Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 35
Filtrar
1.
J Virol ; 93(23)2019 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-31554689

RESUMEN

In 2010, sporadic cases of avian leukosis virus (ALV)-like bursal lymphoma, also known as spontaneous lymphoid leukosis (LL)-like tumors, were identified in two commercial broiler breeder flocks in the absence of exogenous ALV infection. Two individual ALV subgroup E (ALV-E) field strains, designated AF227 and AF229, were isolated from two different breeder farms. The role of these ALV-E field isolates in development of and the potential joint impact in conjunction with a Marek's disease virus (MDV) vaccine (SB-1) were further characterized in chickens of an experimental line and commercial broiler breeders. The experimental line 0.TVB*S1, commonly known as the rapid feathering-susceptible (RFS) line, of chickens lacks all endogenous ALV and is fully susceptible to all subgroups of ALV, including ALV-E. Spontaneous LL-like tumors occurred following infection with AF227, AF229, and a reference ALV-E strain, RAV60, in RFS chickens. Vaccination with serotype 2 MDV, SB-1, in addition to AF227 or AF229 inoculation, significantly enhanced the spontaneous LL-like tumor incidence in the RFS chickens. The spontaneous LL-like tumor incidence jumped from 14% by AF227 alone to 42 to 43% by AF227 in combination with SB-1 in the RFS chickens under controlled conditions. RNA-sequencing analysis of the LL-like lymphomas and nonmalignant bursa tissues of the RFS line of birds identified hundreds of differentially expressed genes that are reportedly involved in key biological processes and pathways, including signaling and signal transduction pathways. The data from this study suggested that both ALV-E and MDV-2 play an important role in enhancement of the spontaneous LL-like tumors in susceptible chickens. The underlying mechanism may be complex and involved in many chicken genes and pathways, including signal transduction pathways and immune system processes, in addition to reported viral genes.IMPORTANCE Lymphoid leukosis (LL)-like lymphoma is a low-incidence yet costly and poorly understood disease of domestic chickens. The observed unique characteristics of LL-like lymphomas are that the incidence of the disease is chicken line dependent; pathologically, it appeared to mimic avian leukosis but is free of exogenous ALV infection; inoculation of the nonpathogenic ALV-E or MDV-2 (SB-1) boosts the incidence of the disease; and inoculation of both the nonpathogenic ALV-E and SB-1 escalates it to much higher levels. This study was designed to test the impact of two new ALV-E isolates, recently derived from commercial broiler breeder flocks, in combination with the nonpathogenic SB-1 on LL-like lymphoma incidences in both an experimental egg layer line of chickens and a commercial broiler breeder line of chickens under a controlled condition. Data from this study provided an additional piece of experimental evidence on the potency of nonpathogenic ALV-E, MDV-2, and ALV-E plus MDV-2 in boosting the incidence of LL-like lymphomas in susceptible chickens. This study also generated the first piece of genomic evidence that suggests host transcriptomic variation plays an important role in modulating LL-like lymphoma formation.


Asunto(s)
Virus de la Leucosis Aviar/aislamiento & purificación , Leucosis Aviar/complicaciones , Leucosis Aviar/virología , Coinfección/virología , Linfoma/complicaciones , Linfoma/virología , Enfermedad de Marek/complicaciones , Enfermedades de las Aves de Corral/virología , Secuencia de Aminoácidos , Animales , Virus de la Leucosis Aviar/genética , Pollos/virología , Susceptibilidad a Enfermedades , Regulación Viral de la Expresión Génica , Genotipo , Herpesvirus Gallináceo 3 , Incidencia , Enfermedad de Marek/virología , Vacunas contra la Enfermedad de Marek , Análisis de Secuencia de ADN , Transducción de Señal , Transcriptoma , Vacunación , Vacunas Virales
2.
Sensors (Basel) ; 20(11)2020 Jun 11.
Artículo en Inglés | MEDLINE | ID: mdl-32545168

RESUMEN

High-throughput plant phenotyping in controlled environments (growth chambers and glasshouses) is often delivered via large, expensive installations, leading to limited access and the increased relevance of "affordable phenotyping" solutions. We present two robot vectors for automated plant phenotyping under controlled conditions. Using 3D-printed components and readily-available hardware and electronic components, these designs are inexpensive, flexible and easily modified to multiple tasks. We present a design for a thermal imaging robot for high-precision time-lapse imaging of canopies and a Plate Imager for high-throughput phenotyping of roots and shoots of plants grown on media plates. Phenotyping in controlled conditions requires multi-position spatial and temporal monitoring of environmental conditions. We also present a low-cost sensor platform for environmental monitoring based on inexpensive sensors, microcontrollers and internet-of-things (IoT) protocols.


Asunto(s)
Monitoreo del Ambiente , Plantas , Fenotipo
3.
Immunogenetics ; 70(10): 693-694, 2018 11.
Artículo en Inglés | MEDLINE | ID: mdl-29982918

RESUMEN

The Figure 3 in the original version of this article was incorrectly published. In this article the top panel of Figure 3 that describes the amino acid sequence alignment is now added. The original article has been corrected.

4.
Immunogenetics ; 70(9): 599-611, 2018 09.
Artículo en Inglés | MEDLINE | ID: mdl-29947944

RESUMEN

The function of the chicken's major histocompatibility complex (MHC or B complex) class I major (BF2) and minor (BF1) glycoproteins is compared for their expression, ability to present viral antigens to cytotoxic T lymphocytes (CTLs), and interaction with natural killer (NK) cells. MHC-restricted CTLs recognized virus antigen in the context of the BF2*21 major glycoprotein but not the BF1*21 minor glycoprotein. Marek's disease virus (MDV), a large DNA virus known to reduce the cell surface expression of class I glycoprotein, reduced the expression of BF2 glycoprotein while BF1glycoprotein expressions are remained as no change or slight increase. In addition, the expression of BF1*21 class I glycoprotein protected target cells from NK cell lysis while the expression of the BF2*21 class I glycoprotein enhanced NK cell lysis of target cells. Therefore, BF1 and BF2 provide two different cellular immune functions; BF1 negatively regulates the NK cell killing activity and BF2 restricts the antigen specific CTL immune response.


Asunto(s)
Pollos/genética , Antígenos de Histocompatibilidad Clase I/genética , Células Asesinas Naturales/inmunología , Linfocitos T Citotóxicos/inmunología , Animales , Virus de la Leucosis Aviar/inmunología , Virus de la Leucosis Aviar/patogenicidad , Línea Celular , Embrión de Pollo , Pollos/inmunología , Epítopos/metabolismo , Regulación de la Expresión Génica/inmunología , Genes MHC Clase I , Herpesvirus Gallináceo 2/patogenicidad , Antígenos de Histocompatibilidad Clase I/inmunología , Interacciones Huésped-Parásitos/inmunología
5.
Avian Dis ; 59(1): 122-9, 2015 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-26292545

RESUMEN

Over the last five decades, the pathogenicity of the Marek's disease virus (MDV) has evolved from the relatively mild strains (mMDV) observed in the 1960s to the more severe very-virulent-plus strains currently observed in today's outbreaks. The use of vaccines to control Marek's disease (MD), but not the infection cycle, is thought to be the major influence on the evolution of MDV. Selection for genetic resistance to MD has also been employed by the industry to control MD in the commercial setting but the role of host genetics on the evolution of MDV has been difficult to investigate in the field. To investigate the influence of vaccination and host resistance we developed a laboratory model to control and assess the effects of virus and animal genetics on MDV evolution. A bacterial artificial chromosome-derived MDV (Md5B40BAC) was used for in vivo passage (IVP) through turkey herpesvirus (HVT)-vaccinated resistant (MHC-B21) and susceptible (MHC-B13) genetic chicken lines. During IVP in the vaccinated susceptible line, the disease incidence increased from 23% MD in the first IVP to 53% MD during the fifth IVP. In the vaccinated resistant line, disease incidence increased from 0% MD during the first IVP to 29% MD during the fifth IVP. Although the IVP isolates remained relatively mild in the vaccinated resistant chicken line (29% MD) they increased from 0% to 63% MD when used to challenge the vaccinated susceptible chickens. There was no corresponding increase in disease incidence when the virus passed in the vaccinated susceptible genetic line was used to challenge the vaccinated resistant line. From this series of experiments we show that a cloned MDV (Md5B40BAC) can be selected by serial IVP to induce greater disease incidence in vaccinated chickens. This increase in disease incidence occurs in both susceptible and resistant chicken lines but is more easily observed in the susceptible line. Not surprisingly, both host genetics and vaccination play a role in selecting for increased MDV virulence. Our results suggest that the progressive increase in MDV virulence is partially masked as it circulates through vaccinated resistant genetic lines, but by applying this virus to less-resistant genetic lines, virus evolution can be clearly observed. We would predict that the introduction of more-resistant genetic lines into a commercial house contaminated with MDV circulating through susceptible lines would be less likely to produce vaccine breaks than placing susceptible lines into a house in which previously the MDV was circulating through resistant genetic lines.


Asunto(s)
Pollos , Herpesvirus Meleágrido 1/inmunología , Complejo Mayor de Histocompatibilidad , Mardivirus/genética , Enfermedad de Marek/virología , Vacunas Virales/inmunología , Animales , Evolución Biológica , Mardivirus/patogenicidad , Organismos Libres de Patógenos Específicos , Virulencia
6.
Avian Dis ; 59(2): 255-62, 2015 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-26473676

RESUMEN

Laryngotracheitis (LT) is a highly contagious respiratory disease of chickens that produces significant economic losses to the poultry industry. Traditionally, LT has been controlled by administration of modified live vaccines. In recent years, the use of recombinant DNA-derived vaccines using turkey herpesvirus (HVT) and fowlpox virus has expanded, as they protect not only against the vector used but also against LT. However, HVT-based vaccines confer limited protection against challenge, with emergent very virulent plus Marek's disease virus (vv+MDV). Serotype 1 vaccines have been proven to be the most efficient against vv+MDV. In particular, deletion of oncogene MEQ from the oncogenic vvMDV strain Md5 (BACδMEQ) resulted in a very efficient vaccine against vv+MDV. In this work, we have developed two recombinant vaccines against MD and LT by using BACδMEQ as a vector that carries either the LT virus (LTV) gene glycoprotein B (gB; BACΔMEQ-gB) or LTV gene glycoprotein J (gJ; BACδMEQ-gJ). We have evaluated the protection that these recombinant vaccines confer against MD and LT challenge when administered alone or in combination. Our results demonstrated that both bivalent vaccines (BACΔMEQ-gB and BACδMEQ-gJ) replicated in chickens and were safe to use in commercial meat-type chickens bearing maternal antibodies against MDV. BACΔMEQ-gB protected as well as a commercial recombinant (r)HVT-LT vaccine against challenge with LTV. However, BACδMEQ-gJ did not protect adequately against LT challenge or increase protection conferred by BACΔMEQ-gB when administered in combination. On the other hand, both BACΔMEQ-gB and BACδMEQ-gJ, administered alone or in combination, protected better against an early challenge with vv+MDV strain 648A than commercial strains of rHVT-LT or CVI988. Our results open a new avenue in the development of recombinant vaccines by using serotype 1 MDV as vectors.


Asunto(s)
Pollos , Infecciones por Herpesviridae/veterinaria , Herpesvirus Gallináceo 1/inmunología , Mardivirus/clasificación , Enfermedad de Marek/prevención & control , Vacunas Virales/inmunología , Animales , Femenino , Infecciones por Herpesviridae/prevención & control , Mardivirus/inmunología , Proyectos Piloto , Vacunas de ADN , Replicación Viral
7.
Protein Sci ; 33(3): e4898, 2024 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-38358135

RESUMEN

Structural genomics consortia established that protein crystallization is the primary obstacle to structure determination using x-ray crystallography. We previously demonstrated that crystallization propensity is systematically related to primary sequence, and we subsequently performed computational analyses showing that arginine is the most overrepresented amino acid in crystal-packing interfaces in the Protein Data Bank. Given the similar physicochemical characteristics of arginine and lysine, we hypothesized that multiple lysine-to-arginine (KR) substitutions should improve crystallization. To test this hypothesis, we developed software that ranks lysine sites in a target protein based on the redundancy-corrected KR substitution frequency in homologs. This software can be run interactively on the worldwide web at https://www.pxengineering.org/. We demonstrate that three unrelated single-domain proteins can tolerate 5-11 KR substitutions with at most minor destabilization, and, for two of these three proteins, the construct with the largest number of KR substitutions exhibits significantly enhanced crystallization propensity. This approach rapidly produced a 1.9 Å crystal structure of a human protein domain refractory to crystallization with its native sequence. Structures from Bulk KR-substituted domains show the engineered arginine residues frequently make hydrogen-bonds across crystal-packing interfaces. We thus demonstrate that Bulk KR substitution represents a rational and efficient method for probabilistic engineering of protein surface properties to improve crystallization.


Asunto(s)
Lisina , Proteínas , Humanos , Lisina/química , Cristalización , Proteínas/genética , Aminoácidos/química , Cristalografía por Rayos X , Arginina/metabolismo
8.
Avian Dis ; 57(2 Suppl): 474-82, 2013 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-23901764

RESUMEN

Since the first report of a polyneuritis in chickens by Joseph Marek in 1907, the clinical nature of the disease has changed. Over the last five decades, the pathogenicity of the Marek's disease virus (MDV) has continued to evolve from the relatively mild strains observed in the 1960s to the more severe strains labeled very virulent plus currently observed in today's outbreaks. To understand the influence of host genetics, specifically the major histocompatibility complex (MHC), on virus evolution, a bacterial artificial chromosome-derived MDV (Md5B40BAC) was passed in vivo through resistant (MHC-B21) and susceptible (MHC-B13) Line 0 chickens. Criteria for selecting virus isolates for in vivo passage were based on virus replication in white blood cells 21 days after challenge and evaluation of MD pathology at necropsy. In the MHC-B13-susceptible line the Md5B40BAC virulence consistently increased from 18% Marek's disease (MD) after in vivo passage 1 (B13-IVP1 Md5B40BAC) to 94% MD after B13-IVP5 Md5B40BAC challenge. In the MHC-B21-resistant line MD virulence fluctuated from 28% at B21-IVP1 Md5B40BAC to a high of 65% in B21-IVP2 Md5B40BAC back to a low of 23% in B21-IVP5 Md5B40BAC-challenged chicks. Although the B21-IVP5 Md5B40BAC isolates were relatively mild in the MHC-B21 chicken line (56% MDV), they were highly virulent in the MHC-B13 line (100% MDV). From this series of experiments it would appear that MDV evolution toward greater virulence occurs in both susceptible and resistant MHC haplotypes, but the resulting increase in pathogenicity is constrained by the resistant MHC haplotype.


Asunto(s)
Evolución Biológica , Pollos , Herpesvirus Gallináceo 2/genética , Herpesvirus Gallináceo 2/patogenicidad , Complejo Mayor de Histocompatibilidad , Enfermedad de Marek/genética , Enfermedades de las Aves de Corral/genética , Animales , Cromosomas Artificiales Bacterianos/genética , Evolución Molecular , Femenino , Citometría de Flujo/veterinaria , Masculino , Enfermedad de Marek/virología , Reacción en Cadena de la Polimerasa/veterinaria , Enfermedades de las Aves de Corral/virología , Pase Seriado/veterinaria , Virulencia
9.
Sci Rep ; 13(1): 5355, 2023 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-37005445

RESUMEN

Genetically resistant or susceptible chickens to Marek's disease (MD) have been widely used models to identify the molecular determinants of these phenotypes. However, these prior studies lacked the basic identification and understanding of immune cell types that could be translated toward improved MD control. To gain insights into specific immune cell types and their responses to Marek's disease virus (MDV) infection, we used single-cell RNA sequencing (scRNAseq) on splenic cells from MD resistant and susceptible birds. In total, 14,378 cells formed clusters that identified various immune cell types. Lymphocytes, specifically T cell subtypes, were the most abundant with significant proportional changes in some subtypes upon infection. The largest number of differentially expressed genes (DEG) response was seen in granulocytes, while macrophage DEGs differed in directionality by subtype and line. Among the most DEG in almost all immune cell types were granzyme and granulysin, both associated with cell-perforating processes. Protein interactive network analyses revealed multiple overlapping canonical pathways within both lymphoid and myeloid cell lineages. This initial estimation of the chicken immune cell type landscape and its accompanying response will greatly aid efforts in identifying specific cell types and improving our knowledge of host response to viral infection.


Asunto(s)
Herpesvirus Gallináceo 2 , Enfermedad de Marek , Animales , Pollos/genética , Susceptibilidad a Enfermedades , Bazo/metabolismo
10.
Avian Dis ; 55(2): 293-301, 2011 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-21793448

RESUMEN

The mechanism of Marek's disease (MD) vaccination to prevent the lymphoproliferative disease in chickens is not well understood. It is generally recognized that vaccination prevents disease, including the induction of T-cell tumors, but it does not prevent the pathogenic virus from infecting and replicating in the vaccinated host, nor does it prevent bird to bird spread of the oncogenic virus. The stage at which the vaccinated immune system intervenes in the process from infection to the induction of tumors remains obscure. Using a transplantable tumor induced by the Md5 strain of MD virus (MDV), we show that CVI988 vaccination does not prevent the induction of transplantable tumors in the 15I(5) x 7(1) chicken line. A monoclonal tumor with a V beta 1 T-cell receptor spectratype of 207 base pairs was used to follow the transplantable tumor in serial passages in vivo. This transplantable tumor could be passed in vaccinated birds. The length of time between vaccination and challenge (5 to 12 days) had little or no influence on the ability to transfer the tumor. There was variability in the manifestation of the disease produced by the transplanted tumor. Some chickens presented as normal but were still capable of transmitting the transplanted tumor to newly vaccinated recipients via their blood. This indicates that some chickens can control, but not eliminate, the tumor. The variables inducing health or disease in the challenged chickens remain obscure, but environmental or other factors likely depress the immune system allowing the tumor to overwhelm the immune system.


Asunto(s)
Pollos , Vacunas contra la Enfermedad de Marek/inmunología , Enfermedad de Marek/prevención & control , Animales , Neoplasias Hepáticas/patología , Neoplasias Hepáticas/virología , Mardivirus/patogenicidad , Organismos Libres de Patógenos Específicos , Virulencia
11.
Virus Genes ; 40(3): 410-20, 2010 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-20229182

RESUMEN

The Marek's disease virus (MDV, Gallid herpesvirus 2) genome encodes approximately 110 open reading frames (ORFs). Many of these ORFs are annotated based purely on homology to other herpesvirus genes, thus, direct experiments are needed to verify the gene products, especially the hypothetical or MDV-specific ORFs, and characterize their biological function, particularly with respect to pathogenicity in chickens. Previously, a comprehensive two-hybrid assay screen revealed nine specific chicken-MDV protein-protein interactions. In order to characterize the role of hypothetical MDV proteins R-LORF10 and LORF4, which were shown to interact with major histocompatibility complex (MHC) class II beta chain and Ii (invariant or gamma) chain, respectively, recombinant MDVs derived from virulent MDV-BAC clone rMd5-B40 were generated. Recombinant MDV rMd5DeltaR-LORF10 lacked part of the promoter and the first 17 amino acids in both copies of R-LORF10, and rMd5mLORF4 had point mutations in LORF4 that disrupted the start codon and introduced a premature stop codon without altering the amino acid sequence of overlapping ORF UL1, which encodes glycoprotein L (gL). Mutations in either R-LORF10 or LORF4 neither prevent MDV reconstitution from modified MDV-BACs nor significantly alter virus growth rate in vitro. However, MDV generated from rMd5DeltaR-LORF10 had reduced virulence compared to the parental MDV. Surprisingly, MDV with the LORF4 mutations had significantly higher overall MD incidence as measured by mortality, tumor production, and MD symptoms in infected chickens. These results indicate R-LORF10 and LORF4 encode real products, and are involved in MDV virulence although their mechanisms, especially with respect to modulation of MHC class II cell surface expression, are not clearly understood.


Asunto(s)
Técnicas de Inactivación de Genes , Mardivirus/genética , Mardivirus/patogenicidad , Enfermedad de Marek/patología , Enfermedades de las Aves de Corral/patología , Proteínas Virales/genética , Factores de Virulencia/genética , Animales , Células Cultivadas , Pollos , Eliminación de Gen , Mardivirus/crecimiento & desarrollo , Enfermedad de Marek/mortalidad , Enfermedad de Marek/virología , Mutación Puntual , Enfermedades de las Aves de Corral/mortalidad , Enfermedades de las Aves de Corral/virología , Análisis de Supervivencia , Virulencia
12.
Avian Dis ; 54(1 Suppl): 572-5, 2010 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-20521696

RESUMEN

The chicken's major histocompatibility complex (MHC) haplotype has profound influence on the resistance or susceptibility to certain pathogens. For example, the B21 MHC haplotype confers resistance to Marek's disease (MD). However, non-MHC genes are also important in disease resistance. For example, lines 6 and 7 both express the B2 MHC haplotype, but differ in non-MHC genes. Line 6, but not line 7, is highly resistant to tumors induced by the Marek's disease herpesviruses and avian leukosis retroviruses. Recently, survival in the field by Thai indigenous chickens to H5N1 high-pathogenicity avian influenza (HPAI) outbreaks was attributed to the B21 MHC haplotype, whereas the B13 MHC haplotype was associated with high mortality in the field. To determine the influence of the MHC haplotype on HPAI resistance, a series of MHC congenic white leghorn chicken lines (B2, B12, B13, B19, and B21) and lines with different background genes but with the same B2 MHC haplotype (Line 63 and 7(1)) were intranasally challenged with low dose (10 mean chicken lethal doses) of reverse-genetics-derived rg-A/chicken/Indonesia/7/2003 (H5N1) HPAI virus. None of the lines were completely resistant to lethal effects of the challenge, as evidenced by mortality rates ranging from 40% to 100%. The B21 line had mortality of 40% and 70%, and the B13 line had mortality of 60% and 100% in two separate trials. In addition, the mean death times varied greatly between groups, ranging from 3.7 to 6.9 days, suggesting differences in pathogenesis. The data show that the MHC has some influence on resistance to AI, but less than previously proposed, and non-MHC background genes may have a bigger influence on resistance than the MHC.


Asunto(s)
Pollos , Predisposición Genética a la Enfermedad , Subtipo H5N1 del Virus de la Influenza A/inmunología , Gripe Aviar/inmunología , Complejo Mayor de Histocompatibilidad/genética , Animales , Haplotipos , Gripe Aviar/virología
13.
Front Psychol ; 11: 565416, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33424678

RESUMEN

In resistance training, the use of predicting proximity to momentary task failure (MF, i.e., maximum effort), and repetitions in reserve scales specifically, is a growing approach to monitoring and controlling effort. However, its validity is reliant upon accuracy in the ability to predict MF which may be affected by congruence of the perception of effort compared with the actual effort required. The present study examined participants with at least 1 year of resistance training experience predicting their proximity to MF in two different experiments using a deception design. Within each experiment participants performed four trials of knee extensions with single sets (i.e., bouts of repetitions) to their self-determined repetition maximum (sdRM; when they predicted they could not complete the next repetition if attempted and thus would reach MF if they did) and MF (i.e., where despite attempting to do so they could not complete the current repetition). For the first experiment (n = 14) participants used loads equal to 70% of a one repetition maximum (1RM; i.e., the heaviest load that could be lifted for a single repetition) performed in a separate baseline session. Aiming to minimize participants between day variability in repetition performances, in the second separate experiment (n = 24) they used loads equal to 70% of their daily isometric maximum voluntary contraction (MVC). Results suggested that participants typically under predicted the number of repetitions they could perform to MF with a meta-analytic estimate across experiments of 2.0 [95%CIs 0.0 to 4.0]. Participants with at least 1 year of resistance training experience are likely not adequately accurate at gauging effort in submaximal conditions. This suggests that perceptions of effort during resistance training task performance may not be congruent with the actual effort required. This has implications for controlling, programming, and manipulating the actual effort in resistance training and potentially on the magnitude of desired adaptations such as improvements in muscular hypertrophy and strength.

14.
Avian Dis ; 52(3): 433-40, 2008 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-18939631

RESUMEN

Endogenous avian leukosis virus (ALVE) and the ALVE receptor (TVB*S1) status of six commercial chicken lines supplying specific-pathogen-free eggs were analyzed. All commercial chicken lines are certified free of the avian leukosis virus (ALV) by screening for expression of the p27 protein using the standard enzyme-linked immunosorbent assay. The commercial chicken lines A, E, and F contained replication competent ALVE inserts. Line A was fixed for ALVE21, and lines E and F were segregating for ALVE10. In addition, ALVE1 was detected in all the chicken lines. Chicken lines B, D, and F were essentially fixed for the TVB*S1 allele that confers susceptibility to ALVE, whereas lines A, C, B, and E were resistant, containing either the TVB*S3 or TVB*R alleles. The results show that lines selected to be ALV p27 negative give rise to two different genotypes. One genotype lacks the TVB*S1 receptor for ALVE. Chicken lines with the TVB*S1 negative genotype can retain replication competent endogenous virus inserts such as ALVE2, 10, or 21 and still display the p27 negative phenotype. These replication competent ALVE viruses are phenotypically p27 negative in the absence of the TVB*S1 receptor because their chromosomal integration sites restrict transcription and subsequent production of the p27 protein and virus particles to levels below the detection limit. If the TVB*S1 receptor is present, the limited production of ALVE virus particles reinfects and integrates into more productive chromosomal locations in the cell. Increased production of infective virus particles and detectable levels of p27 follow this reinfection and integration into more active regions of the cells genome. The other genotype observed in the commercial lines retains the ALVE receptor (TVB*S1) but either lacks replication competent inserts or expresses the envelope encoded protein from defective inserts such as ALVE3 or ALVE6. In this phenotype, the env-coded glycoprotein encoded by the defective inserts binds to the TVB*S1 receptor and blocks the reinfection of the replication competent ALVE virus. This receptor interference stops reinfection and subsequent production of detectable virus particles and the p27 protein. Mixtures of different p27 negative phenotypes can result in the p27 positive phenotype and ALVE virus production. For example, mixtures of ALVE receptor positive (TVB*S1) but ALVE negative (p27 negative and envelope negative) chick embryo fibroblasts (CEFs) with fibroblasts that are receptor negative but ALVE positive could generate cells expressing high levels of p27 and ALVE virus. In this situation, the undetectable levels of ALVE virus from the receptor negative CEFs would infect and integrate into the receptor positive CEFs and produce detectable levels of ALVE virus. The implications of these findings for vaccine manufacturers and regulatory agencies are discussed.


Asunto(s)
Virus de la Leucosis Aviar/genética , Pollos/metabolismo , Pollos/virología , Susceptibilidad a Enfermedades/virología , Receptores Virales/metabolismo , Replicación Viral/genética , Animales , Southern Blotting , Cruzamiento/métodos , Cartilla de ADN/genética , Huevos/virología , Ensayo de Inmunoadsorción Enzimática , Genotipo , Polimorfismo de Longitud del Fragmento de Restricción , Receptores Virales/genética , Organismos Libres de Patógenos Específicos
15.
Avian Dis ; 50(3): 380-5, 2006 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-17039837

RESUMEN

Commercial Marek's disease (MD) vaccines produced by two manufacturers were tested for possible contamination with avian leukosis virus (ALV). Samples of MD vaccines manufactured by two companies (A and B) were received from a breeder company; samples were also received directly from vaccine company B. Using virus isolation tests, samples initially tested positive for subgroup E (endogenous) ALV. However, upon repassage, the vaccines also tested positive for exogenous ALV. The isolated exogenous ALV proved to be a subgroup A virus, as determined by flow cytometry using polyclonal chicken antibodies specific for various subgroups of ALV, and by DNA sequencing of the envelope glygoprotein (gp85). The exogenous ALV isolated from MD vaccines was inoculated in chickens from ADOL lines 15I(5) x 7(1) and 0 to determine its pathogenicity and compare it with that of Rous-associated-virus-1 (RAV-1), the prototype strain of ALV-A. Each chicken from each line was inoculated with approximately 10,000 infectious units of RAV-1 or the ALV-A isolated from vaccines termed B-39 virus at 7th day of embryonation. At hatch, and at 4, 8, and 16 wk of age, chickens were tested for viremia and cloacal shedding; chickens were also observed for ALV-induced tumors within 16 wk of age. Viremia and cloacal shedding results suggest that chickens from both lines were susceptible to infection with either virus. Within 16 wk of age, the proportion of ALV tumors induced by strain B-39 in line 0 and line 15I5 x 7(1) chickens was 0% and 12%, respectively, compared with 62% and 67% in chickens inoculated with RAV-1. The data indicate that commercial MD vaccines produced by two manufacturers were contaminated with endogenous subgroup E and an exogenous subgroup A ALV. Further, data from biological characterization suggest that the ALV-A isolated from commercial MD vaccines is of low oncogenicity, compared with that of RAV-1. GenBank accession numbers: The gp85 gene sequences of ALV isolated from commercial Marek's disease vaccines have been deposited in GenBank and assigned the following accession numbers: A46 subgroup A, DQ412726 ; B53 subgroup A, DQ412727; A46 subgroup E, DQ412728; B53 subgroup E, DQ412729.


Asunto(s)
Virus de la Leucosis Aviar/aislamiento & purificación , Leucosis Aviar/virología , Pollos/virología , Contaminación de Medicamentos , Vacunas contra la Enfermedad de Marek/análisis , Animales , Virus de la Leucosis Aviar/patogenicidad , Datos de Secuencia Molecular , Enfermedades de las Aves de Corral/virología , Virulencia
16.
Avian Dis ; 50(4): 572-8, 2006 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-17274296

RESUMEN

Infection of broiler chickens with subgroup J avian leukosis virus (ALV) results in the induction of myeloid tumors. However, although egg-type chickens are susceptible to infection with ALV-J, the tumor incidence is very low, and on rare occasions the tumors observed are of the myeloid lineage. We recently described the isolation of an ALV (AF115-4) from commercial egg-type chickens suffering from myeloid leukosis. AF115-4 was initially identified as an ALV-J isolate based on PCR analysis of the long terminal repeat (LTR). However, further characterization of the viral envelope indicated that the virus is recombinant with subgroups B envelope and J LTR. Here we further characterize this recombinant virus at both the molecular and biological levels. We show that the AF115-4 isolate expresses a recombinant envelope glycoprotein encoded by a subgroup B gp85 region and a subgroup E gp37 region. The host range ofAF115-4 was analyzed using cells resistant to infection by subgroups A/B, J, or E; this shows that no ALV-J was present in the isolates obtained from the affected chickens. Additional antigenic characterization of AF115-4 using chicken sera specific for subgroups B or J indicated that no ALV-J was present in the samples examined. Inoculation of AF 115-4 into ALV-susceptible 1515 X 71 chickens resulted in the induction of lymphoid leukosis but not the expected myeloid leukosis affecting the commercial chickens. These results suggest that differences in the genetic makeup of the chickens from which AF115-4 was isolated and the line 1515 X 71 used in the present experiments may be responsible for the observed differences in pathogenicity. In addition, the results suggest that ALV-J continues to evolve by recombination, generating new viruses with different pathological properties.


Asunto(s)
Virus de la Leucosis Aviar/genética , Secuencias Repetidas Terminales/genética , Proteínas del Envoltorio Viral/genética , Secuencia de Aminoácidos , Animales , Leucosis Aviar/virología , Secuencia de Bases , Línea Celular , Pollos/virología , Datos de Secuencia Molecular , Enfermedades de las Aves de Corral/virología , Proteínas del Envoltorio Viral/química
17.
FEMS Microbiol Ecol ; 92(12)2016 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-27604255

RESUMEN

Marek's disease (MD) is an important neoplastic disease of chickens caused by Marek ': s disease virus (MDV), a highly oncogenic alphaherpesvirus. In this study using two chicken lines, one resistant and another susceptible to MD, splenic T cells and cecal microbiome were profiled to gain a better understanding of primary differences in these lines. The percent of splenic CD4+ T cells were similar regardless of MDV challenge status in both bird lines. In contrast, CD8αα profiles were different (P < 0.005) between chicken lines under naïve status and under MDV challenge, suggesting that CD8αα T cells play a key role in mediating MDV infection. Microbiome composition was different between naïve resistant (Blautia spp.) and susceptible birds (Streptococcus spp.) (P < 0.05) during initial colonization. With MDV challenge, both chicken lines showed lower numbers of beneficial Faecalibacterium spp. and increased number of Lactobacillus spp. Metabolic profiles between naïve chicken types were similar but with MDV challenge, there were differences in metabolism in both chicken lines, with amino acid metabolism impacted in resistant birds and lipid metabolism in susceptible birds. These results provide insights into immune response and potential interplay with the microbiome during infection with an oncogenic virus.

18.
Avian Dis ; 49(2): 199-206, 2005 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-16094823

RESUMEN

Marek's disease virus (MDV) is an alpha-herpesvirus that causes rapid development of T-cell lymphomas in chickens. MDV-encoded vIL-8 is homologous to the cellular IL-8 gene, and its function in MDV pathogenesis has yet to be determined. Using overlapping cosmid clone-based technology, we have generated an MDV vIL-8 deletion mutant virus, rMd5/delta vIL-8. In vivo experiments with this mutant virus demonstrated that deletion of vIL-8 results in attenuation of the virus and induction of significantly less gross tumor, both in viscera and nerves, when compared to the parental virus. Reintroduction of the vIL-8 gene in the genome of the mutant virus restored the virulence of the virus to the wild-type levels, indicating that vIL-8 plays a role in MDV-induced pathogenesis. In this study, we show that there is a significant difference in the reduction of B cells and activation of T cells in the spleen cells of chickens inoculated with parental rMd5 and vIL-8 deletion mutant virus. These results indicate that vIL-8 is involved in the early phase of pathogenesis, presumably by attracting target cells to the initial site of infection. In addition, protection studies with the vIL-8 mutant virus showed that this mildly virulent virus protects susceptible maternal antibody-positive viruses at a higher level than the commonly used serotype 1 CVI988 vaccine. These results confirm the potential of partially attenuated viruses as vaccines against very virulent plus strains and the usefulness of recombinant DNA technology to generate the next generation of MDV vaccines.


Asunto(s)
Pollos , Eliminación de Gen , Herpesvirus Gallináceo 2/patogenicidad , Inmunidad Celular/inmunología , Interleucina-8/genética , Enfermedad de Marek/prevención & control , Enfermedades de las Aves de Corral/prevención & control , Animales , Anticuerpos Monoclonales/inmunología , Citometría de Flujo/veterinaria , Herpesvirus Gallináceo 2/genética , Inmunohistoquímica/veterinaria , Enfermedad de Marek/inmunología , Enfermedades de las Aves de Corral/inmunología , Enfermedades de las Aves de Corral/virología , Virulencia
19.
Virology ; 475: 88-95, 2015 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-25462349

RESUMEN

Marek׳s disease virus (MDV) is a widespread α-herpesvirus of chickens that causes T cell tumors. Acute, but not latent, MDV infection has previously been shown to lead to downregulation of cell-surface MHC class I (Virology 282:198-205 (2001)), but the gene(s) involved have not been identified. Here we demonstrate that an MDV gene, MDV012, is capable of reducing surface expression of MHC class I on chicken cells. Co-expression of an MHC class I-binding peptide targeted to the endoplasmic reticulum (bypassing the requirement for the TAP peptide transporter) partially rescued MHC class I expression in the presence of MDV012, suggesting that MDV012 is a TAP-blocking MHC class I immune evasion protein. This is the first unique non-mammalian MHC class I immune evasion gene identified, and suggests that α-herpesviruses have conserved this function for at least 100 million years.


Asunto(s)
Regulación de la Expresión Génica/inmunología , Antígenos de Histocompatibilidad Clase I/metabolismo , Evasión Inmune/genética , Mardivirus/genética , Proteínas Virales/metabolismo , Secuencia de Aminoácidos , Animales , Anticuerpos Antivirales/inmunología , Línea Celular , Pollos , Evasión Inmune/fisiología , Mardivirus/metabolismo , Datos de Secuencia Molecular , Proteínas Virales/genética
20.
Avian Dis ; 48(3): 471-87, 2004 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-15529969

RESUMEN

Marek's disease (MD) is caused by Marek's disease virus (MDV), a highly cell-associated alphaherpesvirus. MD is primarily characterized by lymphocyte infiltration of the nerves and the development of lymphomas in visceral organs, muscle, and skin. MDV encodes two phosphoproteins, pp24 and pp38, that are highly expressed during lytic infection. These proteins were initially identified in MDV-induced tumors but are now known to be linked primarily to MDV lytic infection. Despite the recent characterization of a pp38 deletion mutant MDV, the functions of these phosphoproteins remain unknown. The goal of this work was to construct recombinant MDVs having direct fusions of a marker gene, the green fluorescent protein (GFP), to pp38 in order to study the expression patterns and localization of this protein during stages of MDV infection. We report the construction of two recombinant viruses, one having the enhanced green fluorescent protein (eGFP) fused in-frame to the pp38 open reading frame (ORF) (RB1Bpp38/eGFP) and the other having soluble-modified GFP (smGFP) downstream but out-of-frame with pp38 (RB1Bpp38/smGFP). During construction of RB1Bpp38/eGFP, an ORF located downstream of pp38 (LORF12) was partially deleted. In RB1Bpp38/smGFP, however, LORF12 and its immediate 5' upstream sequence was left intact. This report describes the construction, cell culture, and in vivo characterization of RB1Bpp38/eGFP and RB1Bpp38/smGFP. Structural analysis showed that the virus stocks of RB1Bpp38/eGFP and RB1Bpp38/smGFP had incorporated the GFP cassette and were free of contaminating parent virus (RB1B). Moreover, RB1Bpp38/eGFP and RB1Bpp38/smGFP contained two and three and four and five copies of the 132-bp repeats, respectively. Expression analysis showed that the transcription of genes in RB1Bpp38/eGFP-and RB1Bpp38/smGFP-infected chicken embryo fibroblasts (CEFs) were similar to RB1B-infected CEFs, with the notable exception of deletion of a LORF12-specific transcript in RB1Bpp38/ eGFP-infected cells. In CEFs, RB1Bpp38/eGFP and RB1Bpp38/smGFP showed comparable one-step growth kinetics to parental virus (RB1B). RB1Bpp38/eGFP and RB1Bpp38/smGFP, however, showed quite distinct growth characteristics in vivo. Two independent clones of RB1Bpp38/eGFP were highly attenuated, whereas RB1Bpp38/smGFP exhibited pathogenesis similar to parent virus and retained oncogenicity. Our results suggest that the RB1Bpp38/eGFP phenotype could be due to an interference with an in vivo-specific pp38 function via GFP direct fusion, to the deletion of LORF12, or to a targeting of the immune response to eGFP. Because deletion of pp38 was recently found not to fully attenuate very virulent MDV strain MD-5, it is possible that deletion of LORF12 may be at least partially responsible for the attenuation of RB1Bpp38/eGFP. The construction of these viruses and the establishment of cell lines from RB1Bpp38/smGFP provide useful tools for the study of MDV lyric infection in cell culture and in vivo, in studies of the reactivation of MDV from latency, and in the functional analysis of LORF12.


Asunto(s)
Antígenos Virales/metabolismo , Pollos/virología , Genes Virales , Proteínas Fluorescentes Verdes/metabolismo , Mardivirus/metabolismo , Enfermedad de Marek/metabolismo , Fosfoproteínas/metabolismo , Enfermedades de las Aves de Corral/virología , Secuencia de Aminoácidos , Animales , Antígenos Virales/genética , Northern Blotting , Southern Blotting , Cartilla de ADN , Componentes del Gen , Eliminación de Gen , Expresión Génica , Técnicas de Transferencia de Gen , Mardivirus/genética , Técnicas de Amplificación de Ácido Nucleico , Fosfoproteínas/genética , Plásmidos/genética , Enfermedades de las Aves de Corral/metabolismo , Proteínas Recombinantes de Fusión/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA