Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 29
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.
Mol Cell Biol ; 6(11): 4088-92, 1986 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-3025631

RESUMO

A high frequency (greater than or equal to 65%) of thymomas induced by mink cell focus-forming virus 69L1 in AKR/J mice contain proviral integrations which are clustered 0.7-kilobase upstream of the c-myc oncogene predominantly in the opposite transcriptional orientation. Blot hybridization experiments showed that on the average there was only a twofold elevation of steady-state c-myc RNA in the thymomas as compared with levels in normal AKR/J thymocytes. Such an increase would not appear to be sufficient as a mechanism of oncogene activation in this system. In contrast, S1 nuclease analysis of transcripts initiated from the two known c-myc promoters indicated a strong shift in promoter usage in virtually all thymomas tested. In normal thymus the ratio of transcripts initiated from the proximal promoter P1 to the distal promoter P2 was 0.2 to 0.3. In contrast, most of the thymomas tested (18 of 23) showed an average P1/P2 ratio of 1.2 regardless of whether or not proviral integrations could be detected within a 21-kilobase EcoRI fragment containing the three c-myc exons. We conclude that alterations in P1/P2 ratios are good indicators of c-myc deregulation in thymic lymphomas.


Assuntos
Vírus da Leucemia Murina/patogenicidade , Linfoma/microbiologia , Vírus Indutores de Focos em Células do Vison/patogenicidade , Oncogenes , Timoma/microbiologia , Neoplasias do Timo/microbiologia , Animais , Genes Virais , Linfoma/genética , Camundongos , Camundongos Endogâmicos AKR , Hibridização de Ácido Nucleico , Timoma/genética , Neoplasias do Timo/genética
3.
Trends Microbiol ; 5(2): 74-82, 1997 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-9108934

RESUMO

Moloney murine leukemia virus is a prototypical simple retrovirus that has been an extremely useful model for leukemogenesis. Important steps in leukemogenesis include proviral activation of cellular proto-oncogenes, generation of mink cell focus-inducing recombinants, and early (preleukemic) virus-induced changes in hematopoiesis.


Assuntos
Leucemia Experimental/etiologia , Vírus da Leucemia Murina de Moloney/patogenicidade , Infecções por Retroviridae/etiologia , Infecções Tumorais por Vírus/etiologia , Animais , Cocarcinogênese , Regulação Neoplásica da Expressão Gênica , Camundongos , Vírus Indutores de Focos em Células do Vison/genética , Vírus Indutores de Focos em Células do Vison/patogenicidade , Vírus da Leucemia Murina de Moloney/genética , Pré-Leucemia/etiologia , Proto-Oncogenes , Recombinação Genética
4.
J Leukoc Biol ; 64(4): 441-50, 1998 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-9766624

RESUMO

Malignant histiocytosis sarcoma virus (MHSV) arose as a recombinant of c-Harvey-ras murine sarcoma virus (Ha-MuSV) and Friend mink cell focus-forming virus (F-MCFV). It is a defective acute transforming retrovirus that, along with Friend murine leukemia helper virus (F-MuLV), induces malignant histiocytosis (MH) in susceptible adult mice. We have assessed the in vivo susceptibility to MHSV in inbred homozygous, F1 hybrid, congenic, and recombinant inbred (RI) mice. We have shown that: (1) in vivo resistance to MHSV is multigenic, regulated by MHC and non-MHC genes in a different fashion than with F-MCFV, F-MuLV, or Ha-MuSV; (2) using BXD RI mice, the resistance phenotype is linked with 95.8% probability to two linked loci, Pmv-9 and Iapls3-14, on chromosome 13 (homologous to the area of human chromosome 5 for which a chromosomal break point at position 5q35 is associated with human MH); and (3) CD4+ T cells are critical for MHSV resistance.


Assuntos
Mapeamento Cromossômico , Sarcoma Histiocítico/genética , Sarcoma Histiocítico/virologia , Complexo Principal de Histocompatibilidade , Infecções por Retroviridae/imunologia , Animais , Linfócitos T CD4-Positivos/imunologia , Aberrações Cromossômicas , Cromossomos Humanos Par 5 , Cruzamentos Genéticos , Feminino , Vírus da Leucemia Murina de Friend/genética , Vírus da Leucemia Murina de Friend/patogenicidade , Ligação Genética , Predisposição Genética para Doença , Sarcoma Histiocítico/imunologia , Homozigoto , Humanos , Imunidade Inata/genética , Camundongos , Camundongos Congênicos , Camundongos Endogâmicos , Camundongos Mutantes , Camundongos SCID , Vírus Indutores de Focos em Células do Vison/genética , Vírus Indutores de Focos em Células do Vison/patogenicidade , Fenótipo , Recombinação Genética , Infecções por Retroviridae/genética , Vírus do Sarcoma Murino/genética , Vírus do Sarcoma Murino/patogenicidade
5.
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
6.
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
7.
J Virol ; 73(1): 234-41, 1999 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-9847326

RESUMO

The oncogenic potential of many nonacute retroviruses is dependent on the duplication of the enhancer sequences present in the unique 3' (U3) region of the long terminal repeat (LTR). In a molecular clone (MCF 247-W) of the murine leukemia virus MCF 247, a leukemogenic mink cell focus-inducing (MCF) virus, the U3 enhancer sequences are tandemly repeated in the LTR. We mutated the enhancer region of MCF 247-W to test the hypothesis that the duplicated enhancer sequences of this virus have a sequence-specific and/or a stereospecific role in enhancer function required for transformation. In one virus, we inserted 14 nucleotide bp into the novel sequence generated at the junction of the two enhancers to generate an MCF virus with an interrupted enhancer region. In the second virus, only one copy of the enhancer sequences was present. This second virus also lacked the junction sequence present between the two enhancers of MCF 247-W. Both viruses were less leukemogenic and had a longer mean latency period than MCF 247-W. These data indicate that the sequence generated at the junction of the two enhancers and/or the stereospecific arrangement of the two enhancer elements are required for the full oncogenic potential of MCF 247-W. We analyzed proviral LTRs within the c-myc locus in tumor DNAs from mice injected with the MCF virus with the interrupted enhancer region. Some of the proviral LTRs integrated upstream of c-myc contain enhancer regions that are larger than those of the injected virus. These results are consistent with the suggestion that the virus with an interrupted enhancer changes in vivo to perform its role in the transformation of T cells.


Assuntos
Elementos Facilitadores Genéticos , Vírus Indutores de Focos em Células do Vison/genética , Vírus Indutores de Focos em Células do Vison/patogenicidade , Animais , Sequência de Bases , Genes myc , Leucemia Experimental/etiologia , Camundongos , Camundongos Endogâmicos AKR , Dados de Sequência Molecular , Sequências Repetidas Terminais
8.
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
9.
Virology ; 168(1): 90-100, 1989 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-2535909

RESUMO

California wild mouse-derived ecotropic virus Cas-Br-M induces a spongiform encephalopathy and a wide variety of hematopoietic neoplasms on inoculation of neonatal mice. We isolated a MCF virus [Ns-6(186) MCF] from a thymic T-cell lymphoma developing in a NFS mouse inoculated with Cas-Br-M virus. Biologically cloned NS-6(186) MCF virus, in contrast to previously studied MCF viruses, was found to induce thymic or nonthymic T-cell lymphomas with high efficiency in the absence of ecotropic helper virus. Comparison of the restriction endonuclease maps derived from Cas-Br-M and NS-6(186) MCF revealed differences only in the env region, between 5.8 and 7.8 kb from the 5' end. Two biologically active molecular clones of the NS-6(186) MCF (clone 15 with two LTRs and clone 19 with 1 LTR) were studied. Although both clones exhibited similar in vitro activities, clone 15-derived virus induced only T-cell lymphomas with short latency whereas clone 19-derived virus induced a wide variety of neoplasms with a significantly longer latency. Nucleotide sequence analysis established that the U3 region of each of the two LTRs of clone 15 has a 53-bp duplication which includes "enhancer elements," but that the single LTR of clone 19 has no such duplication.


Assuntos
DNA Viral/genética , Vírus da Leucemia Murina/genética , Leucemia Experimental/microbiologia , Vírus Indutores de Focos em Células do Vison/genética , Sequência de Aminoácidos , Animais , Sequência de Bases , Células Cultivadas , Clonagem Molecular , Citometria de Fluxo , Camundongos , Vírus Indutores de Focos em Células do Vison/patogenicidade , Dados de Sequência Molecular , Sequências Repetitivas de Ácido Nucleico , Mapeamento por Restrição , Transfecção
10.
J Virol ; 63(3): 1284-92, 1989 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-2536834

RESUMO

Oncogenic mink cell focus-forming (MCF) viruses, such as MCF 247, show a positive correlation between the ability to replicate efficiently in the thymus and a leukemogenic phenotype. Other MCF viruses, such as MCF 30-2, replicate to high titers in thymocytes and do not accelerate the onset of leukemia. We used these two MCF viruses with different biological phenotypes to distinguish the effect of specific viral genes and genetic determinants on thymotropism and leukemogenicity. Our goal was to identify the viral sequences that distinguish thymotropic, nonleukemogenic viruses such as MCF 30-2 from thymotropic, leukemogenic viruses such as MCF 247. We cloned MCF 30-2, compared the genetic hallmarks of MCF 30-2 with those of MCF 247, constructed a series of recombinants, and tested the ability of recombinant viruses to replicate in the thymus and to induce leukemia. The results established that (i) MCF 30-2 and MCF 247 differ in the numbers of copies of the enhancer sequences in the long terminal repeats. (ii) The thymotropic phenotype of both viruses is independent of the number of copies of the enhancer sequences. (iii) The oncogenic phenotype of MCF 247 is correlated with the presence in the virus of duplicated enhancer sequences or with the presence of an enhancer with a specific sequence. These results show that the pathogenic phenotypes of MCF viruses are dissociable from the thymotropic phenotype and depend, at least in part, upon the enhancer sequences. On the basis of these results, we suggest that the molecular mechanisms by which the enhancer sequences determine thymotropism are different from those that determine oncogenicity.


Assuntos
Elementos Facilitadores Genéticos , Vírus da Leucemia Murina/genética , Vírus Indutores de Focos em Células do Vison/genética , Timo/microbiologia , Sequência de Aminoácidos , Sequência de Bases , Clonagem Molecular , Engenharia Genética , Leucemia Experimental/genética , Vírus Indutores de Focos em Células do Vison/patogenicidade , Dados de Sequência Molecular , Mapeamento de Nucleotídeos , Sequências Reguladoras de Ácido Nucleico , Proteínas dos Retroviridae/genética , Proteínas do Envelope Viral/genética
11.
Virology ; 183(1): 343-50, 1991 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-2053287

RESUMO

The replication-defective Friend spleen focus-forming virus (F-SFFV) induces acute erythroblastosis in adult mice. The envelope-related (env) gene and LTR are the only functional elements of the viral genome. The env-coded glycoprotein gp55 has been shown to be responsible for target cell specificity and for the short latency of the disease caused by SFFV. This molecule closely resembles the env coded proteins gp70 + p15E of mink cell focus inducing viruses (MCFV). The only substantial differences between these two env genes are a large deletion spanning 585 nucleotides in the middle of the F-SFFV gene and a frameshift mutation near the 3' end leading to a modified and shortened membrane anchor in the mature protein. To determine if the large deletion and/or the frameshift mutation are capable of changing the properties of a nonpathogenic MCFV into those of an acutely pathogenic SFFV we introduced these changes into the env gene of an MCFV. The results show that the mutated MCFV is as acutely pathogenic as F-SFFV. We therefore conclude that the modified membrane anchor of gp55 and the change caused by the large deletion are the essential determinants of the high pathogenicity of SFFV.


Assuntos
Clonagem Molecular , Produtos do Gene env/genética , Vírus Formadores de Foco no Baço/genética , Animais , Células Cultivadas , Clonagem Molecular/métodos , Mutação da Fase de Leitura , Genes env , Leucemia Experimental/genética , Leucemia Experimental/patologia , Camundongos , Camundongos Endogâmicos DBA , Vírus Indutores de Focos em Células do Vison/genética , Vírus Indutores de Focos em Células do Vison/patogenicidade , Ratos , Sequências Repetitivas de Ácido Nucleico , Vírus Formadores de Foco no Baço/patogenicidade , Transfecção
12.
J Virol ; 61(9): 2782-92, 1987 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-3039169

RESUMO

The leukemogenic membrane glycoprotein of Friend spleen focus-forming virus (SFFV) has an apparent Mr of 55,000 (gp55), is encoded by a recombinant env gene, and occurs on cell surfaces and in intracellular organelles. There is evidence that the amino-terminal region of gp55 forms a dualtropic-specific domain that is connected to the remainder of the glycoprotein by a proline-rich linker (C. Machida, R. Bestwick, B. Boswell, and D. Kabat, Virology 144:158-172, 1985). Using the colinear form of a cloned polycythemic strain of SFFV proviral DNA, we constructed seven in-phase env mutants by insertion of linkers and by a deletion. The mutagenized SFFVs were transfected into fibroblasts and were rescued by superinfection with a helper murine leukemia virus. Four of the mutants cause erythroblastosis. These include one with a 6-base-pair (bp) insert in the ecotropic-related sequence near the 3' end of the gene, two with a 12- or 18-bp insert in the region that encodes the proline-rich linker, and one with a 6-bp insert in the dualtropic-specific region. The other mutants (RI, Sm1, and Sm2) are nonpathogenic and contain lesions in dualtropic-specific region. The other mutants (RI, Sm1, and Sm2) are nonpathogenic and contain lesions in dualtropic-specific sequences that are highly conserved among strains of SFFV. A pathogenic revertant (RI-rev) was isolated from one mouse that developed erythroblastosis 3 weeks after infection with RI. RI-rev contains a second-site env mutation that affects the same domain as the primary mutation does and that increases the size of the encoded glycoprotein. All pathogenic SFFVs encode glycoproteins that are expressed on cell surfaces, whereas the nonpathogenic glycoproteins are exclusively intracellular. The pathogenic SFFVs also specifically cause a weak interference to superinfection by dualtropic MuLVs. These results are compatible with the multidomain model for the structure of gp55 and suggest that processing of gp55 to plasma membranes is required for pathogenesis. The amino-terminal region of gp55 binds to dualtropic murine leukemia virus receptors, and this interaction is preserved in the SFFV mutants that cause erythroblastosis.


Assuntos
Glicoproteínas/fisiologia , Vírus da Leucemia Murina/patogenicidade , Proteínas de Membrana/fisiologia , Receptores Virais/metabolismo , Vírus Formadores de Foco no Baço/patogenicidade , Animais , Anticorpos Monoclonais/imunologia , Feminino , Glicoproteínas/análise , Glicoproteínas/genética , Camundongos , Vírus Indutores de Focos em Células do Vison/patogenicidade , Modelos Biológicos , Mutação , Vírus Formadores de Foco no Baço/genética , Interferência Viral , Replicação Viral
13.
J Virol ; 67(7): 3763-70, 1993 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-8510205

RESUMO

We analyzed viral recombination events that occur during the preleukemic period in AKR mice. We tagged a molecular chimera between the nonleukemogenic virus Akv and the leukemogenic mink cell focus-inducing (MCF) virus MCF 247 with an amber suppressor tRNA gene, supF. We injected the supF-tagged chimeric virus that contains all of the genes of MCF 247 except the envelope gene, which in turn is derived from Akv, into newborn AKR mice to evaluate its pathogenic potential. Approximately the same percentage of animals developed leukemia with similar latent periods when injected with either the tagged or nontagged virus. DNA from tumors induced in AKR mice by the tagged chimeric virus was analyzed by Southern blotting with the supF gene as a probe. One set of tumors contained the injected supF-tagged virus. Two kinds of supF-tagged proviruses were found in a second set of tumors. One group of supF-tagged viruses had a restriction map consistent with that of the injected virus, while the other group of proviruses had restriction maps that suggested that the proviruses had acquired an MCF virus-like envelope gene by recombination with endogenous viral sequences. These results demonstrate that injected viruses recombine in vivo with endogenous viral sequences. Furthermore, the progression to leukemia was accelerated in mice that develop tumors containing proviruses with an MCF virus env gene, emphasizing the importance of the role of the MCF virus env gene product in transformation.


Assuntos
Vírus AKR da Leucemia Murina/genética , Leucemia Experimental/microbiologia , Camundongos Endogâmicos AKR/microbiologia , Vírus Indutores de Focos em Células do Vison/genética , Animais , Southern Blotting , Genes env , Leucemia Experimental/genética , Linfoma/genética , Linfoma/microbiologia , Camundongos , Vírus Indutores de Focos em Células do Vison/patogenicidade , Recombinação Genética , Mapeamento por Restrição , Timoma/genética , Timoma/microbiologia
14.
J Virol ; 71(7): 5287-94, 1997 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-9188597

RESUMO

Several murine leukemia viruses (MuLV) induce neurologic disease in susceptible mice. To identify features of central nervous system (CNS) infection that correlate with neurovirulence, we compared two neurovirulent MuLV, Fr98 and Fr98/SE, with a nonneurovirulent MuLV, Fr54. All three viruses utilize the polytropic receptor and are coisogenic, each containing a different envelope gene within a common genetic background. Both Fr98 and Fr98/SE induce a clinical neurologic disease characterized by hyperexcitability and ataxia yet differ in incubation period, 16 to 30 and 30 to 60 days, respectively. Fr54 infects the CNS but fails to induce clinical signs of neurologic disease. In this study, we compared the histopathology, regional virus distribution, and cell tropism in the brain, as well as the relative CNS viral burdens. All three viruses induced similar histopathologic effects, characterized by intense reactive astrogliosis and microglial activation associated with minimal vacuolar degeneration. The infected target cells for each virus consisted primarily of endothelial and microglial cells, with rare oligodendrocytes. Infection localized predominantly in white matter tracts of the cerebellum, internal capsule, and corpus callosum. The only feature that correlated with relative neurovirulence was viral burden as measured by both viral CA protein expression in cerebellar homogenates and quantification of infected cells. Interestingly, Fr54 (nonneurovirulent) and Fr98/SE (slow disease) had similar viral burdens at 3 weeks postinoculation, suggesting that they entered the brain with comparable efficiencies. However, spread of Fr98/SE within the brain thereafter exceeded that of Fr54, reaching levels of viral burden comparable to that seen for Fr98 (rapid disease) at 3 weeks. These results suggest that the determinants of neurovirulence in the envelope gene may influence the efficiency of virus spread within the brain and that a critical number of infected cells may be required for induction of clinical neurologic disease.


Assuntos
Encéfalo/virologia , Leucemia Experimental/virologia , Microglia/virologia , Vírus Indutores de Focos em Células do Vison/isolamento & purificação , Infecções por Retroviridae/virologia , Infecções Tumorais por Vírus/virologia , Animais , Encéfalo/citologia , Leucemia Experimental/patologia , Camundongos , Camundongos Endogâmicos , Vírus Indutores de Focos em Células do Vison/patogenicidade , Infecções por Retroviridae/patologia , Infecções Tumorais por Vírus/patologia , Carga Viral , Virulência
15.
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
16.
J Virol ; 65(1): 132-7, 1991 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-1985193

RESUMO

The 3' half of the env gene of the dualtropic Friend mink cell focus-forming virus was modified by replacing the restriction enzyme fragment of the genome DNA with the corresponding fragment of the acutely leukemogenic, polycythemia-inducing strain of Friend spleen focus-forming virus (F-SFFVP) genome DNA. Replacement with the fragment of F-SFFVP env containing the 585-bp deletion, the 6-bp duplication, and the single-base insertion converted the resulting chimeric genome so that the mutant had a pathogenic activity like that of F-SFFVP. Replacement with the fragment containing only the 585-bp deletion did not result in a pathogenic virus. However, when this virus pseudotyped by Friend murine leukemia virus was passaged in newborn DBA/2 mice, we could recover weakly pathogenic viruses with a high frequency. Molecular analysis of the genome of the recovered virus revealed the presence of a single-base insertion in the same T5 stretch where the wild-type F-SFFV env has the single-base insertion. These results provided evidence that the unique genomic structures present in the 3' half of F-SFFV env are the sole determinants that distinguish the pathogenicity of F-SFFV from that of Friend mink cell focus-forming virus. The importance of the dualtropic env-specific sequence present in the 5' half of F-SFFV env for the pathogenic activity was evaluated by constructing a mutant F-SFFV genome in which this sequence was replaced by the ecotropic env sequence of Friend murine leukemia virus and by examining its pathogenicity. The results indicated that the dualtropic env-specific sequence was essential to pathogenic activity.


Assuntos
Vírus da Leucemia Murina de Friend/genética , Genes env , Vírus Indutores de Focos em Células do Vison/genética , Vírus Formadores de Foco no Baço/genética , Animais , Clonagem Molecular , DNA Viral/genética , DNA Viral/isolamento & purificação , Feminino , Vírus da Leucemia Murina de Friend/patogenicidade , Leucemia Experimental/microbiologia , Masculino , Camundongos , Camundongos Endogâmicos DBA , Camundongos Endogâmicos , Vírus Indutores de Focos em Células do Vison/patogenicidade , Plasmídeos , Recombinação Genética , Sequências Repetitivas de Ácido Nucleico , Mapeamento por Restrição , Vírus Formadores de Foco no Baço/patogenicidade
17.
J Immunol ; 148(9): 2947-52, 1992 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-1349324

RESUMO

AKR mice are highly susceptible to spontaneous T cell lymphomagenesis and thymus removal at the age of 1 to 3 mo greatly reduces its development. Twelve-mo-old AKR mice thymectomized at young age were shown previously to carry potential lymphoma cells that could be triggered to develop into B cell lymphomas (80 to 100%) after removal from their host "restrictive" environment into young histocompatible hosts. Additional attempts were made to terminate the potential lymphoma cell dormant state in 12-mo-old thymectomized AKR mice. Replenishment of some deficiencies caused by thymectomy at a young age, including a s.c. syngeneic thymus graft or a single injection of the dual tropic recombinant virus isolates DTV-71 or MCF-247 into 12-mo-old thymectomized AKR mice resulted in Ly-1+ pre-B or B cell lymphoma development in 80 to 98% of these treated mice. In vivo elimination of T cell subsets by administration of cyclosporin A or by mAb expressed on Th cells (anti-CD4) or cytotoxic T cells (anti-CD8) stimulated the progression of dormant potential lymphoma cells towards B cell lymphoma development. The most striking results were observed after administration of anti-CD8 mAb: 90 to 100% of these treated mice developed Ly-1+ B cell lymphomas within 80 days. The effect of rIL-2 on dormant PLC was also tested. Administration of rIL-2 to 12-mo-old thymectomized mice terminated tumor dormancy in 94% of the treated mice within 66 days. Tests of the resulting B lymphomas for dual tropic recombinant virus/mink cell focus-inducing virus infection indicated that the breakdown of tumor dormancy did not result from development of pathogenic class I mink cell focus-inducing viruses. These results suggest that T cell subsets and/or their products are involved in the proliferation arrest of potential lymphoma cells present in thymectomized AKR mice.


Assuntos
Linfoma de Células B/etiologia , Timo/imunologia , Animais , Southern Blotting , Antígenos CD8/fisiologia , Interleucina-2/farmacologia , Linfoma de Células B/genética , Linfoma de Células B/microbiologia , Camundongos , Camundongos Endogâmicos AKR , Vírus Indutores de Focos em Células do Vison/patogenicidade , Polimorfismo de Fragmento de Restrição , Proteínas Recombinantes/farmacologia , Mapeamento por Restrição , Subpopulações de Linfócitos T/imunologia , Timectomia
18.
Proc Natl Acad Sci U S A ; 88(24): 11138-42, 1991 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-1763029

RESUMO

Calorie restriction suppresses mammary proviral mRNA expression and protooncogene activation in breast tumor-prone C3H/Ou mice while inhibiting tumor formation. To determine whether the beneficial effects of chronic energy-intake restriction (CEIR) can be extended to an organ site of retrovirus-induced tumorigenesis where the dynamics of growth and sexual maturity are not paramount as they are in breast tissue, calorie restriction of 40% was imposed on thymic lymphoma-prone AKR mice when 4 weeks old. Recombination between various murine leukemia virus (MuLV) mRNAs, resulting in the generation of an 8.4-kilobase genomic-length transcript with mink cytopathic focus-forming (MCF) characteristics, is considered the proximal retroviral event in AKR lymphomagenesis. Thymic expression of subgenomic MCF MuLV mRNA was uniformly suppressed among 6- and 8-week-old CEIR mice (P less than 0.02). This suppression of MuLV transcription preceded a 25% reduction in the appearance of genomic-length MCF transcripts among CEIR mice and a 28% reduction in cumulative lymphoma mortality. The latency to median tumor incidence was extended greater than 3 months by calorie restriction, and median lifespan was extended approximately 50%. Survival curves for the full-fed and CEIR dietary cohorts were found to be significantly different (P less than 0.0001), with full-fed mice experiencing a 3 times greater risk of lymphoma mortality. These findings extend the known range of pathologic states influenced by CEIR in inbred mice and show that retroviral mechanisms involved in generation of lymphoid malignancy can be significantly impaired by calorie restriction.


Assuntos
Dieta Redutora , Regulação Viral da Expressão Gênica , Leucemia Experimental/microbiologia , Leucemia Experimental/prevenção & controle , Linfoma/microbiologia , Linfoma/prevenção & controle , Vírus Indutores de Focos em Células do Vison/genética , Transcrição Gênica , Animais , Peso Corporal , Ingestão de Energia , Feminino , Expressão Gênica , Genoma Viral , Camundongos , Camundongos Endogâmicos AKR , Vírus Indutores de Focos em Células do Vison/patogenicidade , Sondas RNA , RNA Mensageiro/genética , RNA Mensageiro/isolamento & purificação , RNA Viral/genética , RNA Viral/isolamento & purificação , Timo/microbiologia
19.
J Virol ; 65(3): 1273-85, 1991 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-1847454

RESUMO

The spontaneous leukemias of AKR mice are caused by mink cell focus-forming (MCF) viruses. These viruses are generated by recombination between several endogenous murine retroviruses. The virological events leading to the generation of the leukemogenic agent were investigated by using an oligonucleotide specific for the U3 region of the leukemogenic virus and env-reactive oligonucleotide probes specific for the different classes of endogenous murine leukemia virus. It was shown that (i) the leukemogenic MCF virus is formed by recombination between at least three different endogenous sequences; (ii) the U3 donor for the leukemogenic virus is the inducible xenotropic virus Bxv-1; (iii) all spontaneous tumors contain viruses with duplicated enhancer regions in their long terminal repeats; (iv) enhancer duplication is a somatic event, since Bxv-1 contains only one copy; (v) the first recombinant virus detectable in mass populations of thymocytes by Southern hybridization analysis contains all structural features of the ultimate leukemogenic virus; and (vi) the multiple novel viruses in a given tumor represent progeny of the same unique recombination events. On the basis of these results, an analysis of the virological events leading to AKR thymomas is presented.


Assuntos
Vírus da Leucemia Murina/genética , Vírus Indutores de Focos em Células do Vison/genética , Provírus/genética , Timoma/microbiologia , Neoplasias do Timo/microbiologia , Animais , Sequência de Bases , Clonagem Molecular , DNA Viral/genética , DNA Viral/isolamento & purificação , Vírus da Leucemia Murina/patogenicidade , Camundongos , Camundongos Endogâmicos AKR , Vírus Indutores de Focos em Células do Vison/isolamento & purificação , Vírus Indutores de Focos em Células do Vison/patogenicidade , Dados de Sequência Molecular , Sondas de Oligonucleotídeos , Provírus/patogenicidade , Recombinação Genética , Sequências Repetitivas de Ácido Nucleico , Mapeamento por Restrição
20.
J Virol ; 61(6): 1861-6, 1987 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-3033317

RESUMO

Despite the high degree of homology (91%) between the nucleotide sequences of the Friend-mink cell focus-forming (MCF) and the Moloney murine leukemia virus (MuLV) genomic long terminal repeats (LTRs), the pathogenicities determined by the LTR sequences of the two viruses are quite different. Friend-MCF MuLV is an erythroid leukemia virus, and Moloney MuLV is a lymphoid leukemia virus. To map the LTR sequences responsible for the different disease specificities, we constructed nine viruses with LTRs recombinant between the Friend-MCF and Moloney MuLVs. Analysis of the leukemia induced with the recombinant viruses showed that a 195-base-pair nucleotide sequence, including a 75-base-pair nucleotide Moloney enhancer, is responsible for the tissue-specific leukemogenicity of Moloney MuLV. However, not only the enhancer but also its downstream sequences appear to be necessary. The Moloney virus enhancer and its downstream sequence exerted a dominant effect over that of the Friend-MCF virus, but the enhancer sequence alone did not. The results that three of the nine recombinant viruses induced both erythroid and lymphoid leukemias supported the hypothesis that multiple viral genetic determinants control both the ability to cause leukemia and the type of leukemia induced.


Assuntos
Vírus da Leucemia Murina de Friend/genética , Vírus da Leucemia Murina/genética , Leucemia Eritroblástica Aguda/genética , Leucemia Experimental/genética , Leucemia Linfoide/genética , Vírus Indutores de Focos em Células do Vison/genética , Vírus da Leucemia Murina de Moloney/genética , Sequências Repetitivas de Ácido Nucleico , Animais , Sequência de Bases , DNA Recombinante/fisiologia , DNA Viral/fisiologia , Elementos Facilitadores Genéticos , Vírus da Leucemia Murina de Friend/patogenicidade , Regulação da Expressão Gênica , Genes Virais , Camundongos , Vírus Indutores de Focos em Células do Vison/patogenicidade , Vírus da Leucemia Murina de Moloney/patogenicidade , Especificidade de Órgãos , Recombinação Genética , Homologia de Sequência do Ácido Nucleico
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA