

RESUMEN

Influenza A virus (IAV) populations harbor large subpopulations of defective-interfering particles characterized by internally deleted viral genomes. These internally deleted genomes have demonstrated the ability to suppress infectivity and boost innate immunity, rendering them promising for therapeutic and immunogenic applications. In this study, we aimed to investigate the diversity and complexity of the internally deleted IAV genomes within a panel of plaque-purified avian influenza viruses selected for their enhanced interferon-inducing phenotypes. Our findings unveiled that the abundance and diversity of internally deleted viral genomes were contingent upon the viral subculture and plaque purification processes. We observed a heightened occurrence of internally deleted genomes with distinct junctions in viral clones exhibiting enhanced interferon-inducing phenotypes, accompanied by additional truncation in the nonstructural 1 protein linker region (NS1Δ76-86). Computational analyses suggest the internally deleted IAV genomes can encode a broad range of carboxy-terminally truncated and intrinsically disordered proteins with variable lengths and amino acid composition. Further research is imperative to unravel the underlying mechanisms driving the increased diversity of internal deletions within the genomes of viral clones exhibiting enhanced interferon-inducing capacities and to explore their potential for modulating cellular processes and immunity.


Asunto(s)
Virus de la Influenza A , Gripe Humana , Animales , Humanos , Interferones/genética , Inmunidad Innata , ARN Viral/genética , Genoma Viral , Proteínas no Estructurales Virales/metabolismo , Replicación Viral/genética
2.
Avian Dis ; 67(1): 94-101, 2023 03.
Artículo en Inglés | MEDLINE | ID: mdl-37140117

RESUMEN

Despite the essential role of innate immunity in defining the outcome of viral infections, the roles played by different components of the avian innate immune system are poorly delineated. Here, we investigated the potential implication of avian toll-like receptor (TLR) 3 (TLR3) and melanoma differentiation-associated (MDA) gene 5 (MDA5) receptors of double-stranded RNA (dsRNA) in induction of the interferon pathway and avian orthoavulavirus 1 (AOAV-1) replication in chicken-origin DF-1 fibroblast cells. TLR3 and MDA5 knockout (KO) DF-1 cells were generated using our avian-specific CRISPR/Cas9 system and stimulated with a synthetic dsRNA ligand polyinosinic:polycytidylic acid [poly(I:C)] or infected with AOAV-1 (previously known as Newcastle disease virus). Poly(I:C) treatment in cell culture media resulted in significant upregulation of interferon (IFN)α, IFNß, and Mx1 gene expression in wild type (WT) DF-1 cells but not in TLR3-MDA5 double KO cells. Interestingly, poly(I:C) treatment induced rapid cell degeneration in WT and MDA5 KO cells, but not in TLR3 knockout or TRL3-MDA5 double knockout (DKO) cells, directly linking poly(I:C)-induced cell degeneration to TLR3-mediated host response. The double knockout cells supported significantly higher replication of AOAV-1 virus than did the WT cells. However, no correlation between the level of virus replication and type I IFN response was observed. Our study suggests that innate immune response is host- and pathogen specific, and further investigation is needed to understand the relevance of dsRNA receptor-mediated immune responses in viral replication and pathogenesis in avian species.


Nota de investigación- En bloqueo de los genes TLR3 y MDA5 en las células DF-1 mejoran la replicación de Ortoavulavirus aviar 1. A pesar del papel esencial de la inmunidad innata en la definición del resultado de las infecciones virales, las funciones que desempeñan los diferentes componentes del sistema inmunitario innato aviar no están completamente definidas. En este estudio se investigó el posible papel del receptor aviar tipo toll (TLR) número 3 (TLR3) y los receptores de ARN de doble cadena (dsRNA del gene asociado a la diferenciación de melanoma (MDA) número 5 (MDA5) en la inducción de la vía del interferón y en la replicación del Ortoavulavirus 1 (AOAV-1) en células de fibroblastos DF-1 de origen en pollo. Las células DF-1 con los genes TLR3 y MDA5 bloqueado (KO) se generaron utilizando nuestro sistema CRISPR/Cas9 específico para aves y se estimularon con un ligando de dsRNA sintético poliinosínico: ácido policitidílico [poli(I:C)] o se infectaron con AOAV-1 (anteriormente conocido como el virus de la enfermedad de Newcastle). El tratamiento con poli(I:C) en medios de cultivo celular resultó en una regulación positiva significativa de la expresión génica de interferón (IFN)α, IFNß y Mx1 en células DF-1 de tipo silvestre (WT) pero no en células con doble bloqueo TLR3-MDA5 (DKO). Curiosamente, el tratamiento con poli(I:C) indujo una rápida degeneración celular en las células silvestres (WT) y las células con el gene MDA5 bloqueado, pero no en las células con bloqueo del gene TLR3 o con las células con doble bloqueo de TRL3-MDA5, lo que vincula directamente la degeneración celular inducida por poli(I:C) con la respuesta de la huésped mediada por TLR3. Las células con doble bloqueo soportaron una replicación significativamente mayor del Ortoavulavirus 1 que las células silvestres. Sin embargo, no se observó correlación entre el nivel de replicación del virus y la respuesta de IFN tipo I. Este estudio sugiere que la respuesta inmune innata es específica del huésped y del patógeno, y se necesita más investigación para comprender la relevancia de las respuestas inmunes mediadas por el receptor dsRNA en la replicación viral y en la patogénesis en las especies aviares.


Asunto(s)
Enfermedades de las Aves de Corral , Receptor Toll-Like 3 , Animales , Receptor Toll-Like 3/genética , Receptor Toll-Like 3/metabolismo , Virus de la Enfermedad de Newcastle/genética , Inmunidad Innata , ARN Bicatenario , Interferones/genética
3.
Vaccine ; 41(11): 1848-1858, 2023 03 10.
Artículo en Inglés | MEDLINE | ID: mdl-36669965

RESUMEN

Emerging avian influenza viruses pose a high risk to poultry production, necessitating the need for more broadly protective vaccines. Live attenuated influenza vaccines offer excellent protective efficacies but their use in poultry farms is discouraged due to safety concerns related to emergence of reassortant viruses. Vaccination of chicken embryos inside eggs (in ovo) induces early immunity in young chicks while reduces the safety concerns related to the use of live vaccines on farms. However, in ovo vaccination using influenza viruses severely affects the egg hatchability. We previously engineered a high interferon-inducing live attenuated influenza vaccine candidate with an enhanced protective efficacy in chickens. Here, we asked whether we could further modify this high interferon-inducing vaccine candidate to develop an in ovo-compatible live attenuated influenza vaccine. We first showed that the enhanced interferon responses induced by the vaccine is not enough to attenuate the virus in ovo. To reduce the pathogenicity of the virus for chicken embryos, we replaced the hemagglutinin cleavage site of the H7 vaccine virus (PENPKTR/GL) with that of the H6-subtype viruses (PQIETR/GL) and disrupted the ribosomal frameshifting site responsible for viral polymerase acidic X protein expression. In ovo vaccination of chickens with up to 105 median egg infectious dose of the modified vaccine had minimal effects on hatchability while protecting the chickens against a heterologous challenge virus at two weeks of age. This study demonstrates that targeted genetic mutations can be applied to further attenuate and enhance the safety of live attenuated influenza vaccines to develop future in ovo vaccines for poultry.


Asunto(s)
Vacunas contra la Influenza , Gripe Aviar , Embrión de Pollo , Animales , Pollos , Hemaglutininas , Proteínas Virales/genética , Vacunas Atenuadas , Interferones , Glicoproteínas Hemaglutininas del Virus de la Influenza/genética , Anticuerpos Antivirales
4.
Avian Dis ; 65(3): 419-428, 2021 09.
Artículo en Inglés | MEDLINE | ID: mdl-34427417

RESUMEN

Toll-like receptor 3 (TLR3) and melanoma differentiation-associated gene 5 (MDA5) are double-stranded RNA (dsRNA)-recognizing receptors that mediate innate immune responses to virus infection. However, the roles played by these receptors in the pathogenesis of avian viruses are poorly understood. In this study, we generated TLR3 and MDA5 single knockout (SKO) and TLR3-MDA5 double knockout (DKO) quail fibroblast cells and examined dsRNA receptor-mediated innate immune responses in vitro. The knockout cells were then stimulated with a synthetic dsRNA ligand polyinosinic:polycytidylic acid [poly(I:C)] or influenza A virus. Endosomal stimulation of TLR3 by adding poly(I:C) in cell culture media or cytoplasmic stimulation of MDA5 by transfecting poly(I:C) resulted in significant increases of TLR3, MDA5, interferon (IFN) ß, and interleukin 8 gene expression levels in wild type (WT) cells. Endosomal poly(I:C) treatment induced a higher level expression of most of the genes tested in MDA5 SKO cells compared with WT cells, but not in TLR3 SKO and DKO cells. Cytoplasmic transfection of poly(I:C) led to significant upregulation of all four genes in WT, TLR3 SKO, and MDA5 SKO cells at 8 hr posttransfection and negligible gene expression changes in TLR3-MDA5 DKO cells. Upon infection with a strain of influenza virus with compromised IFN antagonistic capability, WT cells produced the highest amount of biologically active type I IFN followed by TLR3 SKO and MDA5 SKO cells. DKO cells did not produce detectable amounts of type I IFN. However, the IFN did not induce an antiviral state fast enough to block virus replication, even in WT cells under the experimental conditions employed. In summary, our data demonstrate that TLR3 and MDA5 are the key functional dsRNA receptors in quail and imply their coordinated roles in the induction of innate immune responses upon virus infection.


Evaluación de las respuestas inmunitarias mediadas por TLR3 y MDA5 utilizando células de fibroblastos de codorniz con genes eliminados. El receptor tipo Toll 3 (TLR3) y el gene 5 asociado a la diferenciación de melanoma (MDA5) son receptores de reconocimiento de ARN de doble cadena (dsRNA) que median las respuestas inmunitarias innatas a la infección por virus. Sin embargo, no se conocen bien las funciones que desempeñan estos receptores en la patogenia de los virus aviares. En este estudio, se generaron células de fibroblastos de codorniz con eliminación simple de los genes TLR3 y MDA5 (SKO) y eliminación doble de los genes TLR3-MDA5 (DKO) y se examinaron las respuestas inmunitarias innatas mediadas por el receptor de dsRNA in vitro. Posteriormente, las células con genes eliminados se estimularon con un ligando sintético de ARN de doble cadena poliinosínico: ácido policitidílico [poli (I: C)] o con el virus de la influenza A. La estimulación endosómica de TLR3 mediante la adición de poli(I: C) en medios de cultivo celular, o la estimulación citoplásmica de MDA5 mediante la transfección de poli(I: C), dieron como resultado aumentos significativos de los niveles de expresión de los genes para TLR3, MDA5, interferón (IFN) ß e interleucina 8 en células de tipo silvestre (WT). El tratamiento con poli(I: C) endosómico indujo un nivel de expresión más alto de la mayoría de los genes analizados en las células con eliminación simple del gene MDA5 en comparación con las células silvestres, pero no en las células con eliminación simple del gene TLR3 y con eliminación doble de genes. La transfección citoplásmica de poli(I: C) condujo a una regulación positiva significativa de los cuatro genes en las células silvestres, en las células con eliminación simple del gene TLR3 y en las células con eliminación simple del gene MDA5 a las ocho horas posteriores a la transfección y cambios insignificantes en la expresión de genes en las células con eliminación doble de los genes TLR3 y MDA5. Durante la infección con una cepa del virus de la influenza con una capacidad antagonista para IFN comprometida, las células silvestres produjeron la mayor cantidad de IFN de tipo I biológicamente activo, seguidas de las células con eliminación simple del gene TLR3 y de las células con eliminación simple del gene MDA5. Las células con eliminación doble de genes no produjeron cantidades detectables de IFN de tipo I. Sin embargo, el IFN no indujo un estado antiviral lo suficientemente rápido como para bloquear la replicación del virus, incluso en células silvestres bajo las condiciones experimentales empleadas. En resumen, los datos de este estudio demuestran que TLR3 y MDA5 son los receptores de ARN de doble cadena funcionales clave en la codorniz e implican sus funciones coordinadas en la inducción de respuestas inmunitarias innatas durante la infección por virus.


Asunto(s)
Codorniz , Receptor Toll-Like 3 , Animales , Fibroblastos , Inmunidad Innata , Poli I-C/farmacología , Receptor Toll-Like 3/genética
5.
PeerJ ; 9: e11806, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34327060

RESUMEN

Turkey respiratory and gut microbiota play important roles in promoting health and production performance. Loss of microbiota homeostasis due to pathogen infection can worsen the disease or predispose the bird to infection by other pathogens. While turkeys are highly susceptible to influenza viruses of different origins, the impact of influenza virus infection on turkey gut and respiratory microbiota has not been demonstrated. In this study, we investigated the relationships between low pathogenicity avian influenza (LPAI) virus replication, cytokine gene expression, and respiratory and gut microbiota disruption in specific-pathogen-free turkeys. Differential replication of two LPAI H5N2 viruses paralleled the levels of clinical signs and cytokine gene expression. During active virus shedding, there was significant increase of ileal and nasal bacterial contents, which inversely corresponded with bacterial species diversity. Spearman's correlation tests between bacterial abundance and local viral titers revealed that LPAI virus-induced dysbiosis was strongest in the nasal cavity followed by trachea, and weakest in the gut. Significant correlations were also observed between cytokine gene expression levels and relative abundances of several bacteria in tracheas of infected turkeys. For example, interferon γ/λ and interleukin-6 gene expression levels were correlated positively with Staphylococcus and Pseudomonas abundances, and negatively with Lactobacillus abundance. Overall, our data suggest a potential relationship where bacterial community diversity and enrichment or depletion of several bacterial genera in the gut and respiratory tract are dependent on the level of LPAI virus replication. Further work is needed to establish whether respiratory and enteric dysbiosis in LPAI virus-infected turkeys is a result of host immunological responses or other causes such as changes in nutritional uptake.

6.
PLoS One ; 16(1): e0241732, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33406075

RESUMEN

Characterization of poultry microbiota is becoming increasingly important due to the growing need for microbiome-based interventions to improve poultry health and production performance. However, the lack of standardized protocols for sampling, sample processing, DNA extraction, sequencing, and bioinformatic analysis can hinder data comparison between studies. Here, we investigated how the DNA extraction process affects microbial community compositions and diversity metrics in different chicken respiratory sample types including choanal and tracheal swabs, nasal cavity and tracheal washes, and lower respiratory lavage. We did a side-by-side comparison of the performances of Qiagen DNeasy blood and tissue (BT) and ZymoBIOMICS DNA Miniprep (ZB) kits. In general, samples extracted with the BT kit yielded higher concentrations of total DNA while those extracted with the ZB kit contained higher numbers of bacterial 16S rRNA gene copies per unit volume. Therefore, the samples were normalized to equal amounts of 16S rRNA gene copies prior to sequencing. For each sample type, all predominant bacterial taxa detected in samples extracted with one kit were present in replicate samples extracted with the other kit and did not show significant differences at the class level. However, a few differentially abundant shared taxa were observed at family and genus levels. Furthermore, between-kit differences in alpha and beta diversity metrics at the amplicon sequence variant level were statistically indistinguishable. Therefore, both kits perform similarly in terms of 16S rRNA gene-based poultry microbiome analysis for the sample types analyzed in this study.


Asunto(s)
Pollos/microbiología , ADN Bacteriano , ADN Ribosómico , Microbiota , ARN Ribosómico 16S , Juego de Reactivos para Diagnóstico , Sistema Respiratorio/microbiología , Animales , ADN Bacteriano/química , ADN Bacteriano/genética , ADN Bacteriano/aislamiento & purificación , ADN Ribosómico/genética , ADN Ribosómico/aislamiento & purificación , ARN Ribosómico 16S/genética , ARN Ribosómico 16S/aislamiento & purificación
7.
Poult Sci ; 99(12): 6513-6524, 2020 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-33248566

RESUMEN

Toll-like receptor 3 (TLR3) induces host innate immune response on recognition of viral double-stranded RNA (dsRNA). Although several studies of avian TLR3 have been reported, none of these studies used a gene knockout (KO) model to directly assess its role in inducing the immune response and effect on other dsRNA receptors. In this study, we determined the coding sequence of quail TLR3, identified isoforms, and generated TLR3 KO quail fibroblast (QT-35) cells using a CRISPR/Cas9 system optimized for avian species. The TLR3-mediated immune response was studied by stimulating the wild-type (WT) and KO QT-35 cells with synthetic dsRNA or polyinosinic:polycytidylic acid [poly(I:C)] or infecting the cells with different RNA viruses such as influenza A virus, avian reovirus, and vesicular stomatitis virus. The direct poly(I:C) treatment significantly increased IFN-ß and IL-8 gene expression along with the cytoplasmic dsRNA receptor, melanoma differentiation-associated gene 5 (MDA5), in WT cells, whereas no changes in all corresponding genes were observed in KO cells. We further confirmed the antiviral effects of poly(I:C)-induced TLR3-mediated immunity by demonstrating significant reduction of virus titer in poly(I:C)-treated WT cells, but not in TLR3 KO cells. On virus infection, varying levels of IFN-ß, IL-8, TLR3, and MDA5 gene upregulation were observed depending on the viruses. No major differences in gene expression level were observed between WT and TLR3 KO cells, which suggests a relatively minor role of TLR3 in sensing and exerting immune response against the viruses tested in vitro. Our data show that quail TLR3 is an important endosomal dsRNA receptor responsible for regulation of type I interferon and proinflammatory cytokine, and affect the expression of MDA5, another dsRNA receptor, most likely through cytokine-mediated communication.


Asunto(s)
Aves , Inmunidad , Isoformas de Proteínas , Receptor Toll-Like 3 , Animales , Aves/inmunología , Células Cultivadas , Fibroblastos/inmunología , Inmunidad/inmunología , Poli I-C/farmacología , Isoformas de Proteínas/inmunología , Codorniz/inmunología , Receptor Toll-Like 3/química , Receptor Toll-Like 3/inmunología
8.
Avian Dis ; 64(3): 277-285, 2020 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-33205170

RESUMEN

Although poultry microbiome discoveries are increasing due to the potential impact on poultry performance, studies examining the poultry respiratory microbiome are challenging because of the low microbial biomass and uniqueness of the avian respiratory tract, making it difficult to sample enough material for microbial analysis. Invasive sampling techniques requiring euthanasia are currently used to increase microbial mass for the analysis, thus making it impossible to sample individual birds longitudinally. In this study, we compared invasive (nasal wash, upper tracheal wash, lower tracheal wash, and lower respiratory lavage) and noninvasive (tracheal and choanal swabs) respiratory sampling techniques in two independent experiments by using 4-wk-old chickens. We first established the experimental baseline of respiratory microbiota by using invasive techniques to enable reasonable comparisons between sampling methods and between experiments. Although noninvasive sampling (live-bird swabs) resulted in lower 16S ribosomal RNA gene copy numbers compared with invasive sampling, live swabs were able to detect the dominant microbes captured by invasive techniques. Nevertheless, swabs from euthanatized birds were more reflective of the microbiota captured through invasive methods than live swab. Furthermore, from two separate experiments, we also demonstrated that respiratory microbiota sampling is highly reproducible, especially in the trachea and lower respiratory tract. Our study provides new insights and perspectives on decision making when sampling and studying poultry respiratory microbiota.


Asunto(s)
Bacterias/aislamiento & purificación , Pollos/microbiología , Microbiota , Sistema Respiratorio/microbiología , Manejo de Especímenes/veterinaria , Animales , Bacterias/genética , ARN Bacteriano/análisis , ARN Ribosómico 16S/análisis , Análisis de Secuencia de ADN/veterinaria , Manejo de Especímenes/instrumentación , Manejo de Especímenes/métodos
9.
J Virol ; 95(2)2020 12 22.
Artículo en Inglés | MEDLINE | ID: mdl-33115873

RESUMEN

Influenza A viruses continue to circulate among wild birds and poultry worldwide, posing constant pandemic threats to humans. Effective control of emerging influenza viruses requires new broadly protective vaccines. Live attenuated influenza vaccines with truncations in nonstructural protein 1 (NS1) have shown broad protective efficacies in birds and mammals, which correlate with the ability to induce elevated interferon responses in the vaccinated hosts. Given the extreme diversity of influenza virus populations, we asked if we could improve an NS1-truncated live attenuated influenza vaccine developed for poultry (PC4) by selecting viral subpopulations with enhanced interferon-inducing capacities. Here, we deconstructed a de novo population of PC4 through plaque isolation, created a large library of clones, and assessed their interferon-inducing phenotypes. While most of the clones displayed the parental interferon-inducing phenotype in cell culture, few clones showed enhanced interferon-inducing phenotypes in cell culture and chickens. The enhanced interferon-inducing phenotypes were linked to either a deletion in NS1 (NS1Δ76-86) or a substitution in polymerase basic 2 protein (PB2-D309N). The NS1Δ76-86 deletion disrupted the putative eukaryotic translation initiation factor 4GI-binding domain and promoted the synthesis of biologically active interferons. The PB2-D309N substitution enhanced the early transcription of interferon mRNA, revealing a novel role for the 309D residue in suppression of interferon responses. We combined these mutations to engineer a novel vaccine candidate that induced additive amounts of interferons and stimulated protective immunity in chickens. Therefore, viral subpopulation screening approaches can guide the design of live vaccines with strong immunostimulatory properties.IMPORTANCE Effectiveness of NS1-truncated live attenuated influenza vaccines relies heavily on their ability to induce elevated interferon responses in vaccinated hosts. Influenza viruses contain diverse particle subpopulations with distinct phenotypes. We show that live influenza vaccines can contain underappreciated subpopulations with enhanced interferon-inducing phenotypes. The genomic traits of such virus subpopulations can be used to further improve the efficacy of the current live vaccines.


Asunto(s)
Virus de la Influenza A/inmunología , Vacunas contra la Influenza/inmunología , Interferones/inmunología , ARN Polimerasa Dependiente del ARN/genética , Proteínas no Estructurales Virales/genética , Proteínas Virales/genética , Animales , Anticuerpos Antivirales/inmunología , Línea Celular , Pollos , Inmunidad Innata , Virus de la Influenza A/genética , Vacunas contra la Influenza/administración & dosificación , Vacunas contra la Influenza/genética , Gripe Aviar/inmunología , Gripe Aviar/prevención & control , Interferones/genética , Mutación , Fenotipo , ARN Polimerasa Dependiente del ARN/inmunología , Vacunación/veterinaria , Vacunas Atenuadas/administración & dosificación , Vacunas Atenuadas/genética , Vacunas Atenuadas/inmunología , Proteínas no Estructurales Virales/inmunología , Proteínas Virales/inmunología
10.
Appl Environ Microbiol ; 86(12)2020 06 02.
Artículo en Inglés | MEDLINE | ID: mdl-32276973

RESUMEN

Communities of gut bacteria (microbiota) are known to play roles in resistance to pathogen infection and optimal weight gain in turkey flocks. However, knowledge of turkey respiratory microbiota and its link to gut microbiota is lacking. This study presents a 16S rRNA gene-based census of the turkey respiratory microbiota (nasal cavity and trachea) alongside gut microbiota (cecum and ileum) in two identical commercial Hybrid Converter turkey flocks raised in parallel under typical field commercial conditions. The flocks were housed in adjacent barns during the brood stage and in geographically separated farms during the grow-out stage. Several bacterial taxa, primarily Staphylococcus, that were acquired in the respiratory tract at the beginning of the brood stage persisted throughout the flock cycle. Late-emerging predominant taxa in the respiratory tract included Deinococcus and Corynebacterium Tracheal and nasal microbiota of turkeys were identifiably distinct from one another and from gut microbiota. Nevertheless, gut and respiratory microbiota changed in parallel over time and appeared to share many taxa. During the brood stage, the two flocks generally acquired similar gut and respiratory microbiota, and their average body weights were comparable. However, there were qualitative and quantitative differences in microbial profiles and body weight gain trajectories after the flocks were transferred to geographically separated grow-out farms. Lower weight gain corresponded to the emergence of Deinococcus and Ornithobacterium in the respiratory tract and Fusobacterium and Parasutterella in gut. This study provides an overview of turkey microbiota under field conditions and suggests several hypotheses concerning the respiratory microbiome.IMPORTANCE Turkey meat is an important source of animal protein, and the industry around its production contributes significantly to the agricultural economy. The microorganisms present in the gut of turkeys are known to impact bird health and flock performance. However, the respiratory microbiota in turkeys is entirely unexplored. This study has elucidated the microbiota of respiratory tracts of turkeys from two commercial flocks raised in parallel throughout a normal flock cycle. Further, the study suggests that bacteria originating in the gut or in poultry house environments influence respiratory communities; consequently, they induce poor performance, either directly or indirectly. Future attempts to develop microbiome-based interventions for turkey health should delimit the contributions of respiratory microbiota and aim to limit disturbances to those communities.


Asunto(s)
Ciego/microbiología , Íleon/microbiología , Microbiota , Cavidad Nasal/microbiología , Tráquea/microbiología , Pavos/microbiología , Aumento de Peso , Animales , Fenómenos Fisiológicos Bacterianos , Trayectoria del Peso Corporal , Microbioma Gastrointestinal , Masculino
11.
Annu Rev Anim Biosci ; 8: 247-267, 2020 02 15.
Artículo en Inglés | MEDLINE | ID: mdl-31479617

RESUMEN

The concept of influenza A virus (IAV) subpopulations emerged approximately 75 years ago, when Preben von Magnus described "incomplete" virus particles that interfere with the replication of infectious virus. It is now widely accepted that infectious particles constitute only a minor portion of biologically active IAV subpopulations. The IAV quasispecies is an extremely diverse swarm of biologically and genetically heterogeneous particle subpopulations that collectively influence the evolutionary fitness of the virus. This review summarizes the current knowledge of IAV subpopulations, focusing on their biologic and genomic diversity. It also discusses the potential roles IAV subpopulations play in virus pathogenesis and live attenuated influenza vaccine development.


Asunto(s)
Virus de la Influenza A/genética , Virus de la Influenza A/patogenicidad , Animales , Genoma Viral , Humanos , Virus de la Influenza A/inmunología , Vacunas contra la Influenza/genética , Vacunas contra la Influenza/inmunología , Gripe Humana/inmunología , Gripe Humana/prevención & control , Gripe Humana/virología , Infecciones por Orthomyxoviridae/virología , Vacunas Atenuadas
12.
Vaccine ; 37(43): 6454-6462, 2019 10 08.
Artículo en Inglés | MEDLINE | ID: mdl-31506195

RESUMEN

Development of a broadly reactive influenza vaccine that can provide protection against emerging type A influenza viruses is a big challenge. We previously demonstrated that a vaccine displaying the extracellular domain of the matrix protein 2 (M2e) on the surface loops of norovirus P-particle (M2eP) can partially protect chickens against several subtypes of avian influenza viruses. In the current study, a chimeric vaccine containing a conserved peptide from the subunit 2 of hemagglutinin (HA) glycoprotein (HA2) and Arabidopsis thaliana cyanase protein (AtCYN) (HA2-AtCYN vaccine) was evaluated in 2-weeks-old chickens. Depending on the route of administration, the HA2-AtCYN vaccine was shown to induce various levels of HA2-specific IgA in tears as well as serum IgG, which were associated with partial protection of chickens against tracheal shedding of a low pathogenicity H5N2 challenge virus. Furthermore, intranasal administration with a combination of HA2-AtCYN and M2eP vaccines resulted in enhanced protection compared to each vaccine alone. Simultaneous intranasal administration of the vaccines did not interfere with secretory IgA induction by each vaccine. Additionally, significantly higher M2eP-specific proliferative responses were observed in peripheral blood mononuclear cells of all M2eP-vaccinated groups when compared with the mock-vaccinated group. Although tripling the number of M2e copies did not enhance the protective efficacy of the chimeric vaccine, it significantly reduced immunodominance of P-particle epitopes without affecting the robustness of M2e-specific immune responses. Taken together, our data suggests that mucosal immunization of chickens with combinations of mechanistically different cross-subtype-conserved vaccines has the potential to enhance the protective efficacy against influenza virus challenge.


Asunto(s)
Glicoproteínas Hemaglutininas del Virus de la Influenza/inmunología , Vacunas contra la Influenza/inmunología , Gripe Aviar/prevención & control , Norovirus , Proteínas de la Matriz Viral/inmunología , Animales , Anticuerpos Antivirales/análisis , Anticuerpos Antivirales/sangre , Arabidopsis/enzimología , Liasas de Carbono-Nitrógeno/genética , Liasas de Carbono-Nitrógeno/inmunología , Pollos/inmunología , Protección Cruzada , Epítopos/inmunología , Inmunoglobulina A/análisis , Inmunoglobulina G/sangre , Subtipo H5N2 del Virus de la Influenza A , Vacunas contra la Influenza/administración & dosificación , Organismos Libres de Patógenos Específicos , Vacunas Sintéticas/administración & dosificación , Vacunas Sintéticas/inmunología , Proteínas de la Matriz Viral/genética
13.
Vet Microbiol ; 235: 170-179, 2019 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-31383299

RESUMEN

Turkey arthritis reovirus (TARV) infections have been recognized since 2011 to cause disease and significant economic losses to the U.S. turkey industry. Reoviral arthritis has been reproduced in commercial-origin turkeys. However, determination of pathogenesis or vaccine efficacy in these turkeys can be complicated by enteric reovirus strains and other pathogens that ubiquitously exist at subclinical levels among commercial turkey flocks. In this study, turkeys from a specific-pathogen-free (SPF) flock were evaluated for use as a turkey reoviral arthritis model. One-day-old or 1-week-old poults were orally inoculated with TARV (O'Neil strain) and monitored for disease onset and progression. A gut isolate of turkey reovirus (MN1 strain) was also tested for comparison. Disease was observed only in TARV-infected birds. Features of reoviral arthritis in SPF turkeys included swelling of hock joints, tenosynovitis, distal tibiotarsal cartilage erosion, and gait defects (lameness). Moreover, TARV infection resulted in a significant depression of body weights during the early times post-infection. Age-dependent susceptibility to TARV infection was unclear. TARV was transmitted to all sentinel birds, which manifested high levels of tenosynovitis and tibiotarsal cartilage erosion. Simulation of stressful conditions by dexamethasone treatment did not affect the viral load or exacerbate the disease. Collectively, the clinical and pathological features of reoviral arthritis in the SPF turkey model generally resembled those induced in commercial turkeys under field and/or experimental conditions. The SPF turkey reoviral arthritis model will be instrumental in evaluation of TARV pathogenesis and reoviral vaccine efficacy.


Asunto(s)
Artritis/veterinaria , Modelos Animales de Enfermedad , Infecciones por Reoviridae/veterinaria , Organismos Libres de Patógenos Específicos , Pavos , Animales , Artritis/virología
14.
Appl Environ Microbiol ; 85(9)2019 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-30824436

RESUMEN

The digestive and respiratory tracts of chickens are colonized by bacteria that are believed to play important roles in the overall health and performance of the birds. Most of the current research on the commensal bacteria (microbiota) of chickens has focused on broilers and gut microbiota, and less attention has been given to layers and respiratory microbiota. This research bias has left significant gaps in our knowledge of the layer microbiome. This study was conducted to define the core microbiota colonizing the upper respiratory tract (URT) and lower intestinal tract (LIT) in commercial layers under field conditions. One hundred eighty-one chickens were sampled from a flock of >80,000 birds at nine times to collect samples for 16S rRNA gene-based bacterial metabarcoding. Generally, the body site and age/farm stage had very dominant effects on the quantity, taxonomic composition, and dynamics of core bacteria. Remarkably, ileal and URT microbiota were compositionally more related to each other than to that from the cecum. Unique taxa dominated in each body site yet some taxa overlapped between URT and LIT sites, demonstrating a common core. The overlapping bacteria also contained various levels of several genera with well-recognized avian pathogens. Our findings suggest that significant interaction exists between gut and respiratory microbiota, including potential pathogens, in all stages of the farm sequence. The baseline data generated in this study can be useful for the development of effective microbiome-based interventions to enhance production performance and to prevent and control disease in commercial chicken layers.IMPORTANCE The poultry industry is faced with numerous challenges associated with infectious diseases and suboptimal performance of flocks. As microbiome research continues to grow, it is becoming clear that poultry health and production performance are partly influenced by nonpathogenic symbionts that occupy different habitats within the bird. This study has defined the baseline composition and overlaps between respiratory and gut bacteria in healthy, optimally performing chicken layers across all stages of the commercial farm sequence. Consequently, the study has set the groundwork for the development of interventions that seek to enhance production performance and to prevent and control infectious diseases through the modulation of gut and respiratory bacteria.


Asunto(s)
Bacterias/aislamiento & purificación , Pollos/microbiología , Tracto Gastrointestinal Inferior/microbiología , Microbiota , Sistema Respiratorio/microbiología , Factores de Edad , Crianza de Animales Domésticos , Animales , Bacterias/clasificación , Código de Barras del ADN Taxonómico/veterinaria , Microbioma Gastrointestinal , ARN Bacteriano/análisis , ARN Ribosómico 16S/análisis
15.
Vaccine ; 37(10): 1356-1364, 2019 02 28.
Artículo en Inglés | MEDLINE | ID: mdl-30691981

RESUMEN

Avian influenza in poultry continues to be a great concern worldwide, and the currently licensed inactivated influenza vaccines are not effective against the novel strains of influenza virus that continue to emerge in the field. This warrants the development of more broadly protective influenza vaccines or vaccination regimens. Live attenuated influenza vaccines (LAIVs) and subunit vaccines derived from viral peptides, such as the highly conserved ectodomain of influenza virus matrix protein 2 (M2e), can offer a more broadly reactive immune response. In chickens, we previously showed that a chimeric norovirus P particle containing M2e (M2eP) could provide partial but broad immunity, when administered as a standalone vaccine, and also enhanced the protective efficacy of inactivated vaccine when used in a combination regimen. We also demonstrated that a naturally-selected NS1-truncated H7N3 LAIV (pc4-LAIV) was highly efficacious against antigenically distant heterologous H7N2 low pathogenicity avian influenza virus challenge, especially when used as the priming vaccine in a prime-boost vaccination regimen. In this study, we investigated the cross-subtype protective efficacy of pc4-LAIV in conjunction with M2eP using single vaccination, combined treatment, and prime-boost approaches. Chickens vaccinated with pc4-LAIV showed significant reduction of tracheal shedding of a low pathogenicity H5N2 challenge virus. This cross-subtype protective efficacy was further enhanced, during the initial stages of challenge virus replication, in chickens that received a vaccination regimen consisting of priming with pc4-LAIV at 1 day of age and boosting with M2eP. Further, H5N2-specific serum IgG and pc4-LAIV-specific hemagglutination-inhibition antibody titers were enhanced in LAIV-primed and M2eP boost-vaccinated chickens. Taken together, our data point to the need of further investigation into the benefits of combined and prime-boost vaccination schemes utilizing LAIV and epitope-based vaccines, to develop more broadly protective vaccination regimens.


Asunto(s)
Anticuerpos Antivirales/sangre , Protección Cruzada , Vacunas contra la Influenza/inmunología , Gripe Aviar/prevención & control , Norovirus , Proteínas de la Matriz Viral/inmunología , Animales , Anticuerpos Neutralizantes/sangre , Pollos , Pruebas de Inhibición de Hemaglutinación , Esquemas de Inmunización , Inmunización Secundaria , Subtipo H5N2 del Virus de la Influenza A , Subtipo H7N2 del Virus de la Influenza A , Subtipo H7N3 del Virus de la Influenza A , Vacunas contra la Influenza/administración & dosificación , Vacunas Atenuadas/administración & dosificación , Vacunas Atenuadas/inmunología , Vacunas de Subunidad/administración & dosificación , Vacunas de Subunidad/inmunología , Proteínas de la Matriz Viral/genética
16.
PLoS One ; 13(4): e0195285, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29624615

RESUMEN

Outbreaks of novel highly pathogenic avian influenza viruses have been reported in poultry species in the United States since 2014. These outbreaks have proven the limitations of biosecurity control programs, and new tools are needed to reinforce the current avian influenza control arsenal. Some enzootic countries have implemented inactivated influenza vaccine (IIV) in their control programs, but there are serious concerns that a long-term use of IIV without eradication may result in the selection of novel antigenically divergent strains. A broadly protective vaccine is needed, such as live-attenuated influenza vaccine (LAIV). We showed in our previous studies that pc4-LAIV (a variant that encodes a C-terminally truncated NS1 protein) can provide significant protection against heterologous challenge virus in chickens vaccinated at 2-4 weeks of age through upregulation of innate and adaptive immune responses. The current study was conducted to compare the performances of pc4-LAIV and IIV in young chickens vaccinated at 1 day of age. A single dose of pc4-LAIV was able to induce stronger innate and mucosal IgA responses and protect young immunologically immature chickens better than a single dose of IIV. Most importantly, when 1-day-old chickens were intranasally primed with pc4-LAIV and subcutaneously boosted with IIV three weeks later, they showed a rapid, robust, and highly cross-reactive serum antibody response and a high level of mucosal IgA antibody response. This vaccination regimen warrants further optimization to increase its range of protection.


Asunto(s)
Pollos/inmunología , Vacunas contra la Influenza/administración & dosificación , Gripe Aviar/inmunología , Gripe Aviar/prevención & control , Enfermedades de las Aves de Corral/inmunología , Enfermedades de las Aves de Corral/prevención & control , Animales , Animales Recién Nacidos , Anticuerpos Antivirales/biosíntesis , Anticuerpos Antivirales/sangre , Anticuerpos Antivirales/genética , Antígenos Virales/genética , Reacciones Cruzadas , Inmunidad Innata/genética , Inmunidad Mucosa/genética , Inmunización Secundaria/métodos , Inmunización Secundaria/veterinaria , Virus de la Influenza A/genética , Virus de la Influenza A/inmunología , Vacunas contra la Influenza/genética , Vacunas contra la Influenza/inmunología , Vacunación/métodos , Vacunación/veterinaria , Vacunas Atenuadas/administración & dosificación , Vacunas Atenuadas/genética , Vacunas Atenuadas/inmunología , Vacunas de Productos Inactivados/administración & dosificación , Vacunas de Productos Inactivados/genética , Vacunas de Productos Inactivados/inmunología
17.
Appl Environ Microbiol ; 84(12)2018 06 15.
Artículo en Inglés | MEDLINE | ID: mdl-29625981

RESUMEN

Defining the baseline bacterial microbiome is critical to understanding its relationship with health and disease. In broiler chickens, the core microbiome and its possible relationships with health and disease have been difficult to define, due to high variability between birds and flocks. Presented here are data from a large, comprehensive microbiota-based study in commercial broilers. The primary goals of this study included understanding what constitutes the core bacterial microbiota in the broiler gastrointestinal, respiratory, and barn environments; how these core players change across age, geography, and time; and which bacterial taxa correlate with enhanced bird performance in antibiotic-free flocks. Using 2,309 samples from 37 different commercial flocks within a vertically integrated broiler system and metadata from these and an additional 512 flocks within that system, the baseline bacterial microbiota was defined using 16S rRNA gene sequencing. The effects of age, sample type, flock, and successive flock cycles were compared, and results indicate a consistent, predictable, age-dependent bacterial microbiota, irrespective of flock. The tracheal bacterial microbiota of broilers was comprehensively defined, and Lactobacillus was the dominant bacterial taxon in the trachea. Numerous bacterial taxa were identified, which were strongly correlated with broiler chicken performance across multiple tissues. While many positively correlated taxa were identified, negatively associated potential pathogens were also identified in the absence of clinical disease, indicating that subclinical dynamics occur that impact performance. Overall, this work provides necessary baseline data for the development of effective antibiotic alternatives, such as probiotics, for sustainable poultry production.IMPORTANCE Multidrug-resistant bacterial pathogens are perhaps the greatest medical challenge we will face in the 21st century and beyond. Antibiotics are necessary in animal production to treat disease. As such, animal production is a contributor to the problem of antibiotic resistance. Efforts are underway to reduce antibiotic use in animal production. However, we are also challenged to feed the world's increasing population, and sustainable meat production is paramount to providing a safe and quality protein source for human consumption. In the absence of antibiotics, alternative approaches are needed to maintain health and prevent disease, and probiotics have great promise as one such approach. This work paves the way for the development of alternative approaches to raising poultry by increasing our understandings of what defines the poultry microbiome and of how it can potentially be modulated to improve animal health and performance.


Asunto(s)
Bacterias/clasificación , Pollos/microbiología , Microbiota , Aves de Corral/microbiología , Animales , Antibacterianos , Bacterias/aislamiento & purificación , Pollos/crecimiento & desarrollo , Industria de Alimentos , ARN Ribosómico 16S/genética , Tráquea/microbiología
18.
PLoS One ; 12(2): e0171174, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28151964

RESUMEN

The current inactivated influenza vaccines provide satisfactory protection against homologous viruses but limited cross-protection against antigenically divergent strains. Consequently, there is a need to develop more broadly protective vaccines. The highly conserved extracellular domain of the matrix protein 2 (M2e) has shown promising results as one of the components of a universal influenza vaccine in different animal models. As an approach to overcome the limited, strain specific, protective efficacy of inactivated influenza vaccine (IIV), a combination of recombinant M2e expressed on the surface of norovirus P particle (M2eP) and IIV was tested in chickens. Co-immunization of birds with both vaccines did not affect the production of M2e-specific IgG antibodies compared to the group vaccinated with M2eP alone. However, the co-immunized birds developed significantly higher pre-challenge hemagglutination inhibition antibody titers against the homologous IIV antigen and heterologous challenge virus. These combined vaccine groups also had cross reactive antibody responses against different viruses (H5, H6, and H7 subtypes) compared to the IIV alone vaccinated group. Upon intranasal challenge with homologous and heterologous viruses, the combined vaccine groups showed greater reduction in viral shedding in tracheal swabs compared to those groups receiving IIV alone. Moreover, M2eP antisera from vaccinated birds were able to bind to the native M2 expressed on the surface of whole virus particles and infected cells, and inhibit virus replication in vitro. Our results support the potential benefit of supplementing IIV with M2eP, to expand the vaccine cross protective efficacy.


Asunto(s)
Vacunas contra la Influenza/administración & dosificación , Gripe Aviar/prevención & control , Norovirus/inmunología , Animales , Anticuerpos Antivirales/sangre , Pollos , Protección Cruzada , Perros , Pruebas de Inhibición de Hemaglutinación , Inmunoglobulina G/sangre , Virus de la Influenza A/clasificación , Virus de la Influenza A/inmunología , Gripe Aviar/inmunología , Gripe Aviar/virología , Células de Riñón Canino Madin Darby , Vacunas de Productos Inactivados/administración & dosificación , Vacunas Sintéticas/administración & dosificación , Proteínas de la Matriz Viral/inmunología , Esparcimiento de Virus
19.
PLoS One ; 11(6): e0156603, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27257989

RESUMEN

Influenza virus mutants that encode C-terminally truncated NS1 proteins (NS1-truncated mutants) are attractive candidates for avian live attenuated influenza vaccine (LAIV) development because they are both attenuated and immunogenic in chickens. We previously showed that a high protective efficacy of NS1-truncated LAIV in chickens corresponds with induction of high levels of type I interferon (IFN) responses in chicken embryonic fibroblast cells. In this study, we investigated the relationship between induction of IFN and IFN-stimulated gene responses in vivo and the immunogenicity and protective efficacy of NS1-truncated LAIV. Our data demonstrates that accelerated antibody induction and protective efficacy of NS1-truncated LAIV correlates well with upregulation of IFN-stimulated genes. Further, through oral administration of recombinant chicken IFN alpha in drinking water, we provide direct evidence that type I IFN can promote rapid induction of adaptive immune responses and protective efficacy of influenza vaccine in chickens.


Asunto(s)
Vacunas contra la Influenza/inmunología , Gripe Aviar/prevención & control , Interferones/inmunología , Proteínas no Estructurales Virales/inmunología , Inmunidad Adaptativa , Administración Oral , Animales , Anticuerpos Antivirales/sangre , Pollos , Gripe Aviar/virología , Interferones/farmacología , Mutación , Proteínas Recombinantes/inmunología , Vacunas Atenuadas/inmunología , Proteínas no Estructurales Virales/genética
20.
PLoS One ; 10(3): e0118934, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25790187

RESUMEN

Mutants of influenza virus that encode C-terminally truncated NS1 proteins (NS1-truncated mutants) characteristically induce high interferon responses. The dual activity of interferon in blocking virus replication and enhancing the development of adaptive immune responses makes these mutants promising as self-adjuvanting live-attenuated influenza vaccine (LAIV) candidates. Yet, among the NS1-truncated mutants, the length of NS1 is not directly correlated with the interferon-inducing efficiency, the level of attenuation, or effectiveness as LAIV. Using quantitative in vitro biologically active particle subpopulation analysis as a tool to identify potential LAIV candidates from a pool of NS1-truncated mutants, we previously predicted that a NS1-truncated mutant pc2, which was less effective as a LAIV in chickens, would be sufficiently effective as a LAIV in mammalian hosts. In this study, we confirmed that pc2 protected mice and pigs against heterologous virus challenge in terms of preventing clinical signs and reducing virus shedding. pc2 expresses a unique SLSYSINWRH motif at the C-terminus of its truncated NS1. Deletion of the SLSYSINWRH motif led to ~821-fold reduction in the peak yield of type I interferon induced in murine cells. Furthermore, replacement of the SLSYSINWRH motif with the wildtype MVKMDQAIMD sequence did not restore the interferon-inducing efficiency. The diminished interferon induction capacity in the absence of the SLSYSINWRH motif was similar to that observed in other mutants which are less effective LAIV candidates. Remarkably, pc2 induced 16-fold or more interferon in human lung and monkey kidney cells compared to the temperature-sensitive, cold-adapted Ann Arbor virus that is currently used as a master backbone for LAIVs such as FluMist. Although the mechanism by which the SLSYSINWRH motif regulates the vaccine properties of pc2 has not been elucidated, this motif has potential use in engineering self-adjuvanting NS1-truncated-based LAIVs.


Asunto(s)
Vacunas contra la Influenza/inmunología , Vacunas contra la Influenza/farmacología , Interferón Tipo I/inmunología , Orthomyxoviridae/genética , Proteínas no Estructurales Virales/genética , Secuencias de Aminoácidos/genética , Análisis de Varianza , Animales , Cartilla de ADN/genética , Técnicas In Vitro , Vacunas contra la Influenza/genética , Ratones , Mutagénesis , Orthomyxoviridae/inmunología , Ingeniería de Proteínas/métodos , Porcinos , Esparcimiento de Virus/efectos de los fármacos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...