Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
PLoS Pathog ; 20(2): e1011928, 2024 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-38324558

RESUMO

The subgroup J avian leukosis virus (ALV-J), a retrovirus, uses its gp85 protein to bind to the receptor, the chicken sodium hydrogen exchanger isoform 1 (chNHE1), facilitating viral invasion. ALV-J is the main epidemic subgroup and shows noteworthy mutations within the receptor-binding domain (RBD) region of gp85, especially in ALV-J layer strains in China. However, the implications of these mutations on viral replication and transmission remain elusive. In this study, the ALV-J layer strain JL08CH3-1 exhibited a more robust replication ability than the prototype strain HPRS103, which is related to variations in the gp85 protein. Notably, the gp85 of JL08CH3-1 demonstrated a heightened binding capacity to chNHE1 compared to HPRS103-gp85 binding. Furthermore, we showed that the specific N123I mutation within gp85 contributed to the enhanced binding capacity of the gp85 protein to chNHE1. Structural analysis indicated that the N123I mutation primarily enhanced the stability of gp85, expanded the interaction interface, and increased the number of hydrogen bonds at the interaction interface to increase the binding capacity between gp85 and chNHE1. We found that the N123I mutation not only improved the viral replication ability of ALV-J but also promoted viral shedding in vivo. These comprehensive data underscore the notion that the N123I mutation increases receptor binding and intensifies viral replication.


Assuntos
Vírus da Leucose Aviária , Leucose Aviária , Doenças das Aves Domésticas , Animais , Vírus da Leucose Aviária/genética , Vírus da Leucose Aviária/química , Mutação , Galinhas , Isoformas de Proteínas/genética , Proteínas do Envelope Viral/genética
2.
Infect Genet Evol ; 89: 104721, 2021 04.
Artigo em Inglês | MEDLINE | ID: mdl-33444858

RESUMO

Since 2005, subgroup J avian leukosis virus (ALV-J) infection has been present in yellow chickens in Guangdong, China, causing severe economic losses to the local poultry industry. ALV-J is a rapidly evolving retrovirus. To investigate the molecular characteristics of ALV-J isolates from yellow breeder chickens in Guangdong, 17 virus strains were isolated from 6549 anticoagulants from clinically healthy birds between 2016 and 2019, and completely sequenced and phylogenetically analyzed. Phylogenetic analysis of the gp85 gene showed that all isolated viruses were divided into three different branches. Notably, 41.2% (7/17) of the isolates shared a novel G2598A nucleotide mutation in the pol gene and caused the stop codon to be advanced by 8 positions. Nearly 200 nucleotides were deleted from the redundant TM (rTM) region in all strains, but all retained an intact direct repeat (DR1). 82.4% (14/17) of isolates contained a complete E element. Additionally, 29.4% (5/17) of isolates detected an 11 bp deletion in U3 region, and the AIB REP1 transcription factor is missing. The study indicated that ALV-J infection had still been prevalent in the yellow breeder chicken farms in Guangdong, and the genetic background of the strains is diverse. This study provides the latest data on the molecular characteristics of ALV-J, which will help to reveal the evolution trend of ALV-J and develop relevant prevention and control measures.


Assuntos
Vírus da Leucose Aviária/genética , Galinhas/genética , Galinhas/virologia , Regiões 3' não Traduzidas , Sequência de Aminoácidos , Animais , Vírus da Leucose Aviária/química , Vírus da Leucose Aviária/classificação , China , DNA Viral/genética , Genes Virais , Variação Genética , Filogenia , Doenças das Aves Domésticas/prevenção & controle , Doenças das Aves Domésticas/virologia , Fatores de Transcrição/metabolismo , Sequenciamento Completo do Genoma
3.
J Virol ; 92(1)2018 01 01.
Artigo em Inglês | MEDLINE | ID: mdl-29070685

RESUMO

Chicken Na+/H+ exchanger type I (chNHE1), a multispan transmembrane protein, is a cellular receptor of the subgroup J avian leukosis virus (ALV-J). To identify the functional determinants of chNHE1 responsible for the ALV-J receptor activity, a series of chimeric receptors was created by exchanging the extracellular loops (ECL) of human NHE1 (huNHE1) and chNHE1 and by ECL replacement with a hemagglutinin (HA) tag. These chimeric receptors then were used in binding and entry assays to map the minimal ALV-J gp85-binding domain of chNHE1. We show that ECL1 of chNHE1 (chECL1) is the critical functional ECL that interacts directly with ALV-J gp85; ECL3 is also involved in ALV-J gp85 binding. Amino acid residues 28 to 39 of the N-terminal membrane-proximal region of chECL1 constitute the minimal domain required for chNHE1 binding of ALV-J gp85. These residues are sufficient to mediate viral entry into ALV-J nonpermissive cells. Point mutation analysis revealed that A30, V33, W38, and E39 of chECL1 are the key residues mediating the binding between chNHE1 and ALV-J gp85. Further, the replacement of residues 28 to 39 of huNHE1 with the corresponding chNHE1 residues converted the nonfunctional ALV-J receptor huNHE1 to a functional one. Importantly, soluble chECL1 and huECL1 harboring chNHE1 residues 28 to 39 both could effectively block ALV-J infection. Collectively, our findings indicate that residues 28 to 39 of chNHE1 constitute a domain that is critical for receptor function and mediate ALV-J entry.IMPORTANCE chNHE1 is a cellular receptor of ALV-J, a retrovirus that causes infections in chickens and serious economic losses in the poultry industry. Until now, the domains determining the chNHE1 receptor function remained unknown. We demonstrate that chECL1 is critical for receptor function, with residues 28 to 39 constituting the minimal functional domain responsible for chNHE1 binding of ALV-J gp85 and efficiently mediating ALV-J cell entry. These residues are located in the membrane-proximal region of the N terminus of chECL1, suggesting that the binding site of ALV-J gp85 on chNHE1 is probably located on the apex of the molecule; the receptor-binding mode might be different from that of retroviruses. We also found that soluble chECL1, as well as huECL1 harboring chNHE1 residues 28 to 39, effectively blocked ALV-J infection. These findings contribute to a better understanding of the ALV-J infection mechanism and also provide new insights into the control strategies for ALV-J infection.


Assuntos
Aminoácidos/química , Vírus da Leucose Aviária/metabolismo , Receptores Virais/metabolismo , Trocadores de Sódio-Hidrogênio/química , Trocadores de Sódio-Hidrogênio/metabolismo , Ligação Viral , Internalização do Vírus , Aminoácidos/metabolismo , Animais , Leucose Aviária/virologia , Vírus da Leucose Aviária/química , Vírus da Leucose Aviária/genética , Galinhas , Humanos , Mutação Puntual , Receptores Virais/genética , Trocadores de Sódio-Hidrogênio/genética
4.
Vaccine ; 35(48 Pt B): 6685-6690, 2017 12 04.
Artigo em Inglês | MEDLINE | ID: mdl-29054728

RESUMO

The hypervariable antigenicity and immunosuppressive features of avian leukosis virus subgroup J (ALV-J) has led to great challenges to develop effective vaccines. Epitope vaccine will be a perspective trend. Previously, we identified a variant antigenic neutralizing epitope in hypervariable region 1 (hr1) of ALV-J, N-LRDFIA/E/TKWKS/GDDL/HLIRPYVNQS-C. BLAST analysis showed that the mutation of A, E, T and H in this epitope cover 79% of all ALV-J strains. Base on this data, we designed a multi-variant epitope ensemble vaccine comprising the four mutation variants linked with glycine and serine. The recombinant multi-variant epitope gene was expressed in Escherichia coli BL21. The expressed protein of the variant multi-variant epitope gene can react with positive sera and monoclonal antibodies of ALV-J, while cannot react with ALV-J negative sera. The multi-variant epitope vaccine that conjugated Freund's adjuvant complete/incomplete showed high immunogenicity that reached the titer of 1:64,000 at 42 days post immunization and maintained the immune period for at least 126 days in SPF chickens. Further, we demonstrated that the antibody induced by the variant multi-variant ensemble epitope vaccine recognized and neutralized different ALV-J strains (NX0101, TA1, WS1, BZ1224 and BZ4). Protection experiment that was evaluated by clinical symptom, viral shedding, weight gain, gross and histopathology showed 100% chickens that inoculated the multi-epitope vaccine were well protected against ALV-J challenge. The result shows a promising multi-variant epitope ensemble vaccine against hypervariable viruses in animals.


Assuntos
Vírus da Leucose Aviária/imunologia , Leucose Aviária/prevenção & controle , Epitopos/genética , Epitopos/imunologia , Vacinas Virais/imunologia , Animais , Anticorpos Monoclonais/imunologia , Anticorpos Antivirais/sangue , Variação Antigênica/genética , Variação Antigênica/imunologia , Leucose Aviária/imunologia , Vírus da Leucose Aviária/química , Vírus da Leucose Aviária/genética , Galinhas/imunologia , Epitopos/química , Adjuvante de Freund , Imunogenicidade da Vacina , Mutação , Testes de Neutralização , Doenças das Aves Domésticas/prevenção & controle , Proteínas do Envelope Viral/imunologia , Vacinas Virais/administração & dosagem
5.
Arch Virol ; 160(4): 995-1004, 2015 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-25655260

RESUMO

Avian leukosis virus subgroup J (ALV-J) is an avian oncogenic retrovirus that has caused severe economic losses in China. Gp85 protein is the main envelope protein and the most variable structural protein of ALV-J. It is also involved in virus neutralization. In this study, a specific monoclonal antibody, 4A3, was produced against the ALV-J gp85 protein. Immunofluorescence assays showed that 4A3 could react with different strains of ALV-J, including the British prototype isolate HPRS103, the American strains, an early Chinese broiler isolate, and layer isolates. A linear epitope on the gp85 protein was identified using a series of partially overlapping fragments spanning the gp85-encoding gene and subjecting them to western blot analysis. The results indicated that (134)AEAELRDFI(142) was the minimal linear epitope that could be recognized by mAb 4A3. Enzyme-linked immunosorbent assay (ELISA) revealed that chicken anti-ALV-J sera and mouse anti-ALV-J gp85 sera could also recognize the minimal linear epitope. Alignment analysis of amino acid sequences indicated that the epitope was highly conserved among 34 ALV-J strains. Furthermore, the epitope was not conserved among subgroup A and B of avian leukosis virus (ALV). Taken together, the mAb and the identified epitope may provide valuable tools for the development of new diagnostic methods for ALV-J.


Assuntos
Vírus da Leucose Aviária/imunologia , Leucose Aviária/virologia , Epitopos de Linfócito B/química , Epitopos de Linfócito B/imunologia , Doenças das Aves Domésticas/virologia , Proteínas do Envelope Viral/química , Proteínas do Envelope Viral/imunologia , Sequência de Aminoácidos , Animais , Anticorpos Monoclonais/imunologia , Anticorpos Antivirais/imunologia , Leucose Aviária/imunologia , Vírus da Leucose Aviária/química , Vírus da Leucose Aviária/genética , Vírus da Leucose Aviária/isolamento & purificação , Galinhas , Mapeamento de Epitopos , Epitopos de Linfócito B/genética , Feminino , Camundongos , Camundongos Endogâmicos BALB C , Dados de Sequência Molecular , Doenças das Aves Domésticas/imunologia , Especificidade da Espécie , Proteínas do Envelope Viral/genética
6.
Biosens Bioelectron ; 45: 40-5, 2013 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-23454341

RESUMO

A novel sandwich-type electrochemical immunosensor was fabricated for ultrasensitive detection of subgroup J of avian leukosis virus (ALVs-J) by employing ß-cyclodextrin-ferrocene (CD-Fc) host-guest complex multifunctional Fe3O4 nanospheres as labels and ß-cyclodextrin functional graphene sheets (CD-GS) nanocomposite as sensor platform. The sensitivity was greatly improved based on the triple amplification strategy. Firstly, the CD-GS improved the electron transfer rate as well as increasing the surface area to capture a large amount of primary antibodies (Ab1). Secondly, the CD on the Fe3O4 surface with strong recognition capability could form stable CD-Fc host-guest inclusion complex and provided larger free room for the conjugation of secondary antibodies (Ab2) and glucose oxidase (GOD). Finally, the conjugated GOD exhibited extraordinary electrochemical biocatalysis towards the reduction reaction of Fc(+) by glucose. Under the optimized conditions, the electrochemical immunosensor exhibited a wide working range from 10(2.27)-10(3.50) TCID50/mL (TCID50: 50% tissue culture infective dose) with a low detection limit of 10(2.19) TCID50/mL (S/N=3). The selectivity, reproducibility, and stability are acceptable. The assay was evaluated for real avian serum sample, receiving satisfactory results. This new type of triple amplification strategy may provide potential applications for the clinic application.


Assuntos
Vírus da Leucose Aviária/isolamento & purificação , Técnicas Biossensoriais/métodos , Grafite/química , Proteínas Virais/isolamento & purificação , Animais , Anticorpos/química , Anticorpos/imunologia , Vírus da Leucose Aviária/química , Aves/sangue , Aves/virologia , Compostos Ferrosos/química , Imunoensaio/métodos , Limite de Detecção , Metalocenos , Nanosferas/química , Proteínas Virais/sangue , Proteínas Virais/imunologia
7.
Virology ; 421(1): 42-50, 2011 Dec 05.
Artigo em Inglês | MEDLINE | ID: mdl-21982594

RESUMO

Integrase (IN) is the enzyme responsible for the integration of the retroviral genome into the host cell DNA. Herein, three mutants of conserved residues (V79, S85 and I146) of the central core domain (CCD) of an Avian Sarcoma/Leukemia Virus IN were analyzed in vitro. Our data revealed (i) the inability of S85T mutant to form dimers and tetramers in the absence of DNA and (ii) a slightly reduced ability of V79A IN in tetramers formation. Surprisingly, both mutants were still able to efficiently achieve concerted DNA integration. This could be explained by the ability of the two mutants to form complexes in the presence of DNA. These data suggest a strong structural role of the region encompassing V79 and S85 residues (ß2/ß3 turn-ß3 strands) following binding to viral DNA and highlight the dynamic nature of IN.


Assuntos
Vírus da Leucose Aviária/enzimologia , Integrases/química , Integrases/metabolismo , Mutação , Proteínas Virais/química , Proteínas Virais/metabolismo , Sequência de Aminoácidos , Vírus da Leucose Aviária/química , Vírus da Leucose Aviária/genética , Vírus da Leucose Aviária/fisiologia , Dimerização , Integrases/genética , Modelos Moleculares , Dados de Sequência Molecular , Multimerização Proteica , Estrutura Terciária de Proteína , Alinhamento de Sequência , Proteínas Virais/genética , Integração Viral
8.
Wei Sheng Wu Xue Bao ; 51(12): 1663-8, 2011 Dec.
Artigo em Chinês | MEDLINE | ID: mdl-22379808

RESUMO

OBJECTIVE: It was reported that subgroup J avian leukosis virus strain NX0101 activates PI3K/Akt pathway during early infection in DF-1 cells. Whether there is YXXM motif in the amino acid sequence of NX0101 and the function of YXXM motif were studied. METHODS: The presence of internal transmembrane domains in the envelope protein of NX0101 was analyzed by Tmpred. Point mutation was introduced to change the YXXM motif in the NX0101 strain to FXXA. The plasmid containing the full genome of NX0101 with mutation within the YXXM motif in pMD18-T vector was constructed and transfected into DF-1 cells. Viral replication levels of NX0101 strain and the mutation one were tested and compared by real-time PCR and ELISA. RESULTS: The amino acid sequence of NX0101 strain had one YXXM motif (amino acids 554-557) in the cytoplasmic tail of envelope protein. The mutated NX0101 strain (Y554F, M557A) was rescued by reverse genetics technique. Viral replication of the mutated NX0101 strain was significantly lower than that of NX0101 strain in the level of either RNA or protein synthesis. CONCLUSION: The results revealed that the (Y)XXM motif was important for virus NX0101 replication in DF-1 cells.


Assuntos
Vírus da Leucose Aviária/fisiologia , Leucose Aviária/virologia , Doenças das Aves Domésticas/virologia , Proteínas do Envelope Viral/química , Proteínas do Envelope Viral/metabolismo , Replicação Viral , Motivos de Aminoácidos , Sequência de Aminoácidos , Animais , Vírus da Leucose Aviária/química , Vírus da Leucose Aviária/genética , Sequência de Bases , Galinhas , Dados de Sequência Molecular , Proteínas do Envelope Viral/genética
9.
Bing Du Xue Bao ; 26(1): 53-7, 2010 Jan.
Artigo em Chinês | MEDLINE | ID: mdl-20329559

RESUMO

By inoculation of blood samples in DF-1 (C/E) cell culture, an exogenous avian leukosis virus (ALV) strain SDAU09C2 was isolated from a breeder farm of Chinese native breed "Luhua" in Shandong province. Comparisons of the amino acid sequence of env gene gp85 from the isolate with those from other ALV reference strains of different subgroups indicated that SDAU09C2 had the highest gp85 identity to two reference strains of subgroup B of 92.5%. Its gp85 identity to other chicken ALV subgroups A, C, D, E was in the range of 73.2%-87.9%. The identity to subgroup J was only 30.3%-32.4%. This is the first report on isolation and identification of ALV-B and its gp85 from Chinese native breed chickens.


Assuntos
Vírus da Leucose Aviária/isolamento & purificação , Leucose Aviária/virologia , Galinhas , Doenças das Aves Domésticas/virologia , Sequência de Aminoácidos , Animais , Vírus da Leucose Aviária/química , Vírus da Leucose Aviária/classificação , Vírus da Leucose Aviária/genética , Cruzamento , Feminino , Dados de Sequência Molecular , Filogenia , Proteínas do Envelope Viral/química , Proteínas do Envelope Viral/genética
10.
Mol Biol (Mosk) ; 43(6): 1077-81, 2009.
Artigo em Russo | MEDLINE | ID: mdl-20088385

RESUMO

Fluorescent properties of 2-aminopurine included in the sequence RNA, are now used for the study of structural dynamics and local changes of structure retroviral RNA. We used 2-aminopurine for studying the conformational states of unpaired loop adenine of avian leukosis virus RNA upon the interaction with the aminoglycoside antibiotics. It was shown, that intensity of 2-aminopurine fluorescence for monomer hairpin RNA has greater value in comparison with both RNA dimers. Comparing the 2-aminopurine fluorescence of RNA dimers, it has found out that the intensity of fluorescence for extended dimer significantly lower than the kissing loop dimer RNA. This can be explained by the fact that the stacking contacts forms more compact structure of a loop in extended dimer concerning those in structure of kissing loop dimer. When the binding of aminogycoside antibiotics with kissing loop dimer RNA it was observed that only the tobramycin increases nearly three times the intensity of 2-aminopurine fluorescence. Results of work testify that it is possible to detect local changes in the complexes of retroviral RNA with ligands, using the fluorescence 2-aminopurine.


Assuntos
Aminoglicosídeos/química , Antibacterianos/química , Vírus da Leucose Aviária/química , Conformação de Ácido Nucleico , RNA Viral/química , Aminopirina/química , Vírus da Leucose Aviária/genética , Vírus da Leucose Aviária/metabolismo , RNA Viral/genética , RNA Viral/metabolismo
11.
Mol Biol (Mosk) ; 42(1): 131-7, 2008.
Artigo em Russo | MEDLINE | ID: mdl-18389630

RESUMO

The dimerization of genomic retroviral RNA is well studied for several groups of viruses, the dimerization of human immunodeficiency (HIV) RNA being investigated in more detail. Regions of dimerization apparently involve the short sequences RNA which are directly responsible for the formation of two type dimers: kissing loop-loop (KD) and linear (LD). The 5'-end sequences from RNA avian viruses, where the dimers are basically formed, considerably differ from those of HIV. However, as it was described earlier, the mechanism of dimerization of RNA from human immunodeficiency and from avian leukosis viruses are identical. The fluorescence of adenine analogue 2-aminopurine (2-AP) incorporated into loop sequence of short fragments RNA ALV was used for analysis of dimers formation. Using the temperature dependence of fluorescence intensity 2-AP we have determined RNA melting temperature under various conditions for KD RNA ALV formed by two strands. Effects of magnesium and aminoglycoside antibiotic paromomycin on stabilization of kissing loop-loop dimer RNA have been studied. Under the experimental conditions KD RNA ALV was found to have the stability at the magnesium concentration higher than 1 mM and at paromomycin concentration higher than 2.5 mkM.


Assuntos
2-Aminopurina/química , Regiões 5' não Traduzidas/química , Vírus da Leucose Aviária/química , RNA de Cadeia Dupla/química , RNA Viral/química , 2-Aminopurina/metabolismo , Regiões 5' não Traduzidas/metabolismo , Antibacterianos/química , Vírus da Leucose Aviária/metabolismo , Dimerização , HIV/química , HIV/metabolismo , Temperatura Alta , Paromomicina/química , RNA de Cadeia Dupla/metabolismo , RNA Viral/metabolismo , Espectrometria de Fluorescência
12.
Mol Biol (Mosk) ; 39(1): 147-54, 2005.
Artigo em Russo | MEDLINE | ID: mdl-15773559

RESUMO

Dimerization of two copies of genomic RNA is a necessary step of retroviral replication. In the case of human immunodeficiency virus type 1 (HIV-1) the process is explored in many details. It is proved that conserved stem-loop structure is an essential element in RNA dimerization. Similar model of two-step dimerization mechanism can be considered for avian sarcoma and leukosis virus group (ASLV) in spite of the absence of homology between dimer initiation site (DIS) of ASLV and that of HIV-1. In this paper, short RNA fragments of two viruses: avian sarcoma virus CT-10 and avian leukosis virus HPRS-103 have been chosen in order to investigate the structural requirements of dimerization process and compare them to that of HIV-1. The rate of spontaneous transition from loose to tight dimer was studied as a function of stem length and temperature. Although both types of dimers were observed for both avian retroviruses chosen, fragments of CT-10 requires much higher RNA concentration to form loose dimer. In spite of identical sequence of the loops (5'-A-CUGCAG-3') avian sarcoma virus CT-10 RNA fragments dimerization was greatly impaired. The differences can be explained by deletion of adenine 271 in avian sarcoma virus CT-10 in the stem and by resulting shortening of the self-complementary loop.


Assuntos
Vírus da Leucose Aviária/genética , Vírus do Sarcoma Aviário/genética , Genoma Viral , RNA Viral/genética , Animais , Vírus da Leucose Aviária/química , Vírus do Sarcoma Aviário/química , Sequência de Bases , Dimerização , Dados de Sequência Molecular , Conformação de Ácido Nucleico , RNA Viral/química
13.
J Virol ; 79(6): 3488-99, 2005 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-15731243

RESUMO

The avian sarcoma/leukosis virus (ASLV) is activated for fusion by a two-step mechanism. For ASLV subgroup A (ASLV-A), association with its receptor (Tva) at neutral pH converts virions to a form that can bind target membranes and, in some assays, induce the lipid-mixing stage of fusion. Low pH is necessary to complete the fusion reaction. ASLV-A env (EnvA) exists on the viral surface as a trimer of heterodimers consisting of receptor binding (SU-A) and fusion-mediating (TM-A) subunits. As the receptor binding and fusion-mediating functions reside in separate subunits, we hypothesize that SU-A and TM-A are conformationally coupled. To begin to understand the effect of the binding of a soluble 47-residue domain of the receptor (sTva) on this coupling and the subsequent function of low pH, we prepared recombinant proteins representing full-length SU-A and a nested set of deletion mutant proteins. Full-length SU-A binds sTva with high affinity, but even small deletions at either the N or the C terminus severely impair sTva binding. We have purified the full-length SU-A subunit and characterized its interactions with sTva and the subsequent effect of low pH on the complex. sTva binds SU-A with an apparent KD of 3 pM. Complex formation occludes hydrophobic surfaces and tryptophan residues and leads to a partial loss of alpha-helical structure in SU-A. Low pH does not alter the off rate for the complex, further alter the secondary structure of SU-A, or induce measurable changes in tryptophan environment. The implications of these findings for fusion are discussed.


Assuntos
Vírus da Leucose Aviária/química , Vírus do Sarcoma Aviário/química , Conformação Proteica , Receptores Virais/metabolismo , Proteínas do Envelope Viral/química , Proteínas do Envelope Viral/metabolismo , Vírus da Leucose Aviária/fisiologia , Proteínas Aviárias , Vírus do Sarcoma Aviário/fisiologia , Dicroísmo Circular , Concentração de Íons de Hidrogênio , Fusão de Membrana , Ligação Proteica , Estrutura Terciária de Proteína , Subunidades Proteicas/química , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Deleção de Sequência , Solubilidade , Espectrometria de Fluorescência , Ressonância de Plasmônio de Superfície , Proteínas do Envelope Viral/genética
14.
Virology ; 326(1): 171-81, 2004 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-15262505

RESUMO

We used enzymatic digestion and mass spectrometry to identify the sites of glycosylation on the SU component of the Avian Sarcoma/Leukosis virus (ASLV) Envelope Glycoprotein (Subgroup A). The analysis was done with an SU(A)-rIgG fusion protein that binds the cognate receptor (Tva) specifically. PNGase F removed all the carbohydrate from the SU(A)-rIgG fusion. PNGase F is specific for N-linked carbohydrates; this shows that all the carbohydrate on SU(A) is N-linked. There are 10 modified aspargines in SU(A) (N17, N59, N80, N97, N117, N196, N230, N246, N254, and N330). All conform to the consensus site for N-linked glycosylation NXS/T. There is one potential glycosylation site (N236) that is not modified. Removing most of the carbohydrate from the mature SU(A)-rIgG by PNGase F treatment greatly reduces the ability of the protein to bind Tva, suggesting that carbohydrate may play a direct role in receptor binding.


Assuntos
Vírus da Leucose Aviária/química , Vírus do Sarcoma Aviário/química , Proteínas do Envelope Viral/química , Sequência de Aminoácidos , Animais , Vírus da Leucose Aviária/metabolismo , Proteínas Aviárias , Vírus do Sarcoma Aviário/metabolismo , Sítios de Ligação , Calreticulina/metabolismo , Metabolismo dos Carboidratos , Carboidratos/química , Linhagem Celular , Galinhas , Glicosilação , Espectrometria de Massas , Dados de Sequência Molecular , Fragmentos de Peptídeos/química , Peptídeo-N4-(N-acetil-beta-glucosaminil) Asparagina Amidase/farmacologia , Receptores Virais/metabolismo , Proteínas do Envelope Viral/metabolismo , Proteínas Virais de Fusão/química , Proteínas Virais de Fusão/metabolismo
15.
Virology ; 272(1): 106-11, 2000 Jun 20.
Artigo em Inglês | MEDLINE | ID: mdl-10873753

RESUMO

Avian leukosis virus, subgroup J (ALV-J), has a wide host range, preferentially infecting meat-type birds, and produces a high incidence of myelocytomatosis and nephromas. Using the published sequences from HPRS-103 (ALV-J isolated in 1989 in Great Britain), we designed a set of PCR primers that amplified proviral DNA from nine U.S. field samples. The primers were specific for ALV-J, not amplifying DNA from uninfected cells or cells infected with ALV subgroups A-E. These primers expanded a 2.4-kb fragment that encompasses gp85, gp37, the E element, and most of the 3' LTR. We also developed a set of PCR primers that amplified a 2.1-kb fragment from ALV-J-infected cells and a 1.6-kb fragment from uninfected ev- chicken embryo fibroblasts (Line 0). Upon cloning and DNA sequencing, we determined that the 2.1- and 1.6-kb fragments contained ALV-J gp85- and gp37-like sequences. Comparison of the amino acid sequences demonstrated that the Line 0 sequences were 97.5% identical with the gp85 and gp37 of HPRS-103 and somewhat less identical with the other nine U.S. isolates. This suggests that the envelope genes of ALV-J may have arisen as a result of a recombination event between exogenous ALV and Line 0-like sequences in the chicken. Phylogenetic analysis also showed that the U.S. field isolates were closely related to one another and more distantly related to the European HPRS-103. The pattern of mutations in the U.S. field isolates suggests that the U.S. strains are slowly drifting away from their progenitor Line 0-like sequences. The development of effective vaccines and diagnostic tests is likely to become more problematic as the viruses continue to mutate.


Assuntos
Animais Domésticos/virologia , Vírus da Leucose Aviária/classificação , Vírus da Leucose Aviária/genética , Produtos do Gene env/genética , Genes Virais/genética , Variação Genética/genética , Sequência de Aminoácidos , Animais , Vírus da Leucose Aviária/química , Linhagem Celular , Embrião de Galinha , Galinhas/virologia , Clonagem Molecular , Primers do DNA/genética , Fibroblastos/citologia , Fibroblastos/virologia , Produtos do Gene env/química , Dados de Sequência Molecular , Mutação/genética , Filogenia , Reação em Cadeia da Polimerase , Recombinação Genética/genética , Alinhamento de Sequência , Estados Unidos
16.
J Virol ; 74(8): 3731-9, 2000 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-10729148

RESUMO

Short hydrophobic regions referred to as fusion peptide domains (FPDs) at or near the amino terminus of the membrane-anchoring subunit of viral glycoproteins are believed to insert into the host membrane during the initial stage of enveloped viral entry. Avian sarcoma and leukosis viruses (ASLV) are unusual among retroviruses in that the region in the envelope glycoprotein (EnvA) proposed to be the FPD is internal and contains a centrally located proline residue. To begin analyzing the function of this region of EnvA, 20 substitution mutations were introduced into the putative FPD. The mutant envelope glycoproteins were evaluated for effects on virion incorporation, receptor binding, and infection. Interestingly, most of the single-substitution mutations had little effect on any of these processes. In contrast, a bulky hydrophobic substitution for the central proline reduced viral titers 15-fold without affecting virion incorporation or receptor binding, whereas substitution of glycine for the proline had only a nominal effect on EnvA function. Similar to other viral FPDs, the putative ASLV FPD has been modeled as an amphipathic helix where most of the bulky hydrophobic residues form a patch on one face of the helix. A series of alanine insertion mutations designed to interrupt the hydrophobic patch on the helix had differential effects on infectivity, and the results of that analysis together with the results observed with the substitution mutations suggest no correlation between maintenance of the hydrophobic patch and glycoprotein function.


Assuntos
Vírus da Leucose Aviária , Vírus do Sarcoma Aviário , Mutação , Proteínas do Envelope Viral/química , Proteínas do Envelope Viral/genética , Motivos de Aminoácidos , Sequência de Aminoácidos , Vírus da Leucose Aviária/química , Vírus da Leucose Aviária/genética , Vírus da Leucose Aviária/patogenicidade , Vírus do Sarcoma Aviário/química , Vírus do Sarcoma Aviário/genética , Vírus do Sarcoma Aviário/patogenicidade , Vírus do Sarcoma Aviário/fisiologia , Linhagem Celular , Ensaio de Imunoadsorção Enzimática , Epitopos , Dados de Sequência Molecular , Mutagênese Insercional , Mutação Puntual , Receptores Virais/metabolismo , Deleção de Sequência , Proteínas do Envelope Viral/metabolismo , Vírion/metabolismo
17.
Oncogene ; 9(5): 1307-20, 1994 May.
Artigo em Inglês | MEDLINE | ID: mdl-8152791

RESUMO

The evolution of oncogene-transducing retroviruses was followed by studying the genomes of five new, erbB carrying retroviruses. These viruses, isolated from cells of one chicken infected with Rous Associated virus 1 (RAV-1), had captured c-erbB sequences as a consequence of RAV-1 integration into the host genome. Their genome structures were distinct; however, their v-erbB genes had sustained identical 5' and 3' deletions and the v-erbB-env junctions were identical at the nucleotide level. The results therefore strongly suggest that all five viruses originate from the same capture event. Sequence analyses of the v-erbB genes from three of these viruses revealed that one of them had undergone no further mutation and lacked detectable capacity to transform cells, therefore probably representing an 'early' form of transducing virus. The two other v-erbB genes contained distinct mutations and differed in their potential to induce fibroblast- and erythroblast transformation; they therefore probably represent later derivatives of the virus that captured the erbB oncogene. The data suggest that the initial retrovirus rapidly underwent many alterations after capture of c-erbB sequences, already in the RAV-1 infected bird as well as during subsequent in vitro isolation procedures. The changes involve both major rearrangements of the genome as well as point mutations that activated the erbB oncogene.


Assuntos
Vírus da Leucose Aviária/genética , Transformação Celular Viral/genética , Deleção de Genes , Genoma Viral , Mutação/genética , Proto-Oncogenes/genética , RNA Viral/genética , Sequência de Aminoácidos , Animais , Vírus da Leucose Aviária/química , Vírus da Leucose Aviária/crescimento & desenvolvimento , Vírus da Leucose Aviária/metabolismo , Sequência de Bases , Embrião de Galinha , Galinhas , Receptores ErbB/análise , Receptores ErbB/genética , Receptores ErbB/metabolismo , Rearranjo Gênico , Genes gag/genética , Leucemia Eritroblástica Aguda/genética , Leucemia Eritroblástica Aguda/microbiologia , Dados de Sequência Molecular , Proteínas Proto-Oncogênicas/análise , Proteínas Proto-Oncogênicas/genética , Proteínas Proto-Oncogênicas/metabolismo , Provírus/química , Provírus/genética , RNA Viral/química , Ativação Viral
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...