Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 90
Filtrar
1.
J Virol ; 93(21)2019 11 01.
Artigo em Inglês | MEDLINE | ID: mdl-31391270

RESUMO

Equine infectious anemia virus (EIAV) is an equine lentivirus similar to HIV-1, targets host immune cells, and causes a life-long infection in horses. The Chinese live EIAV vaccine is attenuated from long-term passaging of a highly virulent strain in vitro The parent pathogenic strain (EIAVDLV34) induces a host inflammatory storm to cause severe pathological injury of animals. However, the vaccine strain (EIAVDLV121) induces a high level of apoptosis to eliminate infected cells. To investigate how these processes are regulated, we performed a comparative proteomics analysis and functional study in equine monocyte-derived macrophages (eMDMs) and found that the divergent mitochondrial protein expression profiles caused by EIAV strains with different virulence led to disparate mitochondrial function, morphology, and metabolism. This in turn promoted the distinct transformation of macrophage inflammatory polarization and intrinsic apoptosis. In EIAVDLV34-infected cells, a high level of glycolysis and increased mitochondrial fragmentation were induced, resulting in the M1-polarized proinflammatory-type transformation of macrophages and the subsequent production of a strong inflammatory response. Following infection with EIAVDLV121, the infected cells were transformed into M2-polarized anti-inflammatory macrophages by inhibition of glycolysis. In this case, a decrease in the mitochondrial membrane potential and impairment of the electron transport chain led to increased levels of apoptosis and reactive oxygen species. These results correlated with viral pathogenicity loss and may help provide an understanding of the key mechanism of lentiviral attenuation.IMPORTANCE Following viral infection, the working pattern and function of the cell can be transformed through the impact on mitochondria. It still unknown how the mitochondrial response changes in cells infected with viruses in the process of virulence attenuation. EIAVDLV121 is the only effective lentiviral vaccine for large-scale use in the world. EIAVDLV34 is the parent pathogenic strain. Unlike EIAVDLV34-induced inflammation storms, EIAVDLV121 can induce high levels of apoptosis. For the first time, we found that, after the mitochondrial protein expression profile is altered, EIAVDLV34-infected cells are transformed into M1-polarized-type macrophages and cause inflammatory injury and that the intrinsic apoptosis pathway is activated in EIAVDLV121-infected cells. These studies shed light on how the mitochondrial protein expression profile changes between cells infected by pathogenic lentivirus strains and cells infected by attenuated lentivirus strains to drive different cellular responses, especially from inflammation to apoptosis.


Assuntos
Anemia Infecciosa Equina/patologia , Vírus da Anemia Infecciosa Equina/patogenicidade , Proteínas Mitocondriais/metabolismo , Animais , Apoptose , Células Cultivadas , Anemia Infecciosa Equina/metabolismo , Anemia Infecciosa Equina/virologia , Glicólise , Cavalos , Inflamação , Macrófagos/metabolismo , Macrófagos/patologia , Macrófagos/virologia , Mitocôndrias/metabolismo , Mitocôndrias/patologia , Proteínas Mitocondriais/genética , Proteômica , Espécies Reativas de Oxigênio , Vacinas Atenuadas , Vacinas Virais , Virulência
2.
Viruses ; 11(4)2019 04 24.
Artigo em Inglês | MEDLINE | ID: mdl-31022927

RESUMO

As the only widely used live lentiviral vaccine, the equine infectious anima virus (EIAV) attenuated vaccine was developed by in vitro passaging of a virulent strain for 121 generations. In our previous study, we observed that the attenuated vaccine was gradually selected under increased environmental pressure at the population level (termed a quasispecies). To further elucidate the potential correlation between viral quasispecies evolution and pathogenesis, a systematic study was performed by sequencing env using several methods. Some key mutations were identified within Env, and we observed that increased percentages of these mutations were accompanied by an increased passage number and attenuated virulence. Phylogenetic analysis revealed that env mutations related to the loss of virulence might have occurred evolutionarily. Among these mutations, deletion of amino acid 236 in the V4 region of Env resulted in the loss of one N-glycosylation site that was crucial for virulence. Notably, the 236-deleted sequence represented a "vaccine-specific" mutation that was also found in wild EIAVLN40 strains based on single genome amplification (SGA) analysis. Therefore, our results suggest that the EIAV attenuated vaccine may originate from a branch of quasispecies of EIAVLN40. Generally, the presented results may increase our understanding of the attenuation mechanism of the EIAV vaccine and provide more information about the evolution of other lentiviruses.


Assuntos
Evolução Molecular , Genoma Viral , Vírus da Anemia Infecciosa Equina/genética , Vírus da Anemia Infecciosa Equina/patogenicidade , Quase-Espécies , Proteínas do Envelope Viral , Animais , Anemia Infecciosa Equina/prevenção & controle , Cavalos , Mutação , Filogenia , Análise de Sequência , Deleção de Sequência , Vacinas Atenuadas , Vacinas Virais , Virulência/genética
3.
Viruses ; 10(8)2018 08 17.
Artigo em Inglês | MEDLINE | ID: mdl-30126090

RESUMO

Lentiviruses are infectious agents of a number of animal species, including sheep, goats, horses, monkeys, cows, and cats, in addition to humans. As in the human case, the host immune response fails to control the establishment of chronic persistent infection that finally leads to a specific disease development. Despite intensive research on the development of lentivirus vaccines, it is still not clear which immune responses can protect against infection. Viral mutations resulting in escape from T-cell or antibody-mediated responses are the basis of the immune failure to control the infection. The innate immune response provides the first line of defense against viral infections in an antigen-independent manner. Antiviral innate responses are conducted by dendritic cells, macrophages, and natural killer cells, often targeted by lentiviruses, and intrinsic antiviral mechanisms exerted by all cells. Intrinsic responses depend on the recognition of the viral pathogen-associated molecular patterns (PAMPs) by pathogen recognition receptors (PRRs), and the signaling cascades leading to an antiviral state by inducing the expression of antiviral proteins, including restriction factors. This review describes the latest advances on innate immunity related to the infection by animal lentiviruses, centered on small ruminant lentiviruses (SRLV), equine infectious anemia virus (EIAV), and feline (FIV) and bovine immunodeficiency viruses (BIV), specifically focusing on the antiviral role of the major restriction factors described thus far.


Assuntos
Regulação da Expressão Gênica/imunologia , Imunidade Inata , Fatores Reguladores de Interferon/imunologia , Infecções por Lentivirus/imunologia , Receptores de Reconhecimento de Padrão/imunologia , Animais , Gatos , Bovinos , Células Dendríticas/imunologia , Células Dendríticas/virologia , Cabras , Cavalos , Vírus da Imunodeficiência Bovina/imunologia , Vírus da Imunodeficiência Bovina/patogenicidade , Vírus da Imunodeficiência Felina/imunologia , Vírus da Imunodeficiência Felina/patogenicidade , Vírus da Anemia Infecciosa Equina/imunologia , Vírus da Anemia Infecciosa Equina/patogenicidade , Fatores Reguladores de Interferon/genética , Células Matadoras Naturais/imunologia , Células Matadoras Naturais/virologia , Infecções por Lentivirus/genética , Infecções por Lentivirus/virologia , Macrófagos/imunologia , Macrófagos/virologia , Moléculas com Motivos Associados a Patógenos/imunologia , Receptores de Reconhecimento de Padrão/genética , Ovinos , Linfócitos T/imunologia , Linfócitos T/virologia
4.
Transbound Emerg Dis ; 64(6): 2104-2112, 2017 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-28299895

RESUMO

Equine infectious anaemia virus (EIAV) is a lentivirus with an almost worldwide distribution that causes persistent infections in equids. Technical limitations have restricted genetic analysis of EIAV field isolates predominantly to gag sequences resulting in very little published information concerning the extent of inter-strain variation in pol, env and the three ancillary open reading frames (ORFs). Here, we describe the use of long-range PCR in conjunction with next-generation sequencing (NGS) for rapid molecular characterization of all viral ORFs and known transcription factor binding motifs within the long terminal repeat of two EIAV isolates from the 2006 Italian outbreak. These isolates were from foals believed to have been exposed to the same source material but with different clinical histories: one died 53 days post-infection (SA) while the other (DE) survived 5 months despite experiencing multiple febrile episodes. Nucleotide sequence identity between the isolates was 99.358% confirming infection with the same EIAV strain with most differences comprising single nucleotide polymorphisms in env and the second exon of rev. Although the synonymous:non-synonymous nucleotide substitution ratio was approximately 2:1 in gag and pol, the situation is reversed in env and ORF3 suggesting these sequences are subjected to host-mediated selective pressure. EIAV proviral quasispecies complexity in vivo has not been extensively investigated; however, analysis suggests it was relatively low in SA at the time of death. These results highlight advantages of NGS for molecular characterization of EIAV namely it avoids potential artefacts generated by traditional composite sequencing strategies and can provide information about viral quasispecies complexity.


Assuntos
Anemia Infecciosa Equina/virologia , Variação Genética , Sequenciamento de Nucleotídeos em Larga Escala/veterinária , Vírus da Anemia Infecciosa Equina/genética , Sequência de Aminoácidos , Animais , Biologia Computacional , Anemia Infecciosa Equina/epidemiologia , Feminino , Cavalos , Vírus da Anemia Infecciosa Equina/isolamento & purificação , Vírus da Anemia Infecciosa Equina/patogenicidade , Masculino , Mutação , Fases de Leitura Aberta/genética , Reação em Cadeia da Polimerase/veterinária , Polimorfismo de Nucleotídeo Único , Quase-Espécies , Alinhamento de Sequência/veterinária , Análise de Sequência de DNA/veterinária
5.
Arch Virol ; 161(10): 2667-72, 2016 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-27383210

RESUMO

Equine infectious anemia virus (EIAV) is a member of the genus Lentivirus of the family Retroviridae. Horses are the most susceptible equids to EIAV infection and are therefore the primary hosts of this virus. In contrast, infected donkeys do not develop clinically active equine infectious anemia (EIA). This phenomenon is similar to what has been observed with HIV-1, which fails to induce AIDS in non-human primates. Interestingly, Shen et al. developed a donkey-tropic pathogenic virus strain (EIAVDV117, DV117) by serially passaging a horse-tropic pathogenic strain, EIAVLN40 (LN40), in donkeys. LN40, which was generated by passaging a field isolate in horses, displayed enhanced virulence in horses but caused no clinical symptoms in donkeys. Infection with DV117 induced acute EIA in nearly 100 % of donkeys. Genomic analysis of DV117 revealed a significantly higher frequency of A-to-G substitutions when compared to LN40. Furthermore, detailed analysis of dinucleotide editing showed that A-to-G mutations had a preference for 5'TpA and 5'ApA. These results strongly implicated the activity of the adenosine deaminase, ADAR1, in this type of mutation. Further investigation demonstrated that overexpression of donkey ADAR1 increased A-to-G mutations within the genome of EIAV. Together with our previous finding that multiple mutations in multiple genes are generated in DV117 during its adaptation from horses to donkeys, the present study suggests that ADAR1-induced A-to-G mutations occur during virus adaption to related new hosts contributing to the alteration of EIAV host tropism.


Assuntos
Adaptação Biológica , Adenosina Desaminase/metabolismo , Vírus da Anemia Infecciosa Equina/genética , Vírus da Anemia Infecciosa Equina/patogenicidade , RNA de Cadeia Dupla/metabolismo , Animais , Equidae , Cavalos , Mutação Puntual , Análise de Sequência de DNA , Inoculações Seriadas
6.
Vet Immunol Immunopathol ; 170: 30-40, 2016 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-26832985

RESUMO

The live equine infectious anemia virus (EIAV) vaccine strain EIAVDLV121 was developed by in vitro attenuation of a virulent strain, EIAVLN40, in the 1970s, and it has been demonstrated to induce protective immunity under laboratory and natural EIAV infection conditions. The detailed biological features of this attenuated virus remain to be further investigated. Experimental inoculation with EIAVDLV121 did not result in clinical symptoms even with immunosuppressive treatment in our previous studies. Here, we further investigated whether the replication of the vaccine strain EIAVDLV121 in experimentally infected horses causes histopathological lesions to develop in the targeted organs. Both the lungs and the spleen have been demonstrated to support EIAV replication. By evaluating the gross macroscopic and histological changes, we found that EIAVDLV121 did not cause detectable histopathological lesions and that it replicated several hundred times more slowly than its parental virulent strain, EIAVLN40, in tissues. Immunochemical assays of these tissues indicated that the primary target cells of EIAVDLV121 were monocytes/macrophages, but that EIAVLN40 also infected alveolar epithelial cells and vascular endothelial cells. In addition, both of these viral strains promoted the up- and down-regulation of the expression of various cytokines and chemokines, implicating the potential involvement of these cellular factors in the pathological outcomes of EIAV infection and host immune responses. Taken together, these results demonstrate that the EIAV vaccine strain does not cause obvious histopathological lesions or clinical symptoms and that it induces a unique cytokine response profile. These features are considered essential for EIAVDLV121 to function as an effective live vaccine.


Assuntos
Anemia Infecciosa Equina/patologia , Vírus da Anemia Infecciosa Equina/patogenicidade , Vacinas Atenuadas/efeitos adversos , Vacinas Virais/efeitos adversos , Replicação Viral , Animais , Citocinas/biossíntese , Anemia Infecciosa Equina/prevenção & controle , Anemia Infecciosa Equina/virologia , Cavalos , Vírus da Anemia Infecciosa Equina/imunologia , Pulmão/patologia , Masculino , Baço/patologia , Vacinas Atenuadas/imunologia , Vacinas Virais/imunologia
7.
Retrovirology ; 13: 9, 2016 Feb 03.
Artigo em Inglês | MEDLINE | ID: mdl-26842878

RESUMO

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.


Assuntos
Anemia Infecciosa Equina/prevenção & controle , Evolução Molecular , Vírus da Anemia Infecciosa Equina/imunologia , Vacinas Virais/imunologia , Animais , China , Equidae , Cavalos , Vírus da Anemia Infecciosa Equina/genética , Vírus da Anemia Infecciosa Equina/patogenicidade , Dados de Sequência Molecular , Mutação , Análise de Sequência de DNA , Inoculações Seriadas , Vacinas Atenuadas/genética , Vacinas Atenuadas/imunologia , Vacinas Atenuadas/isolamento & purificação , Vacinas Virais/genética , Vacinas Virais/isolamento & purificação
8.
Math Biosci ; 270(Pt B): 237-48, 2015 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-25865935

RESUMO

Equine infectious anemia virus (EIAV) is a lentivirus in the retrovirus family that infects horses and ponies. Two strains, referred to as the sensitive strain and the resistant strain, have been isolated from an experimentally-infected pony. The sensitive strain is vulnerable to neutralization by antibodies whereas the resistant strain is neutralization-insensitive. The sensitive strain mutates to the resistant strain. EIAV may infect healthy target cells via free virus or alternatively, directly from an infected target cell through cell-to-cell transfer. The proportion of transmission from free-virus or from cell-to-cell transmission is unknown. A system of ordinary differential equations (ODEs) is formulated for the virus-cell dynamics of EIAV. In addition, a Markov chain model and a branching process approximation near the infection-free equilibrium (IFE) are formulated. The basic reproduction number R0 is defined as the maximum of two reproduction numbers, R0s and R0r, one for the sensitive strain and one for the resistant strain. The IFE is shown to be globally asymptotically stable for the ODE model in a special case when the basic reproduction number is less than one. In addition, two endemic equilibria exist, a coexistence equilibrium and a resistant strain equilibrium. It is shown that if R0>1, the infection persists with at least one of the two strains. However, for small infectious doses, the sensitive strain and the resistant strain may not persist in the Markov chain model. Parameter values applicable to EIAV are used to illustrate the dynamics of the ODE and the Markov chain models. The examples highlight the importance of the proportion of cell-to-cell versus free-virus transmission that either leads to infection clearance or to infection persistence with either coexistence of both strains or to dominance by the resistant strain.


Assuntos
Anemia Infecciosa Equina/transmissão , Vírus da Anemia Infecciosa Equina/patogenicidade , Modelos Teóricos , Animais , Cavalos
9.
Proteomics ; 15(11): 1843-58, 2015 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-25684102

RESUMO

Similar to the well-studied viruses human immunodeficiency virus (HIV)-1 and simian immunodeficiency virus (SIV), equine infectious anemia virus (EIAV) is another member of the Lentivirus genus in the family Retroviridae. Previous studies revealed that interactions between EIAV and the host resulted in viral evolution in pathogenicity and immunogenicity, as well as adaptation to the host. Proteomic analysis has been performed to examine changes in protein expression and/or modification in host cells infected with viruses and has revealed useful information for virus-host interactions. In this study, altered protein expression in equine monocyte-derived macrophages (eMDMs, the principle target cell of EIAV in vivo) infected with the EIAV pathogenic strain EIAV(DLV34) (DLV34) was examined using 2D-LC-MS/MS coupled with the iTRAQ labeling technique. The expression levels of 210 cellular proteins were identified to be significantly upregulated or downregulated by infection with DLV34. Alterations in protein expression were confirmed by examining the mRNA levels of eight selected proteins using quantitative real-time reverse-transcription PCR, and by verifying the levels of ten selected proteins using parallel reaction monitoring (PRM). Further analysis of GO and Kyoto Encyclopedia of Genes and Genomes (KEGG)-Pathway enrichment demonstrated that these differentially expressed proteins are primarily related to the biological processes of oxidative phosphorylation, protein folding, RNA splicing, and ubiquitylation. Our results can facilitate a better understanding of the host response to EIAV infection and the cellular processes required for EIAV replication and pathogenesis.


Assuntos
Interações Hospedeiro-Patógeno , Vírus da Anemia Infecciosa Equina/patogenicidade , Macrófagos/metabolismo , Macrófagos/virologia , Proteoma/análise , Sequência de Aminoácidos , Animais , Células Cultivadas , Anemia Infecciosa Equina/metabolismo , Ontologia Genética , Cavalos , Dados de Sequência Molecular , Proteoma/genética , Proteoma/metabolismo , Reprodutibilidade dos Testes , Reação em Cadeia da Polimerase Via Transcriptase Reversa/métodos , Espectrometria de Massas em Tandem , Replicação Viral
10.
Protein Sci ; 24(5): 633-42, 2015 May.
Artigo em Inglês | MEDLINE | ID: mdl-25559821

RESUMO

Equine lentivirus receptor 1 (ELR1) has been identified as a functional cellular receptor for equine infectious anemia virus (EIAV). Herein, recombinant ELR1 and EIAV surface glycoprotein gp90 were respectively expressed in Drosophila melanogaster S2 cells, and purified to homogeneity by Ni-NTA affinity chromatography and gel filtration chromatography. Gel filtration chromatography and analytical ultracentrifugation (AUC) analyses indicated that both ELR1 and gp90 existed as individual monomers in solution and formed a complex with a stoichiometry of 1:1 when mixed. The structure of ELR1 was first determined with the molecular replacement method, which belongs to the space group P42 21 2 with one molecule in an asymmetric unit. It contains eight antiparallel ß-sheets, of which four are in cysteine rich domain 1 (CRD1) and two are in CRD2 and CRD3, respectively. Alignment of ELR1 with HVEM and CD134 indicated that Tyr61, Leu70, and Gly72 in CRD1 of ELR1 are important residues for binding to gp90. Isothermal titration calorimetry (ITC) experiments further confirmed that Leu70 and Gly72 are the critical residues.


Assuntos
Lentivirus Equinos/química , Glicoproteínas de Membrana/química , Estrutura Secundária de Proteína , Receptores Virais/química , Proteínas Recombinantes/química , Animais , Drosophila melanogaster , Anemia Infecciosa Equina/genética , Anemia Infecciosa Equina/virologia , Cavalos/virologia , Vírus da Anemia Infecciosa Equina/patogenicidade , Glicoproteínas de Membrana/biossíntese , Glicoproteínas de Membrana/genética , Complexos Multiproteicos/química , Proteínas Recombinantes/biossíntese , Proteínas Recombinantes/genética
11.
Virology ; 476: 364-371, 2015 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-25589239

RESUMO

Adenosine deaminases that act on RNA (ADARs) have been reported to be functional on various viruses. ADAR1 may exhibit antiviral or proviral activity depending on the type of virus. Human immunodeficiency virus (HIV)-1 is the most well-studied lentivirus with respect to its interaction with ADAR1, and variable results have been reported. In this study, we demonstrated that equine ADAR1 (eADAR1) was a positive regulator of equine infectious anemia virus (EIAV), another lentivirus of the Retroviridae family. First, eADAR1 significantly promoted EIAV replication, and the enhancement of viral protein expression was associated with the long terminal repeat (LTR) and Rev response element (RRE) regions. Second, the RNA binding domain 1 of eADAR1 was essential only for enhancing LTR-mediated gene expression. Third, in contrast with APOBEC proteins, which have been shown to reduce lentiviral infectivity, eADAR1 increased the EIAV infectivity. This study indicated that eADAR1 was proviral rather than antiviral for EIAV.


Assuntos
Adenosina Desaminase/metabolismo , Anemia Infecciosa Equina/enzimologia , Vírus da Anemia Infecciosa Equina/fisiologia , Vírus da Anemia Infecciosa Equina/patogenicidade , RNA de Cadeia Dupla/metabolismo , RNA Viral/metabolismo , Replicação Viral , Adenosina Desaminase/genética , Animais , Linhagem Celular , Anemia Infecciosa Equina/genética , Anemia Infecciosa Equina/virologia , Cavalos , Interações Hospedeiro-Patógeno , Vírus da Anemia Infecciosa Equina/genética , Estrutura Terciária de Proteína , RNA de Cadeia Dupla/genética , RNA Viral/química , RNA Viral/genética , Sequências Repetidas Terminais , Virulência
12.
Virology ; 443(2): 321-8, 2013 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-23763769

RESUMO

The contribution of S2 accessory gene of equine infectious anemia virus (EIAV) to the virulence of pathogenic strains was investigated in the present study by reverse mutation of all four consensus S2 mutation sites in an attenuated EIAV proviral strain, FDDV3-8, to the corresponding sequences of a highly pathogenic strain DV117. The S2 reverse-mutated recombinant strain FDDVS2r1-2-3-4 replicated with similar kinetics to FDDV3-8 in cultivated target cells. In contrast to the results of other studies of EIAV with dysfunctional S2, reverse mutation of S2 only transiently and moderately increased the plasma viral load of inoculated horses, and induction of transient immunosuppression did not boost viral pathogenicity. In addition, inoculation of FDDVS2r1-2-3-4 induced partial protection to a challenge pathogenic virus. These results suggest that the attenuated EIAV vaccine strain with multiple mutations in multiple genes will not easily revert to a virulent phenotype.


Assuntos
Anemia Infecciosa Equina/patologia , Vírus da Anemia Infecciosa Equina/patogenicidade , Mutação , Vacinas Atenuadas/genética , Proteínas Virais/genética , Vacinas Virais/genética , Sequência de Aminoácidos , Animais , Anemia Infecciosa Equina/virologia , Cavalos/virologia , Vírus da Anemia Infecciosa Equina/genética , Dados de Sequência Molecular , Vacinas Atenuadas/imunologia , Carga Viral , Vacinas Virais/imunologia , Virulência/genética , Replicação Viral
13.
PLoS One ; 8(6): e66093, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23785473

RESUMO

Lentiviral envelope (Env) antigenic variation and associated immune evasion present major obstacles to vaccine development. The concept that Env is a critical determinant for vaccine efficacy is well accepted, however defined correlates of protection associated with Env variation have yet to be determined. We reported an attenuated equine infectious anemia virus (EIAV) vaccine study that directly examined the effect of lentiviral Env sequence variation on vaccine efficacy. The study identified a significant, inverse, linear correlation between vaccine efficacy and increasing divergence of the challenge virus Env gp90 protein compared to the vaccine virus gp90. The report demonstrated approximately 100% protection of immunized ponies from disease after challenge by virus with a homologous gp90 (EV0), and roughly 40% protection against challenge by virus (EV13) with a gp90 13% divergent from the vaccine strain. In the current study we examine whether the protection observed when challenging with the EV0 strain could be conferred to animals via chimeric challenge viruses between the EV0 and EV13 strains, allowing for mapping of protection to specific Env sequences. Viruses containing the EV13 proviral backbone and selected domains of the EV0 gp90 were constructed and in vitro and in vivo infectivity examined. Vaccine efficacy studies indicated that homology between the vaccine strain gp90 and the N-terminus of the challenge strain gp90 was capable of inducing immunity that resulted in significantly lower levels of post-challenge virus and significantly delayed the onset of disease. However, a homologous N-terminal region alone inserted in the EV13 backbone could not impart the 100% protection observed with the EV0 strain. Data presented here denote the complicated and potentially contradictory relationship between in vitro virulence and in vivo pathogenicity. The study highlights the importance of structural conformation for immunogens and emphasizes the need for antibody binding, not neutralizing, assays that correlate with vaccine protection.


Assuntos
Anemia Infecciosa Equina/prevenção & controle , Vírus da Anemia Infecciosa Equina/genética , Vírus da Anemia Infecciosa Equina/imunologia , Proteínas do Envelope Viral/genética , Proteínas do Envelope Viral/imunologia , Vacinas Virais/imunologia , Animais , Linhagem Celular , Anemia Infecciosa Equina/imunologia , Anemia Infecciosa Equina/virologia , Ordem dos Genes , Genoma Viral , Cavalos , Imunidade Celular , Imunidade Humoral , Vírus da Anemia Infecciosa Equina/patogenicidade , Provírus/genética , Recombinação Genética , Carga Viral , Vacinas Virais/genética , Virulência/genética
14.
Viral Immunol ; 25(4): 324-8, 2012 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-22746986

RESUMO

Determining mechanisms of viral escape to particular epitopes recognized by virus-neutralizing antibody can facilitate characterization of host-neutralizing antibody responses as type- versus group-specific, and provides necessary information for vaccine development. Our study reveals that a single N-glycan located in the 5' region of the Wyoming wild-type equine infectious anemia virus (EIAV) principal neutralizing domain (PND) accounts for the differences in neutralization phenotype observed between PND variants, while variations in charged amino acids within the PND do not appear to play a key role in viral escape. Site-directed mutagenesis and peptide mapping of a conserved epitope to neutralizing antibody in the 3' region of the PND showed rapid selective pressure for acquisition of a 5' PND N-glycan responsible for defining the specificity of the neutralizing-antibody response.


Assuntos
Anticorpos Neutralizantes/imunologia , Especificidade de Anticorpos/imunologia , Epitopos/imunologia , Anemia Infecciosa Equina/imunologia , Evasão da Resposta Imune/imunologia , Vírus da Anemia Infecciosa Equina/imunologia , Sequência de Aminoácidos , Animais , Anticorpos Neutralizantes/biossíntese , Mapeamento de Epitopos , Anemia Infecciosa Equina/virologia , Cavalos , Vírus da Anemia Infecciosa Equina/genética , Vírus da Anemia Infecciosa Equina/patogenicidade , Dados de Sequência Molecular , Mutagênese Sítio-Dirigida , Testes de Neutralização
15.
J Virol ; 85(24): 13322-32, 2011 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-21957284

RESUMO

Human immunodeficiency virus type 1 (HIV-1) Gag is the main structural protein driving assembly and release of virions from infected cells. Gag alone is capable of self-assembly in vitro, but host factors have been shown to play a role in efficient viral replication and particle morphogenesis within the living cell. In a series of affinity purification experiments, we identified the cellular protein Lyric to be an HIV-1 Gag-interacting protein. Lyric was previously described to be an HIV-inducible gene and is involved in various signaling pathways. Gag interacts with endogenous Lyric via its matrix (MA) and nucleocapsid (NC) domains. This interaction requires Gag multimerization and Lyric amino acids 101 to 289. Endogenous Lyric is incorporated into HIV-1 virions and is cleaved by the viral protease. Gag-Lyric interaction was also observed for murine leukemia virus and equine infectious anemia virus, suggesting that it represents a conserved feature among retroviruses. Expression of the Gag binding domain of Lyric increased Gag expression levels and viral infectivity, whereas expression of a Lyric mutant lacking the Gag binding site resulted in lower Gag expression and decreased viral infectivity. The results of the current study identify Lyric to be a cellular interaction partner of HIV-1 Gag and hint at a potential role in regulating infectivity. Further experiments are needed to elucidate the precise role of this interaction.


Assuntos
Moléculas de Adesão Celular/metabolismo , HIV-1/patogenicidade , Interações Hospedeiro-Patógeno , Mapeamento de Interação de Proteínas , Produtos do Gene gag do Vírus da Imunodeficiência Humana/metabolismo , Humanos , Vírus da Anemia Infecciosa Equina/patogenicidade , Vírus da Leucemia Murina/patogenicidade , Proteínas de Membrana , Ligação Proteica , Proteínas de Ligação a RNA
16.
Virus Res ; 160(1-2): 274-82, 2011 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-21782860

RESUMO

The attenuated equine infectious anemia virus (EIAV) vaccine was the first attenuated lentivirus vaccine to be used in a large-scale application and has been used to successfully control the spread of equine infectious anemia (EIA) in China. To better understand the potential role of cytokines in the pathogenesis of EIAV infection and resulting immune response, we used branched DNA technology to compare the mRNA expression levels of 12 cytokines and chemokines, including IL-1α, IL-1ß, IL-4, IL-10, TNF-α, IFN-γ, IP-10, IL-8, MIP-1α, MIP-1ß, MCP-1, and MCP-2, in equine monocyte-derived macrophages (eMDMs) infected with the EIAV(DLV121) vaccine strain or the parental EIAV(DLV34) pathogenic strain. Infection with EIAV(DLV34) and EIAV(DLV121) both caused changes in the mRNA levels of various cytokines and chemokines in eMDMs. In the early stage of infection with EIAV(DLV34) (0-24h), the expression of the pro-inflammatory cytokines TNF-α and IL-1ß were significantly up-regulated, while with EIAV(DLV121), expression of the anti-inflammatory cytokine IL-4 was markedly up-regulated. The effects on the expression of other cytokines and chemokines were similar between these two strains of virus. During the first 4 days after infection, the expression level of IL-4 in cells infected with the pathogenic strain were significantly higher than that in cells infected with the vaccine strain, but the expression of IL-1α and IL-1ß induced by the vaccine strain was significantly higher than that observed with the pathogenic strain. In addition, after 4 days of infection with the pathogenic strain, the expression levels of 5 chemokines, but not IP-10, were markedly increased in eMDMs. In contrast, the vaccine strain did not up-regulate these chemokines to this level. Contrary to our expectation, induced apoptosis in eMDMs infected with the vaccine strain was significantly higher than that infected with the pathogenic strain 4 days and 6 days after infection. Together, these results contribute to a greater understanding of the pathogenesis of EIAV and of the mechanisms by which the immune response is induced after EIAV infection.


Assuntos
Apoptose , Citocinas/biossíntese , Vírus da Anemia Infecciosa Equina/imunologia , Vírus da Anemia Infecciosa Equina/patogenicidade , Macrófagos/imunologia , Macrófagos/virologia , Vacinas Virais/imunologia , Animais , China , Citocinas/genética , Perfilação da Expressão Gênica , Cavalos , Vacinas Atenuadas/imunologia
17.
J Virol ; 85(19): 10421-4, 2011 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-21752904

RESUMO

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.


Assuntos
Anticorpos Neutralizantes/imunologia , Vírus da Anemia Infecciosa Equina/crescimento & desenvolvimento , Vírus da Anemia Infecciosa Equina/imunologia , Mutação , Animais , Anticorpos Antivirais/imunologia , Linhagem Celular , Vírus da Anemia Infecciosa Equina/genética , Vírus da Anemia Infecciosa Equina/patogenicidade , Testes de Neutralização , Virulência
18.
Virus Res ; 158(1-2): 235-45, 2011 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-21539871

RESUMO

Preliminary studies revealed that the gene of the gp45 transmembrane protein (TM) of the attenuated equine infectious anemia virus (EIAV) vaccine strain EIAV(FDDV13) had a high frequency of a premature stop codon at position 261W, which generated a 154-residue truncation at the C-terminus. EIAV(FDDV-TM36), a recombinant virus with the TM truncated at the intracytoplasmic (CT) domain due to the presence of a stop codon, was constructed based on EIAV(FDDV)3-8, which is a proviral derivative of the vaccine. EIAV(FDDV-TM36) had a significantly reduced replication capability compared to EIAV(FDDV)3-8 in equine or donkey monocyte-derived macrophages and a decreased ability to induce apoptosis. However, both viruses raised a similar plasma viral load in inoculated horses and did not induce clinical symptoms of EIA. To further compare the in vivo behavior between EIAV(FDDV-TM36) and EIAV(FDDV)3-8, inoculated horses were transiently immunosuppressed with dexamethasone. While three of the four horses inoculated with EIAV(FDDV)3-8 demonstrated significant increases in viral loads after the drug treatment, none of the four horses inoculated with EIAV(FDDV-TM36) showed a statistically increased plasma viral load. Significantly increased neutralizing antibody levels were also observed in the group of horses inoculated with EIAV(FDDV)3-8, but not EIAV(FDDV-TM36), after immunosuppression. Our results indicate that although the CT truncation of TM decreased viral replication in cultivated equine and donkey macrophages, the primary target cell of EIAV, and did not influence the plasma viral load of inoculated hosts, it weakened the potential pathogenicity of the vaccine. The host immunity is presumably responsible for the equal in vivo replication levels of viruses with either the CT-truncated or prototype TM.


Assuntos
Vírus da Anemia Infecciosa Equina/patogenicidade , Deleção de Sequência , Proteínas do Envelope Viral/genética , Vacinas Virais/efeitos adversos , Replicação Viral , Animais , Anticorpos Neutralizantes/sangue , Anticorpos Antivirais/sangue , Códon sem Sentido , Equidae , Cavalos , Vírus da Anemia Infecciosa Equina/genética , Monócitos/virologia , Vacinas Atenuadas/efeitos adversos , Vacinas Atenuadas/genética , Vacinas Atenuadas/imunologia , Vacinas Virais/genética , Vacinas Virais/imunologia , Virulência
19.
Virus Genes ; 42(2): 220-8, 2011 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-21369830

RESUMO

The Chinese equine infectious anemia virus (EIAV) virulent strain EIAV(LN40) is derived from a naturally occurring virus by continuously passing in horses for 16 generations. Its genome sequence is 23% different from that of the American strains or the Japanese strains, and the variation of envelope gp90 surface unit (SU) is as high as 41%. In this study, evolutions of the EIAV(LN40) gp90 gene in four infected horses were analyzed. Results showed that new quasispecies arose in the early stage of infection in all EIAV(LN40)-infected horses. These quasispecies belonged to branches different from EIAV(LN40) in a phylogenetic tree. In contrast, the gp90 sequences of viruses isolated after disease onset remained in the same phylogenetic branch as EIAV(LN40), with some having exactly the same sequences. The glycosylation sites 191NSSN and 237NNTW in the V3 and V4 region present or absent simultaneously in most of the predicted amino acid sequences. Changes in the glycosylation sites within V3, V4, and V5 regions are usually associated with the disease status. Glycosylation sites (191NSSN, 237NNTW, and 280NDTS) within these three regions were present in EIAV(LN40) and most of the quasispecies isolated after, but not before disease onset. These unique evolutionary characteristics of SU have not been reported for EIAV and other lentiviruses. Our results provide a reference for a further understanding of the mechanism underlying the persistent infection and escape from immune surveillance of EIAV.


Assuntos
Evolução Molecular , Glicoproteínas/genética , Cavalos/virologia , Vírus da Anemia Infecciosa Equina/genética , Proteínas do Envelope Viral/genética , Sequência de Aminoácidos , Substituição de Aminoácidos , Animais , Glicoproteínas/isolamento & purificação , Glicosilação , Vírus da Anemia Infecciosa Equina/patogenicidade , Dados de Sequência Molecular , Filogenia , RNA Viral/genética , Análise de Sequência de RNA , Deleção de Sequência , Proteínas do Envelope Viral/isolamento & purificação
20.
Virology ; 410(1): 96-106, 2011 Feb 05.
Artigo em Inglês | MEDLINE | ID: mdl-21094511

RESUMO

To investigate essential factors that determine the efficacy of vaccines against lentiviruses, an effective attenuated equine infectious anemia virus (EIAV) vaccine strain and a proviral derivative of the vaccine were compared with respect to differences in inducing protective immunity. Although these two strains replicated equally well in vitro and in vivo, the proviral strain induced significantly less protection from disease and infection caused by viral challenge and significantly lower specific neutralizing capability. These findings indicated that the proviral strain had lost the ability to stimulate immune protection compared to the parental vaccine strain. A further analysis of the envelope gp90 gene variation revealed that compared to the proviral strain, the vaccine strain displayed a wide sequence diversity in immunogen composition. Thus, we inferred that the differences in immunogen composition might be the major cause for the failure of the proviral derivative to elicit the immune protection induced by the parental strain.


Assuntos
Anemia Infecciosa Equina/prevenção & controle , Vírus da Anemia Infecciosa Equina/genética , Vírus da Anemia Infecciosa Equina/imunologia , Provírus/imunologia , Vacinas Virais , Animais , Anticorpos Antivirais , Clonagem Molecular , Anemia Infecciosa Equina/virologia , Feminino , Cavalos , Vírus da Anemia Infecciosa Equina/patogenicidade , Masculino , Vacinas Atenuadas , Carga Viral , Virulência
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA