Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 133
Filtrar
1.
J Virol ; 84(4): 1874-80, 2010 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-20007266

RESUMO

The human retrovirus XMRV (xenotropic murine leukemia virus-related virus) is associated with prostate cancer, but a causal relationship has not been established. Here, we have used cultured fibroblast and epithelial cell lines to test the hypothesis that XMRV might have direct transforming activity but found only rare transformation events, suggestive of indirect transformation, even when the target cells expressed the human Xpr1 cell entry receptor for XMRV. Characterization of cells from three transformed foci showed that all were infected with and produced XMRV, and one produced a highly active transforming virus, presumably generated by recombination between XMRV and host cell nucleic acids. Given the sequence similarity of XMRV to mink cell focus-forming (MCF) viruses and the enhanced leukemogenic activity of the latter, we tested XMRV for related MCF-like cytopathic activities in cultured mink cells but found none. These results indicate that XMRV has no direct transforming activity but can activate endogenous oncogenes, resulting in cell transformation. As part of these experiments, we show that XMRV can infect and be produced at a high titer from human HT-1080 fibrosarcoma cells that express TRIM5alpha (Ref1), showing that XMRV is resistant to TRIM5alpha restriction. In addition, XMRV poorly infects NIH 3T3 cells expressing human Xpr1 but relatively efficiently infects BALB 3T3 cells expressing human Xpr1, showing that XMRV is a B-tropic virus and that its infectivity is regulated by the Fv1 mouse locus.


Assuntos
Neoplasias da Próstata/virologia , Retroviridae/patogenicidade , Animais , Fatores de Restrição Antivirais , Proteínas de Transporte/genética , Proteínas de Transporte/fisiologia , Linhagem Celular , Linhagem Celular Tumoral , Transformação Celular Neoplásica , Transformação Celular Viral , Efeito Citopatogênico Viral , Cães , Humanos , Vírus da Leucemia Murina/patogenicidade , Masculino , Camundongos , Vírus Indutores de Focos em Células do Vison/patogenicidade , Células NIH 3T3 , Neoplasias da Próstata/genética , Neoplasias da Próstata/fisiopatologia , Ratos , Receptores Acoplados a Proteínas G/genética , Receptores Acoplados a Proteínas G/fisiologia , Receptores Virais/genética , Receptores Virais/fisiologia , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Especificidade da Espécie , Proteínas com Motivo Tripartido , Ubiquitina-Proteína Ligases , Receptor do Retrovírus Politrópico e Xenotrópico
2.
J Virol ; 78(21): 12071-4, 2004 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-15479849

RESUMO

We previously demonstrated that mink cells undergo apoptosis after MCF13 murine leukemia virus (MLV) infection. In this study, we observed that virus-infected mink epithelial cells had significantly larger amounts of steady-state levels of MCF13 MLV envelope precursor protein (gPr80(env)) than did Mus dunni fibroblasts, which are resistant to virus-induced cytopathicity. Infection of mink cells with the noncytopathic NZB-9 MLV did not result in the accumulation of gPr80(env). MCF13 MLV infection of mink cells produced low cell surface expression of envelope glycoprotein and less efficient spread of infectious virus. Western blot analysis of mink epithelial cells infected with MCF13 MLV showed an increase in GRP78/BiP, which was not observed for either mink cells infected with NZB-9 MLV or M. dunni fibroblasts infected with MCF13 MLV. MCF13 MLV infection of mink cells also resulted in a significant upregulation of CHOP/GADD153. These results indicate that the accumulation of MCF13 MLV gPr80(env) triggers endoplasmic reticulum stress, which may mediate apoptosis in mink epithelial cells.


Assuntos
Apoptose , Retículo Endoplasmático/fisiologia , Vírus Indutores de Focos em Células do Vison/patogenicidade , Animais , Proteínas Estimuladoras de Ligação a CCAAT/análise , Chaperona BiP do Retículo Endoplasmático , Células Epiteliais/patologia , Produtos do Gene env/metabolismo , Proteínas de Choque Térmico/análise , Vison , Chaperonas Moleculares/análise , Fator de Transcrição CHOP , Fatores de Transcrição/análise
3.
J Virol ; 78(10): 5088-96, 2004 May.
Artigo em Inglês | MEDLINE | ID: mdl-15113890

RESUMO

Upon inoculation into AKR mice, mink cell focus-forming murine leukemia virus (MCF MLV) accelerates thymic lymphoma formation. During the preleukemic phase of disease, we observed the induction of apoptosis in thymic lymphocytes. A similar induction of apoptosis was observed for cultured mink epithelial cells after MCF13 MLV infection. In this study, the relevance of viral pathogenicity to cell killing was determined by testing the susceptibility of various cell types from different species to lymphomagenic MLVs. We observed that the cytopathic effect of lymphomagenic MLVs was restricted to mink cells. Southern blot analysis of MLV-infected cells revealed an accumulation of the linear form of unintegrated viral DNA, particularly in mink cells after MCF13 MLV infection. Thus, a strong correlation was observed between viral superinfection, which results in the accumulation of high levels of unintegrated viral DNA, and cell killing. Immunoblot analysis for MCF13 MLV-infected mink epithelial cells did not show a significant change in total p53 levels or its phosphorylated form at Ser-15 compared with that in mock-treated cells. Moreover, a time course analysis for mink epithelial cells infected with MCF13 MLV did not reveal mitochondrial depolarization or a significant change in Bax levels. These results demonstrate that MCF13 MLV induces apoptosis preferentially in cells in which superinfection occurs, and the mechanism involved is independent of p53 activation and mitochondrial damage.


Assuntos
Apoptose , Dano ao DNA , Vírus Indutores de Focos em Células do Vison/patogenicidade , Mitocôndrias/fisiologia , Proteína Supressora de Tumor p53/fisiologia , Animais , Camundongos , Vison , Células NIH 3T3
4.
Viral Immunol ; 16(2): 203-12, 2003.
Artigo em Inglês | MEDLINE | ID: mdl-12828871

RESUMO

In genetically susceptible strains, such as C57BL/6 (B6) mice, LP-BM5 causes murine AIDS (MAIDS). LP-BM5 is a complex mixture of murine leukemia viruses (MuLV) that includes replication competent ecotropic (BM5eco) and mink cell focus inducing (MCF), and replication defective (BM5d) MuLV. At present, for the BM5eco virus, sequence information on only the gag region is available. In this paper, we describe for the first time the sequencing of the entire BM5eco viral genome as well as analysis of homology with two other previously sequenced and well-characterized MuLVs, Emv-11 and Emv-2, the latter constituting the parental virus for BM5eco. We propose that the detailed sequence comparisons herein provide cogent evidence that BM5eco utilizes variations in cytotoxic T lymphocytes (CTL) epitopes as an immune escape mechanism. This CTL evasion mechanism may contribute substantially to the underlying prototypic susceptibility of B6 mice to LP-BM5-induced MAIDS.


Assuntos
Vírus Auxiliares , Epitopos Imunodominantes/genética , Vírus da Leucemia Murina/genética , Síndrome de Imunodeficiência Adquirida Murina/virologia , Seleção Genética , Linfócitos T Citotóxicos/imunologia , Sequência de Aminoácidos , Animais , Citotoxicidade Imunológica , Vírus Defeituosos , Vírus Auxiliares/genética , Vírus Auxiliares/imunologia , Vírus da Leucemia Murina/imunologia , Camundongos , Vírus Indutores de Focos em Células do Vison , Dados de Sequência Molecular , Análise de Sequência de DNA
5.
Artigo em Inglês | MEDLINE | ID: mdl-12675894

RESUMO

Between late December 1999 and late April 2000, three locally bred Friesian calves (ageing 25, 28 and 35 days) in a dairy farm, at Al-Ahsa locality of the eastern region of Saudi Arabia showed dullness and inappetence. Their rectal temperatures ranged between 41 and 41.5 degrees C. One to 2 days later and onwards, the calves showed lacrimation, nasal discharge, salivation, oedema of the head, conjunctivitis, exo-ophthalmia and corneal opacity. One calf showed diarrhoea. The superficial lymph nodes were oedematous and swollen. The calves died after 7, 5 and 8 days, respectively, following the onset of the disease. Calves and rabbits were experimentally infected with materials from the naturally infected calves. The rabbits showed fever, mild conjunctivitis and one rabbit showed wet faeces. The experimentally inoculated calves showed rise in temperature and mild symptoms but none of them died. The virus from the naturally infected calves and from the experimentally infected rabbits was identified as malignant catarrhal fever (MCF) virus using both the complement fixation test and the fluorescent antibody test, employing a reference anti-serum against the WC 11 strain of MCF virus. Serological survey for MCF antibodies in cattle, sheep and goats from the affected farm revealed that 54% of the examined animals were positive. The situation of MCF in Saudi Arabia was discussed in relation to sheep and wild game. This paper constitutes the first report of MCF in Saudi Arabia and the Gulf region.


Assuntos
Anticorpos Antivirais/sangue , Herpesviridae/imunologia , Febre Catarral Maligna/epidemiologia , Vírus Indutores de Focos em Células do Vison/imunologia , Animais , Animais Recém-Nascidos , Bovinos , Testes de Fixação de Complemento/veterinária , Imunofluorescência/veterinária , Cabras , Herpesviridae/isolamento & purificação , Febre Catarral Maligna/etiologia , Febre Catarral Maligna/patologia , Vírus Indutores de Focos em Células do Vison/isolamento & purificação , Coelhos , Arábia Saudita/epidemiologia , Ovinos
6.
J Virol ; 77(6): 3460-9, 2003 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-12610121

RESUMO

During ongoing C-type retrovirus infection, the probability of leukemia caused by insertional gene activation is markedly increased by the emergence of recombinant retroviruses that repeatedly infect host cells. The murine mink cell focus-inducing (MCF) viruses with this property have acquired characteristic changes in the N-terminal domain of their envelope glycoprotein that specify binding to a different receptor than the parental ecotropic virus. In this report, we show that MCF virus infection occurs through binding to this receptor (termed Syg1) and, remarkably, by a second mechanism that does not utilize the Syg1 receptor. By the latter route, the N-terminal domain of the ecotropic virus glycoprotein expressed on the cell surface in a complex with its receptor activates the fusion mechanism of the MCF virus in trans. The rate of MCF virus spread through a population of permissive human cells was increased by establishment of trans activation, indicating that Syg1 receptor-dependent and -independent pathways function in parallel. Also, trans activation shortened the interval between initial infection and onset of cell-cell fusion associated with repeated infection of the same cell. Our findings indicate that pathogenic retrovirus infection may be initiated by virus binding to cell receptors or to the virus envelope glycoprotein of other viruses expressed on the cell surface. Also, they support a broader principle: that cooperative virus-virus interactions, as well as virus-host interactions, shape the composition and properties of the retrovirus quasispecies.


Assuntos
Fusão de Membrana , Vírus Indutores de Focos em Células do Vison/metabolismo , Vírus Indutores de Focos em Células do Vison/patogenicidade , Receptores Virais/metabolismo , Transdução de Sinais , Proteínas do Envelope Viral/metabolismo , Animais , Fusão Celular , Linhagem Celular , Cricetinae , Regulação Viral da Expressão Gênica , Humanos , Camundongos , Ativação Transcricional , Proteínas do Envelope Viral/química , Proteínas do Envelope Viral/genética , Receptor do Retrovírus Politrópico e Xenotrópico
7.
J Virol ; 77(5): 3326-33, 2003 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-12584358

RESUMO

The ability of mink cell focus-inducing (MCF) viruses to induce thymomas is determined, in part, by transcriptional enhancers in the U3 region of their long terminal repeats (LTRs). To elucidate sequence motifs important for enhancer function in vivo, we injected newborn mice with MCF 1dr (supF), a weakly pathogenic, molecularly tagged (supF) MCF virus containing only one copy of a sequence that is present as two copies (known as the directly repeated [DR] sequence) in the U3 region of MCF 247 and analyzed LTRs from supF-tagged proviruses in two resulting thymomas. Tagged proviruses integrated upstream and in the reverse transcriptional orientation relative to c-myc provided the focus of our studies. These proviruses are thought to contribute to thymoma induction by enhancer-mediated deregulation of c-myc expression. The U3 region in a tagged LTR in one thymoma was cloned and sequenced. Relative to MCF 1dr (supF), the cloned U3 region contained an insertion of 140 bp derived predominantly from the DR sequence of the injected virus. The inserted sequence contains predicted binding sites for transcription factors known to regulate the U3 regions of various murine leukemia viruses. Similar constellations of binding sites were duplicated in two proviral LTRs integrated upstream from c-myc in a second thymoma. We replaced the U3 sequences in an infectious molecular clone of MCF 247 with the cloned proviral U3 sequences from the first thymoma and generated an infectious chimeric virus, MCF ProEn. When injected into neonatal AKR mice, MCF ProEn was more pathogenic than the parental virus, MCF 1dr (supF), as evidenced by the more rapid onset and higher incidence of thymomas. Molecular analyses of the resultant thymomas indicated that the U3 region of MCF ProEn was genetically stable. These data suggest that the arrangement and/or redundancy of transcription factor binding sites generated by specific U3 sequence duplications are important to the biological events mediated by MCF proviruses integrated near c-myc that contribute to transformation.


Assuntos
Transformação Celular Viral , Elementos Facilitadores Genéticos/genética , Vírus Indutores de Focos em Células do Vison/patogenicidade , Sequências Repetidas Terminais/genética , Timoma/virologia , Neoplasias do Timo/virologia , Animais , Animais Recém-Nascidos , Sítios de Ligação , Regulação Viral da Expressão Gênica , Genes myc/genética , Camundongos , Camundongos Endogâmicos AKR , Vírus Indutores de Focos em Células do Vison/genética , Dados de Sequência Molecular , Recombinação Genética , Infecções por Retroviridae/virologia , Fatores de Transcrição/metabolismo , Infecções Tumorais por Vírus/virologia
8.
Gene Ther ; 9(4): 303-6, 2002 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-11896470

RESUMO

To achieve high transgene expression in the liver, we have compared the reporter gene expression among various murine retroviral long terminal repeats (LTRs) or leader sequences in vitro. Transient reporter gene expression assays revealed the highest gene expression by the polycythemic strain of spleen focus-forming virus (SFFVp) LTR in differentiated hepatocellular carcinoma cell lines, HuH-7 and PLC/PRF/5. However, remarkable difference was not observed among LTRs in other types of human liver tumor cell lines. Essentially the same results were obtained by infecting these cells with a series of retroviral vectors. Repression of transgene expression was observed by the leader sequences from Moloney murine leukemia virus (MoMLV), but not from mouse embryonic stem cell virus (MESV). Strengths of the promoters were further compared in murine hepatocytes in vivo. Although the proportions of genomic integration were almost the same, higher gene expression was observed by the FMEV-type vector, which contained the SFFVp LTR and the MESV leader, in comparison with that by the MoMLV-based vector. Thus, FMEV-type vectors may represent a novel type of vectors for human gene therapy with hepatocytes.


Assuntos
Vetores Genéticos , Hepatócitos/metabolismo , Retroviridae/genética , Transgenes , Animais , Expressão Gênica , Genes Reporter , Humanos , Camundongos , Vírus Indutores de Focos em Células do Vison/genética , Vírus da Leucemia Murina de Moloney/genética , Vírus Formadores de Foco no Baço/genética , Sequências Repetidas Terminais/genética , Células Tumorais Cultivadas
9.
J Virol ; 76(3): 1527-32, 2002 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-11773427

RESUMO

We have molecularly cloned a feline leukemia virus (FeLV) (clone 33) from a domestic cat with acute myeloid leukemia (AML). The long terminal repeat (LTR) of this virus, like the LTRs present in FeLV proviruses from other cats with AML, contains an unusual structure in its U3 region upstream of the enhancer (URE) consisting of three tandem direct repeats of 47 bp. To test the disease potential and specificity of this unique FeLV LTR, we replaced the U3 region of the LTR of the erythroleukemia-inducing Friend murine leukemia virus (F-MuLV) with that of FeLV clone 33. When the resulting virus, F33V, was injected into newborn mice, almost all of the mice eventually developed hematopoietic malignancies, with a significant percentage being in the myeloid lineage. This is in contrast to mice injected with an F-MuLV recombinant containing the U3 region of another FeLV that lacks repetitive URE sequences, none of which developed myeloid malignancies. Examination of tumor proviruses from F33V-infected mice failed to detect any changes in FeLV U3 sequences other than that in the URE. Like F-MuLV-infected mice, those infected with the F-MuLV/FeLV recombinants were able to generate and replicate mink cell focus-inducing viruses. Our studies are consistent with the idea that the presence of repetitive sequences upstream of the enhancer in the LTR of FeLV may favor the activation of this promoter in myeloid cells and contribute to the development of malignancies in this hematopoietic lineage.


Assuntos
DNA Viral , Vírus da Leucemia Murina de Friend/genética , Vírus da Leucemia Felina/genética , Sequências Repetidas Terminais , Animais , Sequência de Bases , Gatos , Linhagem Celular , Clonagem Molecular , Vírus da Leucemia Murina de Friend/fisiologia , Leucemia Eritroblástica Aguda/virologia , Leucemia Experimental/virologia , Leucemia Mieloide/virologia , Linfoma/virologia , Camundongos , Vírus Indutores de Focos em Células do Vison/genética , Dados de Sequência Molecular , Recombinação Genética , Retroviridae , Infecções por Retroviridae/virologia , Análise de Sequência de DNA , Homologia de Sequência do Ácido Nucleico , Infecções Tumorais por Vírus/virologia
10.
J Virol ; 76(1): 78-87, 2002 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-11739673

RESUMO

Mink cell focus-inducing (MCF) viruses induce T-cell lymphomas in AKR/J strain mice. MCF 247, the prototype of this group of nonacute murine leukemia viruses, transforms thymocytes, in part, by insertional mutagenesis and enhancer-mediated dysregulation of cellular proto-oncogenes. The unique 3' (U3) regions in the long terminal repeats of other murine leukemia viruses contain transcription factor binding sites known to be important for enhancer function and for the induction of T-cell lymphomas. Although transcription factor binding sites important for the biological properties of MCF 247 have not been identified, pathogenesis studies from our laboratory suggested to us that binding sites for Ikaros, a lymphoid-cell-restricted transcriptional regulator, affect the biological properties of MCF 247. In this report, we demonstrate that Ikaros binds to predicted sites in U3 sequences of MCF 247 and that site-directed mutations in these sites greatly diminish this binding in vitro. Consistent with these findings, ectopic expression of Ikaros in murine cells that do not normally express this protein significantly increases transcription from the viral promoter in transient gene expression assays. Moreover, site-directed mutations in specific Ikaros-binding sites reduce this activity in T-cell lines that express Ikaros endogenously. To determine whether the Ikaros-binding sites are functional in vivo, we inoculated newborn mice with a variant MCF virus containing a mutant Ikaros-binding site. The variant virus replicated in thymocytes less efficiently and induced lymphomas with a delayed onset compared to the wild-type virus. These data are consistent with the hypothesis that the Ikaros-binding sites in the U3 region of MCF 247 are functional and cooperate with other DNA elements for optimal enhancer function in vivo.


Assuntos
Proteínas de Ligação a DNA , Vírus Indutores de Focos em Células do Vison/patogenicidade , Fatores de Transcrição/fisiologia , Regiões 3' não Traduzidas/genética , Regiões 3' não Traduzidas/metabolismo , Células 3T3 , Animais , Animais Recém-Nascidos , Sítios de Ligação , Linhagem Celular , Humanos , Fator de Transcrição Ikaros , Células Jurkat , Leucemia Experimental/virologia , Camundongos , Camundongos Endogâmicos AKR , Vírus Indutores de Focos em Células do Vison/genética , Mutagênese Sítio-Dirigida , Ligação Proteica , Infecções por Retroviridae/virologia , Fatores de Transcrição/metabolismo , Transcrição Gênica , Infecções Tumorais por Vírus/virologia
11.
J Virol ; 75(13): 6007-15, 2001 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-11390602

RESUMO

Induction of apoptosis by different types of pathogenic retroviruses is an important step in disease development. We have observed that infection of thymic lymphocytes by the mink cell focus-forming murine leukemia virus (MCF MLV) during the preleukemic period resulted in an enhancement of apoptosis of these cells. To further study the ability of MCF MLVs to induce apoptosis and the role of this process in viral pathogenesis, we have developed an in vitro system of virus-induced apoptosis. MCF13 MLV infection of mink epithelial cells resulted in the production of cytopathic foci. In contrast, infection of mink cells with the 4070A amphotropic MLV did not produce any cytopathic effects. Staining of MCF13 MLV-infected cells with propidium iodide and annexin V-fluorescein isothiocyanate indicated that virus-induced cell death was due to apoptosis. At 6 days postinfection, the percentage of apoptotic MCF13 MLV-infected cells was 27% compared with 2 to 3% for mock- or amphotropic MLV-infected cells, representing a 9- to 14-fold difference. Assays for caspase-3 activation confirmed the detection by flow cytometry of apoptosis of MCF13 MLV-infected cells. Large amounts of unintegrated linear viral DNA were detectable by Southern blot analysis during the acute phase of infection, which indicated that MCF13 MLV is able to superinfect mink cells. Unintegrated viral DNA of only the linear form was detectable in thymic lymphocytes isolated from MCF13 MLV-inoculated mice during the preleukemic period. These results indicated that the ability of MCF13 MLV to induce apoptosis is correlated with its ability to superinfect cells and that this occurs as an early step in thymic lymphoma development.


Assuntos
Apoptose , Vírus Indutores de Focos em Células do Vison/fisiologia , Vison/virologia , Animais , DNA Viral/análise , DNA Viral/fisiologia , Células Epiteliais/virologia , Glicoproteínas/fisiologia , Receptores Virais/fisiologia
12.
Virology ; 283(1): 121-31, 2001 Apr 25.
Artigo em Inglês | MEDLINE | ID: mdl-11312668

RESUMO

Long terminal repeat (LTR) sequences are important determinants of mink cell focus-forming (MCF) murine leukemia virus pathogenesis. These sequences include the enhancer and sequences between the enhancer and promoter (DEN). In a previous study we showed that a virus missing the DEN region in its LTR was severely attenuated in its ability to induce thymic lymphoma. In this study we observed that a virus with an LTR consisting of DEN but no enhancer sequences was pathogenic. We compared the pathogenicity of this DEN virus with other LTR mutant MCF13 viruses that contained a single enhancer (1R) or a single enhancer plus DEN (1R + DEN). All LTR mutant viruses generated thymic lymphoma, however, at a much lower incidence and with a longer latency compared with wild-type (WT) MCF13 virus. DEN virus replication in the thymus was the lowest compared with the 1R and 1R + DEN viruses. Viral replication in a different thymic subpopulation could not explain the decreased pathogenicity of the LTR mutant viruses compared with WT virus. However, lower levels of mutant virus replication in the thymus compared with WT during the preleukemic period may contribute to the attenuation of pathogenicity. The phenotype of tumors induced by the mutant viruses was similar and differed from tumors induced by WT virus by the presence of CD3(-)CD4(-)CD8(-) cells. Analysis of LTR sequences of infectious virus rescued from tumors induced by the 1R and 1R + DEN viruses showed that amplification of enhancer sequences had occurred during tumor development. The lack of DEN virus expression by tumor cells led us to propose that DEN sequences may play a role at an early step in tumorigenesis.


Assuntos
Elementos Facilitadores Genéticos/genética , Vírus Indutores de Focos em Células do Vison/patogenicidade , Regiões Promotoras Genéticas/genética , Sequências Repetidas Terminais/genética , Animais , Citometria de Fluxo , Leucemia Experimental/virologia , Linfoma/virologia , Camundongos , Vírus Indutores de Focos em Células do Vison/genética , Mutação , Infecções por Retroviridae/virologia , Timo/citologia , Timo/imunologia , Timo/virologia , Neoplasias do Timo/virologia , Infecções Tumorais por Vírus/virologia , Virulência , Replicação Viral
13.
Dev Biol (Basel) ; 106: 163-7; discussion 167-8, 253-63, 2001.
Artigo em Inglês | MEDLINE | ID: mdl-11761228

RESUMO

Using neoplastic cell lines as substrates for vaccine development could inadvertently result in viral-viral or viral-cellular interactions whose biological consequences are unclear. In this review, the generation of mink cell focus-inducing (MCF) retroviruses in the mouse is discussed as a model for understanding how viral-viral and viral-cellular interactions can result in the generation of new retroviruses with pathological consequences.


Assuntos
Leucemia Experimental/virologia , Fusão de Membrana , Vírus Indutores de Focos em Células do Vison/fisiologia , Modelos Biológicos , Animais , Camundongos , Vírus Indutores de Focos em Células do Vison/patogenicidade , Células Tumorais Cultivadas
14.
J Virol ; 74(17): 8119-26, 2000 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-10933722

RESUMO

In a previous study we identified the subpopulations of thymus cells that were infected by the lymphomagenic MCF13 murine leukemia virus (MLV) (F. K. Yoshimura, T. Wang, and M. Cankovic, J. Virol. 73:4890-4898, 1999) and observed an effect on thymus size by virus infection. In this report we describe our results which demonstrate that MCF13 MLV infection of thymuses reduced the number of T lymphocytes in this organ. Histological examination showed diffuse lymphocyte depletion, which was most striking in the CD4(+) CD8(+) lymphocyte-enriched cortical zone. Consistent with this, flow cytometric analysis showed that the lymphocytes which were depleted were predominantly the immature CD3(-) CD4(+) CD8(+) and CD3(+) CD4(+) CD8(+) cells. A comparison of the percentages of live, apoptotic, and dead cells of the gp70(+) and gp70(-) thymic lymphocytes suggested that this effect on thymus cellularity is a result of virus infection. Studies of the survival of thymic T lymphocytes in culture showed that cells from MCF13 MLV-inoculated mice underwent greater apoptosis and death than cells from control animals. Assays for apoptosis included 7-amino-actinomycin D staining, DNA fragmentation, and cleavage of caspase-3 and poly(ADP-ribose) polymerase proenzymes. Our results suggest that apoptosis of thymic lymphocytes by virus infection is an important step in the early stages of MCF13 MLV tumorigenesis.


Assuntos
Apoptose , Leucemia Experimental/patologia , Vírus Indutores de Focos em Células do Vison/imunologia , Infecções por Retroviridae/patologia , Linfócitos T/virologia , Infecções Tumorais por Vírus/patologia , Animais , Linfócitos T CD4-Positivos/patologia , Linfócitos T CD4-Positivos/virologia , Linfócitos T CD8-Positivos/patologia , Linfócitos T CD8-Positivos/virologia , Caspase 3 , Caspases/metabolismo , Células Cultivadas , Fragmentação do DNA , Precursores Enzimáticos/metabolismo , Citometria de Fluxo , Leucemia Experimental/imunologia , Leucemia Experimental/virologia , Camundongos , Camundongos Endogâmicos AKR , Poli(ADP-Ribose) Polimerases/metabolismo , Infecções por Retroviridae/imunologia , Infecções por Retroviridae/virologia , Linfócitos T/patologia , Timo/patologia , Timo/virologia , Infecções Tumorais por Vírus/imunologia , Infecções Tumorais por Vírus/virologia
15.
J Virol ; 74(15): 6832-7, 2000 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-10888622

RESUMO

Historically, Swiss Webster mice of the CFW subline, both inbred and random-bred stocks, have been considered to have a low spontaneous occurrence of hematopoietic system tumors, and previous reports of infectious expression of murine leukemia viruses (MuLVs) have been rare and unremarkable. In marked contrast, in the present study of CFW mice from one source observed by two laboratories over a 2-year period, nearly 60% developed tumors, 85% of which were lymphomas, the majority of B-cell origin. All tumors tested expressed ecotropic MuLVs, and most expressed mink cell focus-inducing (MCF) MuLVs. Among normal mice of weanling to advanced age, over one-half were positive for ecotropic virus in tail or lymphoid tissues, and MCF virus was frequently present in lymphoid tissue, less often in tail. Patterns of ecotropic proviral integration indicated that natural infection occurred by both genetic and exogenous routes. Lymphomas were induced in NIH Swiss mice infected as neonates with tissue culture-propagated MuLVs isolated from normal and tumor tissue of CFW mice.


Assuntos
Vírus da Leucemia Murina/isolamento & purificação , Linfoma de Células B/virologia , Camundongos/virologia , Infecções por Retroviridae/virologia , Infecções Tumorais por Vírus/virologia , Animais , Linhagem Celular , Vírus da Leucemia Murina/genética , Vírus da Leucemia Murina/patogenicidade , Leucemia Experimental/patologia , Leucemia Experimental/virologia , Vírus Indutores de Focos em Células do Vison/genética , Vírus Indutores de Focos em Células do Vison/isolamento & purificação , Vírus Indutores de Focos em Células do Vison/patogenicidade , Infecções por Retroviridae/patologia , Infecções Tumorais por Vírus/patologia
16.
Hum Gene Ther ; 11(2): 271-84, 2000 Jan 20.
Artigo em Inglês | MEDLINE | ID: mdl-10680841

RESUMO

A novel murine stromal cell line, HESS-M28, was established, which supports the expansion of human CD34+CD38- cells more than 300-fold in vitro in the presence of human IL-3 and SCF. These cells were used in an attempt to evaluate cis-acting elements of retroviral vectors in human primitive hematopoietic cells. Cord blood cells were cultured on top of the mixed cell layers of the stromal cell line, HESS-M28, and retroviral vector-producing cells. The FMEV-type vector SF/Lyt contained the spleen focus-forming virus U3 and the MESV primer-binding site (PBS), while MO3/Lyt contained the U3 region and PBS from Mo-MuLV. After transduction by the FMEV-type and Mo-MuLV-based vectors, expression of the marker gene murine CD8 (mCD8) was examined in CD34-, CD34+, and CD34+CD38- cells. In CD34+ and CD34+CD38- cells, expression of mCD8 was higher with the FMEV-type vector, SF/Lyt, compared with the cells transduced by the Mo-MuLV-based vector MO3/Lyt, although the expression was comparable in CD34- cells. Expression of marker genes was also confirmed in long-term culture-initiating cells (LTC-ICs) and SCID-repopulating cells (SRCs).


Assuntos
Antígenos CD34/genética , Antígenos CD , Antígenos de Diferenciação/genética , Técnicas de Cultura de Células/métodos , Vírus da Leucemia Murina de Friend/genética , Vetores Genéticos/genética , Células-Tronco Hematopoéticas/metabolismo , Vírus Indutores de Focos em Células do Vison/genética , Vírus da Leucemia Murina de Moloney/genética , NAD+ Nucleosidase/genética , Retroviridae/genética , ADP-Ribosil Ciclase , ADP-Ribosil Ciclase 1 , Animais , Antígenos CD8/genética , Ciclo Celular , Linhagem Celular , DNA Viral/metabolismo , Sangue Fetal/metabolismo , Citometria de Fluxo , Expressão Gênica , Técnicas de Transferência de Genes , Humanos , Imunofenotipagem , Canamicina Quinase/genética , Glicoproteínas de Membrana , Camundongos , Camundongos Endogâmicos C3H , Camundongos Endogâmicos NOD , Camundongos SCID
17.
Arch Virol ; 144(11): 2207-12, 1999.
Artigo em Inglês | MEDLINE | ID: mdl-10603174

RESUMO

We report the complete nucleotide sequence of the potent T-lymphomagenic murine retrovirus SL3-3. The non-LTR regions of the virus show 98% sequence identity to the endogenous ecotropic Akv murine leukemia virus. While the region encoding the surface envelope protein is completely identical to that of Akv, a approximately 200 nucleotide stretch in the integrase encoding region upstream of env is similar to the sequence of mink cell focus forming (MCF) viruses and shows a complete match with the mouse retrovirus 10A1. The history of SL3-3 may therefore include recombination involving an Akv-like virus and a virus with MCF-like sequences.


Assuntos
Genes env , Vírus da Leucemia Murina/genética , Vírus Indutores de Focos em Células do Vison/genética , Animais , Sequência de Bases , DNA Viral , Camundongos , Dados de Sequência Molecular
18.
J Virol ; 73(7): 5671-80, 1999 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-10364317

RESUMO

One hallmark of murine leukemia virus (MuLV) leukemogenesis in mice is the appearance of env gene recombinants known as mink cell focus-inducing (MCF) viruses. The site(s) of MCF recombinant generation in the animal during Moloney MuLV (M-MuLV) infection is unknown, and the exact roles of MCF viruses in disease induction remain unclear. Previous comparative studies between M-MuLV and an enhancer variant, Mo+PyF101 MuLV, suggested that MCF generation or early propagation might take place in the bone marrow under conditions of efficient leukemogenesis. Moreover, M-MuLV induces disease efficiently following both intraperitoneal (i.p.) and subcutaneous (s.c.) inoculation but leukemogenicity by Mo+PyF101 M-MuLV is efficient following i.p. inoculation but attenuated upon s. c. inoculation. Time course studies of MCF recombinant appearance in the bone marrow, spleen, and thymus of wild-type and Mo+PyF101 M-MuLV i.p.- and s.c.-inoculated mice were carried out by performing focal immunofluorescence assays. Both the route of inoculation and the presence of the PyF101 enhancer sequences affected the patterns of MCF generation or early propagation. The bone marrow was a likely site of MCF recombinant generation and/or early propagation following i.p. inoculation of M-MuLV. On the other hand, when the same virus was inoculated s.c., the primary site of MCF generation appeared to be the thymus. Also, when Mo+PyF101 M-MuLV was inoculated i.p., MCF generation appeared to occur primarily in the thymus. The time course studies indicated that MCF recombinants are not involved in preleukemic changes such as splenic hyperplasia. On the other hand, MCFs were detected in tumors from Mo+PyF101 M-MuLV s. c.-inoculated mice even though they were largely undetectable at preleukemic times. These results support a role for MCF recombinants late in disease induction.


Assuntos
Elementos Facilitadores Genéticos , Variação Genética , Leucemia Experimental/virologia , Vírus Indutores de Focos em Células do Vison/fisiologia , Vírus da Leucemia Murina de Moloney/fisiologia , Infecções por Retroviridae/virologia , Infecções Tumorais por Vírus/virologia , Injeções Intraperitoneais , Injeções Subcutâneas , Vírus da Leucemia Murina de Moloney/genética , Provírus
19.
J Virol ; 73(5): 3595-602, 1999 May.
Artigo em Inglês | MEDLINE | ID: mdl-10196250

RESUMO

The common virus integration site (VIS) Evi11 was recently identified within the gene encoding the hematopoietic G-protein-coupled peripheral cannabinoid receptor Cnr2 (also referred to as Cb2). Here we show that Cnr2 is a frequent target (12%) for insertion of Cas-Br-M murine leukemia virus (MuLV) in primary tumors in NIH/Swiss mice. Multiple provirus insertions in Evi11 were cloned and shown to be located within the 3' untranslated region of the candidate proto-oncogene Cnr2. These results suggest that proviral insertion in the Cnr2 gene is an important step in Cas-Br-M MuLV-induced leukemogenesis in NIH/Swiss mice. To isolate Evi11/Cnr2 collaborating proto-oncogenes, we searched for novel common VISs in the Cas-Br-M MuLV-induced primary tumors and identified a novel frequent common VIS, Evi12 (14%). Interestingly, 54% of the Evi11/Cnr2-rearranged primary tumors contained insertions in Evi12 as well, which suggests cooperative action of the target genes in these two common VISs in leukemogenesis. By interspecific backcross analysis it was shown that Evi12 resides on mouse chromosome 10 in a region that shares homology with human chromosomes 12q and 19p. Sequence analysis demonstrated that Evi12 is located upstream of the gene encoding the molecular chaperone Tra1/Grp94, which was previously mapped to mouse chromosome 10 and human chromosome 12q22-24. Thus, Tra1/Grp94 is a candidate target gene for retroviral activation or inactivation in Evi12. However, Northern and Western blot analyses did not provide evidence that proviral insertion had altered the expression of Tra1/Grp94. Additional studies are required to determine whether Tra1/Grp94 or another candidate proto-oncogene in Evi12 is involved in leukemogenesis.


Assuntos
Canabinoides , Proteínas de Transporte , Proteínas de Choque Térmico HSP70/genética , Proteínas de Membrana/genética , Vírus Indutores de Focos em Células do Vison/genética , Proteínas de Transferência de Fosfolipídeos , Proteínas/genética , Receptores de Droga/genética , Integração Viral , Células 3T3 , Animais , Sequência de Bases , Sítios de Ligação , Feminino , Genes Virais , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Dados de Sequência Molecular , Regiões Promotoras Genéticas , Proto-Oncogene Mas , Receptores de Canabinoides , Células Tumorais Cultivadas
20.
J Virol ; 73(5): 3733-6, 1999 May.
Artigo em Inglês | MEDLINE | ID: mdl-10196266

RESUMO

The Asian mouse Mus castaneus is resistant to infection by the polytropic mink cell focus-inducing (MCF) subgroup of murine leukemia viruses (MuLVs). Genetic crosses showed this recessive resistance to be governed by a single gene that maps at or near the gene encoding the polytropic viral receptor, Rmc1. To investigate this resistance, we mated M. castaneus with mice carrying the wild mouse Sxv variant of the Rmc1 receptor that allows infection by xenotropic as well as polytropic virus. Unlike other F1 hybrids of M. castaneus, these F1 mice were resistant to both xenotropic and polytropic classes of MuLVs. Analysis of backcrossed progeny of the F1 hybrids mated to Sxv mice indicates that resistance is due to inheritance of two M. castaneus genes. Cells from individual backcross mice were also examined for cell surface antigen by fluorescence-activated cell sorter analysis with monoclonal antibodies reactive with xenotropic or MCF virus env glycoproteins. A correlation was observed between virus resistance and antigen, suggesting that virus resistance is due to expression of endogenous viral envelope genes that interfere with infection by exogenous virus. Since the inbred strain Rmc1 receptor remains functional in the presence of these M. castaneus genes, and since M. castaneus contains multiple copies of xenotropic MuLV env genes, we suggest that these resistance genes control expression of xenotropic env glycoprotein that interferes with exogenous virus in cells containing the Sxv variant of Rmc1.


Assuntos
Proteínas de Membrana , Vírus Indutores de Focos em Células do Vison/imunologia , Muridae/imunologia , Receptores Virais/metabolismo , Animais , Células Cultivadas , Feminino , Imunidade Inata , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos DBA , Vírus Indutores de Focos em Células do Vison/metabolismo , Muridae/genética , Receptores Acoplados a Proteínas G , Receptores Virais/genética , Proteínas do Envelope Viral/imunologia , Receptor do Retrovírus Politrópico e Xenotrópico
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...