Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Resultados 1 - 20 de 31
Filtrar
1.
Immunity ; 46(1): 133-147, 2017 01 17.
Artículo en Inglés | MEDLINE | ID: mdl-28087239

RESUMEN

Immuno-surveillance networks operating at barrier sites are tuned by local tissue cues to ensure effective immunity. Site-specific commensal bacteria provide key signals ensuring host defense in the skin and gut. However, how the oral microbiome and tissue-specific signals balance immunity and regulation at the gingiva, a key oral barrier, remains minimally explored. In contrast to the skin and gut, we demonstrate that gingiva-resident T helper 17 (Th17) cells developed via a commensal colonization-independent mechanism. Accumulation of Th17 cells at the gingiva was driven in response to the physiological barrier damage that occurs during mastication. Physiological mechanical damage, via induction of interleukin 6 (IL-6) from epithelial cells, tailored effector T cell function, promoting increases in gingival Th17 cell numbers. These data highlight that diverse tissue-specific mechanisms govern education of Th17 cell responses and demonstrate that mechanical damage helps define the immune tone of this important oral barrier.


Asunto(s)
Encía/inmunología , Inmunidad Mucosa/inmunología , Vigilancia Inmunológica/inmunología , Mucosa Bucal/inmunología , Células Th17/inmunología , Animales , Citometría de Flujo , Encía/microbiología , Humanos , Masticación , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Microbiota , Mucosa Bucal/microbiología , Reacción en Cadena en Tiempo Real de la Polimerasa
2.
FASEB J ; 33(5): 6226-6238, 2019 05.
Artículo en Inglés | MEDLINE | ID: mdl-30794439

RESUMEN

Pulmonary airway epithelial cells (AECs) form a critical interface between host and environment. We investigated the role of the circadian clock using mice bearing targeted deletion of the circadian gene brain and muscle ARNT-like 1 (Bmal1) in AECs. Pulmonary neutrophil infiltration, biomechanical function, and responses to influenza infection were all disrupted. A circadian time-series RNA sequencing study of laser-captured AECs revealed widespread disruption in genes of the core circadian clock and output pathways regulating cell metabolism (lipids and xenobiotics), extracellular matrix, and chemokine signaling, but strikingly also the gain of a novel rhythmic transcriptome in Bmal1-targeted cells. Many of the rhythmic components were replicated in primary AECs cultured in air-liquid interface, indicating significant cell autonomy for control of pulmonary circadian physiology. Finally, we found that metabolic cues dictate phasing of the pulmonary clock and circadian responses to immunologic challenges. Thus, the local circadian clock in AECs is vital in lung health by coordinating major cell processes such as metabolism and immunity.-Zhang, Z. Hunter, L., Wu, G., Maidstone, R., Mizoro, Y., Vonslow, R., Fife, M., Hopwood, T., Begley, N., Saer, B., Wang, P., Cunningham, P., Baxter, M., Durrington, H., Blaikley, J. F., Hussell, T., Rattray, M., Hogenesch, J. B., Gibbs, J., Ray, D. W., Loudon, A. S. I. Genome-wide effect of pulmonary airway epithelial cell-specific Bmal1 deletion.


Asunto(s)
Factores de Transcripción ARNTL/genética , Células Epiteliales Alveolares/metabolismo , Transcriptoma , Células Epiteliales Alveolares/microbiología , Animales , Células Cultivadas , Relojes Circadianos , Femenino , Eliminación de Gen , Humanos , Metabolismo de los Lípidos , Masculino , Ratones , Ratones Endogámicos C57BL , Infecciones por Orthomyxoviridae/genética , Infecciones por Orthomyxoviridae/inmunología , Xenobióticos/metabolismo
3.
BMC Genomics ; 20(1): 272, 2019 Apr 05.
Artículo en Inglés | MEDLINE | ID: mdl-30952207

RESUMEN

BACKGROUND: The interferon-induced transmembrane (IFITM) protein family comprises a class of restriction factors widely characterised in humans for their potent antiviral activity. Their biological activity is well documented in several animal species, but their genetic variation and biological mechanism is less well understood, particularly in avian species. RESULTS: Here we report the complete sequence of the domestic chicken Gallus gallus IFITM locus from a wide variety of chicken breeds to examine the detailed pattern of genetic variation of the locus on chromosome 5, including the flanking genes ATHL1 and B4GALNT4. We have generated chIFITM sequences from commercial breeds (supermarket-derived chicken breasts), indigenous chickens from Nigeria (Nsukka) and Ethiopia, European breeds and inbred chicken lines from the Pirbright Institute, totalling of 206 chickens. Through mapping of genetic variants to the latest chIFITM consensus sequence our data reveal that the chIFITM locus does not show structural variation in the locus across the populations analysed, despite spanning diverse breeds from different geographic locations. However, single nucleotide variants (SNVs) in functionally important regions of the proteins within certain groups of chickens were detected, in particular the European breeds and indigenous birds from Ethiopia and Nigeria. In addition, we also found that two out of four SNVs located in the chIFITM1 (Ser36 and Arg77) and chIFITM3 (Val103) proteins were simultaneously under positive selection. CONCLUSIONS: Together these data suggest that IFITM genetic variation may contribute to the capacities of different chicken populations to resist virus infection.


Asunto(s)
Antígenos de Diferenciación/genética , Evolución Molecular , Sitios Genéticos , Marcadores Genéticos , Polimorfismo de Nucleótido Simple , Selección Genética , Secuencia de Aminoácidos , Animales , Pollos , Mapeo Cromosómico , Variaciones en el Número de Copia de ADN , Genoma , Análisis de Secuencia de ADN , Homología de Secuencia
4.
Eur J Immunol ; 48(5): 855-860, 2018 05.
Artículo en Inglés | MEDLINE | ID: mdl-29400409

RESUMEN

The requirement to remove apoptotic cells is equally important in homeostasis and inflammatory disease. In particular, during viral infections large quantities of infected cells undergo apoptosis and need to be efficiently cleared by phagocytes to prevent secondary necrosis. Although specific roles of several apoptotic cell sensors, such as the TAM (Tyro3, Axl, MerTK) receptor family, have been characterized in mouse models, little is known about their regulation and involvement in apoptotic cell uptake (efferocytosis) by human macrophages under inflammatory conditions. We show that whereas pro-inflammatory stimuli consistently downregulated MerTK expression in human monocyte-derived macrophages (MDMs), stimuli indicative of a viral infection, interferon-α (IFN-α) and the TLR3 ligand poly(I:C), specifically induced Axl expression and promoted binding of the bridging molecule Gas6. Axl induction by IFN-α and poly(I:C) was associated with higher MDM efferocytic capacity compared to cells treated with other pro-inflammatory stimuli, such as LPS and IFN-γ. While MerTK blocking antibody uniformly suppressed apoptotic cell uptake by MDMs, Axl blocking antibody significantly reduced efferocytosis by poly(I:C)-stimulated MDMs, but not by resting MDMs. Our observations demonstrate that Axl induction during viral infections contributes to maintaining macrophage capacity to engulf apoptotic cells, which may have important consequences for resolution of anti-viral immune responses.


Asunto(s)
Apoptosis/inmunología , Macrófagos/inmunología , Fagocitosis/inmunología , Proteínas Proto-Oncogénicas/metabolismo , Proteínas Tirosina Quinasas Receptoras/metabolismo , Tirosina Quinasa c-Mer/metabolismo , Línea Celular Tumoral , Humanos , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Interferón-alfa/inmunología , Células Jurkat , Macrófagos/virología , Poli I-C/inmunología , Tirosina Quinasa del Receptor Axl
5.
BMC Genomics ; 19(1): 624, 2018 Aug 22.
Artículo en Inglés | MEDLINE | ID: mdl-30134833

RESUMEN

BACKGROUND: The new genomic technologies have provided novel insights into the genetics of interactions between vectors, viruses and hosts, which are leading to advances in the control of arboviruses of medical importance. However, the development of tools and resources available for vectors of non-zoonotic arboviruses remains neglected. Biting midges of the genus Culicoides transmit some of the most important arboviruses of wildlife and livestock worldwide, with a global impact on economic productivity, health and welfare. The absence of a suitable reference genome has hindered genomic analyses to date in this important genus of vectors. In the present study, the genome of Culicoides sonorensis, a vector of bluetongue virus (BTV) in the USA, has been sequenced to provide the first reference genome for these vectors. In this study, we also report the use of the reference genome to perform initial transcriptomic analyses of vector competence for BTV. RESULTS: Our analyses reveal that the genome is 189 Mb, assembled in 7974 scaffolds. Its annotation using the transcriptomic data generated in this study and in a previous study has identified 15,612 genes. Gene expression analyses of C. sonorensis females infected with BTV performed in this study revealed 165 genes that were differentially expressed between vector competent and refractory females. Two candidate genes, glutathione S-transferase (gst) and the antiviral helicase ski2, previously recognized as involved in vector competence for BTV in C. sonorensis (gst) and repressing dsRNA virus propagation (ski2), were confirmed in this study. CONCLUSIONS: The reference genome of C. sonorensis has enabled preliminary analyses of the gene expression profiles of vector competent and refractory individuals. The genome and transcriptomes generated in this study provide suitable tools for future research on arbovirus transmission. These provide a valuable resource for these vector lineage, which diverged from other major Dipteran vector families over 200 million years ago. The genome will be a valuable source of comparative data for other important Dipteran vector families including mosquitoes (Culicidae) and sandflies (Psychodidae), and together with the transcriptomic data can yield potential targets for transgenic modification in vector control and functional studies.


Asunto(s)
Virus de la Lengua Azul/fisiología , Lengua Azul/transmisión , Ceratopogonidae/genética , Ceratopogonidae/virología , Genoma de los Insectos , Insectos Vectores , Animales , Lengua Azul/inmunología , Lengua Azul/virología , Virus de la Lengua Azul/inmunología , Ceratopogonidae/inmunología , Evolución Molecular , Perfilación de la Expresión Génica , Interacciones Huésped-Patógeno/genética , Interacciones Huésped-Patógeno/inmunología , Inmunidad Innata/genética , Insectos Vectores/genética , Insectos Vectores/fisiología , Anotación de Secuencia Molecular , Análisis de Secuencia de ADN , Transcriptoma/genética
6.
BMC Genomics ; 18(1): 419, 2017 05 30.
Artículo en Inglés | MEDLINE | ID: mdl-28558694

RESUMEN

BACKGROUND: Interferon inducible transmembrane (IFITM) proteins are effectors of the immune system widely characterized for their role in restricting infection by diverse enveloped and non-enveloped viruses. The chicken IFITM (chIFITM) genes are clustered on chromosome 5 and to date four genes have been annotated, namely chIFITM1, chIFITM3, chIFITM5 and chIFITM10. However, due to poor assembly of this locus in the Gallus Gallus v4 genome, accurate characterization has so far proven problematic. Recently, a new chicken reference genome assembly Gallus Gallus v5 was generated using Sanger, 454, Illumina and PacBio sequencing technologies identifying considerable differences in the chIFITM locus over the previous genome releases. METHODS: We re-sequenced the locus using both Illumina MiSeq and PacBio RS II sequencing technologies and we mapped RNA-seq data from the European Nucleotide Archive (ENA) to this finalized chIFITM locus. Using SureSelect probes capture probes designed to the finalized chIFITM locus, we sequenced the locus of a different chicken breed, namely a White Leghorn, and a turkey. RESULTS: We confirmed the Gallus Gallus v5 consensus except for two insertions of 5 and 1 base pair within the chIFITM3 and B4GALNT4 genes, respectively, and a single base pair deletion within the B4GALNT4 gene. The pull down revealed a single amino acid substitution of A63V in the CIL domain of IFITM2 compared to Red Jungle fowl and 13, 13 and 11 differences between IFITM1, 2 and 3 of chickens and turkeys, respectively. RNA-seq shows chIFITM2 and chIFITM3 expression in numerous tissue types of different chicken breeds and avian cell lines, while the expression of the putative chIFITM1 is limited to the testis, caecum and ileum tissues. CONCLUSIONS: Locus resequencing using these capture probes and RNA-seq based expression analysis will allow the further characterization of genetic diversity within Galliformes.


Asunto(s)
Galliformes/genética , Sitios Genéticos/genética , Variación Genética , Análisis de Secuencia de ARN , Animales
7.
Avian Pathol ; 46(2): 173-180, 2017 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-27624876

RESUMEN

Infectious bronchitis virus (IBV) causes infectious bronchitis in poultry, a respiratory disease that is a source of major economic loss to the poultry industry. Detection and the study of the molecular pathogenesis of the virus often involve the use of real-time quantitative PCR assays (qPCR). To account for error within the experiments, the levels of target gene transcription are normalized to that of suitable reference genes. Despite publication of the MIQE (Minimum Information for Publication of Quantitative Real-Time PCR Experiments) guidelines in 2009, single un-tested reference genes are often used for normalization of qPCR assays in avian research studies. Here, we use the geNorm algorithm to identify suitable reference genes in different avian cell types during infection with apathogenic and pathogenic strains of IBV. We discuss the importance of selecting an appropriate experimental sample subset for geNorm analysis, and show the effect that this selection can have on resultant reference gene selection. The effects of inappropriate normalization on the transcription pattern of a cellular signalling gene, AKT1, and the interferon-inducible, MX1, were studied. We identify the possibility of the misinterpretation of qPCR data when an inappropriate normalization strategy is employed. This is most notable when measuring the transcription of AKT1, where changes are minimal during infection.


Asunto(s)
Pollos/virología , Infecciones por Coronavirus/veterinaria , Virus de la Bronquitis Infecciosa/aislamiento & purificación , Enfermedades de las Aves de Corral/virología , Reacción en Cadena en Tiempo Real de la Polimerasa/veterinaria , Animales , Infecciones por Coronavirus/virología , Virus de la Bronquitis Infecciosa/genética , Riñón/virología , Reacción en Cadena en Tiempo Real de la Polimerasa/métodos , Estándares de Referencia , Organismos Libres de Patógenos Específicos
8.
Immunogenetics ; 66(7-8): 427-38, 2014 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-24863340

RESUMEN

The interleukin-1 gene family encodes a group of related proteins that exhibit a remarkable pleiotropy in the context of health and disease. The set of indispensable functions they control suggests that these genes should be found in all eukaryotic species. The ligands and receptors of this family have been primarily characterised in man and mouse. The genomes of most non-mammalian animal species sequenced so far possess all of the IL-1 receptor genes found in mammals. Yet, strikingly, very few of the ligands are identifiable in non-mammalian genomes. Our recent identification of two further IL-1 ligands in the chicken warranted a critical reappraisal of the evolution of this vitally important cytokine family. This review presents substantial data gathered across multiple, divergent metazoan genomes to unambiguously trace the origin of these genes. With the hypothesis that all of these genes, both ligands and receptors, were formed in a single ancient ancestor, extensive database mining revealed sufficient evidence to confirm this. It therefore suggests that the emergence of mammals is unrelated to the expansion of the IL-1 family. A thorough review of this cytokine family in the chicken, the most extensively studied amongst non-mammalian species, is also presented.


Asunto(s)
Proteínas Aviares/genética , Pollos/genética , Pollos/inmunología , Interleucina-1/genética , Animales , Evolución Molecular , Humanos , Ligandos , Ratones , Familia de Multigenes , Filogenia , Receptores de Interleucina-1/genética , Vertebrados/genética , Vertebrados/inmunología
9.
J Immunol ; 189(2): 539-50, 2012 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-22689884

RESUMEN

The human IL-1 family contains 11 genes encoded at three separate loci. Nine, including IL-1R antagonist (IL-1RN), are present at a single locus on chromosome 2, whereas IL-18 and IL-33 lie on chromosomes 11 and 9, respectively. There are currently only two known orthologs in the chicken, IL-1ß and IL-18, which are encoded on chromosomes 22 and 24, respectively. Two novel chicken IL-1 family sequences were identified from expressed sequence tag libraries, representing secretory and intracellular (icIL-1RN) structural variants of the IL-1RN gene, as seen in mammals. Two further putative splice variants (SVs) of both chicken IL-1RN (chIL-1RN) structural variants were also isolated. Alternative splicing of human icIL-1RN gives three different transcripts; there are no known SVs for human secretory IL-1RN. The chicken icIL-1RN SVs differ from those found in human icIL-1RN in terms of the rearrangements involved. In mammals, IL-1RN inhibits IL-1 activity by physically occupying the IL-1 type I receptor. Both full-length structural variants of chIL-1RN exhibited biological activity similar to their mammalian orthologs in a macrophage cell line bioassay. The four SVs, however, were not biologically active. The chicken IL-1 family is more fragmented in the genome than those of mammals, particularly in that the large multigene locus seen in mammals is absent. This suggests differential evolution of the family since the divergence of birds and mammals from a common ancestor, and makes determination of the full repertoire of chicken IL-1 family members more challenging.


Asunto(s)
Proteína Antagonista del Receptor de Interleucina 1/química , Proteína Antagonista del Receptor de Interleucina 1/genética , Empalme Alternativo/inmunología , Secuencia de Aminoácidos , Animales , Secuencia de Bases , Células COS , Línea Celular , Pollos , Chlorocebus aethiops , Clonación Molecular , ADN Complementario/aislamiento & purificación , Células HEK293 , Humanos , Proteína Antagonista del Receptor de Interleucina 1/fisiología , Mamíferos , Isoformas de Proteínas/química , Isoformas de Proteínas/genética
10.
BMC Vet Res ; 10: 77, 2014 Mar 31.
Artículo en Inglés | MEDLINE | ID: mdl-24685104

RESUMEN

In the past decade, two pathogens transmitted by Culicoides biting midges (Diptera: Ceratopogonidae), bluetongue virus and Schmallenberg virus, have caused serious economic losses to the European livestock industry, most notably affecting sheep and cattle. These outbreaks of arboviral disease have highlighted large knowledge gaps on the biology and ecology of indigenous Culicoides species. With these research gaps in mind, and as a means of assessing what potential disease outbreaks to expect in the future, an international workshop was held in May 2013 at Wageningen University, The Netherlands. It brought together research groups from Belgium, France, Germany, Spain, Switzerland, United Kingdom and The Netherlands, with diverse backgrounds in vector ecology, epidemiology, entomology, virology, animal health, modelling, and genetics. Here, we report on the key findings of this workshop.


Asunto(s)
Virus de la Lengua Azul/fisiología , Lengua Azul/transmisión , Infecciones por Bunyaviridae/transmisión , Ceratopogonidae/virología , Orthobunyavirus/fisiología , Animales , Bovinos/virología , Enfermedades de los Bovinos/transmisión , Enfermedades de los Bovinos/virología , Enfermedades Transmisibles Emergentes/veterinaria , Educación , Europa (Continente) , Ovinos/virología
11.
BMC Genomics ; 14: 398, 2013 Jun 13.
Artículo en Inglés | MEDLINE | ID: mdl-23763846

RESUMEN

BACKGROUND: Detecting genetic variation is a critical step in elucidating the molecular mechanisms underlying phenotypic diversity. Until recently, such detection has mostly focused on single nucleotide polymorphisms (SNPs) because of the ease in screening complete genomes. Another type of variant, copy number variation (CNV), is emerging as a significant contributor to phenotypic variation in many species. Here we describe a genome-wide CNV study using array comparative genomic hybridization (aCGH) in a wide variety of chicken breeds. RESULTS: We identified 3,154 CNVs, grouped into 1,556 CNV regions (CNVRs). Thirty percent of the CNVs were detected in at least 2 individuals. The average size of the CNVs detected was 46.3 kb with the largest CNV, located on GGAZ, being 4.3 Mb. Approximately 75% of the CNVs are copy number losses relatively to the Red Jungle Fowl reference genome. The genome coverage of CNVRs in this study is 60 Mb, which represents almost 5.4% of the chicken genome. In particular large gene families such as the keratin gene family and the MHC show extensive CNV. CONCLUSIONS: A relative large group of the CNVs are line-specific, several of which were previously shown to be related to the causative mutation for a number of phenotypic variants. The chance that inter-specific CNVs fall into CNVRs detected in chicken is related to the evolutionary distance between the species. Our results provide a valuable resource for the study of genetic and phenotypic variation in this phenotypically diverse species.


Asunto(s)
Pollos/genética , Variaciones en el Número de Copia de ADN , Genoma , Animales , Cruzamiento , Análisis por Conglomerados , Hibridación Genómica Comparativa , Biología Computacional/métodos , Femenino , Ligamiento Genético , Masculino
12.
BMC Genomics ; 14: 59, 2013 Jan 28.
Artículo en Inglés | MEDLINE | ID: mdl-23356797

RESUMEN

BACKGROUND: High density (HD) SNP genotyping arrays are an important tool for genetic analyses of animals and plants. Although the chicken is one of the most important farm animals, no HD array is yet available for high resolution genetic analysis of this species. RESULTS: We report here the development of a 600 K Affymetrix® Axiom® HD genotyping array designed using SNPs segregating in a wide variety of chicken populations. In order to generate a large catalogue of segregating SNPs, we re-sequenced 243 chickens from 24 chicken lines derived from diverse sources (experimental, commercial broiler and layer lines) by pooling 10-15 samples within each line. About 139 million (M) putative SNPs were detected by mapping sequence reads to the new reference genome (Gallus_gallus_4.0) of which ~78 M appeared to be segregating in different lines. Using criteria such as high SNP-quality score, acceptable design scores predicting high conversion performance in the final array and uniformity of distribution across the genome, we selected ~1.8 M SNPs for validation through genotyping on an independent set of samples (n = 282). About 64% of the SNPs were polymorphic with high call rates (>98%), good cluster separation and stable Mendelian inheritance. Polymorphic SNPs were further analysed for their population characteristics and genomic effects. SNPs with extreme breach of Hardy-Weinberg equilibrium (P < 0.00001) were excluded from the panel. The final array, designed on the basis of these analyses, consists of 580,954 SNPs and includes 21,534 coding variants. SNPs were selected to achieve an essentially uniform distribution based on genetic map distance for both broiler and layer lines. Due to a lower extent of LD in broilers compared to layers, as reported in previous studies, the ratio of broiler and layer SNPs in the array was kept as 3:2. The final panel was shown to genotype a wide range of samples including broilers and layers with over 100 K to 450 K informative SNPs per line. A principal component analysis was used to demonstrate the ability of the array to detect the expected population structure which is an important pre-investigation step for many genome-wide analyses. CONCLUSIONS: This Affymetrix® Axiom® array is the first SNP genotyping array for chicken that has been made commercially available to the public as a product. This array is expected to find widespread usage both in research and commercial application such as in genomic selection, genome-wide association studies, selection signature analyses, fine mapping of QTLs and detection of copy number variants.


Asunto(s)
Pollos/genética , Técnicas de Genotipaje/instrumentación , Polimorfismo de Nucleótido Simple/genética , Animales , Artefactos , Biología Computacional , Frecuencia de los Genes , Masculino , Reproducibilidad de los Resultados , Análisis de Secuencia
14.
BMC Genomics ; 13: 146, 2012 Apr 24.
Artículo en Inglés | MEDLINE | ID: mdl-22531008

RESUMEN

BACKGROUND: Chicken meat and eggs can be a source of human zoonotic pathogens, especially Salmonella species. These food items contain a potential hazard for humans. Chickens lines differ in susceptibility for Salmonella and can harbor Salmonella pathogens without showing clinical signs of illness. Many investigations including genomic studies have examined the mechanisms how chickens react to infection. Apart from the innate immune response, many physiological mechanisms and pathways are reported to be involved in the chicken host response to Salmonella infection. The objective of this study was to perform a meta-analysis of diverse experiments to identify general and host specific mechanisms to the Salmonella challenge. RESULTS: Diverse chicken lines differing in susceptibility to Salmonella infection were challenged with different Salmonella serovars at several time points. Various tissues were sampled at different time points post-infection, and resulting host transcriptional differences investigated using different microarray platforms. The meta-analysis was performed with the R-package metaMA to create lists of differentially regulated genes. These gene lists showed many similarities for different chicken breeds and tissues, and also for different Salmonella serovars measured at different times post infection. Functional biological analysis of these differentially expressed gene lists revealed several common mechanisms for the chicken host response to Salmonella infection. The meta-analysis-specific genes (i.e. genes found differentially expressed only in the meta-analysis) confirmed and expanded the biological functional mechanisms. CONCLUSIONS: The meta-analysis combination of heterogeneous expression profiling data provided useful insights into the common metabolic pathways and functions of different chicken lines infected with different Salmonella serovars.


Asunto(s)
Pollos/genética , Pollos/metabolismo , Salmonelosis Animal/genética , Salmonella/metabolismo , Animales , Movimiento Celular , Biología Computacional , Perfilación de la Expresión Génica , Análisis de Secuencia por Matrices de Oligonucleótidos , Fosforilación , Complejo de la Endopetidasa Proteasomal/metabolismo , Proteolisis , Salmonella/genética , Salmonelosis Animal/metabolismo , Salmonelosis Animal/microbiología , Ubiquitina/metabolismo
15.
J Virol ; 85(21): 11146-58, 2011 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-21865384

RESUMEN

Marek's disease virus (MDV) is a highly contagious oncogenic alphaherpesvirus that causes disease that is both a cancer model and a continuing threat to the world's poultry industry. This comprehensive gene expression study analyzes the host response to infection in both resistant and susceptible lines of chickens and inherent expression differences between the two lines following the infection of the host. A novel pathogenicity mechanism, involving the downregulation of genes containing HIC1 transcription factor binding sites as early as 4 days postinfection, was suggested from this analysis. HIC1 drives antitumor mechanisms, suggesting that MDV infection switches off genes involved in antitumor regulation several days before the expression of the MDV oncogene meq. The comparison of the gene expression data to previous QTL data identified several genes as candidates for involvement in resistance to MD. One of these genes, IRG1, was confirmed by single nucleotide polymorphism analysis to be involved in susceptibility. Its precise mechanism remains to be elucidated, although the analysis of gene expression data suggests it has a role in apoptosis. Understanding which genes are involved in susceptibility/resistance to MD and defining the pathological mechanisms of the disease gives us a much greater ability to try to reduce the incidence of this virus, which is costly to the poultry industry in terms of both animal welfare and economics.


Asunto(s)
Predisposición Genética a la Enfermedad , Factores de Transcripción de Tipo Kruppel/metabolismo , Mardivirus/inmunología , Mardivirus/patogenicidad , Enfermedad de Marek/genética , Enfermedad de Marek/inmunología , Animales , Pollos , Perfilación de la Expresión Génica , Factores de Transcripción de Tipo Kruppel/genética , Estados Unidos
16.
Dev Comp Immunol ; 114: 103850, 2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-32918930

RESUMEN

The human IL-1 receptor family is comprised of 11 membrane bound or soluble receptors and the IL-18 binding protein (IL-18BP). These receptors are dispersed across seven genomic loci, with the majority at a single locus. Direct orthologues were identified in the chicken at conserved genomic loci; however, the IL-18BP remained absent from the first four builds of the chicken genome sequence. Subsequent assemblies identified the gene at a locus syntenic with mammals; however, these predicted sequences differed between genome builds and contained multiple errors. A partial IL-18BP-like sequence in the NCBI EST database was used to clone the full-length cDNA. A splice variant, which lacks the exon that encodes part of the signal peptide, was also cloned. Human IL-18BP is differentially spliced to produce a number of variants, which are all secreted. By contrast, the spliced chicken isoform was predicted to be intracellular, and we identified similar variants with the same exon missing in a limited number of divergent vertebrate species. Mammalian and viral IL-18BPs inhibit IL-18 activity by directly binding to this cytokine. Full-length and intracellular chicken IL-18BPs were equally effective at inhibiting IL-18-mediated IFN-γ release from an avian B-cell line. Analysis of the predicted chIL-18BP protein sequence revealed two crucial residues, which account for 50% of the binding affinity between human IL-18 and IL-18BP, are conserved in the chicken and a fowlpox-encoded homologue, fpv214. This suggests specific fowlpox viruses used in humans as a vaccine vector have the potential to dampen anti-viral host immune responses.


Asunto(s)
Proteínas Aviares/genética , Linfocitos B/inmunología , Pollos/inmunología , Virus de la Viruela de las Aves de Corral/metabolismo , Péptidos y Proteínas de Señalización Intercelular/genética , Interleucina-18/metabolismo , Isoformas de Proteínas/genética , Proteínas Virales/metabolismo , Animales , Proteínas Aviares/metabolismo , Línea Celular , Clonación Molecular , Virus de la Viruela de las Aves de Corral/genética , Sitios Genéticos/genética , Vectores Genéticos/genética , Interacciones Huésped-Patógeno , Inmunomodulación , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Interferón gamma/metabolismo , Activación de Linfocitos , Mamíferos , Unión Proteica , Sintenía , Proteínas Virales/genética
17.
Sci Rep ; 11(1): 1623, 2021 01 12.
Artículo en Inglés | MEDLINE | ID: mdl-33436657

RESUMEN

Campylobacter is the leading cause of bacterial foodborne gastroenteritis worldwide. Handling or consumption of contaminated poultry meat is a key risk factor for human campylobacteriosis. One potential control strategy is to select poultry with increased resistance to Campylobacter. We associated high-density genome-wide genotypes (600K single nucleotide polymorphisms) of 3000 commercial broilers with Campylobacter load in their caeca. Trait heritability was modest but significant (h2 = 0.11 ± 0.03). Results confirmed quantitative trait loci (QTL) on chromosomes 14 and 16 previously identified in inbred chicken lines, and detected two additional QTLs on chromosomes 19 and 26. RNA-Seq analysis of broilers at the extremes of colonisation phenotype identified differentially transcribed genes within the QTL on chromosome 16 and proximal to the major histocompatibility complex (MHC) locus. We identified strong cis-QTLs located within MHC suggesting the presence of cis-acting variation in MHC class I and II and BG genes. Pathway and network analyses implicated cooperative functional pathways and networks in colonisation, including those related to antigen presentation, innate and adaptive immune responses, calcium, and renin-angiotensin signalling. While co-selection for enhanced resistance and other breeding goals is feasible, the frequency of resistance-associated alleles was high in the population studied and non-genetic factors significantly influenced Campylobacter colonisation.


Asunto(s)
Campylobacter/fisiología , Pollos/genética , Resistencia a la Enfermedad/genética , Carácter Cuantitativo Heredable , Transcriptoma , Inmunidad Adaptativa/genética , Animales , Estudio de Asociación del Genoma Completo , Genotipo , Antígenos de Histocompatibilidad Clase I/genética , Antígenos de Histocompatibilidad Clase I/metabolismo , Antígenos de Histocompatibilidad Clase II/genética , Antígenos de Histocompatibilidad Clase II/metabolismo , Inmunidad Innata/genética , Polimorfismo de Nucleótido Simple , Enfermedades de las Aves de Corral/microbiología
18.
Genes (Basel) ; 11(8)2020 08 10.
Artículo en Inglés | MEDLINE | ID: mdl-32785186

RESUMEN

The coronaviruses are a large family of enveloped RNA viruses that commonly cause gastrointestinal or respiratory illnesses in the infected host. Avian coronavirus infectious bronchitis virus (IBV) is a highly contagious respiratory pathogen of chickens that can affect the kidneys and reproductive systems resulting in bird mortality and decreased reproductivity. The interferon-inducible transmembrane (IFITM) proteins are activated in response to viral infections and represent a class of cellular restriction factors that restrict the replication of many viral pathogens. Here, we characterize the relative mRNA expression of the chicken IFITM genes in response to IBV infection, in vivo, ex vivo and in vitro using the pathogenic M41-CK strain, the nephropathogenic QX strain and the nonpathogenic Beaudette strain. In vivo we demonstrate a significant upregulation of chIFITM1, 2, 3 and 5 in M41-CK- and QX-infected trachea two days post-infection. In vitro infection with Beaudette, M41-CK and QX results in a significant upregulation of chIFITM1, 2 and 3 at 24 h post-infection. We confirmed a differential innate response following infection with distinct IBV strains and believe that our data provide new insights into the possible role of chIFITMs in early IBV infection.


Asunto(s)
Pollos/genética , Pollos/virología , Infecciones por Coronavirus/veterinaria , Interacciones Huésped-Patógeno/genética , Proteínas de la Membrana/genética , Animales , Infecciones por Coronavirus/genética , Regulación Viral de la Expresión Génica , Interacciones Huésped-Patógeno/fisiología , Virus de la Bronquitis Infecciosa/patogenicidad , Virus de la Bronquitis Infecciosa/fisiología , Técnicas de Cultivo de Órganos , Enfermedades de las Aves de Corral/etiología , Enfermedades de las Aves de Corral/genética , Enfermedades de las Aves de Corral/virología , Carga Viral , Tropismo Viral
19.
Artículo en Inglés | MEDLINE | ID: mdl-32984077

RESUMEN

Exposure to respiratory pathogens is a leading cause of exacerbations of airway diseases such as asthma and chronic obstructive pulmonary disease (COPD). Pellino-1 is an E3 ubiquitin ligase known to regulate virally-induced inflammation. We wished to determine the role of Pellino-1 in the host response to respiratory viruses in health and disease. Pellino-1 expression was examined in bronchial sections from patients with GOLD stage two COPD and healthy controls. Primary bronchial epithelial cells (PBECs) in which Pellino-1 expression had been knocked down were extracellularly challenged with the TLR3 agonist poly(I:C). C57BL/6 Peli1-/- mice and wild type littermates were subjected to intranasal infection with clinically-relevant respiratory viruses: rhinovirus (RV1B) and influenza A. We found that Pellino-1 is expressed in the airways of normal subjects and those with COPD, and that Pellino-1 regulates TLR3 signaling and responses to airways viruses. In particular we observed that knockout of Pellino-1 in the murine lung resulted in increased production of proinflammatory cytokines IL-6 and TNFα upon viral infection, accompanied by enhanced recruitment of immune cells to the airways, without any change in viral replication. Pellino-1 therefore regulates inflammatory airway responses without altering replication of respiratory viruses.


Asunto(s)
Infecciones por Picornaviridae , Enfermedad Pulmonar Obstructiva Crónica , Virosis , Animales , Humanos , Ratones , Ratones Endogámicos C57BL , Proteínas Nucleares , Rhinovirus , Ubiquitina-Proteína Ligasas/genética
20.
Gen Comp Endocrinol ; 163(1-2): 83-91, 2009 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-18957294

RESUMEN

Despite occupying the same habitats as mammals, having similar ranges of body mass and longevity, and facing similar pathogen challenges, birds have a different repertoire of organs, cells, molecules and genes of the immune system when compared to mammals. In other words, birds are not "mice with feathers", at least not in terms of their immune systems. Here we discuss differences between immune gene repertoires of birds and mammals, particularly those known to play a role in immune-endocrine interactions in mammals. If we are to begin to understand immune-endocrine interactions in the chicken, we need to understand these repertoires and also the biological function of the proteins encoded by these genes. We also discuss developments in our ability to understand the function of dendritic cells in the chicken; the function of these professional antigen-presenting cells is affected by stress in mammals. With regard to the endocrine system, we describe relevant chicken pituitary-adrenal hormones, and review recent findings on the expression of their receptors, as these receptors play a crucial role in modulating immune-endocrine interactions. Finally, we review the (albeit limited) work that has been carried out to understand immune-endocrine interactions in the chicken in the post-genome era.


Asunto(s)
Pollos/fisiología , Sistema Endocrino/metabolismo , Sistema Inmunológico/metabolismo , Animales , Quimiocinas/metabolismo , Pollos/inmunología , Pollos/metabolismo , Citocinas/metabolismo , Modelos Biológicos , Receptores Toll-Like/metabolismo
SELECCIÓN DE REFERENCIAS
Detalles de la búsqueda