Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 33
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Viruses ; 11(6)2019 05 30.
Artigo em Inglês | MEDLINE | ID: mdl-31151254

RESUMO

The initial step of retrovirus entry-the interaction between the virus envelope glycoprotein trimer and a cellular receptor-is complex, involving multiple, noncontiguous determinants in both proteins that specify receptor choice, binding affinity and the ability to trigger conformational changes in the viral glycoproteins. Despite the complexity of this interaction, retroviruses have the ability to evolve the structure of their envelope glycoproteins to use a different cellular protein as receptors. The highly homologous subgroup A to E Avian Sarcoma and Leukosis Virus (ASLV) glycoproteins belong to the group of class 1 viral fusion proteins with a two-step triggering mechanism that allows experimental access to intermediate structures during the fusion process. We and others have taken advantage of replication-competent ASLVs and exploited genetic selection strategies to force the ASLVs to naturally evolve and acquire envelope glycoprotein mutations to escape the pressure on virus entry and still yield a functional replicating virus. This approach allows for the simultaneous selection of multiple mutations in multiple functional domains of the envelope glycoprotein that may be required to yield a functional virus. Here, we review the ASLV family and experimental system and the reverse engineering approaches used to understand the evolution of ASLV receptor usage.


Assuntos
Vírus da Leucose Aviária/genética , Vírus do Sarcoma Aviário/genética , Evolução Molecular , Receptores Virais/genética , Genética Reversa , Animais , Vírus do Sarcoma Aviário/classificação , Galinhas/virologia , Mutação , Sarcoma Aviário , Proteínas do Envelope Viral/genética , Internalização do Vírus , Replicação Viral
2.
Biotechnol Bioeng ; 86(6): 650-60, 2004 Jun 20.
Artigo em Inglês | MEDLINE | ID: mdl-15137076

RESUMO

The association of cell surface receptors with sterol-sphingolipid-enriched microdomains of the plasma membrane, so-called lipid rafts, may affect the receptor-mediated entry and trafficking dynamics of viruses. A model retrovirus, subgroup A avian sarcoma and leukosis virus (ASLV-A), can initiate infection by binding to either of two forms of the tumor virus subgroup A (TVA) receptor, a lipid-raft-associated glycosylphosphatidylinositol (GPI)-anchored receptor (TVA800) or a transmembrane receptor (TVA950). Narayan et al. previously found that virus particles bound to TVA950 were more rapidly internalized than virions bound to TVA800, and the internalization via TVA950 exhibited biphasic kinetics. To explore potential molecular mechanisms for these results we developed a mathematical model that accounts for internalization of viruses through cellular pits, trafficking to an endosomal compartment where fusion occurs, and viral DNA synthesis. By fitting the model to experimental data we found that viruses bound to TVA950 were internalized up to 2.6-fold more rapidly than viruses bound to TVA800. Two- to threefold greater lateral diffusivities of transmembrane proteins, relative to GPI-anchored proteins, observed in other systems, suggest that the internalization rate of ASLV-A is diffusion-limited. Furthermore, by allowing for recycling of internalized TVA950-bound viruses back to the cell surface, we can account for the observed biphasic internalization kinetics. This mechanism is also consistent with the observed slower rate of DNA synthesis for viruses that enter via TVA950. Overall, the model provides a means to generate new experimentally testable hypotheses and sets a foundation for building a quantitative and integrated understanding of viral entry, trafficking, and intracellular dynamics.


Assuntos
Microdomínios da Membrana/fisiologia , Retroviridae/fisiologia , Animais , Vírus da Leucose Aviária/fisiologia , Vírus do Sarcoma Aviário/classificação , Vírus do Sarcoma Aviário/fisiologia , Transporte Biológico , Aves/virologia , Fusão Celular , Difusão , Endocitose , Cinética , Modelos Estatísticos , Estrutura Terciária de Proteína , Receptores Virais/química , Receptores Virais/metabolismo , Vírion/metabolismo
3.
Anim Genet ; 35(3): 176-81, 2004 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-15147387

RESUMO

Avian sarcoma and leucosis viruses (ASLV) are classified into six major subgroups (A to E and J) according to the properties of the viral envelope proteins and the usage of cellular receptors for virus entry. Subgroup A and B receptors are identified molecularly and their genomic positions TVA and TVB are mapped. The subgroup C receptor is unknown, its genomic locus TVC is reported to be genetically linked to TVA, which resides on chicken chromosome 28. In this study, we used two chicken inbred lines that carry different alleles coding for resistance (TVC(R) and sensitivity (TVC(S)) to infection by subgroup C viruses. A backross population of these lines was tested for susceptibility to subgroup C infection and genotyped for markers from chicken chromosome 28. We confirmed the close linkage between TVA and TVC loci. Further, we have described the position of TVC on chromosome 28 relative to markers from the consensus map of the chicken genome.


Assuntos
Vírus do Sarcoma Aviário/genética , Galinhas/genética , Mapeamento Cromossômico , Fenótipo , Receptores Virais/genética , Proteínas do Envelope Viral/genética , Alelos , Animais , Vírus do Sarcoma Aviário/classificação , Galinhas/virologia , Cruzamentos Genéticos , Primers do DNA , Repetições de Microssatélites/genética
4.
Poult Sci ; 83(4): 638-49, 2004 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-15109061

RESUMO

The chicken major histocompatibility (B) complex (MHC) affects disease outcome significantly. One of the best characterized systems of MHC control is the response to the oncogenic retrovirus, Rous sarcoma virus (RSV). Genetic selection altered the tumor growth pattern, either regressively or progressively, with the data suggesting control by one or a few loci. Particular MHC genotypes determine RSV tumor regression or progression indicating the crucial B complex role in Rous sarcoma outcome. Analysis of inbred lines, their crosses, congenic lines, and noninbred populations has revealed the anti-RSV response of many B complex haplotypes. Tumor growth disparity among lines identical at the MHC but differing in their background genes suggested a non-MHC gene contribution to tumor fate. Genetic complementation in tumor growth has also been demonstrated for MHC and non-MHC genes. RSV tumor expansion reflects both tumor cell proliferation and viral replication generating new tumor cells. In addition, the B complex controls tumor growth induced by a subviral DNA construct encoding only the RSV v-src oncogene. Immunity to subsequent tumors and metastasis also exhibit MHC control. Genotypes that regressed either RSV or v-src DNA primary tumors had enhanced protection against subsequent homologous challenge. Regressor B genotypes had lower tumor metastasis compared with progressor types. Together, the data indicate that B complex control of RSV tumor fate is strongly defined by the response to a v-src-determined function. Differential RSV tumor outcomes among various B genotypes may include immune recognition of a tumor-specific antigen or immune system influences on viral replication.


Assuntos
Vírus do Sarcoma Aviário/imunologia , Complexo Principal de Histocompatibilidade/imunologia , Doenças das Aves Domésticas/imunologia , Sarcoma Aviário/imunologia , Animais , Vírus do Sarcoma Aviário/classificação , Galinhas , Cruzamentos Genéticos , Feminino , Dosagem de Genes , Masculino , Doenças das Aves Domésticas/genética , Doenças das Aves Domésticas/patologia , Sarcoma Aviário/genética , Sarcoma Aviário/patologia
5.
J Virol ; 77(5): 3058-66, 2003 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-12584331

RESUMO

We previously showed that the envelope glycoprotein (EnvA) of avian sarcoma/leukosis virus subtype A (ASLV-A) binds to liposomes at neutral pH following incubation with its receptor, Tva, at >or=22 degrees C. We also provided evidence that ASLV-C fuses with cells at neutral pH. These findings suggested that receptor binding at neutral pH and >or=22 degrees C is sufficient to activate Env for fusion. A recent study suggested that two steps are necessary to activate avian retroviral Envs: receptor binding at neutral pH, followed by exposure to low pH (W. Mothes et al., Cell 103:679-689, 2000). Therefore, we evaluated the requirements for intact ASLV-A particles to bind to target bilayers and fuse with cells. We found that ASLV-A particles bind stably to liposomes in a receptor- and temperature-dependent manner at neutral pH. Using ASLV-A particles biosynthetically labeled with pyrene, we found that ASLV-A mixes its lipid envelope with cells within 5 to 10 min at 37 degrees C. Lipid mixing was neither inhibited nor enhanced by incubation at low pH. Lipid mixing of ASLV-A was inhibited by a peptide designed to prevent six-helix bundle formation in EnvA; the same peptide inhibits virus infection and EnvA-mediated cell-cell fusion (at both neutral and low pHs). Bafilomycin and dominant-negative dynamin inhibited lipid mixing of Sindbis virus (which requires low pH for fusion), but not of ASLV-A, with host cells. Finally, we found that, although EnvA-induced cell-cell fusion is enhanced at low pH, a mutant EnvA that is severely compromised in its ability to support infection still induced massive syncytia at low pH. Our results indicate that receptor binding at neutral pH is sufficient to activate EnvA, such that ASLV-A particles bind hydrophobically to and merge their membranes with target cells. Possible roles for low pH at subsequent stages of viral entry are discussed.


Assuntos
Vírus da Leucose Aviária/fisiologia , Vírus do Sarcoma Aviário/fisiologia , Metabolismo dos Lipídeos , Fusão de Membrana , Células 3T3 , Sequência de Aminoácidos , Animais , Vírus da Leucose Aviária/classificação , Vírus do Sarcoma Aviário/classificação , Fusão Celular , Linhagem Celular , Produtos do Gene env/metabolismo , Concentração de Íons de Hidrogênio , Lipossomos/metabolismo , Fusão de Membrana/efeitos dos fármacos , Camundongos , Dados de Sequência Molecular , Peptídeos/síntese química , Peptídeos/química , Peptídeos/metabolismo , Peptídeos/farmacologia , Vírion/metabolismo
6.
Folia Biol (Praha) ; 48(1): 28-33, 2002.
Artigo em Inglês | MEDLINE | ID: mdl-11871857

RESUMO

Existing variants of the oncogene v-src differ in their transforming potential as well as in the range of their hosts. We compared the protein kinase activities of two Prague C v-Src variants (PRC and H19), reported to be of low oncogenic potential (Plachý et al., 1995), with the highly oncogenic Schmidt-Ruppin A v-Src (SRA). We employed in vitro kinase assays of affinity-purified proteins expressed in rabbit reticulocyte lysate and in S. cerevisiae. In both systems used, the specific kinase activity of the Prague C v-Src kinases amounted to only ca 20% of the activity of SRA. This positions the PRC Src close to activated c-Src, despite the lack of the regulatory C-terminal tail in PRC. We constructed chimeras between PRC and SRA v-Src and tested them for specific kinase activity in S. cerevisiae. Remarkably, the regulatory N-terminal part of PRC, when fused to the SRA-derived kinase domain, lowered the chimeras' PK activity to ca 20%, suggesting that it is the regulatory part of PRC that is responsible for its low phosphotransferase activity.


Assuntos
Vírus do Sarcoma Aviário/enzimologia , Proteína Oncogênica pp60(v-src)/química , Vírus do Sarcoma Aviário/classificação , Vírus do Sarcoma Aviário/genética , Domínio Catalítico , Genes src , Proteína Oncogênica pp60(v-src)/antagonistas & inibidores , Proteína Oncogênica pp60(v-src)/farmacologia , Fosforilação , Processamento de Proteína Pós-Traducional , Estrutura Terciária de Proteína , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/farmacologia , Saccharomyces cerevisiae , Especificidade da Espécie , Relação Estrutura-Atividade
7.
J Virol ; 75(4): 2002-9, 2001 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-11160701

RESUMO

In a previous study, we found avian sarcoma and leukosis virus (ASLV) gag genes in 19 species of birds in the order Galliformes including all grouse and ptarmigan (Tetraoninae) surveyed. Our data suggested that retroviruses had been transmitted horizontally among some host species. To further investigate these elements, we sequenced a replication-defective retrovirus, here named tetraonine endogenous retrovirus (TERV), from Bonasa umbellus (ruffed grouse). This is the first report of a complete, replication-defective ASLV provirus sequence from any bird other than the domestic chicken. We found a replication-defective proviral sequence consisting of putative Gag and Env proteins flanked by long terminal repeats. Reverse transcription-PCR analysis showed that retroviral gag sequences closely related to TERV are transcribed, supporting the hypothesis that TERV is an active endogenous retrovirus. Phylogenetic analyses suggest that TERV may have arisen via recombination between different retroviral lineages infecting birds. Southern blotting using gag probes showed that TERV occurs in tetraonines but not in chickens or ducks, suggesting that integration occurred after the earliest phasianid divergences but prior to the radiation of tetraonine birds.


Assuntos
Vírus da Leucose Aviária/classificação , Vírus do Sarcoma Aviário/classificação , Aves/virologia , Retrovirus Endógenos/classificação , Retrovirus Endógenos/genética , Sequência de Aminoácidos , Animais , Vírus da Leucose Aviária/genética , Vírus do Sarcoma Aviário/genética , Bacteriófago lambda , Sequência de Bases , Evolução Biológica , Produtos do Gene gag/metabolismo , Genes gag , Biblioteca Genômica , Dados de Sequência Molecular , Filogenia , Provírus/genética , Recombinação Genética , Análise de Sequência de DNA , Replicação Viral
8.
J Virol ; 74(9): 3984-95, 2000 May.
Artigo em Inglês | MEDLINE | ID: mdl-10756010

RESUMO

In a study of the evolution and distribution of avian retroviruses, we found avian sarcoma and leukosis virus (ASLV) gag genes in 26 species of galliform birds from North America, Central America, eastern Europe, Asia, and Africa. Nineteen of the 26 host species from whom ASLVs were sequenced were not previously known to contain ASLVs. We assessed congruence between ASLV phylogenies based on a total of 110 gag gene sequences and ASLV-host phylogenies based on mitochondrial 12S ribosomal DNA and ND2 sequences to infer coevolutionary history for ASLVs and their hosts. Widespread distribution of ASLVs among diverse, endemic galliform host species suggests an ancient association. Congruent ASLV and host phylogenies for two species of Perdix, two species of Gallus, and Lagopus lagopus and L. mutus also indicate an old association with vertical transmission and cospeciation for these ASLVs and hosts. An inference of horizontal transmission of ASLVs among some members of the Tetraoninae subfamily (grouse and ptarmigan) is supported by ASLV monophyletic groups reflecting geographic distribution and proximity of hosts rather than host species phylogeny. We provide a preliminary phylogenetic taxonomy for the new ASLVs, in which named taxa denote monophyletic groups.


Assuntos
Vírus da Leucose Aviária/genética , Vírus do Sarcoma Aviário/genética , Aves/virologia , Genes gag , Animais , Vírus da Leucose Aviária/classificação , Vírus do Sarcoma Aviário/classificação , Sequência de Bases , DNA Viral , Dados de Sequência Molecular , Filogenia , Reação em Cadeia da Polimerase/métodos , Análise de Sequência de DNA
9.
Cancer Res ; 53(4): 915-20, 1993 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-8381333

RESUMO

We have developed an oncogene-specific tumor regression system in chickens. Injection s.c. of chicken wing webs with either rASV1702 or rASV157, mutants of Rous sarcoma virus (RSV) that express nonmyristolyated src product containing novel N-terminal domains, results in noninvasive fibrosarcomas that regress fully. The ability of challenge infections with wild-type RSV to form tumors is suppressed. This protective effect was shown to be specific for determinants encoded or induced by v-src (I. Gelman and H. Hanafusa, J. Virol., 61: 2461-2468, 1989). In the current study, we used SC chickens, inbred for major histocompatibility complex Class I haplotype B2/B2, to investigate whether this protection results from active immunity. Preinoculation of chickens with either replication-defective rASV1702 virus or non-virus-producing syngeneic chicken embryo fibroblasts expressing 1702src conferred protection against challenge infections with RSV. Thus, viremia was not required for this protection. Splenic lymphocytes from rASV1702-infected donors could transfer protective immunity against RSV tumor challenge to naive chickens. These lymphocytes were cytotoxic in vitro against RSV- or rASV1702-infected SC-chicken embryo fibroblasts, but not against SC-chicken embryo fibroblasts infected with helper virus, suggesting a specificity for src-encoded or -induced determinants. In contrast, splenic lymphocytes from RSV-infected chickens transferred protective immunity poorly and exhibited low in vitro cytotoxic potential for src determinants, suggesting possible suppression mechanisms. Finally, murine cell lines expressing 157src or 1702src produced tumors in nude mice that failed to regress. Thus, although cells expressing 157src or 1702src are inherently tumorigenic, the tumors they induce most likely regress due to immune mechanisms. These results suggest that 1702src and 157src induce src-specific tumor Ag that potently prime an oncogene-specific protective cellular immunity.


Assuntos
Vírus do Sarcoma Aviário , Transformação Celular Viral/imunologia , Genes src/imunologia , Imunoterapia Adotiva , Linfócitos/imunologia , Sarcoma Aviário/imunologia , Animais , Antígenos de Neoplasias/imunologia , Vírus do Sarcoma Aviário/classificação , Vírus do Sarcoma Aviário/genética , Galinhas , Imunidade Celular , Complexo Principal de Histocompatibilidade , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Sarcoma Aviário/terapia
10.
J Gen Virol ; 73 ( Pt 11): 2995-7, 1992 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-1331300

RESUMO

The host ranges of the Rous sarcoma virus (RSV) pseudotype RSV(HPRS-103) of a novel avian leukosis virus (ALV), strain HPRS-103, and representative RSV pseudotypes of subgroups A to F, have been determined in embryo fibroblasts from 12 avian species. Domestic fowl, red jungle fowl, Sonnerat's jungle fowl and turkey were susceptible to infection by RSV(HPRS-103); ring-necked pheasant, Japanese green pheasant, golden pheasant, Japanese quail, guinea-fowl, Peking duck, Muscovy duck and goose were resistant. The host range pattern of RSV(HPRS-103) differs from those of viruses of subgroups A to G and I, and provides support for placing the HPRS-103 strain of ALV in a new envelope subgroup, designated J.


Assuntos
Vírus do Sarcoma Aviário/patogenicidade , Aves/microbiologia , Proteínas do Envelope Viral/classificação , Animais , Vírus do Sarcoma Aviário/classificação , Células Cultivadas , Embrião não Mamífero/citologia , Fibroblastos/microbiologia , Especificidade da Espécie , Infecções Tumorais por Vírus/epidemiologia , Virulência
11.
Nucleic Acids Res ; 19(24): 6929-34, 1991 Dec 25.
Artigo em Inglês | MEDLINE | ID: mdl-1662367

RESUMO

A study of the secondary structures of the 5'-leader RNA sequences of avian leukosis/sarcoma viruses was conducted using phylogenetic sequence alignment, theoretical structures calculated from base-pairing interactions involving the calculated minimal delta G values, and RNaseT1 sensitivity. The results suggest that all of the avian retroviral RNA leaders may be able to adopt similar conformations. Open reading frames in the leader RNAs may be positioned to facilitate viral activities such as translation and packaging of the genomic RNA into virus particles.


Assuntos
Vírus da Leucose Aviária/genética , Vírus do Sarcoma Aviário/genética , RNA Viral/química , Vírus da Leucose Aviária/classificação , Vírus do Sarcoma Aviário/classificação , Composição de Bases , Sequência de Bases , Exorribonucleases/metabolismo , Regulação Viral da Expressão Gênica/genética , Dados de Sequência Molecular , Conformação de Ácido Nucleico , Fases de Leitura Aberta/genética , Filogenia , Biossíntese de Proteínas/genética , RNA Viral/genética , RNA Viral/metabolismo , Alinhamento de Sequência , Replicação Viral/genética
13.
Virus Res ; 9(2-3): 159-203, 1988 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-2833047

RESUMO

Twelve independent isolates of avian sarcoma viruses (ASVs) can be divided into four groups according to the transforming genes harbored in the viral genomes. The first group is represented by viruses containing the transforming sequence, src, inserted in the viral genome as an independent gene; the other three groups of viruses contain transforming genes fps, yes or ros fused to various length of the truncated structural gene gag. These transforming sequences have been obtained by avian retroviruses from chicken cellular DNA by recombination. The src-containing viruses code for an independent polypeptide, p60src; and the representative fps, yes and ros-containing ASVs code for P140/130gag-fps, P90gag-yes and P68gag-ros fusion polypeptides respectively. All of these transforming proteins are associated with the tyrosine-specific protein kinase activity capable of autophosphorylation and phosphorylating certain foreign substrates. p60src and P68gag-ros are integral cellular membrane proteins and P140/130gag-fps and P90gag-yes are only loosely associated with the plasma membrane. Cells transformed by ASVs contain many newly phosphorylated proteins and in most cases have an elevated level of total phosphotyrosine. However, no definitive correlation between phosphorylation of a particular substrate and transformation has been established except that a marked increase of the tyrosine phosphorylation of a 34,000 to 37,000 dalton protein is observed in most ASV transformed cells. The kinase activity of ASV transforming proteins appears to be essential, but not sufficient for transformation. The N-terminal domain of p60src required for myristylation and membrane binding is also crucial for transformation. By contrast, the gag portion of the FSV P130gag-fps is dispensable for in vitro transformation and removal of it has only an attenuating effect on in vivo tumorigenicity. The products of cellular src, fps and yes proto-oncogenes have been identified and shown to also have tyrosine-specific protein kinase activity. The transforming potential of c-src and c-fps has been studied and shown that certain structural changes are necessary to convert them into transforming genes. Among the cellular proto-oncogenes related to the four ASV transforming genes, c-ros most likely codes for a growth factor receptor-like molecule. It is possible that the oncogene products of ASVs act through certain membrane receptor(s) or enzyme(s), such as protein kinase C, in the process of cell transformation.


Assuntos
Vírus do Sarcoma Aviário/fisiologia , Animais , Vírus do Sarcoma Aviário/classificação , Vírus do Sarcoma Aviário/genética , Genes Virais , Substâncias de Crescimento/genética , Oncogenes , Replicação Viral
14.
Virus Res ; 8(1): 73-7, 1987 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-2821707

RESUMO

Avian RNA tumor virus envelope glycoprotein protects against sarcoma development by an avian sarcoma virus of the same subgroup. Avian RNA tumor viruses, members of the retrovirus family, induce various malignancies in fowl (Weiss et al. (eds.), 1982, RNA Tumor Viruses, Cold Spring Harbor, N.Y.). These viruses consist of a genomic RNA core surrounded by an envelope with embedded glycoproteins, of 85 and 37 kDa. The 85 kDa glycoprotein is antigenically specific for each subgroup as determined by neutralization. The envelope glycoprotein can be removed from the virion with retention of its antigenicity (Duesberg et al., 1970, Virology 41, 631-646). Two fractions of 4-6S and 8S, separated by sedimentation, were shown to retain antigenicity by interference of neutralization of virus by antibody. Thus, the 4-6S and 8S preparations could possibly serve as immunogens. The objective of this study was to determine if such envelope glycoprotein preparations could function as potential vaccines, and if so, whether the protection afforded would be subgroup specific.


Assuntos
Vírus do Sarcoma Aviário/imunologia , Sarcoma Experimental/prevenção & controle , Proteínas do Envelope Viral/imunologia , Vacinas Virais/imunologia , Animais , Vírus do Sarcoma Aviário/classificação , Glicoproteínas/imunologia , Imunização , Especificidade da Espécie
15.
Virology ; 156(1): 163-6, 1987 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-3027964

RESUMO

In order to obtain information on the phylogenies of viral strains which belong to RSV (Rous sarcoma virus) and ALV (avian leukosis virus), the nucleotide sequences of noncoding regions adjacent to the U3 region in two ALV strains, Rous-associated virus 7 (RAV 7) and RAV 50, and three RSV strains, Bratislava 77 (B77), Prague:subgroup B (PR-B), and Schmidt-Ruppin:subgroup B (SR-B) were determined by extension from a common primer. The sequences thus deduced were compared with known sequences of other RSV and ALV strains and the structural features of the newly determined viral genomes were discussed.


Assuntos
Vírus da Leucose Aviária/genética , Vírus do Sarcoma Aviário/genética , Genes Virais , Genes , Proteínas do Envelope Viral/genética , Vírus da Leucose Aviária/classificação , Vírus do Sarcoma Aviário/classificação , Sequência de Bases , Filogenia
17.
J Gen Virol ; 50(1): 211-6, 1980 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-6255086

RESUMO

Simultaneous injection of newborn rabbits with the Schmidt-Ruppin and Prague strains of Rous sarcoma virus regularly induces antibodies which not only recognize the pp60src of both strains but also give positive kinase reaction with extracts of cells infected with either strain.


Assuntos
Anticorpos Antivirais/biossíntese , Vírus do Sarcoma Aviário/imunologia , Fosfoproteínas/imunologia , Proteínas Virais/imunologia , Animais , Vírus do Sarcoma Aviário/classificação , Vírus do Sarcoma Aviário/enzimologia , Transformação Celular Viral , Reações Cruzadas , Imunização , Proteínas Quinases/metabolismo , Coelhos
18.
J Virol ; 29(3): 926-37, 1979 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-221681

RESUMO

The number of different oncoviral env genes that can be expressed by a single chicken embryo fibroblast was investigated. Fibroblasts were infected with one to three subgroups of Rous-associated virus, which is a nontransforming avian oncovirus, then superinfected with a transforming virus, Rous sarcoma virus, of a different subgroup. The subgroups of viruses released by the resulting clones were analyzed. When two viral subgroups were used for preinfection, all the resulting clones produced transforming virus particles having the subgroup of the superinfecting virus, and most clones produced transforming virus particles of all the infecting viral subgroups. However, when cells were preinfected with three viral subgroups, many of the resulting clones did not produce transforming virus particles having the subgroup of the superinfecting virus, and only 1 of 23 clones produced transforming particles of all the infecting viral subgroups. DNA annealing experiments showed that cells infected with three or four viral subgroups had an additional 8 to 20 copies of proviral DNA per cell. Finally, most clones resulting from cells simultaneously infected with three or four viral subgroups were able to produce virus of all infecting subgroups. It appears that the number of exogenous oncoviral env genes that can be expressed by a single cell is limited, and in the range of 4 to 8-20 per cell.


Assuntos
Vírus da Leucose Aviária/crescimento & desenvolvimento , Vírus do Sarcoma Aviário/crescimento & desenvolvimento , Replicação Viral , Animais , Vírus da Leucose Aviária/classificação , Vírus do Sarcoma Aviário/classificação , Transformação Celular Neoplásica , Transformação Celular Viral , Embrião de Galinha , Células Clonais , Técnicas de Cultura , DNA Viral/análise , Fibroblastos
20.
J Virol ; 20(2): 384-90, 1976 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-185423

RESUMO

Chicken fibroblasts derived from the H & N flock, which have been characterized as resistant to subgroup B avian oncornaviruses in focus assays, can be infected in suspension shortly after trypsinization by subgroup B sarcoma and leukosis viruses. Once cells are plated, resistance to infection reappears rapidly. C/BE cell suspensions obtained by treatment with EDTA instead of trypsin are not as sensitive to infection. Late interference established by preinfection with subgroup B leukosis viruses is not overcome by trypsinization. In addition to C/BE H & N chicken cells, C/ABE RPRL line 7 cells can also be infected by subgroup B viruses shortly after trypsinization; however, none of the cell types can be made sensitive to subgroup E infection. These results are discussed in relation to current information on the genetic control of resistance to avian oncornaviruses.


Assuntos
Vírus do Sarcoma Aviário/crescimento & desenvolvimento , Animais , Vírus da Leucose Aviária/crescimento & desenvolvimento , Vírus do Sarcoma Aviário/classificação , Linhagem Celular , Embrião de Galinha , Ácido Edético/farmacologia , Codorniz , Tripsina/farmacologia , Interferência Viral , Replicação Viral
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...