Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 81
Filtrar
Mais filtros








Base de dados
Intervalo de ano de publicação
1.
Viruses ; 13(1)2021 Jan 18.
Artigo em Inglês | MEDLINE | ID: mdl-33477490

RESUMO

The assembly of a hexameric lattice of retroviral immature particles requires the involvement of cell factors such as proteins and small molecules. A small, negatively charged polyanionic molecule, myo-inositol hexaphosphate (IP6), was identified to stimulate the assembly of immature particles of HIV-1 and other lentiviruses. Interestingly, cryo-electron tomography analysis of the immature particles of two lentiviruses, HIV-1 and equine infectious anemia virus (EIAV), revealed that the IP6 binding site is similar. Based on this amino acid conservation of the IP6 interacting site, it is presumed that the assembly of immature particles of all lentiviruses is stimulated by IP6. Although this specific region for IP6 binding may be unique for lentiviruses, it is plausible that other retroviral species also recruit some small polyanion to facilitate the assembly of their immature particles. To study whether the assembly of retroviruses other than lentiviruses can be stimulated by polyanionic molecules, we measured the effect of various polyanions on the assembly of immature virus-like particles of Rous sarcoma virus (RSV), a member of alpharetroviruses, Mason-Pfizer monkey virus (M-PMV) representative of betaretroviruses, and murine leukemia virus (MLV), a member of gammaretroviruses. RSV, M-PMV and MLV immature virus-like particles were assembled in vitro from truncated Gag molecules and the effect of selected polyanions, myo-inostol hexaphosphate, myo-inositol, glucose-1,6-bisphosphate, myo-inositol hexasulphate, and mellitic acid, on the particles assembly was quantified. Our results suggest that the assembly of immature particles of RSV and MLV was indeed stimulated by the presence of myo-inostol hexaphosphate and myo-inositol, respectively. In contrast, no effect on the assembly of M-PMV as a betaretrovirus member was observed.


Assuntos
Membrana Celular/química , Membrana Celular/metabolismo , Interações Hospedeiro-Patógeno , Polieletrólitos/química , Retroviridae/fisiologia , Montagem de Vírus , Alpharetrovirus/fisiologia , Animais , Betaretrovirus/fisiologia , Células Cultivadas , Gammaretrovirus/fisiologia , Produtos do Gene gag/química , Produtos do Gene gag/metabolismo , Polieletrólitos/metabolismo , Retroviridae/ultraestrutura , Vírion
2.
Viruses ; 6(12): 4811-38, 2014 Dec 05.
Artigo em Inglês | MEDLINE | ID: mdl-25490763

RESUMO

Gene therapy using integrating retroviral vectors has proven its effectiveness in several clinical trials for the treatment of inherited diseases and cancer. However, vector-mediated adverse events related to insertional mutagenesis were also observed, emphasizing the need for safer therapeutic vectors. Paradoxically, alpharetroviruses, originally discovered as cancer-causing agents, have a more random and potentially safer integration pattern compared to gammaretro- and lentiviruses. In this review, we provide a short overview of the history of alpharetroviruses and explain how they can be converted into state-of-the-art gene delivery tools with improved safety features. We discuss development of alpharetroviral vectors in compliance with regulatory requirements for clinical translation, and provide an outlook on possible future gene therapy applications. Taken together, this review is a broad overview of alpharetroviral vectors spanning the bridge from their parental virus discovery to their potential applicability in clinical settings.


Assuntos
Alpharetrovirus/fisiologia , Terapia Genética/instrumentação , Neoplasias/terapia , Alpharetrovirus/genética , Animais , Terapia Genética/métodos , Vetores Genéticos/genética , Vetores Genéticos/fisiologia , Humanos , Neoplasias/genética , Integração Viral
3.
Virology ; 450-451: 2-12, 2014 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-24503062

RESUMO

Lymphoproliferative disease virus (LPDV) is an exogenous oncogenic retrovirus that induces lymphoid tumors in some galliform species of birds. Historically, outbreaks of LPDV have been reported from Europe and Israel. Although the virus has previously never been detected in North America, herein we describe the widespread distribution, genetic diversity, pathogenesis, and evolution of LPDV in the United States. Characterization of the provirus genome of the index LPDV case from North America demonstrated an 88% nucleotide identity to the Israeli prototype strain. Although phylogenetic analysis indicated that the majority of viruses fell into a single North American lineage, a small subset of viruses from South Carolina were most closely related to the Israeli prototype. These results suggest that LPDV was transferred between continents to initiate outbreaks of disease. However, the direction (New World to Old World or vice versa), mechanism, and time frame of the transcontinental spread currently remain unknown.


Assuntos
Alpharetrovirus/fisiologia , Doenças Transmissíveis Emergentes/veterinária , Doenças Negligenciadas/veterinária , Doenças das Aves Domésticas/virologia , Infecções por Retroviridae/veterinária , Infecções Tumorais por Vírus/veterinária , Alpharetrovirus/classificação , Alpharetrovirus/genética , Alpharetrovirus/isolamento & purificação , Animais , Carcinogênese , Doenças Transmissíveis Emergentes/virologia , Evolução Molecular , Dados de Sequência Molecular , Doenças Negligenciadas/virologia , Filogenia , Doenças das Aves Domésticas/diagnóstico , Doenças das Aves Domésticas/epidemiologia , Infecções por Retroviridae/virologia , Infecções Tumorais por Vírus/virologia , Perus/virologia , Estados Unidos/epidemiologia
4.
J Virol ; 86(4): 2021-30, 2012 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-22171251

RESUMO

The group of closely related avian sarcoma and leukosis viruses (ASLVs) evolved from a common ancestor into multiple subgroups, A to J, with differential host range among galliform species and chicken lines. These subgroups differ in variable parts of their envelope glycoproteins, the major determinants of virus interaction with specific receptor molecules. Three genetic loci, tva, tvb, and tvc, code for single membrane-spanning receptors from diverse protein families that confer susceptibility to the ASLV subgroups. The host range expansion of the ancestral virus might have been driven by gradual evolution of resistance in host cells, and the resistance alleles in all three receptor loci have been identified. Here, we characterized two alleles of the tva receptor gene with similar intronic deletions comprising the deduced branch-point signal within the first intron and leading to inefficient splicing of tva mRNA. As a result, we observed decreased susceptibility to subgroup A ASLV in vitro and in vivo. These alleles were independently found in a close-bred line of domestic chicken and Indian red jungle fowl (Gallus gallus murghi), suggesting that their prevalence might be much wider in outbred chicken breeds. We identified defective splicing to be a mechanism of resistance to ASLV and conclude that such a type of mutation could play an important role in virus-host coevolution.


Assuntos
Alpharetrovirus/fisiologia , Proteínas Aviárias/genética , Galinhas/genética , Predisposição Genética para Doença , Doenças das Aves Domésticas/genética , Splicing de RNA , Receptores Virais/genética , Sarcoma Aviário/genética , Deleção de Sequência , Alpharetrovirus/genética , Sequência de Aminoácidos , Animais , Proteínas Aviárias/metabolismo , Sequência de Bases , Galinhas/metabolismo , Galinhas/virologia , Íntrons , Dados de Sequência Molecular , Doenças das Aves Domésticas/metabolismo , Doenças das Aves Domésticas/virologia , Receptores Virais/metabolismo , Sarcoma Aviário/metabolismo , Sarcoma Aviário/virologia
5.
Retrovirology ; 8: 96, 2011 Nov 18.
Artigo em Inglês | MEDLINE | ID: mdl-22099981

RESUMO

BACKGROUND: Understanding the mechanism by which viruses enter their target cell is an essential part of understanding their infectious cycle. Previous studies have focussed on the multiplicity of viral envelope proteins that need to bind to their cognate receptor to initiate entry. Avian sarcoma and leukosis virus Envelope protein (ASLV Env) mediates entry via a receptor, Tva, which can be attached to the cell surface either by a phospholipid anchor (Tva800) or a transmembrane domain (Tva950). In these studies, we have now investigated the number of target receptors necessary for entry of ASLV Env-pseudotyped virions. RESULTS: Using titration and modelling experiments we provide evidence that binding of more than one receptor, probably two, is needed for entry of virions via Tva800. However, binding of just one Tva950 receptor is sufficient for successful entry. CONCLUSIONS: The different modes of attachment of Tva800 and Tva950 to the cell membrane have important implications for the utilisation of these proteins as receptors for viral binding and/or uptake.


Assuntos
Alpharetrovirus/fisiologia , Proteínas Aviárias/metabolismo , Receptores Virais/metabolismo , Ligação Viral , Internalização do Vírus , Animais , Linhagem Celular , Membrana Celular/metabolismo , Células HEK293 , Humanos , Camundongos , Modelos Biológicos , Proteínas do Envelope Viral/metabolismo
6.
J Virol ; 84(13): 6626-35, 2010 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-20410274

RESUMO

Accidental insertional activation of proto-oncogenes and potential vector mobilization pose serious challenges for human gene therapy using retroviral vectors. Comparative analyses of integration sites of different retroviral vectors have elucidated distinct target site preferences, highlighting vectors based on the alpharetrovirus Rous sarcoma virus (RSV) as those with the most neutral integration spectrum. To date, alpharetroviral vector systems are based mainly on single constructs containing viral coding sequences and intact long terminal repeats (LTR). Even though they are considered to be replication incompetent in mammalian cells, the transfer of intact viral genomes is unacceptable for clinical applications, due to the risk of vector mobilization and the potentially immunogenic expression of viral proteins, which we minimized by setting up a split-packaging system expressing the necessary viral proteins in trans. Moreover, intact LTRs containing transcriptional elements are capable of activating cellular genes. By removing most of these transcriptional elements, we were able to generate a self-inactivating (SIN) alpharetroviral vector, whose LTR transcriptional activity is strongly reduced and whose transgene expression can be driven by an internal promoter of choice. Codon optimization of the alpharetroviral Gag/Pol expression construct and further optimization steps allowed the production of high-titer self-inactivating vector particles in human cells. We demonstrate proof of principle for the versatility of alpharetroviral SIN vectors for the genetic modification of murine and human hematopoietic cells at a low multiplicity of infection.


Assuntos
Alpharetrovirus/fisiologia , Terapia Genética/métodos , Vetores Genéticos , Sequências Repetidas Terminais/genética , Transgenes , Montagem de Vírus , Alpharetrovirus/genética , Animais , Linhagem Celular , Células Cultivadas , Expressão Gênica , Humanos , Camundongos , Regiões Promotoras Genéticas , RNA Viral/genética
7.
Virology ; 392(1): 94-102, 2009 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-19638332

RESUMO

Integrase (IN) is the enzyme responsible for provirus integration of retroviruses into the host cell genome. We used an Avian Sarcoma and Leukemia Viruses (ASLV) integration assay to investigate the way in which IN integrates substrates mutated or devoid of one or both IN recognition sequences. We found that replacing U5 by non-viral sequences (U5del) or U3 by a mutated sequence (pseudoU3) resulted in two and three fold reduction of two-ended integration (integration of the two ends from a donor DNA) respectively, but had a slight effect on concerted integration (integration of both ends at the same site of target DNA). Further, IN was still able to integrate the viral ends of the double mutant (pseudoU3/U5del) in a two-ended and concerted integration reaction. However, efficiency and accuracy (i.e. fidelity of size duplication and of end cleavage) of integration were reduced.


Assuntos
Alpharetrovirus/genética , Alpharetrovirus/fisiologia , Integrases/genética , Integrases/fisiologia , Integração Viral/genética , Integração Viral/fisiologia , Animais , Sequência de Bases , Sítios de Ligação/genética , Primers do DNA/genética , DNA Viral/genética , DNA Viral/metabolismo , Genes Virais , Técnicas In Vitro , Modelos Biológicos , Mutação , Deleção de Sequência
8.
J Virol ; 82(22): 11419-28, 2008 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-18768966

RESUMO

We recently identified and cloned the receptor for subgroup C avian sarcoma and leukosis viruses [ASLV(C)], i.e., Tvc, a protein most closely related to mammalian butyrophilins, which are members of the immunoglobulin protein family. The extracellular domain of Tvc contains two immunoglobulin-like domains, IgV and IgC, which presumably each contain a disulfide bond important for native function of the protein. In this study, we have begun to identify the functional determinants of Tvc responsible for ASLV(C) receptor activity. We found that the IgV domain of the Tvc receptor is responsible for interacting with the glycoprotein of ASLV(C). Additional experiments demonstrated that a domain was necessary as a spacer between the IgV domain and the membrane-spanning domain for efficient Tvc receptor activity, most likely to orient the IgV domain a proper distance from the cell membrane. The effects on ASLV(C) glycoprotein binding and infection efficiency were also studied by site-directed mutagenesis of the cysteine residues of Tvc as well as conserved amino acid residues of the IgV Tvc domain compared to other IgV domains. In this initial analysis of Tvc determinants important for interacting with ASLV(C) glycoproteins, at least two aromatic amino acid residues in the IgV domain of Tvc, Trp-48 and Tyr-105, were identified as critical for efficient ASLV(C) infection. Interestingly, one or more aromatic amino acid residues have been identified as critical determinants in the other ASLV(A-E) receptors for a proper interaction with ASLV glycoproteins. This suggests that the ASLV glycoproteins may share a common mechanism of receptor interaction with an aromatic residue(s) on the receptor critical for triggering conformational changes in SU that initiate the fusion process required for efficient virus infection.


Assuntos
Alpharetrovirus/fisiologia , Membrana Celular/química , Mapeamento de Interação de Proteínas , Receptores Virais/metabolismo , Ligação Viral , Sequência de Aminoácidos , Substituição de Aminoácidos/genética , Animais , Linhagem Celular , Embrião de Galinha , Galinhas , Dados de Sequência Molecular , Mutagênese Sítio-Dirigida , Ligação Proteica , Estrutura Terciária de Proteína , Receptores Virais/genética , Alinhamento de Sequência , Proteínas do Envelope Viral/metabolismo
9.
Virus Res ; 135(1): 72-82, 2008 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-18420298

RESUMO

During retroviral integration, the viral integrase recognizes the attachment (att) sequence (formed by juxtaposition of two LTRs ends) as the substrate of integration. We have developed a self-deleting Avian Leukosis and Sarcoma Viruses (ALSVs)-based retroviral vector carrying an additional copy of the att sequence, between neo and puro genes. We observed that: (i) the resulting NP3Catt vector was produced at neo and puro titers respectively smaller and higher than that of the parental vector devoid of the att sequence; (ii) 61% of NP3Catt proviruses were flanked by LTRs; most of them were deleted of internal sequences, probably during the reverse transcription step; (iii) 31% of clones were deleted of the whole 5' part of their genome and were flanked, in 5', by the additional att sequence and, in 3', by an LTR. Integration of these last proviruses was often imprecise with respect to the viral ends. At total, 77% of proviruses had lost the packaging signal and were not mobilizable by a replication-competent virus and 92% had lost the selectable gene in a single round of replication. Although still to improve, the att vector could be considered as an interesting new safe retroviral vector for gene transfer experiments.


Assuntos
Alpharetrovirus/enzimologia , Alpharetrovirus/genética , Vetores Genéticos/genética , Integrases/metabolismo , Deleção de Sequência , Integração Viral , Alpharetrovirus/fisiologia , Animais , Sequência de Bases , Linhagem Celular , Técnicas de Transferência de Genes , Vetores Genéticos/química , Integrases/genética , Provírus/enzimologia , Provírus/genética , Provírus/fisiologia , Codorniz , RNA Viral/química , RNA Viral/genética , Sequências Repetidas Terminais , Proteínas Virais/genética , Proteínas Virais/metabolismo , Replicação Viral
10.
J Virol ; 80(2): 562-70, 2006 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-16378958

RESUMO

The ability of many retroviruses to cause disease can be correlated to their cytopathic effect (CPE) in tissue culture characterized by an acute period of cell death and viral DNA accumulation. Here, we show that mutants of a subgroup B avian retrovirus (Alpharetrovirus) cause a very dramatic CPE in certain susceptible avian cells that is coincident with elevated levels of apoptosis, as measured by nuclear morphology, and persistent viral DNA accumulation. These mutants also have a broadly extended host range that includes rodent, cat, dog, monkey, and human cells (31). Previously, we have shown that the mutants exhibit diminished resistance to superinfection. The results presented here have important implications for the process of evolution of retroviruses to use distinct cellular receptors.


Assuntos
Alpharetrovirus/fisiologia , Alpharetrovirus/patogenicidade , Infecções por Retroviridae/virologia , Infecções Tumorais por Vírus/virologia , Alpharetrovirus/genética , Animais , Apoptose , Evolução Biológica , Linhagem Celular , Núcleo Celular/metabolismo , Efeito Citopatogênico Viral , DNA Viral/metabolismo , Mutação , Virulência , Replicação Viral
11.
J Virol ; 79(18): 12035-44, 2005 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-16140779

RESUMO

We have analyzed the placement of sites of integration of avian sarcoma-leukosis virus (ASLV) and human immunodeficiency virus (HIV) DNA in the draft chicken genome sequence, with the goals of assessing species-specific effects on integration and allowing comparison to the distribution of chicken endogenous retroviruses (ERVs). We infected chicken embryo fibroblasts (CEF) with ASLV or HIV and sequenced 863 junctions between host and viral DNA. The relationship with cellular gene activity was analyzed by transcriptional profiling of uninfected or ASLV-infected CEF cells. ASLV weakly favored integration in active transcription units (TUs), and HIV strongly favored active TUs, trends seen previously for integration in human cells. The ERVs, in contrast, accumulated mostly outside TUs, including ERVs related to ASLV. The minority of ERVs present within TUs were mainly in the antisense orientation; consequently, the viral splicing and polyadenylation signals would not disrupt cellular mRNA synthesis. In contrast, de novo ASLV integration sites within TUs showed no orientation bias. Comparing the distribution of de novo ASLV integration sites to ERVs indicated that purifying selection against gene disruption, and not initial integration targeting, probably determined the ERV distribution. Further analysis indicated that ERVs in humans, mice, and rats showed similar distributions, suggesting purifying selection dictated their distributions as well.


Assuntos
Alpharetrovirus/genética , Alpharetrovirus/fisiologia , Galinhas/genética , Galinhas/virologia , HIV-1/genética , HIV-1/fisiologia , Integração Viral/genética , Alpharetrovirus/patogenicidade , Animais , Sítios de Ligação/genética , Cromossomos/genética , Cromossomos/virologia , Clonagem Molecular , DNA/genética , Retrovirus Endógenos/genética , Retrovirus Endógenos/patogenicidade , Retrovirus Endógenos/fisiologia , Genoma , HIV-1/patogenicidade , Humanos , Camundongos , Dados de Sequência Molecular , Ratos , Transcrição Gênica
12.
Virology ; 318(2): 566-81, 2004 Jan 20.
Artigo em Inglês | MEDLINE | ID: mdl-14972525

RESUMO

During replicative cycle of retroviruses, the reverse-transcribed viral DNA is integrated into the cell DNA by the viral integrase (IN) enzyme. The central core domain of IN contains the catalytic site of the enzyme and is involved in binding viral ends and cell DNA as well as dimerization. We previously performed single amino acid substitutions in the core domain of an Avian Leukemia and Sarcoma Virus (ALSV) IN [Arch. Virol. 147 (2002) 1761]. Here, we modeled the resulting IN mutants and analyzed the ability of these mutants to mediate concerted DNA integration in an in vitro assay, and to form dimers by protein-protein cross-linking and size exclusion chromatography. The N197C mutation resulted in the inability of the mutant to perform concerted integration that was concomitant with a loss of IN dimerization. Surprisingly, mutations Q102G and A106V at the dimer interface resulted in mutants with higher efficiencies than the wild-type IN in performing two-ended concerted integration of viral DNA ends. The G139D and A195V mutants had a trend to perform one-ended DNA integration of viral ends instead of two-ended integration. More drastically, the I88L and L135G mutants preferentially mediated nonconcerted DNA integration although the proteins form dimers. Therefore, these mutations may alter the formation of IN complexes of higher molecular size than a dimer that would be required for concerted integration. This study points to the important role of core domain residues in the concerted integration of viral DNA ends as well as in the oligomerization of the enzyme.


Assuntos
Alpharetrovirus/fisiologia , Integrases/metabolismo , Integração Viral , Replicação Viral , Alpharetrovirus/enzimologia , Sequência de Aminoácidos , Domínio Catalítico/genética , Integrases/genética , Modelos Moleculares , Dados de Sequência Molecular , Mutação
13.
J Virol ; 77(3): 1977-83, 2003 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-12525631

RESUMO

The receptor "priming" model for entry of the retrovirus avian sarcoma and leukosis virus (ASLV) predicts that upon binding cell surface receptors, virions are endocytosed and trafficked to acidic endosomes where fusion occurs. To test this model directly, we have now followed subgroup A ASLV (ASLV-A) virions entering cells via either the transmembrane (TVA950) or glycophosphatidylinositol (GPI)-anchored (TVA800) forms of the cellular receptor. Our results suggest that viruses entering via these two forms of receptor are subjected to different intracellular fates, perhaps due to use of different endocytic trafficking pathways to access acidic fusion compartments. Kinetic analyses demonstrated that virus bound to TVA800 was taken up from the cell surface more slowly but then trafficked to the site of fusion more quickly than that entering via TVA950. Furthermore, transiently arresting virions within putative fusion compartments with NH4Cl led to a substantially greater decrease in the infectivity of virions using TVA950 than with those using TVA800. The increased infectivity of virions using TVA800 correlated with the localization of this receptor to lipid rafts, since this effect was abolished by pharmacological disruption of lipid rafts. Together these results suggest that, in the presence of NH4Cl, virus bound to the GPI-anchored receptor may utilize a lipid raft-dependent pathway to accumulate within a fusion compartment where it is more stable than if it enters via the transmembrane receptor. The TVA800/ASLV-A system should prove useful for the molecular analysis of lipid raft-dependent endocytosis and may provide a tool for the biochemical dissection of the poorly understood uncoating step of retroviral replication.


Assuntos
Alpharetrovirus/fisiologia , Microdomínios da Membrana/fisiologia , Receptores Virais/fisiologia , Cloreto de Amônio/farmacologia , Linhagem Celular , Ciclodextrinas/farmacologia , Endocitose , Humanos , Vírion/fisiologia
14.
Blood ; 101(3): 1103-10, 2003 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-12393697

RESUMO

Acute chicken leukemia retroviruses, because of their capacity to readily transform hematopoietic cells in vitro, are ideal models to study the mechanisms governing the cell-type specificity of oncoproteins. Here we analyzed the transformation specificity of 2 acute chicken leukemia retroviruses, the Myb-Ets- encoding E26 virus and the ErbA/ErbB-encoding avian erythroblastosis virus (AEV). While cells transformed by E26 are multipotent (designated "MEP" cells), those transformed by AEV resemble erythroblasts. Using antibodies to separate subpopulations of precirculation yolk sac cells, both viruses were found to induce the proliferation of primitive erythroid progenitors within 2 days of infection. However, while AEV induced a block in differentiation of the cells, E26 induced a gradual shift in their phenotype and the acquisition of the potential for multilineage differentiation. These results suggest that the Myb-Ets oncoprotein of the E26 leukemia virus converts primitive erythroid cells into proliferating definitive-type multipotent hematopoietic progenitors.


Assuntos
Alpharetrovirus/fisiologia , Transformação Celular Viral , Células Precursoras Eritroides/virologia , Células-Tronco Multipotentes/virologia , Células-Tronco Neoplásicas/virologia , Animais , Vírus da Mieloblastose Aviária/fisiologia , Blastoderma/patologia , Blastoderma/virologia , Diferenciação Celular , Linhagem da Célula , Separação Celular/métodos , Embrião de Galinha , Células Precursoras Eritroides/patologia , Células-Tronco Hematopoéticas/patologia , Células-Tronco Hematopoéticas/virologia , Imunofenotipagem , Células-Tronco Multipotentes/patologia , Células-Tronco Neoplásicas/patologia
15.
J Gen Virol ; 83(Pt 10): 2553-2561, 2002 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-12237439

RESUMO

Among the six envelope subgroups of avian leukosis virus (ALV) that infect chickens, subgroups A (ALV-A) and J (ALV-J) are the most pathogenic and widespread among commercial chicken populations. While ALV-A is predominantly associated with lymphoid leukosis (LL) and less frequently with erythroblastosis (EB), ALV-J mainly induces tumours of the myeloid lineage. In order to examine the basis for the lineage specificity of tumour induction by these two ALV subgroups, we constructed two chimeric viruses by substituting the env genes into the reciprocal proviral clones. The chimeric HPRS-103(A) virus carrying the subgroup A env gene is identical to ALV-J prototype virus HPRS-103 except for the env gene, and the chimeric RCAS(J) virus carrying the subgroup J env gene is identical to the parent replication-competent ALV-A vector RCAS except for the env gene. In experimentally inoculated chickens, HPRS-103(A) virus induced LL and EB similar to ALV-A isolates such as RAV-1, while RCAS(J) virus induced myeloid leukosis (ML) and EB, similar to ALV-J, suggesting that the env gene is the major determinant for the lineage-specific oncogenicity. There were genetic differences in susceptibility to tumour induction between line 0 and line 15(I) chickens, indicating that in addition to the env gene, other viral or host factors could also serve as determinants for oncogenicity. Induction of both LL and ML by the two chimeric viruses occurred through the activation of c-myc, while the EB tumours were induced by activation of the c-erbB oncogene.


Assuntos
Alpharetrovirus/fisiologia , Vírus da Leucose Aviária/fisiologia , Leucose Aviária/virologia , Vírus da Mieloblastose Aviária/fisiologia , Leucemia Linfoide/virologia , Leucemia Mieloide/virologia , Proteínas do Envelope Viral/fisiologia , Alpharetrovirus/genética , Animais , Vírus da Leucose Aviária/genética , Vírus da Mieloblastose Aviária/genética , Sequência de Bases , Linhagem da Célula , Embrião de Galinha , DNA Viral , Rearranjo Gênico , Genes myc , Linfócitos , Dados de Sequência Molecular , Células Mieloides , Recombinação Genética , Proteínas do Envelope Viral/genética , Integração Viral
16.
J Soc Biol ; 195(2): 107-13, 2001.
Artigo em Francês | MEDLINE | ID: mdl-11723821

RESUMO

The retinal pigment epithelium (RPE) develops from the same sheet of neuroepithelium as the neuroretina. When infected with MC29, a v-myc expressing virus, the RPE cells can be induced to transdifferentiate and to take a neuroretinal epithelium fate. After a PCR-based differential screening from these cells we have identified three genes of interest. Qath5, a quail basic helix-loop-helix (bHLH) gene that is closely related to the Drosophila atonal, and whose expression is found in the developing neuroretina. A Chx10-related homeobox gene also expressed in the developing neuroretina and HuD, a RNA-binding protein not expressed in the RPE but expressed during neurogenesis. Beside these genes whose function is involved in regulating neuronal differentiation myc also induced a transient Mitf expression. Mitf is expressed in the entire optic cup, later restricted to the pigmented retina. Mitf is involved in the regulation of the pigmented differentiation. We conclude that v-myc can reverse the RPE to the bipotential retinal primordia.


Assuntos
Alpharetrovirus/fisiologia , Proteínas do Olho/biossíntese , Genes myc , Substâncias de Crescimento , Proteína Oncogênica p55(v-myc)/fisiologia , Epitélio Pigmentado Ocular/embriologia , Proteínas de Peixe-Zebra , Alpharetrovirus/genética , Diferenciação Celular , Transformação Celular Viral/genética , Proteínas de Ligação a DNA/biossíntese , Proteínas de Ligação a DNA/genética , Proteínas ELAV , Olho/embriologia , Proteínas do Olho/genética , Perfilação da Expressão Gênica , Regulação da Expressão Gênica no Desenvolvimento , Genes Homeobox , Sequências Hélice-Alça-Hélice , Proteínas de Homeodomínio/biossíntese , Proteínas de Homeodomínio/genética , Fator de Transcrição Associado à Microftalmia , Proteínas do Tecido Nervoso/biossíntese , Proteínas do Tecido Nervoso/genética , Epitélio Pigmentado Ocular/citologia , Reação em Cadeia da Polimerase , Proteínas de Ligação a RNA/biossíntese , Proteínas de Ligação a RNA/genética , Fatores de Transcrição/biossíntese , Fatores de Transcrição/genética
17.
Oncogene ; 19(55): 6472-81, 2000 Dec 18.
Artigo em Inglês | MEDLINE | ID: mdl-11175363

RESUMO

Studies of retroviral-induced oncogenesis in animal systems led to the initial discovery of viral oncogenes and their cellular homologs, and provided critical insights into their role in the neoplastic process. V-ets, the founding member of the ETS oncogene family, was originally identified as part of the fusion oncogene encoded by the avian acute leukemia virus E26 and subsequent analysis of virus induced leukemias led to the initial isolation of two other members of the ETS gene family. PU.1 was identified as a target of insertional activation in the majority of tumors induced by the murine Spleen Focus Forming virus (SFFV), while fli-1 proved to be the target of Friend murine leukemia virus (F-MuLV) in F-MuLV induced erythroleukemia, as well as that of the 10A1 and Graffi viruses. The common features of the erythroid and myeloid diseases induced by these viruses provided the initial demonstration that these and other members of the ETS family play important roles in hematopoietic development as well as disease. This review provides an overview of the role of ETS genes in retrovirally induced neoplasia, their possible mechanisms of action, and how these viral studies relate to current knowledge of the functions of these genes in hematopoiesis.


Assuntos
Transformação Celular Viral/genética , Regulação Neoplásica da Expressão Gênica , Regulação Viral da Expressão Gênica , Família Multigênica , Oncogenes , Proteínas Proto-Oncogênicas , Retroviridae/genética , Células 3T3 , Alpharetrovirus/genética , Alpharetrovirus/fisiologia , Animais , Vírus da Mieloblastose Aviária/genética , Vírus da Mieloblastose Aviária/fisiologia , Galinhas , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/fisiologia , Fibroblastos/metabolismo , Hematopoese/genética , Humanos , Vírus da Leucemia Murina/genética , Vírus da Leucemia Murina/fisiologia , Camundongos , Mutagênese Insercional , Proteína Proto-Oncogênica c-fli-1 , Provírus/genética , Retroviridae/fisiologia , Vírus Formadores de Foco no Baço/genética , Vírus Formadores de Foco no Baço/fisiologia , Transativadores/genética , Transativadores/fisiologia
18.
J Virol ; 74(1): 456-64, 2000 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-10590135

RESUMO

We previously identified a 160-nucleotide packaging signal, MPsi, from the 5' end of the Rous sarcoma virus genome. In this study, we determine the secondary structure of MPsi by using phylogenetic analysis with computer modeling and heterologous packaging assays of point mutants. The results of the in vivo studies are in good agreement with the computer model. Additionally, the packaging studies indicate several structures which are important for efficient packaging, including a single-stranded bulge containing the initiation codon for the short open reading frame, uORF3, as well as adjacent stem structures. Finally, we show that the L3 stem-loop at the 3' end of MPsi is dispensable for packaging, thus identifying an 82-nucleotide minimal packaging signal, microPsi, composed of the O3 stem-loop.


Assuntos
Alpharetrovirus/fisiologia , Conformação de Ácido Nucleico , RNA Viral/química , Montagem de Vírus , Alpharetrovirus/genética , Sequência de Bases , Dados de Sequência Molecular , Mutagênese Sítio-Dirigida , Fases de Leitura Aberta , Filogenia , RNA Viral/genética , Homologia de Sequência do Ácido Nucleico
19.
Virology ; 248(2): 305-11, 1998 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-9721239

RESUMO

The lack of a well-behaved permanent, adherent, nontransformed chicken cell line has made some experiments with avian leukosis-sarcoma viruses (ASLV) and vectors considerably more difficult. The EV-O-derived line, DF-1, supports the efficient replication of subgroups (A), (B), and (C) ASLV, as well as amphotrophic murine leukemia virus and an ASLV-derived vector that has its env gene derived from the env gene from an amphotrophic murine leukemia virus. The cell line responds appropriately to the expression of a transforming oncogene (v-myc) to a growth suppressor gene [p21(waf1)] and can be sorted (using FACS) if infected by an ASLV vector that expresses GFP.


Assuntos
Alpharetrovirus/fisiologia , Linhagem Celular/virologia , Replicação Viral , Animais , Transformação Celular Neoplásica , Galinhas , Inibidor de Quinase Dependente de Ciclina p21 , Ciclinas/fisiologia , Genes myc/fisiologia , Vetores Genéticos , Vírus da Leucemia Murina/fisiologia , Oncogenes/fisiologia , Proteínas do Envelope Viral/fisiologia
20.
J Vet Med Sci ; 57(5): 8O1-11, 1995 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-8593284

RESUMO

Human and animal herpesviruses are able to stimulate gene expression of various human and animal retroviruses and the activation appears to be a common theme. It is of interest why the retroviruses can be commonly activated by herpesviruses despite clear diversities in each mechanism for retrovirus gene expression. In this review, three typical examples of the interactions; human immunodeficiency virus and human herpesviruses, avian retroviruses and Marek's disease virus, and feline immunodeficiency virus and feline herpesvirus type I will be given to respond the above question on the basis of the comparative study of molecular interactions.


Assuntos
Herpesviridae/genética , Herpesviridae/fisiologia , Retroviridae/genética , Retroviridae/fisiologia , Alpharetrovirus/genética , Alpharetrovirus/fisiologia , Animais , Aves , Gatos , Regulação Viral da Expressão Gênica , HIV/genética , HIV/fisiologia , Herpesvirus Galináceo 2/genética , Herpesvirus Galináceo 2/fisiologia , Humanos , Vírus da Imunodeficiência Felina/genética , Vírus da Imunodeficiência Felina/fisiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA