Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 77
Filtrar
Más filtros

Bases de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Proc Natl Acad Sci U S A ; 119(4)2022 01 25.
Artículo en Inglés | MEDLINE | ID: mdl-35042807

RESUMEN

Genomics encompasses the entire tree of life, both extinct and extant, and the evolutionary processes that shape this diversity. To date, genomic research has focused on humans, a small number of agricultural species, and established laboratory models. Fewer than 18,000 of ∼2,000,000 eukaryotic species (<1%) have a representative genome sequence in GenBank, and only a fraction of these have ancillary information on genome structure, genetic variation, gene expression, epigenetic modifications, and population diversity. This imbalance reflects a perception that human studies are paramount in disease research. Yet understanding how genomes work, and how genetic variation shapes phenotypes, requires a broad view that embraces the vast diversity of life. We have the technology to collect massive and exquisitely detailed datasets about the world, but expertise is siloed into distinct fields. A new approach, integrating comparative genomics with cell and evolutionary biology, ecology, archaeology, anthropology, and conservation biology, is essential for understanding and protecting ourselves and our world. Here, we describe potential for scientific discovery when comparative genomics works in close collaboration with a broad range of fields as well as the technical, scientific, and social constraints that must be addressed.


Asunto(s)
Biodiversidad , Evolución Biológica , Genómica/métodos , Animales , Evolución Molecular , Variación Genética/genética , Genoma/genética , Genómica/tendencias , Humanos , Filogenia
2.
BMC Biol ; 21(1): 267, 2023 11 22.
Artículo en Inglés | MEDLINE | ID: mdl-37993882

RESUMEN

BACKGROUND: The red junglefowl, the wild outgroup of domestic chickens, has historically served as a reference for genomic studies of domestic chickens. These studies have provided insight into the etiology of traits of commercial importance. However, the use of a single reference genome does not capture diversity present among modern breeds, many of which have accumulated molecular changes due to drift and selection. While reference-based resequencing is well-suited to cataloging simple variants such as single-nucleotide changes and short insertions and deletions, it is mostly inadequate to discover more complex structural variation in the genome. METHODS: We present a pangenome for the domestic chicken consisting of thirty assemblies of chickens from different breeds and research lines. RESULTS: We demonstrate how this pangenome can be used to catalog structural variants present in modern breeds and untangle complex nested variation. We show that alignment of short reads from 100 diverse wild and domestic chickens to this pangenome reduces reference bias by 38%, which affects downstream genotyping results. This approach also allows for the accurate genotyping of a large and complex pair of structural variants at the K feathering locus using short reads, which would not be possible using a linear reference. CONCLUSIONS: We expect that this new paradigm of genomic reference will allow better pinpointing of exact mutations responsible for specific phenotypes, which will in turn be necessary for breeding chickens that meet new sustainability criteria and are resilient to quickly evolving pathogen threats.


Asunto(s)
Pollos , Genoma , Animales , Pollos/genética , Genotipo , Análisis de Secuencia de ADN , Genómica
3.
PLoS Biol ; 18(3): e3000619, 2020 03.
Artículo en Inglés | MEDLINE | ID: mdl-32134914

RESUMEN

Many livestock and human vaccines are leaky because they block symptoms but do not prevent infection or onward transmission. This leakiness is concerning because it increases vaccination coverage required to prevent disease spread and can promote evolution of increased pathogen virulence. Despite leakiness, vaccination may reduce pathogen load, affecting disease transmission dynamics. However, the impacts on post-transmission disease development and infectiousness in contact individuals are unknown. Here, we use transmission experiments involving Marek disease virus (MDV) in chickens to show that vaccination with a leaky vaccine substantially reduces viral load in both vaccinated individuals and unvaccinated contact individuals they infect. Consequently, contact birds are less likely to develop disease symptoms or die, show less severe symptoms, and shed less infectious virus themselves, when infected by vaccinated birds. These results highlight that even partial vaccination with a leaky vaccine can have unforeseen positive consequences in controlling the spread and symptoms of disease.


Asunto(s)
Herpesvirus Gallináceo 2/patogenicidad , Enfermedad de Marek/transmisión , Vacunas Virales/farmacología , Animales , Pollos , Plumas/virología , Interacciones Huésped-Patógeno , Enfermedad de Marek/etiología , Enfermedad de Marek/mortalidad , Enfermedad de Marek/prevención & control , Vacunación , Carga Viral , Vacunas Virales/administración & dosificación , Virulencia , Esparcimiento de Virus
4.
BMC Genet ; 21(1): 77, 2020 07 16.
Artículo en Inglés | MEDLINE | ID: mdl-32677890

RESUMEN

BACKGROUND: Marek's disease (MD) is a highly neoplastic disease primarily affecting chickens, and remains as a chronic infectious disease that threatens the poultry industry. Copy number variation (CNV) has been examined in many species and is recognized as a major source of genetic variation that directly contributes to phenotypic variation such as resistance to infectious diseases. Two highly inbred chicken lines, 63 (MD-resistant) and 72 (MD-susceptible), as well as their F1 generation and six recombinant congenic strains (RCSs) with varied susceptibility to MD, are considered as ideal models to identify the complex mechanisms of genetic and molecular resistance to MD. RESULTS: In the present study, to unravel the potential genetic mechanisms underlying resistance to MD, we performed a genome-wide CNV detection using next generation sequencing on the inbred chicken lines with the assistance of CNVnator. As a result, a total of 1649 CNV regions (CNVRs) were successfully identified after merging all the nine datasets, of which 90 CNVRs were overlapped across all the chicken lines. Within these shared regions, 1360 harbored genes were identified. In addition, 55 and 44 CNVRs with 62 and 57 harbored genes were specifically identified in line 63 and 72, respectively. Bioinformatics analysis showed that the nearby genes were significantly enriched in 36 GO terms and 6 KEGG pathways including JAK/STAT signaling pathway. Ten CNVRs (nine deletions and one duplication) involved in 10 disease-related genes were selected for validation by using quantitative real-time PCR (qPCR), all of which were successfully confirmed. Finally, qPCR was also used to validate two deletion events in line 72 that were definitely normal in line 63. One high-confidence gene, IRF2 was identified as the most promising candidate gene underlying resistance and susceptibility to MD in view of its function and overlaps with data from previous study. CONCLUSIONS: Our findings provide valuable insights for understanding the genetic mechanism of resistance to MD and the identified gene and pathway could be considered as the subject of further functional characterization.


Asunto(s)
Pollos/genética , Variaciones en el Número de Copia de ADN , Resistencia a la Enfermedad/genética , Enfermedad de Marek/genética , Animales , Pollos/virología , Ontología de Genes , Secuenciación de Nucleótidos de Alto Rendimiento
5.
J Gen Virol ; 100(7): 1132-1139, 2019 07.
Artículo en Inglés | MEDLINE | ID: mdl-31184569

RESUMEN

Marek's disease virus (MDV) is the most well-cited example of vaccine-driven virulence evolution. MDV induces a lymphoproliferative disease in chickens, which is currently controlled by widespread vaccination of flocks. Unfortunately, Marek's disease (MD) vaccines, while effective in preventing tumours, do not prevent viral replication and mutation, which has been hypothesized as the major driving force for increased MDV virulence of field strains during the past 40 years in US commercial flocks. To limit future virulence increases, there is interest in characterizing MDV strain genomes collected over the years and associating genetic variations with variation in virulence. In this study, we characterized 70 MDV genomes with known virulence by complete or targeted DNA sequencing, and identified genetic variants that showed association with virulence. Our results revealed a number of MDV genes as would be expected for a complex trait. In addition, phylogenetic analysis revealed a clear separation of strains that varied by virulence. Interestingly, high virulence isolates from the same farms persisted over years despite eradication attempts, which has implications on control efforts. Given the growing ability to bioengineer the MDV genome, it should be feasible to experimentally test whether these individual variants influence virulence markers alone or combinations. Once validated, these markers may provide an alternative to live bird testing for evaluating virulence of new MDV field strains.


Asunto(s)
Herpesvirus Gallináceo 2/genética , Herpesvirus Gallináceo 2/patogenicidad , Enfermedad de Marek/virología , Enfermedades de las Aves de Corral/virología , Proteínas Virales/genética , Animales , Pollos , Femenino , Genoma Viral , Herpesvirus Gallináceo 2/clasificación , Herpesvirus Gallináceo 2/aislamiento & purificación , Masculino , Filogenia , Estados Unidos , Proteínas Virales/metabolismo , Virulencia
6.
BMC Genomics ; 19(1): 684, 2018 Sep 18.
Artículo en Inglés | MEDLINE | ID: mdl-30227846

RESUMEN

BACKGROUND: Numerous long non-coding RNAs (lncRNAs) have been identified and their roles in gene regulation in humans, mice, and other model organisms studied; however, far less research has been focused on lncRNAs in farm animal species. While previous studies in chickens, cattle, and pigs identified lncRNAs in specific developmental stages or differentially expressed under specific conditions in a limited number of tissues, more comprehensive identification of lncRNAs in these species is needed. The goal of the FAANG Consortium (Functional Annotation of Animal Genomes) is to functionally annotate animal genomes, including the annotation of lncRNAs. As one of the FAANG pilot projects, lncRNAs were identified across eight tissues in two adult male biological replicates from chickens, cattle, and pigs. RESULTS: Comprehensive lncRNA annotations for the chicken, cattle, and pig genomes were generated by utilizing RNA-seq from eight tissue types from two biological replicates per species at the adult developmental stage. A total of 9393 lncRNAs in chickens, 7235 lncRNAs in cattle, and 14,429 lncRNAs in pigs were identified. Including novel isoforms and lncRNAs from novel loci, 5288 novel lncRNAs were identified in chickens, 3732 in cattle, and 4870 in pigs. These transcripts match previously known patterns of lncRNAs, such as generally lower expression levels than mRNAs and higher tissue specificity. An analysis of lncRNA conservation across species identified a set of conserved lncRNAs with potential functions associated with chromatin structure and gene regulation. Tissue-specific lncRNAs were identified. Genes proximal to tissue-specific lncRNAs were enriched for GO terms associated with the tissue of origin, such as leukocyte activation in spleen. CONCLUSIONS: LncRNAs were identified in three important farm animal species using eight tissues from adult individuals. About half of the identified lncRNAs were not previously reported in the NCBI annotations for these species. While lncRNAs are less conserved than protein-coding genes, a set of positionally conserved lncRNAs were identified among chickens, cattle, and pigs with potential functions related to chromatin structure and gene regulation. Tissue-specific lncRNAs have potential regulatory functions on genes enriched for tissue-specific GO terms. Future work will include epigenetic data from ChIP-seq experiments to further refine these annotations.


Asunto(s)
Bovinos/genética , Pollos/genética , Genoma , Especificidad de Órganos , ARN Largo no Codificante/genética , Porcinos/genética , Animales , Animales Domésticos/genética , Perfilación de la Expresión Génica , Regulación de la Expresión Génica , Secuenciación de Nucleótidos de Alto Rendimiento , Masculino , Anotación de Secuencia Molecular
7.
Cytogenet Genome Res ; 156(4): 204-214, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30572327

RESUMEN

Marek's disease (MD) is an infectious disease characterized by lymphomas and high mortality in susceptible chickens. The causative and ubiquitous alpha-herpesvirus known as MD virus (MDV) integrates into host telomeres during early infection through latency, known to be an important phase for oncogenic transformation. Herein, we sought to determine the influence of vaccination and host genetics on the temporal dynamics of MDV-host genome interactions. We studied integration profiles using 2 MD vaccines that vary in protective efficacy in 2 genetic lines that differ in MD resistance/susceptibility. Virus integration of both oncogenic MDV and vaccine strains was observed in both MD susceptible and resistant birds, however, the lines differed in their dynamic telomere-integration profiles. Notably, the resistant host genotype exhibited a smaller percentage of replicating cells with the virus telomere-integrated only phenotype as compared to the susceptible genotype. Vaccination with Rispens, the most protective MD vaccine, also reduced the establishment of the virus telomere-integrated only phenotype, suggesting a significant role of the phenotype in MD lymphoma development. The effect of Rispens vaccination was most dramatic in the susceptible genotype. These results suggest important connections between vaccinal immunity, MDV telomere integration, virus-induced oncogenesis, and virus-host genome interactions in the context of host genetics and disease susceptibility.


Asunto(s)
Pollos/genética , Herpesvirus Gallináceo 2/fisiología , Vacunas contra la Enfermedad de Marek/administración & dosificación , Telómero/virología , Animales , Pollos/virología , Resistencia a la Enfermedad , Genotipo , Herpesvirus Gallináceo 2/efectos de los fármacos , Enfermedad de Marek/prevención & control , Enfermedad de Marek/virología , Vacunas contra la Enfermedad de Marek/farmacología , Enfermedades de las Aves de Corral/prevención & control , Enfermedades de las Aves de Corral/virología , Vacunación , Integración Viral/efectos de los fármacos , Replicación Viral
8.
Development ; 141(15): 3003-12, 2014 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-25053433

RESUMEN

talpid(2) is an avian autosomal recessive mutant with a myriad of congenital malformations, including polydactyly and facial clefting. Although phenotypically similar to talpid(3), talpid(2) has a distinct facial phenotype and an unknown cellular, molecular and genetic basis. We set out to determine the etiology of the craniofacial phenotype of this mutant. We confirmed that primary cilia were disrupted in talpid(2) mutants. Molecularly, we found disruptions in Hedgehog signaling. Post-translational processing of GLI2 and GLI3 was aberrant in the developing facial prominences. Although both GLI2 and GLI3 processing were disrupted in talpid(2) mutants, only GLI3 activator levels were significantly altered in the nucleus. Through additional fine mapping and whole-genome sequencing, we determined that the talpid(2) phenotype was linked to a 1.4 Mb region on GGA1q that contained the gene encoding the ciliary protein C2CD3. We cloned the avian ortholog of C2CD3 and found its expression was ubiquitous, but most robust in the developing limbs and facial prominences. Furthermore, we found that C2CD3 is localized proximal to the ciliary axoneme and is important for docking the mother centriole to the ciliary vesicle and cell membrane. Finally, we identified a 19 bp deletion in talpid(2) C2CD3 that produces a premature stop codon, and thus a truncated protein, as the likely causal allele for the phenotype. Together, these data provide insight into the cellular, molecular and genetic etiology of the talpid(2) phenotype. Our data suggest that, although the talpid(2) and talpid(3) mutations affect a common ciliogenesis pathway, they are caused by mutations in different ciliary proteins that result in differences in craniofacial phenotype.


Asunto(s)
Anomalías Craneofaciales/genética , Proteínas Hedgehog/genética , Proteínas Hedgehog/metabolismo , Factores de Transcripción de Tipo Kruppel/genética , Mutación , Alelos , Animales , Membrana Celular/metabolismo , Núcleo Celular , Centriolos/metabolismo , Embrión de Pollo , Mapeo Cromosómico , Cilios/metabolismo , Codón de Terminación , Fibroblastos/metabolismo , Proteínas Hedgehog/fisiología , Heterocigoto , Fenotipo , Polimorfismo Genético , Procesamiento Proteico-Postraduccional , Análisis de Secuencia de ADN , Transducción de Señal , Proteína Gli2 con Dedos de Zinc
10.
Anim Genet ; 47(5): 528-33, 2016 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-27453069

RESUMEN

The Functional Annotation of Animal Genomes (FAANG) Consortium recently held a Gathering On FAANG (GO-FAANG) Workshop in Washington, DC on October 7-8, 2015. This consortium is a grass-roots organization formed to advance the annotation of newly assembled genomes of domesticated and non-model organisms (www.faang.org). The workshop gathered together from around the world a group of 100+ genome scientists, administrators, representatives of funding agencies and commodity groups to discuss the latest advancements of the consortium, new perspectives, next steps and implementation plans. The workshop was streamed live and recorded, and all talks, along with speaker slide presentations, are available at www.faang.org. In this report, we describe the major activities and outcomes of this meeting. We also provide updates on ongoing efforts to implement discussions and decisions taken at GO-FAANG to guide future FAANG activities. In summary, reference datasets are being established under pilot projects; plans for tissue sets, morphological classification and methods of sample collection for different tissues were organized; and core assays and data and meta-data analysis standards were established.


Asunto(s)
Animales Domésticos/genética , Genoma , Genómica , Animales , Congresos como Asunto , District of Columbia , Cooperación Internacional , Estándares de Referencia
11.
Anim Biotechnol ; 27(4): 238-44, 2016 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-27565867

RESUMEN

Utilizing RNA-seq data, 1,574 candidate genes with alternative splicing were previously identified between two chicken lines that differ in Marek's disease (MD) genetic resistance under control and Marek's disease virus infection conditions. After filtering out 1,530 genes with splice variants in the first or last exon, 44 genes were screened for possible exon loss or gain using PCR and gel electrophoresis. Consequently, 7 genes exhibited visually detectable differential expression of splice variants between lines 6 (MD resistant) and 7 (MD susceptible), and the resultant PCR products verified by DNA sequencing. Birds from inbred line 6 have transcripts that preferentially retain an exon compared to line 7 chickens for ITGB2, SGPL1, and COMMD5. Birds from inbred line 7 have alleles that preferentially retain an exon compared to line 6 for MOCS2. CCBL2 exon 1a is absent and ATAD1 exon 2 is truncated by 87 nucleotides in transcripts expressed by line 7 compared to those from line 6. For CHTF18, line 6 transcripts have an indel mutation with 7 additional nucleotides in exon 21 compared to line 7. The current study validates 7 genes with alternatively spliced isomers between the two chicken lines, which helps provide potential underlying mechanisms for the phenotypic differences.


Asunto(s)
Empalme Alternativo/genética , Pollos/genética , Resistencia a la Enfermedad/genética , Enfermedad de Marek/genética , Animales
12.
BMC Genomics ; 16: 843, 2015 Oct 23.
Artículo en Inglés | MEDLINE | ID: mdl-26492869

RESUMEN

BACKGROUND: Copy number variation (CNV) is a major source of genome polymorphism that directly contributes to phenotypic variation such as resistance to infectious diseases. Lines 63 and 72 are two highly inbred experimental chicken lines that differ greatly in susceptibility to Marek's disease (MD), and have been used extensively in efforts to identify the genetic and molecular basis for genetic resistance to MD. Using next generation sequencing, we present a genome-wide assessment of CNVs that are potentially associated with genetic resistance to MD. METHODS: Three chickens randomly selected from each line were sequenced to an average depth of 20×. Two popular software, CNVnator and Pindel, were used to call genomic CNVs separately. The results were combined to obtain a union set of genomic CNVs in the two chicken lines. RESULTS: A total of 5,680 CNV regions (CNVRs) were identified after merging the two datasets, of which 1,546 and 1,866 were specific to the MD resistant or susceptible line, respectively. Over half of the line-specific CNVRs were shared by 2 or more chickens, reflecting the reduced diversity in both inbred lines. The CNVRs fixed in the susceptible lines were significantly enriched in genes involved in MAPK signaling pathway. We also found 67 CNVRs overlapping with 62 genes previously shown to be strong candidates of the underlying genes responsible for the susceptibility to MD. CONCLUSIONS: Our findings provide new insights into the genetic architecture of the two chicken lines and additional evidence that MAPK signaling pathway may play an important role in host response to MD virus infection. The rich source of line-specific CNVs is valuable for future disease-related association studies in the two chicken lines.


Asunto(s)
Pollos/genética , Variaciones en el Número de Copia de ADN/genética , Resistencia a la Enfermedad/genética , Enfermedad de Marek/genética , Animales , Pollos/virología , Susceptibilidad a Enfermedades , Genoma , Secuenciación de Nucleótidos de Alto Rendimiento , Enfermedad de Marek/virología
13.
BMC Genomics ; 16: 816, 2015 Oct 19.
Artículo en Inglés | MEDLINE | ID: mdl-26481588

RESUMEN

BACKGROUND: Marek's disease (MD) is a lymphoproliferative disease of poultry induced by Marek's disease virus (MDV), a highly oncogenic alphaherpesvirus. Identifying the underlying genes conferring MD genetic resistance is desired for more efficacious control measures including genomic selection, which requires accurately identified genetic markers throughout the chicken genome. METHODS: Hypothesizing that variants located in transcriptional regulatory regions are the main mechanism underlying this complex trait, a genome-wide association study was conducted by genotyping a ~1,000 bird MD resource population derived from experimental inbred layers with SNPs containing 1,824 previously identified allele-specific expression (ASE) SNPs in response to MDV infection as well as 3,097 random SNPs equally spaced throughout the chicken genome. Based on the calculated associations, genomic predictions were determined for 200 roosters and selected sires had their progeny tested for Marek's disease incidence. RESULTS: Our analyses indicate that these ASE SNPs account for more than 83 % of the genetic variance and exhibit nearly all the highest associations. To validate these findings, 200 roosters had their genetic merit predicted from the ASE SNPs only, and the top 30 and bottom 30 ranked roosters were reciprocally mated to random hens. The resulting progeny showed that after only one generation of bidirectional selection, there was a 22 % difference in MD incidence and this approach gave a 125 % increase in accuracy compared to current pedigree-based estimates. CONCLUSIONS: We conclude that variation in transcriptional regulation is the major driving cause for genetic resistance to MD, and ASE SNPs identify the underlying genes and are sufficiently linked to the causative polymorphisms that they can be used for accurate genomic prediction as well as help define the underlying molecular basis. Furthermore, this approach should be applicable to other complex traits.


Asunto(s)
Resistencia a la Enfermedad/genética , Estudio de Asociación del Genoma Completo , Enfermedad de Marek/genética , Sitios de Carácter Cuantitativo/genética , Alelos , Animales , Pollos/genética , Femenino , Expresión Génica , Genotipo , Herpesvirus Gallináceo 2/patogenicidad , Masculino , Enfermedad de Marek/virología , Fenotipo , Polimorfismo de Nucleótido Simple , Elementos Reguladores de la Transcripción/genética
14.
J Virol ; 88(11): 6232-42, 2014 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-24648463

RESUMEN

UNLABELLED: Marek's disease (MD) is a lymphoproliferative disease of chickens caused by the oncogenic Gallid herpesvirus 2, commonly known as Marek's disease virus (MDV). MD vaccines, the primary control method, are often generated by repeated in vitro serial passage of this highly cell-associated virus to attenuate virulent MDV strains. To understand the genetic basis of attenuation, we used experimental evolution by serially passing three virulent MDV replicates generated from an infectious bacterial artificial chromosome (BAC) clone. All replicates became completely or highly attenuated, indicating that de novo mutation, and not selection among quasispecies existing in a strain, is the primary driving force for the reduction in virulence. Sequence analysis of the attenuated replicates revealed 41 to 95 single-nucleotide variants (SNVs) at 2% or higher frequency in each population and several candidate genes containing high-frequency, nonsynonymous mutations. Five candidate mutations were incorporated into recombinant viruses to determine their in vivo effect. SNVs within UL42 (DNA polymerase auxiliary subunit) and UL46 (tegument) had no measurable influence, while two independent mutations in LORF2 (a gene of unknown function) improved survival time of birds but did not alter disease incidence. A fifth SNV located within UL5 (helicase-primase subunit) greatly reduced in vivo viral replication, increased survival time of birds, and resulted in only 0 to 11% disease incidence. This study shows that multiple genes, often within pathways involving DNA replication and transcriptional regulation, are involved in de novo attenuation of MDV and provides targets for the rational design of future MD vaccines. IMPORTANCE: Marek's disease virus (MDV) is a very important pathogen in chickens that costs the worldwide poultry industry $1 billion to $2 billion annually. Marek's disease (MD) vaccines, the primary control method, are often produced by passing virulent strains in cell culture until attenuated. To understand this process, we identified all the changes in the viral genome that occurred during repeated cell passage. We find that a single mutation in the UL5 gene, which encodes a viral protein necessary for DNA replication, reduces disease incidence by 90% or more. In addition, other candidate genes were identified. This information should lead to the development of more effective and rationally designed MD vaccines leading to improved animal health and welfare and lower costs to consumers.


Asunto(s)
ADN Helicasas/genética , ADN Primasa/genética , Herpesvirus Gallináceo 2/patogenicidad , Enfermedad de Marek/prevención & control , Vacunas Atenuadas/genética , Proteínas Virales/genética , Animales , Secuencia de Bases , Evolución Molecular Dirigida , Herpesvirus Gallináceo 2/genética , Técnicas In Vitro , Datos de Secuencia Molecular , Mutación/genética , Polimorfismo de Nucleótido Simple/genética , Análisis de Secuencia de ADN , Pase Seriado/métodos , Virulencia/genética
15.
Avian Pathol ; 44(4): 254-8, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25968878

RESUMEN

Marek's disease virus (MDV) is an oncogenic alphaherpesvirus and the causative agent of Marek's disease (MD), characterized by immunosuppression, paralysis, nerve enlargement and induction of T-cell lymphomas in chickens. Despite widespread usage of vaccines since the 1970s to control MD, more virulent field strains of MDV have emerged that overcome vaccinal protection, necessitating the development of new and more protective MD vaccines. The ∆Meq virus, a recombinant Md5 strain MDV lacking the viral oncogene Meq, is one candidate MD vaccine with great potential but unfortunately it also causes bursal-thymic atrophy (BTA) in maternal antibody negative chickens, raising concerns that impede commercial use as a vaccine. Previously, we identified a point mutation within UL5 that reduced in vivo replication in attenuated viruses. We proposed that introduction of the UL5 point mutation into the ∆Meq virus would reduce in vivo replication and eliminate BTA yet potentially retain high protective abilities. In birds, the ∆Meq+UL5 recombinant MDV had reduced replication compared to the original ∆Meq virus, while weights of lymphoid organs indicated that ∆Meq+UL5 did not induce BTA, supporting the hypothesis that reduction of in vivo replication would also abolish BTA. Vaccine trials of the ∆Meq+UL5 virus compared to other ∆Meq-based viruses and commercial vaccines show that, while the ∆Meq+UL5 does provide vaccinal protection, this protection was also reduced compared to the original ∆Meq virus. Therefore, it appears that a very delicate balance is required between levels of replication able to induce high vaccinal protection, yet not so high as to induce BTA.


Asunto(s)
Pollos/inmunología , ADN Helicasas/inmunología , ADN Primasa/inmunología , Mardivirus/inmunología , Enfermedad de Marek/inmunología , Enfermedades de las Aves de Corral/inmunología , Animales , Atrofia/veterinaria , ADN Helicasas/genética , ADN Primasa/genética , Mardivirus/patogenicidad , Enfermedad de Marek/prevención & control , Enfermedad de Marek/virología , Vacunas contra la Enfermedad de Marek/genética , Vacunas contra la Enfermedad de Marek/inmunología , Mutación Puntual , Enfermedades de las Aves de Corral/prevención & control , Enfermedades de las Aves de Corral/virología , Proteínas Virales/genética , Proteínas Virales/inmunología
16.
Avian Dis ; 59(1): 94-7, 2015 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-26292541

RESUMEN

Marek's disease virus (MDV) is an oncogenic herpesvirus that afflicts chickens with the disease known as Marek's disease (MD). This virus induces tumors, nerve lesions, immunosuppression, and death of affected birds. Vaccines are the primary control method for MD but, due to the periodic evolution of field strains, it is necessary to explore the development of new MD vaccines. MD vaccines are often attenuated MDV strains generated through serial passage in vitro. We previously used experimental evolution of MDV to provide a better understanding of the genetic basis of attenuation. During complete genome sequencing of evolved MDV populations, we identified a point mutation within the UL5 helicase-primase gene and created a UL5 recombinant virus that significantly reduced disease incidence by 89%-100%. To determine if experimental evolution also identifies mutations that provide protective qualities as potential vaccine candidates, we tested the UL5 recombinant virus as a vaccine and compared its protection to commercial herpesvirus of turkey (HVT) and bivalent (HVT + SB-1) vaccines. Both commercial vaccines resulted in higher protection against MD than did the UL5 recombinant virus, although the UL5 virus did provide protection against developing MD in 46%-70% of birds challenged. This indicates that a mutation within the UL5 helicase-primase gene not only reduces virulence but also confers protection against challenge with virulent MDV, providing support that not only can experimental evolution identify candidate mutations involved in attenuation but can also identify potential candidates for use in vaccine development.


Asunto(s)
ADN Helicasas/metabolismo , ADN Primasa/metabolismo , Mardivirus/genética , Enfermedad de Marek/prevención & control , Polimorfismo de Nucleótido Simple , Proteínas Virales/metabolismo , Animales , Pollos , ADN Helicasas/genética , ADN Primasa/genética , ADN Primasa/inmunología , Inmunidad Materno-Adquirida , Mardivirus/clasificación , Vacunas contra la Enfermedad de Marek/inmunología , Subunidades de Proteína , Proteínas Virales/genética , Proteínas Virales/inmunología
17.
Cytogenet Genome Res ; 144(2): 142-54, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25402709

RESUMEN

Marek's disease virus (MDV) is an oncogenic α-herpesvirus that induces Marek's disease characterized by fatal lymphomas in chickens. Here, we explored the timing during pathogenesis when the virus integrates into the host genome, the cell type involved, the role of viral integration on cellular transformation, and tumor clonality. Three immune organs of chicken (thymus, bursa, and spleen) were extracted following infection with either an oncogenic or a non-oncogenic strain of MDV. Using molecular cytogenetics, cells were investigated for viral integration at key time points throughout pathogenesis. Integration profiling of tumors (early to late stage) was conducted. Virus integration was widespread in B and T lymphocytes based on their abundance in bursa and thymus, respectively. Viral replication was detected early after infection as was viral integration into the host genome. Integration is a natural part of the MDV herpesvirus life cycle. In addition, our data using a non-oncogenic virus establish that although integration is a hallmark of tumor cell populations, integration alone is not sufficient for cellular transformation. Our results provide evidence for progression of lineage clonality within tumors. Understanding the features of integration provides insight into the mechanisms of herpesvirus pathology which could lead to disease mitigation strategies.


Asunto(s)
Linfocitos B/virología , Bolsa de Fabricio/virología , Herpesvirus Gallináceo 2/genética , Bazo/virología , Linfocitos T/virología , Timo/virología , Animales , Linaje de la Célula , Pollos , Cruzamientos Genéticos , Perfilación de la Expresión Génica , Genoma Viral , Herpesvirus Gallináceo 2/fisiología , Hibridación Fluorescente in Situ , Fenotipo , Integración Viral
18.
J Virol ; 87(16): 9016-29, 2013 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-23740999

RESUMEN

Marek's disease (MD) is an economically significant disease in chickens that is caused by the highly oncogenic Marek's disease virus (MDV). A major unanswered question is the mechanism of MDV-induced tumor formation. Meq, a bZIP transcription factor discovered in the 1990s, is critically involved in viral oncogenicity, but only a few of its host target genes have been described, impeding our understanding of MDV-induced tumorigenesis. Using chromatin immunoprecipitation-sequencing (ChIP-seq) and microarray analysis, a high-confidence list of Meq binding sites in the chicken genome and a global transcriptome of Meq-responsive genes were generated. Meq binding sites were found to be enriched in the promoter regions of upregulated genes but not in those of downregulated genes. ChIP-seq was also performed for c-Jun, a known heterodimeric partner of Meq. The close location of binding sites of Meq and c-Jun was noted, suggesting cooperativity between these two factors in modulating transcription. Pathway analysis indicated that Meq transcriptionally regulates many genes that are part of several signaling pathways including the extracellular signal-regulated kinase /mitogen-activated protein kinase (ERK/MAPK), Jak-STAT, and ErbB pathways, which are critical for oncogenesis and/or include signaling mediators involved in apoptosis. Meq activates oncogenic signaling cascades by transcriptionally activating major kinases in the ERK/MAPK pathway and simultaneously repressing phosphatases, as verified using inhibitors of MEK and ERK1/2 in a cell proliferation assay. This study provides significant insights into the mechanistic basis of Meq-dependent cell transformation.


Asunto(s)
Transformación Celular Viral , Interacciones Huésped-Patógeno , Mardivirus/patogenicidad , Proteínas Oncogénicas Virales/genética , Proteínas Oncogénicas Virales/metabolismo , Factores de Virulencia/genética , Factores de Virulencia/metabolismo , Animales , Sitios de Unión , Línea Celular , Pollos , Inmunoprecipitación de Cromatina , ADN/metabolismo , Perfilación de la Expresión Génica , Análisis por Micromatrices , Regiones Promotoras Genéticas , Unión Proteica , Análisis de Secuencia de ADN , Transducción de Señal , Transcripción Genética
19.
Genet Sel Evol ; 46: 43, 2014 Jul 07.
Artículo en Inglés | MEDLINE | ID: mdl-25001618

RESUMEN

BACKGROUND: Antimicrobial peptides (AMP) are important elements of the first line of defence against pathogens in animals. NK-lysin is a cationic AMP that plays a critical role in innate immunity. The chicken NK-lysin gene has been cloned and its antimicrobial and anticancer activity has been described but its location in the chicken genome remains unknown. Here, we mapped the NK-lysin gene and examined the distribution of a functionally significant single nucleotide polymorphism (SNP) among different chicken inbred lines and heritage breeds. RESULTS: A 6000 rad radiation hybrid panel (ChickRH6) was used to map the NK-lysin gene to the distal end of chromosome 22. Two additional genes, the adipocyte enhancer-binding protein 1-like gene (AEBP1) and the DNA polymerase delta subunit 2-like (POLD2) gene, are located in the same NW_003779909 contig as NK-lysin, and were thus indirectly mapped to chromosome 22 as well. Previously, we reported a functionally significant SNP at position 271 of the NK-lysin coding sequence in two different chicken breeds. Here, we examined this SNP and found that the A allele appears to be more common than the G allele in these heritage breeds and inbred lines. CONCLUSIONS: The chicken NK-lysin gene mapped to the distal end of chromosome 22. Two additional genes, AEBP1 and POLD2, were indirectly mapped to chromosome 22 also. SNP analyses revealed that the A allele, which encodes a peptide with a higher antimicrobial activity, is more common than the G allele in our tested inbred lines and heritage breeds.


Asunto(s)
Proteínas Aviares/genética , Pollos/genética , Mapeo Cromosómico , Proteolípidos/genética , Alelos , Animales , Cruzamiento , Carboxipeptidasas/genética , Mapeo Cromosómico/veterinaria , Cromosomas/genética , ADN Polimerasa III/genética , Frecuencia de los Genes , Marcadores Genéticos , Genoma , Genotipo , Fenotipo , Polimorfismo de Nucleótido Simple , Proteínas Represoras/genética , Análisis de Secuencia de ADN/veterinaria
20.
Poult Sci ; 93(3): 550-5, 2014 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-24604847

RESUMEN

The Toll-like receptor (TLR) signaling pathway is one of the innate immune defense mechanisms against pathogens in vertebrates and invertebrates. However, the role of TLR in non-MHC genetic resistance or susceptibility to Marek's disease (MD) in the chicken is yet to be elucidated. Chicken embryo fibroblast (CEF) cells from MD susceptible and resistant lines were infected either with Marek's disease virus (MDV) or treated with polyionosinic-polycytidylic acid, a synthetic analog of dsRNA, and the expression of TLR and pro-inflammatory cytokines was studied at 8 and 36 h posttreatment by quantitative reverse transcriptase PCR. Findings of the present study reveal that MDV infection and polyionosinic-polycytidylic acid treatment significantly elevated the mRNA expression of TLR3, IL6, and IL8 in both susceptible and resistant lines. Furthermore, basal expression levels in uninfected CEF for TLR3, TLR7, and IL8 genes were significantly higher in resistant chickens compared with those of susceptible chickens. Our results suggest that TLR3 together with pro-inflammatory cytokines may play a significant role in genetic resistance to MD.


Asunto(s)
Proteínas Aviares/genética , Pollos , Citocinas/genética , Regulación del Desarrollo de la Expresión Génica , Enfermedad de Marek/genética , Enfermedades de las Aves de Corral/genética , Receptores Toll-Like/genética , Animales , Proteínas Aviares/metabolismo , Embrión de Pollo , Citocinas/metabolismo , Susceptibilidad a Enfermedades/inmunología , Susceptibilidad a Enfermedades/metabolismo , Susceptibilidad a Enfermedades/veterinaria , Fibroblastos , Herpesvirus Gallináceo 3/fisiología , Enfermedad de Marek/inmunología , Enfermedad de Marek/virología , Poli I-C/administración & dosificación , Enfermedades de las Aves de Corral/inmunología , Enfermedades de las Aves de Corral/virología , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa/veterinaria , Transducción de Señal , Receptores Toll-Like/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA