Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 264
Filtrar
Más filtros

Tipo del documento
Intervalo de año de publicación
1.
Arch Virol ; 163(10): 2871-2875, 2018 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-29982961

RESUMEN

A codon-optimized equine infectious anemia virus p26 gene was fused to a maltose-binding protein (MBP) and expressed in Escherichia coli for use as an antigen in agar gel immunodiffusion (AGID) and enzyme-linked immunosorbent assay (ELISA) for diagnosis of equine infectious anemia. An analysis of analytical sensitivity and specificity showed that the antigen MBP-p26rec reacted positively with a reference World Organization for Animal Health serum and demonstrated no cross-reaction against sera from vaccinated animals in either test. The diagnostic characteristics were evaluated and presented excellent values. The AGIDrec showed 100% sensitivity and specificity, and the ELISArec showed 100% sensitivity and 99.64% specificity. In addition, MBP-p26rec was stabile after three years of storage at 4 °C, maintaining its immunoreactivity.


Asunto(s)
Ensayo de Inmunoadsorción Enzimática/métodos , Anemia Infecciosa Equina/virología , Inmunodifusión/métodos , Virus de la Anemia Infecciosa Equina/aislamiento & purificación , Proteínas de Unión a Maltosa/análisis , Proteínas del Núcleo Viral/análisis , Animales , Ensayo de Inmunoadsorción Enzimática/instrumentación , Anemia Infecciosa Equina/diagnóstico , Anemia Infecciosa Equina/inmunología , Caballos , Inmunodifusión/instrumentación , Virus de la Anemia Infecciosa Equina/genética , Virus de la Anemia Infecciosa Equina/inmunología , Proteínas de Unión a Maltosa/genética , Proteínas de Unión a Maltosa/inmunología , Proteínas del Núcleo Viral/genética , Proteínas del Núcleo Viral/inmunología
2.
J Immunol ; 196(4): 1943-54, 2016 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-26764037

RESUMEN

MHC class I (MHC I)-restricted virus-specific CTLs are implicated as critical components in the control of this naturally occurring lentivirus and in the protective immune response to the successfully applied attenuated equine infectious anemia virus vaccine in the horse. Nevertheless, the structural basis for how the equine MHC I presents epitope peptides remains unknown. In this study, we investigated the binding of several equine infectious anemia virus-derived epitope peptides by the ability to refold recombinant molecules and by thermal stability, and then by determining the x-ray structure of five peptide-MHC I complexes: equine MHC class I allele (Eqca)-N*00602/Env-RW12, Eqca-N*00602/Gag-GW12, Eqca-N*00602/Rev-QW11, Eqca-N*00602/Gag-CF9, and Eqca-N*00601/Gag-GW12. Although Eqca-N*00601 and Eqca-N*00602 differ by a single amino acid, Eqca-N*00601 exhibited a drastically different peptide presentation when binding a similar CTL epitope, Gag-GW12; the result makes the previously reported function clear to be non-cross-recognition between these two alleles. The structures plus Eqca-N*00602 complexed with a 9-mer peptide are particularly noteworthy in that we illuminated differences in apparent flexibility in the center of the epitope peptides for the complexes with Gag-GW12 as compared with Env-RW12, and a strict selection of epitope peptides with normal length. The featured preferences and unconventional presentations of long peptides by equine MHC I molecules provide structural bases to explain the exceptional anti-lentivirus immunity in the horse. We think that the beneficial reference points could serve as an initial platform for other human or animal lentiviruses.


Asunto(s)
Presentación de Antígeno/inmunología , Epítopos de Linfocito T/inmunología , Antígenos de Histocompatibilidad Clase I/inmunología , Caballos/inmunología , Virus de la Anemia Infecciosa Equina/inmunología , Activación de Linfocitos/inmunología , Alelos , Secuencia de Aminoácidos , Animales , Antígenos Virales/inmunología , Cristalografía por Rayos X , Anemia Infecciosa Equina/inmunología , Datos de Secuencia Molecular , Linfocitos T Citotóxicos/inmunología
3.
PLoS Pathog ; 11(1): e1004610, 2015 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-25569288

RESUMEN

Lentiviral Envelope (Env) antigenic variation and related immune evasion present major hurdles to effective vaccine development. Centralized Env immunogens that minimize the genetic distance between vaccine proteins and circulating viral isolates are an area of increasing study in HIV vaccinology. To date, the efficacy of centralized immunogens has not been evaluated in the context of an animal model that could provide both immunogenicity and protective efficacy data. We previously reported on a live-attenuated (attenuated) equine infectious anemia (EIAV) virus vaccine, which provides 100% protection from disease after virulent, homologous, virus challenge. Further, protective efficacy demonstrated a significant, inverse, linear relationship between EIAV Env divergence and protection from disease when vaccinates were challenged with viral strains of increasing Env divergence from the vaccine strain Env. Here, we sought to comprehensively examine the protective efficacy of centralized immunogens in our attenuated vaccine platform. We developed, constructed, and extensively tested a consensus Env, which in a virulent proviral backbone generated a fully replication-competent pathogenic virus, and compared this consensus Env to an ancestral Env in our attenuated proviral backbone. A polyvalent attenuated vaccine was established for comparison to the centralized vaccines. Additionally, an engineered quasispecies challenge model was created for rigorous assessment of protective efficacy. Twenty-four EIAV-naïve animals were vaccinated and challenged along with six-control animals six months post-second inoculation. Pre-challenge data indicated the consensus Env was more broadly immunogenic than the Env of the other attenuated vaccines. However, challenge data demonstrated a significant increase in protective efficacy of the polyvalent vaccine. These findings reveal, for the first time, a consensus Env immunogen that generated a fully-functional, replication-competent lentivirus, which when experimentally evaluated, demonstrated broader immunogenicity that does not equate to higher protective efficacy.


Asunto(s)
Anemia Infecciosa Equina/prevención & control , Caballos/inmunología , Virus de la Anemia Infecciosa Equina/inmunología , Vacunas Atenuadas/uso terapéutico , Proteínas del Envoltorio Viral/inmunología , Secuencia de Aminoácidos , Animales , Variación Antigénica/inmunología , Secuencia de Bases , Variación Genética , Virus de la Anemia Infecciosa Equina/genética , Datos de Secuencia Molecular , Filogenia , Resultado del Tratamiento , Proteínas del Envoltorio Viral/genética , Vacunas Virales/uso terapéutico
4.
Microb Pathog ; 106: 65-68, 2017 May.
Artículo en Inglés | MEDLINE | ID: mdl-27816678

RESUMEN

Tetherin (BST-2) is an important host restriction factor that can inhibit the release of a diverse array of enveloped viruses from infected cells. Conversely, to facilitate their release and spread, many viruses have evolved various strategies to overcome the antiviral effect of tetherin in a species-specific manner. During the development of an attenuated equine infectious anemia virus (EIAV) vaccine in our laboratory, we found that serial passage of a field-isolated virulent EIAV strains in horse and donkey as well as the cultivated donkey cells, produces several typical EIAV strains, including EIAVDV, EIAVDLV, and EIAVFDDV, which exhibit distinct virulence and replication features in vivo and in vitro. However, the role of host restriction factors in EIAV evolution during the serial passage is not well understood. This study aimed to evaluate whether these newly generated strains adapt differently to donkey tetherin (do-tetherin) based on their virulence. We found that do-tetherin exerts an inhibition on the release of the viral particles produced by all three strains, albeit with varying intensity: EIAVDV < EIAVDLV < EIAVFDDV. Additionally, all three EIAV strains could counteract the restriction mediated by do-tetherin via their envelope proteins (Env) with varying strength: EIAVDV > EIAVDLV > EIAVFDDV. These results indicate that donkey tetherin is involved in shaping of EIAV evolution during serial passage.


Asunto(s)
Antígenos CD/inmunología , Antígenos CD/farmacología , Proteínas Ligadas a GPI/inmunología , Proteínas Ligadas a GPI/farmacología , Virus de la Anemia Infecciosa Equina/efectos de los fármacos , Virus de la Anemia Infecciosa Equina/inmunología , Animales , Evolución Biológica , Células Cultivadas , ADN Viral , Equidae , Células HEK293 , Caballos , Humanos , Inmunidad Innata , Virus de la Anemia Infecciosa Equina/crecimiento & desarrollo , Mutación , Proyectos Piloto , Vacunas Atenuadas/inmunología , Proteínas del Envoltorio Viral/efectos de los fármacos , Proteínas del Envoltorio Viral/genética , Vacunas Virales/inmunología , Virión/efectos de los fármacos , Virulencia , Replicación Viral/efectos de los fármacos
5.
BMC Vet Res ; 13(1): 105, 2017 Apr 14.
Artículo en Inglés | MEDLINE | ID: mdl-28410613

RESUMEN

BACKGROUND: ELISAs are known to have a higher diagnostic sensitivity than the agar gel immunodiffusion (AGID) when employed for serological diagnosis of equine infectious anaemia (EIA). For this purpose, an "in-house" and five commercial ELISAs available in Italy were assessed by the National Reference Centre for EIA for their analytic specificity (Sp); precocity, defined as capability of detecting first antibodies produced during a new infection; precision based on repeatability and reproducibility, estimated from the coefficient of variation (CV); accuracy, estimated from multiple K and relative Sp and sensitivity (Se). Two serum panels, positive for non-equine retroviruses and the most frequent equine viruses, were employed to measure analytic Sp. ELISA precocity was also compared to that of one "in-house" and three commercial AGID kits, employing a panel of sera, collected weekly from horses infected with modified EIA viruses. Precision and accuracy were defined using results of a panel containing positive and negative sera examined in an inter-laboratory trial with the participation of the ten Official Laboratories. Furthermore, a questionnaire was used to assess the appropriateness of each kit for routine use. RESULTS: Analytic Sp was 100%, while the 75th percentile of CVs for positive sera varied from 0.4% to 12.73% for repeatability and from 1.6% to 44.87% for reproducibility. Although CV of the negative serum was constantly high, its outcome was unaltered. Relative Se ranged from 98.2% to 100%, relative Sp was constantly 100% and multiple K ranged from 0.95 to 1. Precocity differed among the assays: three kits detected 4.8% and 42.9% positive samples on 21 days post infection (dpi), all assays detected positive samples on 28 dpi, between 47.6% and 95.2%. Precocity of ELISAs was superior to that of the AGIDs except for two assays. In view of the feedback obtained from the questionnaires, all kits were considered appropriate for routine use. CONCLUSION: All ELISAs having high Se and precocity are preferable as a screening test in EIA surveillance programmes to the AGID tests examined. These two tests can be incorporated in a serial diagnostic pathway to improve the efficacy of a surveillance plan.


Asunto(s)
Ensayo de Inmunoadsorción Enzimática/veterinaria , Anemia Infecciosa Equina/diagnóstico , Virus de la Anemia Infecciosa Equina/inmunología , Animales , Anticuerpos Antivirales/sangre , Ensayo de Inmunoadsorción Enzimática/métodos , Anemia Infecciosa Equina/virología , Caballos , Italia , Reproducibilidad de los Resultados , Sensibilidad y Especificidad
6.
Retrovirology ; 13: 9, 2016 Feb 03.
Artículo en Inglés | MEDLINE | ID: mdl-26842878

RESUMEN

BACKGROUND: The equine infectious anemia virus (EIAV) vaccine is the only attenuated lentiviral vaccine applied on a large scale that has been shown to be effective in controlling the prevalence of EIA in China. This vaccine was developed by successive passaging of a field-isolated virulent strain in different hosts and cultivated cells. To explore the molecular basis for the phenotype alteration of this vaccine strain, we systematically analyzed its genomic evolution during vaccine development. RESULTS: Sequence analysis revealed that the genetic distance between the wild-type strain and six representative strains isolated from key development stages gradually increased with the number of passages. Env gene, but not gag and pol, showed a clear evolutionary flow similar to that of the whole genomes of different generations during the attenuation. Stable mutations were identified in multiple regions of multiple genes along with virus passaging. The adaption of the virus to the growth environment of cultured cells with accumulated genomic and genetic variations was positively correlated with the reduction in pathogenicity and rise of immunogenicity. Statistical analyses revealed significant differences in the frequency of the most stable mutations between in vivo and ex vivo-adapted strains and between virulent and attenuated strains. CONCLUSIONS: These data indicate that EIAV evolution during vaccine development generated an accumulation of mutations under the selective drive force, which helps to better understand the molecular basis of lentivirus pathogenicity and immunogenicity.


Asunto(s)
Anemia Infecciosa Equina/prevención & control , Evolución Molecular , Virus de la Anemia Infecciosa Equina/inmunología , Vacunas Virales/inmunología , Animales , China , Equidae , Caballos , Virus de la Anemia Infecciosa Equina/genética , Virus de la Anemia Infecciosa Equina/patogenicidad , Datos de Secuencia Molecular , Mutación , Análisis de Secuencia de ADN , Pase Seriado , Vacunas Atenuadas/genética , Vacunas Atenuadas/inmunología , Vacunas Atenuadas/aislamiento & purificación , Vacunas Virales/genética , Vacunas Virales/aislamiento & purificación
7.
J Virol ; 89(13): 6945-51, 2015 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-25878104

RESUMEN

Lentivirus escape from neutralizing antibodies (NAbs) is not well understood. In this work, we quantified antibody escape of a lentivirus, using antibody escape data from horses infected with equine infectious anemia virus. We calculated antibody blocking rates of wild-type virus, fitness costs of mutant virus, and growth rates of both viruses. These quantitative kinetic estimates of antibody escape are important for understanding lentiviral control by antibody neutralization and in developing NAb-eliciting vaccine strategies.


Asunto(s)
Anticuerpos Neutralizantes/sangre , Anticuerpos Antivirales/sangre , Anemia Infecciosa Equina/inmunología , Anemia Infecciosa Equina/virología , Evasión Inmune , Virus de la Anemia Infecciosa Equina/inmunología , Animales , Caballos , Virus de la Anemia Infecciosa Equina/genética , Virus de la Anemia Infecciosa Equina/crecimiento & desarrollo , Modelos Teóricos , Mutación
8.
Virus Genes ; 52(6): 814-822, 2016 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-27572122

RESUMEN

The Chinese EIAV vaccine is an attenuated live virus vaccine obtained by serial passage of a virulent horse isolate (EIAVL) in donkeys (EIAVD) and, subsequently, in donkey cells in vitro. In this study, we compare the env gene of the original horse virulent virus (EIAVL) with attenuated strains serially passaged in donkey MDM (EIAVDLV) and donkey dermal cells (EIAVFDDV). Genetic comparisons among parental and attenuated strains found that vaccine strains contained amino acid substitutions/deletions in gp90 that resulted in a loss of three potential N-linked glycosylation sites, designated g5, g9, and g10. To investigate the biological significance of these changes, reverse-mutated viruses were constructed in the backbone of the EIAVFDDV infectious molecular clone (pLGFD3). The resulting virus stocks were characterized for replication efficiency in donkey dermal cells and donkey MDM, and were tested for sensitivity to neutralization using sera from two ponies experimentally infected with EIAVFDDV. Results clearly show that these mutations generated by site-directed mutagenesis resulted in cloned viruses with enhanced resistance to serum neutralizing antibodies that were also able to recognize parental viruses. This study indicates that these mutations played an important role in the attenuation of the EIAV vaccine strains.


Asunto(s)
Sustitución de Aminoácidos , Productos del Gen env/genética , Productos del Gen env/inmunología , Virus de la Anemia Infecciosa Equina/genética , Virus de la Anemia Infecciosa Equina/inmunología , Mutación , Vacunas Virales/genética , Vacunas Virales/inmunología , Secuencia de Aminoácidos , Animales , Anticuerpos Neutralizantes/inmunología , Anticuerpos Antivirales/inmunología , Línea Celular , Células Cultivadas , Productos del Gen env/química , Productos del Gen env/metabolismo , Genoma Viral , Glicosilación , Caballos , Pruebas de Neutralización
9.
Appl Microbiol Biotechnol ; 100(24): 10531-10542, 2016 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-27660181

RESUMEN

The equine infectious anemia virus (EIAV) capsid protein (p26) is one of the major immunogenic proteins during EIAV infection and is widely used for the detection of EIAV antibodies in horses. However, few reports have described the use of EIAV-specific monoclonal antibodies (MAbs) in etiological and immunological detection. Previously, we developed an antigen capture enzyme-linked immunosorbent assay (AC-ELISA) for the quantification of the EIAV p26 protein level. However, the epitopes recognized by the MAbs were not identified, and the utilization of the MAbs needs to be evaluated. In this study, we characterized two monoclonal antibodies (9H8 and 1G11 MAbs) against EIAV p26. Two B-cell epitopes are located in amino acid residues, 73NLDKIAEE81 (HE) and 199KNAMRHLRPEDTLEEKMYAC218 (GE) for the 9H8 and 1G11 MAbs, respectively. The 1G11 epitope (GE) varied among viruses isolated worldwide but can be recognized by anti-EIAV sera from different regions, including China, the USA, and Argentina. Meanwhile, 1G11 MAb could react with the mutants of almost all the EIAV strains. Furthermore, we found that the histidine at position 204 (H204), leucine at position 205 (L205), and aspartic acid at position 209 (D209) of EIAV p26 individually played pivotal roles in binding with the 1G11 MAb. Our results revealed that the GE peptide might be a common B-cell binding epitope of EIAV antibodies. This is also the first report to identify a broad-spectrum monoclonal antibody (1G11) against p26 of EIAV. These findings may provide a useful basis for the development of new diagnostic assays for EIAV.


Asunto(s)
Mapeo Epitopo , Epítopos de Linfocito B/inmunología , Virus de la Anemia Infecciosa Equina/inmunología , Proteínas del Núcleo Viral/inmunología , Anticuerpos Monoclonales/inmunología , Anticuerpos Antivirales/inmunología , Argentina , China , Virus de la Anemia Infecciosa Equina/aislamiento & purificación , Estados Unidos
10.
J Virol ; 88(21): 12296-310, 2014 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-25122784

RESUMEN

UNLABELLED: Viperin is an endoplasmic reticulum (ER)-associated multifunctional protein that regulates virus replication and possesses broad antiviral activity. In many cases, viperin interferes with the trafficking and budding of viral structural proteins by distorting the membrane transportation system. The lentivirus equine infectious anemia virus (EIAV) has been studied extensively. In this study, we examined the restrictive effect of equine viperin (eViperin) on EIAV replication and investigated the possible molecular basis of this restriction to obtain insights into the effect of this cellular factor on retroviruses. We demonstrated that EIAV infection of primary equine monocyte-derived macrophages (eMDMs) upregulated the expression of eViperin. The overexpression of eViperin significantly inhibited the replication of EIAV in eMDMs, and knockdown of eViperin transcription enhanced the replication of EIAV in eMDMs by approximately 45.8%. Further experiments indicated that eViperin restricts EIAV at multiple steps of viral replication. The overexpression of eViperin inhibited EIAV Gag release. Both the α-helix domain and radical S-adenosylmethionine (SAM) domain were required for this activity. However, the essential motifs in SAM were different from those reported for the inhibition of HIV-1 Gag by human viperin. Furthermore, eViperin disrupted the synthesis of both EIAV Env and receptor, which consequently inhibited viral production and entry, respectively, and this disruption was dependent on the eViperin α-helix domain. Using immunofluorescence assays and electron microscopy, we demonstrated that the α-helix domain is responsible for the distortion of the endoplasmic reticulum (ER). Finally, EIAV did not exhibit counteracting eViperin at the protein level. IMPORTANCE: In previous studies, viperin was indicated as restricting virus replications primarily by the inhibition of virus budding. Here, we show that viperin may have multiple antiviral mechanisms, including the reduction of EIAV Gag budding and Env expression, and these activities are dependent on different viperin domains. We especially demonstrate that the overexpression of viperin inhibits EIAV entry by decreasing the level of virus receptor. Therefore, viperin restriction of viruses is determined largely by the dependence of virus on the cellular membrane transportation system.


Asunto(s)
Retículo Endoplásmico/metabolismo , Retículo Endoplásmico/virología , Interacciones Huésped-Patógeno , Virus de la Anemia Infecciosa Equina/inmunología , Virus de la Anemia Infecciosa Equina/fisiología , Proteínas Virales/metabolismo , Replicación Viral , Animales , Células Cultivadas , Retículo Endoplásmico/ultraestructura , Técnica del Anticuerpo Fluorescente , VIH-1 , Caballos , Macrófagos/inmunología , Macrófagos/virología , Microscopía Electrónica , Liberación del Virus
11.
Rev Argent Microbiol ; 47(1): 25-8, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25735214

RESUMEN

The most used and reliable indicator of Equine infectious anemia virus (EIAV) infection is the detection of its specific antibodies in horse serum. In the present study, the performance of two commercial ELISA tests for the detection of EIAV antibodies as well as the potential advantages of their use as an EIAV infection screening tool were evaluated in 302 horse serum samples. Both ELISA assays showed 100% diagnostic sensitivity, and 92.3-94.3% diagnostic specificity. Discordant results were analyzed by immunoblot. The results showed that both ELISA tests are very efficient at detecting EIAV infected animals, allowing to identify a higher number of positive horse cases. Thus, ELISA assays can be useful tools in EIA control and eradication.


Asunto(s)
Anticuerpos Antivirales/sangre , Ensayo de Inmunoadsorción Enzimática , Anemia Infecciosa Equina/diagnóstico , Virus de la Anemia Infecciosa Equina/inmunología , Animales , Caballos , Juego de Reactivos para Diagnóstico
12.
Appl Microbiol Biotechnol ; 98(21): 9073-81, 2014 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-25256618

RESUMEN

An antigen capture enzyme-linked immunosorbent assay (AC-ELISA) was established based on two monoclonal antibodies (mAbs) for the quantification of equine infectious anemia virus (EIAV). Two p26-specific monoclonal antibodies were developed in mice. The mAb 9H8 was coated in microtiter plates as the capture antibody; the other mAb, 1G11, was coupled to horseradish peroxidase (HRP) and used as the detection antibody. The limit of detection for the EIAV p26 protein was 0.98 ng/ml, and the linearity range was 3.9-62.5 ng/ml. The sensitivity of p26 AC-ELISA for the detection of the virus (EIAV infectious clone, FDDVcmv3-8) was the same as that for the purified p26 protein. No cross-reaction with other equine viruses was observed by this method. The intra- and inter-assay coefficients of variation were below 8.3 and 10.3 % for testing p26 and FDDVcmv3-8, respectively. The AC-ELISA was also compared to Western blotting (WB) and reverse transcriptase (RT) assays, validating the sensitivity, accuracy, and reliability of this method. Both the AC-ELISA and RT assay showed good agreement, with a correlation coefficient of R (2) =0.9946. Sample analysis showed that this AC-ELISA is a useful tool for quantifying EIAV p26 in cell lysates and culture medium.


Asunto(s)
Virus de la Anemia Infecciosa Equina/inmunología , Proteínas del Núcleo Viral/análisis , Virología/métodos , Animales , Anticuerpos Monoclonales/aislamiento & purificación , Anticuerpos Antivirales/aislamiento & purificación , Ensayo de Inmunoadsorción Enzimática/métodos , Ratones , Reproducibilidad de los Resultados , Sensibilidad y Especificidad
13.
ScientificWorldJournal ; 2014: 620906, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24523642

RESUMEN

Thermostabilizing effect of heavy water (D2O) or deuterium oxide has been demonstrated previously on several enzymes and vaccines like oral poliovirus vaccine and influenza virus vaccine. In view of the above observations, effect of heavy water on in situ thermostabilization of recombinant p26 protein on enzyme-linked immunosorbent assay (ELISA) for serodiagnosis of equine infectious anemia virus (EIAV) infection was investigated in the present study. The carbonate-bicarbonate coating buffer was prepared in 60% and 80% D2O for coating the p26 protein in 96-well ELISA plate and thermal stability was examined at 4 °C, 37 °C, 42 °C, and 45 °C over a storage time from 2 weeks to 10 months. A set of positive serum (n = 12) consisting of strong, medium, and weak titer strength (4 samples in each category) and negative serum (n = 30) were assessed in ELISA during the study period. At each time point, ELISA results were compared with fresh plate to assess thermal protective effect of D2O. Gradual increase in the stabilizing effect of 80% D2O at elevated temperature (37 °C < 42 °C < 45 °C) was observed. The 80% D2O provides the thermal protection to rp26 protein in ELISA plate up to 2 months of incubation at 45 °C. The findings of the present study have the future implication of adopting cost effective strategies for generating more heat tolerable ELISA reagents with extended shelf life.


Asunto(s)
Óxido de Deuterio , Ensayo de Inmunoadsorción Enzimática/métodos , Anemia Infecciosa Equina/diagnóstico , Virus de la Anemia Infecciosa Equina , Proteínas Recombinantes , Proteínas Virales , Animales , Anticuerpos Antivirales/inmunología , Caballos , Virus de la Anemia Infecciosa Equina/inmunología , Estabilidad Proteica , Sensibilidad y Especificidad , Temperatura , Proteínas Virales/inmunología
14.
J Virol ; 85(19): 10421-4, 2011 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-21752904

RESUMEN

Two variants of equine infectious anemia virus (EIAV) that differed in sensitivity to broadly neutralizing antibody were tested in direct competition assays. No differences were observed in the growth curves and relative fitness scores of EIAVs of principal neutralizing domain variants of groups 1 (EIAV(PND-1)) and 5 (EIAV(PND-5)), respectively; however, the neutralization-resistant EIAV(PND-5) variant was less infectious in single-round replication assays. Infectious center assays indicated similar rates of cell-to-cell spread, which was approximately 1,000-fold more efficient than cell-free infectivity. These data indicate that efficient cell-to-cell spread can overcome the decreased infectivity that may accompany immune escape and should be considered in studies assessing the relative levels of fitness among lentivirus variants, including HIV-1.


Asunto(s)
Anticuerpos Neutralizantes/inmunología , Virus de la Anemia Infecciosa Equina/crecimiento & desarrollo , Virus de la Anemia Infecciosa Equina/inmunología , Mutación , Animales , Anticuerpos Antivirales/inmunología , Línea Celular , Virus de la Anemia Infecciosa Equina/genética , Virus de la Anemia Infecciosa Equina/patogenicidad , Pruebas de Neutralización , Virulencia
15.
J Virol ; 85(13): 6814-8, 2011 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-21543497

RESUMEN

Using the equine infectious anemia virus (EIAV) lentivirus model system, we previously demonstrated protective effects of broadly neutralizing immune plasma in young horses (foals) with severe combined immunodeficiency (SCID). However, in vivo selection of a neutralization-resistant envelope variant occurred. Here, we determined the protective effects of purified immunoglobulin with more potent broadly neutralizing activity. Overall, protection correlated with the breadth and potency of neutralizing activity in vitro. Four of five SCID foals were completely protected against homologous challenge, while partial protection occurred following heterologous challenge. These results support the inclusion of broadly neutralizing antibodies in lentivirus control strategies.


Asunto(s)
Anticuerpos Neutralizantes/inmunología , Anemia Infecciosa Equina/prevención & control , Enfermedades de los Caballos/prevención & control , Inmunoglobulinas/inmunología , Virus de la Anemia Infecciosa Equina/inmunología , Inmunodeficiencia Combinada Grave/veterinaria , Animales , Anticuerpos Neutralizantes/administración & dosificación , Anticuerpos Antivirales/administración & dosificación , Anticuerpos Antivirales/inmunología , Anemia Infecciosa Equina/inmunología , Anemia Infecciosa Equina/virología , Enfermedades de los Caballos/inmunología , Enfermedades de los Caballos/virología , Caballos , Inmunoglobulinas/administración & dosificación , Inmunodeficiencia Combinada Grave/inmunología , Inmunodeficiencia Combinada Grave/virología
16.
Artículo en Inglés | MEDLINE | ID: mdl-22232164

RESUMEN

In order to clarify the structure and the peptide-presentation characteristics of the equine major histocompatibility complex (MHC) class I molecule, a complex of equine MHC class I molecule (ELA-A1 haplotype, 7-6 allele) with mouse ß(2)-microglobulin and the cytotoxic T lymphocyte (CTL) epitope Env-RW12 (RVEDVTNTAEYW) derived from equine infectious anaemia virus (EIAV) envelope protein (residues 195-206) was refolded and crystallized. The crystal, which belonged to space group P2(1), diffracted to 2.3 Å resolution and had unit-cell parameters a = 82.5, b = 71.4, c = 99.8 Å, ß = 102.9°. The crystal structure contained two molecules in the asymmetric unit. These results should help to determine the first equine MHC class I molecule structure presenting an EIAV CTL epitope.


Asunto(s)
Epítopos de Linfocito T/química , Productos del Gen env/química , Antígenos de Histocompatibilidad Clase I/química , Caballos/inmunología , Virus de la Anemia Infecciosa Equina/química , Linfocitos T Citotóxicos/química , Secuencia de Aminoácidos , Animales , Cristalografía por Rayos X , Epítopos de Linfocito T/inmunología , Epítopos de Linfocito T/metabolismo , Expresión Génica , Productos del Gen env/inmunología , Antígenos de Histocompatibilidad Clase I/inmunología , Antígenos de Histocompatibilidad Clase I/metabolismo , Caballos/metabolismo , Virus de la Anemia Infecciosa Equina/inmunología , Datos de Secuencia Molecular , Pliegue de Proteína , Linfocitos T Citotóxicos/inmunología , Linfocitos T Citotóxicos/metabolismo
17.
Immunopharmacol Immunotoxicol ; 34(2): 222-31, 2012 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-21851326

RESUMEN

Cross-reactive immunity occurs when infection with or vaccination against one virus protects against another related family member. A search for homologues of the HIV-1 envelope glycoprotein revealed that it is composed of thousands of intercalating and overlapping viral matches of pentapeptide or longer gapped consensi, belonging to over 70% of the currently sequenced virome, infecting all kingdoms from bacteria to man. It was also highly homologous to proteins from the Visna/Maedi and other ovine viruses, while other proteins (nef/tat/gag/pol) were homologous to proteins from the equine infectious anaemia virus and HTLV-2/HTLV-3 viruses. This phenomenon suggests that horizontal gene transfer from coinfecting RNA and DNA viruses to retroviruses is extensive, providing a route for the subsequent insertion of non-retroviral genes into human and other genomes via retroviral integration. This homology includes all viruses for which vaccines already exist. Cross-reactive immunity may be operative in AIDS, as Vaccinia vaccination decreases viral replication in HIV-1 infected patients' cells, for the CCR5 tropic form. Measles, Dengue virus, or GB virus C infections also decrease the HIV-1 viral load. A resumption of Vaccinia/smallpox vaccination might be expected to have a significant effect on the AIDS pandemic, and a careful study of the potential uses of other existing viral and bacterial vaccines merits close attention. This phenomenon may also be relevant to other recalcitrant viruses, bacteria, and parasites for which no vaccine exists and the armory of existing vaccines may have a role to play in diseases other than those for which they were designed.


Asunto(s)
Genoma Viral/inmunología , Infecciones por VIH/prevención & control , Homología de Secuencia de Aminoácido , Virus Vaccinia/genética , Vacunas Virales/genética , Virus/genética , Productos del Gen env del Virus de la Inmunodeficiencia Humana/genética , Secuencia de Aminoácidos , Virus de la Artritis-Encefalitis Caprina/genética , Virus de la Artritis-Encefalitis Caprina/inmunología , Reacciones Cruzadas/genética , Reacciones Cruzadas/inmunología , Virus del Dengue/genética , Virus del Dengue/inmunología , Epítopos de Linfocito B/genética , Epítopos de Linfocito B/inmunología , Virus GB-C/genética , Virus GB-C/inmunología , Productos del Gen env/genética , Productos del Gen env/inmunología , Productos del Gen gag/genética , Productos del Gen gag/inmunología , Genoma Viral/genética , VIH-1/genética , VIH-1/inmunología , Virus Linfotrópico T Tipo 2 Humano/genética , Virus Linfotrópico T Tipo 2 Humano/inmunología , Virus Linfotrópico T Tipo 3 Humano/genética , Virus Linfotrópico T Tipo 3 Humano/inmunología , Humanos , Virus de la Inmunodeficiencia Felina/genética , Virus de la Inmunodeficiencia Felina/inmunología , Virus de la Anemia Infecciosa Equina/genética , Virus de la Anemia Infecciosa Equina/inmunología , Lentivirus/genética , Lentivirus/inmunología , Virus del Sarampión/genética , Virus del Sarampión/inmunología , Datos de Secuencia Molecular , Virus Vaccinia/inmunología , Vacunas Virales/inmunología , Vacunas Virales/uso terapéutico , Virus/inmunología , Virus Visna-Maedi/genética , Virus Visna-Maedi/inmunología , Productos del Gen env del Virus de la Inmunodeficiencia Humana/inmunología , Productos del Gen gag del Virus de la Inmunodeficiencia Humana/genética , Productos del Gen gag del Virus de la Inmunodeficiencia Humana/inmunología , Productos del Gen nef del Virus de la Inmunodeficiencia Humana/genética , Productos del Gen nef del Virus de la Inmunodeficiencia Humana/inmunología , Productos del Gen tat del Virus de la Inmunodeficiencia Humana/genética , Productos del Gen tat del Virus de la Inmunodeficiencia Humana/inmunología
18.
J Virol ; 84(13): 6536-48, 2010 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-20392850

RESUMEN

Vaccines preventing HIV-1 infection will likely elicit antibodies that neutralize diverse strains. However, the capacity for lentiviruses to escape broadly neutralizing antibodies (NAbs) is not completely understood, nor is it known whether NAbs alone can control heterologous infection. Here, we determined that convalescent immune plasma from a horse persistently infected with equine infectious anemia virus (EIAV) neutralized homologous virus and several envelope variants containing heterologous principal neutralizing domains (PND). Plasma was infused into young horses (foals) affected with severe combined immunodeficiency (SCID), followed by challenge with a homologous EIAV stock. Treated SCID foals were protected against clinical disease, with complete prevention of infection occurring in one foal. In three SCID foals, a novel neutralization-resistant variant arose that was found to preexist at a low frequency in the challenge inoculum. In contrast, SCID foals infused with nonimmune plasma developed acute disease associated with high levels of the predominant challenge virus. Following transfer to an immunocompetent horse, the neutralization-resistant variant induced a single febrile episode and was subsequently controlled in the absence of type-specific NAb. Long-term control was associated with the presence of cytotoxic T lymphocytes (CTL). Our results demonstrate that immune plasma with neutralizing activity against heterologous PND variants can prevent lentivirus infection and clinical disease in the complete absence of T cells. Importantly, however, rare neutralization-resistant envelope variants can replicate in vivo under relatively broad selection pressure, highlighting the need for protective lentivirus vaccines to elicit NAb responses with increased breadth and potency and/or CTL that target conserved epitopes.


Asunto(s)
Anticuerpos Neutralizantes/inmunología , Anticuerpos Antivirales/inmunología , Anemia Infecciosa Equina/inmunología , Sueros Inmunes/inmunología , Virus de la Anemia Infecciosa Equina/inmunología , Animales , Caballos , Virus de la Anemia Infecciosa Equina/genética , Inmunodeficiencia Combinada Grave/complicaciones , Linfocitos T Citotóxicos/inmunología
19.
Arch Virol ; 156(8): 1455-62, 2011 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-21499906

RESUMEN

The attenuated Chinese equine infectious anemia virus (EIAV) vaccine has successfully protected millions of equine animals from EIA disease in China. In this pilot study, to determine whether this attenuated vaccine can induce broadly neutralizing antibodies, we immunized four horses with the attenuated Chinese vaccine strain EIAVFDDV and then observed the evolution of neutralizing antibodies against different EIAV strains. During the vaccination phase, all vaccinees rapidly developed high levels of neutralizing antibodies against the homologous vaccine strain (pLGFD3V), and 3 out of 4 horses showed a gradual increase in serum neutralizing activity against two relatively heterologous virulent variants of the challenge strain (pLGFD3Mu12V and DLV34). After challenge, the three horses that had developed high levels of neutralizing antibodies against pLGFD3Mu12V and DLV34 did not show signs of infection, which was demonstrated by immune suppression, while the one horse producing serum that could only neutralize pLGFD3V developed a febrile episode during the 8-month observation period. To assess whether the broadly neutralizing activity is associated with immune protection, sera drawn on the day of challenge from these four vaccinees and an additional four EIAVFDDV-vaccinated horses were analyzed for neutralizing antibodies against pLGFD3V, pLGFD3Mu12V and DLV34. Although there was no significant correlation between protection from infection and serum neutralizing activity against any of these three viral strains, protection from infection was observed to correlate better with serum neutralizing activity against the two heterologous virulent strains than against the homologous vaccine strain. These data indicate that EIAVFDDV induced broadly neutralizing antibodies, which might confer enhanced protection of vaccinees from infection by the challenge virus.


Asunto(s)
Anticuerpos Neutralizantes/sangre , Anemia Infecciosa Equina/prevención & control , Virus de la Anemia Infecciosa Equina/inmunología , Vacunas Virales/inmunología , Animales , Anemia Infecciosa Equina/sangre , Anemia Infecciosa Equina/inmunología , Caballos , Virus de la Anemia Infecciosa Equina/clasificación , Proyectos Piloto , Vacunas Atenuadas/inmunología
20.
Arch Virol ; 156(2): 353-7, 2011 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-21136127

RESUMEN

A lentiviral vaccine, live attenuated equine infectious anemia virus (EIAV) vaccine, was developed in the 1970s, and this has made tremendous contributions to the control of equine infectious anemia (EIA) in China. Four key virus strains were generated during the attenuation of the EIAV vaccine: the original Liao-Ning strain (EIAV(LN40)), a donkey-adapted virulent strain (EIAV(DV117)), a donkey-leukocyte-attenuated vaccine strain (EIAV(DLV121)), and a fetal donkey dermal cell (FDD)-adapted vaccine strain (EIAV(FDDV13)). In this study, we analyzed the proviral genomes of these four EIAV strains and found a series of consensus substitutions among these strains. These mutations provide useful information for understanding the genetic basis of EIAV attenuation. Our results suggest that multiple mutations in a variety of genes in our attenuated EIAV vaccines not only provide a basis for virulence attenuation and induction of protective immunity but also greatly reduce the risk of reversion to virulence.


Asunto(s)
Anemia Infecciosa Equina/prevención & control , Virus de la Anemia Infecciosa Equina/genética , Virus de la Anemia Infecciosa Equina/inmunología , Vacunas Virales/genética , Sustitución de Aminoácidos , Animales , Antígenos Virales/genética , China , Secuencia de Consenso , Equidae , Anemia Infecciosa Equina/inmunología , Variación Genética , Genoma Viral , Caballos , Virus de la Anemia Infecciosa Equina/patogenicidad , Especificidad de la Especie , Vacunas Atenuadas/genética , Proteínas Virales/genética , Proteínas Virales/inmunología , Virulencia/genética , Virulencia/inmunología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA