Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 37
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
J Evol Biol ; 31(2): 314-322, 2018 02.
Artigo em Inglês | MEDLINE | ID: mdl-29266576

RESUMO

The virulence levels attained by serial passage of pathogens through similar host genotypes are much higher than observed in natural systems; however, it is unknown what keeps natural virulence levels below these empirically demonstrated maximum levels. One hypothesis suggests that host diversity impedes pathogen virulence, because adaptation to one host genotype carries trade-offs in the ability to replicate and cause disease in other host genotypes. To test this hypothesis, with the simplest level of population diversity within the loci of the major histocompatibility complex (MHC), we serially passaged Friend virus complex (FVC) through two rounds, in hosts with either the same MHC genotypes (pure passage) or hosts with different MHC genotypes (alternated passage). Alternated passages showed a significant overall reduction in viral titre (31%) and virulence (54%) when compared to pure passages. Furthermore, a resistant host genotype initially dominated any effects due to MHC diversity; however, when FVC was allowed to adapt to the resistant host genotype, predicted MHC effects emerged; that is, alternated lines show reduced virulence. These data indicate serial exposure to diverse MHC genotypes is an impediment to pathogen adaptation, suggesting genetic variation at MHC loci is important for limiting virulence in a rapidly evolving pathogen and supports negative frequency-dependent selection as a force maintaining MHC diversity in host populations.


Assuntos
Evolução Biológica , Vírus da Leucemia Murina de Friend/patogenicidade , Complexo Principal de Histocompatibilidade , Vírus Formadores de Foco no Baço/patogenicidade , Animais , Variação Genética , Camundongos , Camundongos Endogâmicos BALB C
2.
J Immunol ; 189(5): 2521-9, 2012 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-22821964

RESUMO

The immune system is tasked with defending against a myriad of microbial infections, and its response to a given infectious microbe may be strongly influenced by coinfection with another microbe. It was shown that infection of mice with lactate dehydrogenase-elevating virus (LDV) impairs early adaptive immune responses to Friend virus (FV) coinfection. To investigate the mechanism of this impairment, we examined LDV-induced innate immune responses and found LDV-specific induction of IFN-α and IFN-γ. LDV-induced IFN-α had little effect on FV infection or immune responses, but unexpectedly, LDV-induced IFN-γ production dampened Th1 adaptive immune responses and enhanced FV infection. Two distinct effects were identified. First, LDV-induced IFN-γ signaling indirectly modulated FV-specific CD8+ T cell responses. Second, intrinsic IFN-γ signaling in B cells promoted polyclonal B cell activation and enhanced early FV infection, despite promotion of germinal center formation and neutralizing Ab production. Results from this model reveal that IFN-γ production can have detrimental effects on early adaptive immune responses and virus control.


Assuntos
Imunidade Adaptativa , Regulação para Baixo/imunologia , Interferon gama/fisiologia , Vírus da Leucemia Murina/imunologia , Infecções por Retroviridae/imunologia , Imunidade Adaptativa/genética , Animais , Modelos Animais de Doenças , Regulação para Baixo/genética , Feminino , Vírus da Leucemia Murina de Friend/imunologia , Vírus da Leucemia Murina de Friend/patogenicidade , Interferon gama/deficiência , Interferon gama/genética , Vírus Elevador do Lactato Desidrogenase/imunologia , Vírus Elevador do Lactato Desidrogenase/patogenicidade , Vírus da Leucemia Murina/patogenicidade , Leucemia Experimental/genética , Leucemia Experimental/imunologia , Leucemia Experimental/virologia , Camundongos , Camundongos Congênicos , Camundongos Endogâmicos A , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Mutantes , Infecções por Retroviridae/genética , Infecções por Retroviridae/patologia , Vírus Formadores de Foco no Baço/imunologia , Vírus Formadores de Foco no Baço/patogenicidade , Infecções Tumorais por Vírus/genética , Infecções Tumorais por Vírus/imunologia , Infecções Tumorais por Vírus/virologia
3.
J Virol ; 84(15): 7675-82, 2010 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-20504929

RESUMO

Infection of erythroid cells by Friend spleen focus-forming virus (SFFV) leads to acute erythroid hyperplasia in mice due to expression of its unique envelope glycoprotein, gp55. Erythroid cells expressing SFFV gp55 proliferate in the absence of their normal regulator, erythropoietin (Epo), because of interaction of the viral envelope protein with the erythropoietin receptor and a short form of the receptor tyrosine kinase Stk (sf-Stk), leading to constitutive activation of several signal transduction pathways. Our previous in vitro studies showed that phosphatidylinositol 3-kinase (PI3-kinase) is activated in SFFV-infected cells and is important in mediating the biological effects of the virus. To determine the role of PI3-kinase in SFFV-induced disease, mice deficient in the p85alpha regulatory subunit of class IA PI3-kinase were inoculated with different strains of SFFV. We observed that p85alpha status determined the extent of erythroid hyperplasia induced by the sf-Stk-dependent viruses SFFV-P (polycythemia-inducing strain of SFFV) and SFFV-A (anemia-inducing strain of SFFV) but not by the sf-Stk-independent SFFV variant BB6. Our data also indicate that p85alpha status determines the response of mice to stress erythropoiesis, consistent with a previous report showing that SFFV uses a stress erythropoiesis pathway to induce erythroleukemia. We further showed that sf-Stk interacts with p85alpha and that this interaction depends upon sf-Stk kinase activity and tyrosine 436 in the multifunctional docking site. Pharmacological inhibition of PI3-kinase blocked proliferation of primary erythroleukemia cells from SFFV-infected mice and the erythroleukemia cell lines derived from them. These results indicate that p85alpha may regulate sf-Stk-dependent erythroid proliferation induced by SFFV as well as stress-induced erythroid hyperplasia.


Assuntos
Leucemia Eritroblástica Aguda/virologia , Fosfatidilinositol 3-Quinases/metabolismo , Receptores Proteína Tirosina Quinases/metabolismo , Vírus Formadores de Foco no Baço/patogenicidade , Animais , Linhagem Celular Tumoral , Camundongos , Camundongos Endogâmicos BALB C , Fosfatidilinositol 3-Quinases/deficiência
4.
Retrovirology ; 5: 99, 2008 Nov 04.
Artigo em Inglês | MEDLINE | ID: mdl-18983647

RESUMO

The Friend virus SFFV (Spleen Focus Forming Virus) provokes an acute erythroblastosis in susceptible strains of mice that progresses to overt erythroleukemia by a multi-step process. For virologists, the Friend virus-induced disease has provided deep insights into the host mechanisms influencing susceptibility to retroviral infection and viremia. These insights have contributed to the understanding of HIV and other human retroviral infections. For cell biologists and oncologists, this leukemia has been a powerful experimental model to identify critical oncogenes involved in a multi-stage process, to understand the contribution of host genes to cancer development, and to investigate the mechanisms leading to cell growth autonomy. This model also provided an example of oncogenic reversion since Friend tumor cells can reinitiate their erythroid differentiation program when exposed in vitro to some chemical inducers. This review highlights recent findings demonstrating that the leukemic progression depends on the cooperation of at least two oncogenic events, one interfering with differentiation and one conferring a proliferative advantage. The Friend model of leukemia progression recapitulates the two phases of human acute myeloid leukemia (AML). Coupling of insights from studies on the Friend erythroleukemia with knowledge on AML might allow a better understanding of the molecular mechanisms involved in the evolution of leukemia in mice and men.


Assuntos
Transformação Celular Neoplásica , Leucemia Eritroblástica Aguda/virologia , Oncogenes , Vírus Formadores de Foco no Baço/patogenicidade , Animais , Animais Recém-Nascidos , Diferenciação Celular , Proliferação de Células , Humanos , Leucemia Eritroblástica Aguda/patologia , Leucemia Mieloide Aguda/patologia , Masculino , Camundongos
5.
J Virol ; 80(12): 5678-85, 2006 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-16731906

RESUMO

Infection of mice with Friend spleen focus-forming virus (SFFV) results in a multistage erythroleukemia. In the first stage, the SFFV envelope glycoprotein interacts with the erythropoietin receptor and a short form of the receptor tyrosine kinase sf-Stk, resulting in constitutive activation of signal transducing molecules and the development of erythropoietin (Epo)-independent erythroid hyperplasia and polycythemia. The second stage results from the outgrowth of a rare virus-infected erythroid cell that expresses nonphysiological levels of the myeloid transcription factor PU.1. These cells exhibit a differentiation block and can be grown as murine erythroleukemia (MEL) cell lines. In this study, we examined SFFV MEL cells to determine whether their transformed phenotype was associated with a block in the activation of any Epo signal-transducing molecules. Our studies indicate that Epo- or SFFV-induced activation of STAT1/3 DNA binding activity is blocked in SFFV MEL cells. The block is at the level of tyrosine phosphorylation of STAT1, although Jak2 phosphorylation is not blocked in these cells. In contrast to Epo, alpha interferon can induce STAT1 phosphorylation and DNA binding in SFFV MEL cells. The SFFV-transformed cells were shown to express elevated levels of the hematopoietic phosphatase SHP-1, and treatment of the cells with a phosphatase inhibitor restored STAT1 tyrosine phosphorylation. MEL cells derived from Friend murine leukemia virus (MuLV) or ME26 MuLV-infected mice, which do not express PU.1, express lower levels of SHP-1 and are not blocked in STAT1/3 DNA-binding activity. Our studies suggest that SFFV-infected erythroid cells become transformed when differentiation signals activated by STAT1/3 are blocked due to high SHP-1 levels induced by inappropriate expression of the PU.1 protein.


Assuntos
Transformação Celular Viral , DNA/metabolismo , Eritroblastos/virologia , Peptídeos e Proteínas de Sinalização Intracelular/genética , Proteínas Tirosina Fosfatases/genética , Fator de Transcrição STAT1/metabolismo , Vírus Formadores de Foco no Baço/patogenicidade , Animais , Diferenciação Celular , Eritropoetina/fisiologia , Regulação da Expressão Gênica , Camundongos , Fosforilação , Proteína Tirosina Fosfatase não Receptora Tipo 6 , Proteínas Proto-Oncogênicas/genética , Transativadores/genética
6.
Proc Natl Acad Sci U S A ; 102(43): 15488-93, 2005 Oct 25.
Artigo em Inglês | MEDLINE | ID: mdl-16223879

RESUMO

Friend spleen focus-forming virus (SFFV) causes rapid erythroleukemia in mice due to expression of its unique envelope glycoprotein, gp55. Erythroid cells expressing SFFV gp55 proliferate in the absence of their normal regulator erythropoietin (Epo) because of constitutive activation of Epo signal transduction pathways. Although SFFV infects many cell types, deregulation of cell growth occurs only when SFFV infects erythroid cells, suggesting that these cells express unique proteins that the virus requires to mediate its biological effects. Not only do erythroid cells express the Epo receptor (EpoR), but those from mice susceptible to SFFV-induced erythroleukemia also express a short form of the receptor tyrosine kinase Stk (sf-Stk). In erythroid cells, SFFV gp55 interacts with the EpoR complex and sf-Stk, leading to activation of the kinase and constitutive activation of signal transducing molecules. In this study, we demonstrate that SFFV gp55 can also deregulate the growth of nonerythroid cells when it is coexpressed with sf-Stk. Expression of SFFV gp55 in rodent fibroblasts engineered to express sf-Stk induced their transformation, as demonstrated by focus formation and anchorage-independent growth in vitro. This transformation by SFFV gp55 requires the kinase activity of sf-Stk and the presence of its extracellular domain but not expression of the EpoR or the tyrosine kinase Jak2, which is required for activation of signal transduction pathways through the EpoR. Thus, expression of SFFV gp55 in nonerythroid cells coexpressing sf-Stk results in their uncontrolled growth, demonstrating a previously unrecognized mechanism for retrovirus transformation of rodent fibroblasts and providing insight into SFFV-induced disease.


Assuntos
Transformação Celular Viral , Receptores Proteína Tirosina Quinases/fisiologia , Vírus Formadores de Foco no Baço/patogenicidade , Animais , Fibroblastos/citologia , Janus Quinase 2 , Camundongos , Células NIH 3T3 , Proteínas Tirosina Quinases/fisiologia , Proteínas Proto-Oncogênicas/fisiologia , Receptores Proteína Tirosina Quinases/química , Proteínas do Envelope Viral/fisiologia
7.
J Virol ; 78(9): 4573-81, 2004 May.
Artigo em Inglês | MEDLINE | ID: mdl-15078939

RESUMO

The erythroleukemia-inducing Friend spleen focus-forming virus (SFFV) encodes a unique envelope protein, gp55, which interacts with the erythropoietin (Epo) receptor complex, causing proliferation and differentiation of erythroid cells in the absence of Epo. Susceptibility to SFFV-induced erythroleukemia is conferred by the Fv-2 gene, which encodes a short form of the receptor tyrosine kinase Stk/Ron (sf-Stk) only in susceptible strains of mice. We recently demonstrated that sf-Stk becomes activated by forming a strong interaction with SFFV gp55. To examine the biological consequences of activated sf-Stk on erythroid cell growth, we prepared retroviral vectors which express sf-Stk, either in conjunction with gp55 or alone in a constitutively activated mutant form, and tested them for their ability to induce Epo-independent erythroid colonies ex vivo and disease in mice. Our data indicate that both gp55-activated sf-Stk and the constitutively activated mutant of sf-Stk induce erythroid cells from Fv-2-susceptible and Fv-2-resistant (sf-Stk null) mice to form Epo-independent colonies. Mutational analysis of sf-Stk indicated that a functional kinase domain and 8 of its 12 tyrosine residues are required for the induction of Epo-independent colonies. Further studies demonstrated that coexpression of SFFV gp55 with sf-Stk significantly extends the half-life of the kinase. When injected into Fv-2-resistant mice, neither the gp55-activated sf-Stk nor the constitutively activated mutant caused erythroleukemia. Surprisingly, both Fv-2-susceptible and -resistant mice injected with the gp55-sf-Stk vector developed clinical signs not previously associated with SFFV-induced disease. We conclude that sf-Stk, activated by either point mutation or interaction with SFFV gp55, is sufficient to induce Epo-independent erythroid colonies from both Fv-2-susceptible and -resistant mice but is unable to cause erythroleukemia in Fv-2-resistant mice.


Assuntos
Células Eritroides/metabolismo , Mutação Puntual , Receptores Proteína Tirosina Quinases/metabolismo , Vírus Formadores de Foco no Baço/patogenicidade , Proteínas do Envelope Viral/metabolismo , Animais , Linhagem Celular , Células Eritroides/citologia , Eritropoetina/metabolismo , Vetores Genéticos , Leucemia Eritroblástica Aguda/fisiopatologia , Leucemia Eritroblástica Aguda/virologia , Camundongos , Camundongos Endogâmicos C57BL , Receptores Proteína Tirosina Quinases/genética , Infecções por Retroviridae/fisiopatologia , Infecções por Retroviridae/virologia , Vírus Formadores de Foco no Baço/metabolismo , Infecções Tumorais por Vírus/fisiopatologia , Infecções Tumorais por Vírus/virologia
8.
Microbiol Immunol ; 42(4): 335-9, 1998.
Artigo em Inglês | MEDLINE | ID: mdl-9623923

RESUMO

Friend spleen focus-forming virus (F-SFFV) is a replication-defective acutely leukemogenic mouse retrovirus and encodes an envelope protein (Env)-like membrane glycoprotein (gp55) in its defective env gene, which is responsible for the early stage of the viral leukemogenesis. Gp55 is a modified Env protein and contains a polytropic mink cell focus-inducing (MCF) murine leukemia virus (MuLV) Env gp70-derived sequence in its amino-terminal region. To evaluate the possibility that the presumed binding of gp55 to an MCF MuLV receptor protein has some role in leukemogenesis, we examined the biological activities of a mutant gp55 (XE gp55), which has a xenotropic MuLV Env gp70 amino-terminal region. XE gp55 displayed almost the same biological activities as the wild-type gp55, excluding the above possibility.


Assuntos
Vírus da Leucemia Murina/genética , Proteínas Oncogênicas de Retroviridae/metabolismo , Vírus Formadores de Foco no Baço/patogenicidade , Proteínas do Envelope Viral/metabolismo , Células 3T3 , Sequência de Aminoácidos , Substituição de Aminoácidos , Animais , Ligação Competitiva , Células Cultivadas , Genes env , Camundongos , Camundongos Endogâmicos DBA , Dados de Sequência Molecular , Mutação , Receptores Virais/metabolismo , Proteínas Oncogênicas de Retroviridae/química , Proteínas Oncogênicas de Retroviridae/genética , Vírus Formadores de Foco no Baço/genética , Relação Estrutura-Atividade , Transfecção , Proteínas do Envelope Viral/química , Proteínas do Envelope Viral/genética
9.
J Virol ; 72(5): 3602-9, 1998 May.
Artigo em Inglês | MEDLINE | ID: mdl-9557641

RESUMO

The Friend spleen focus-forming virus (SFFV) env gene encodes a glycoprotein with apparent Mr of 55,000 that binds to erythropoietin receptors (EpoR) to stimulate erythroblastosis. A retroviral vector that does not encode any Env glycoprotein was packaged into retroviral particles and was coinjected into mice in the presence of a nonpathogenic helper virus. Although most mice remained healthy, one mouse developed splenomegaly and polycythemia at 67 days; the virus from this mouse reproducibly caused the same symptoms in secondary recipients by 2 to 3 weeks postinfection. This disease, which was characterized by extramedullary erythropoietin-independent erythropoiesis in the spleens and livers, was also reproduced in long-term bone marrow cultures. Viruses from the diseased primary mouse and from secondary recipients converted an erythropoietin-dependent cell line (BaF3/EpoR) into factor-independent derivatives but had no effect on the interleukin-3-dependent parental BaF3 cells. Most of these factor-independent cell clones contained a major Env-related glycoprotein with an Mr of 60,000. During further in vivo passaging, a virus that encodes an Mr-55,000 glycoprotein became predominant. Sequence analysis indicated that the ultimate virus is a new SFFV that encodes a glycoprotein of 410 amino acids with the hallmark features of classical gp55s. Our results suggest that SFFV-related viruses can form in mice by recombination of retroviruses with genomic and helper virus sequences and that these novel viruses then evolve to become increasingly pathogenic.


Assuntos
Leucemia Eritroblástica Aguda/veterinária , Infecções por Retroviridae/veterinária , Vírus Formadores de Foco no Baço/genética , Infecções Tumorais por Vírus/veterinária , Sequência de Aminoácidos , Animais , Sequência de Bases , Evolução Biológica , Células da Medula Óssea/metabolismo , Linhagem Celular , Células Cultivadas , DNA Viral , Feminino , Leucemia Eritroblástica Aguda/virologia , Camundongos , Camundongos Endogâmicos DBA , Dados de Sequência Molecular , Policitemia/virologia , Receptores da Eritropoetina/metabolismo , Infecções por Retroviridae/virologia , Análise de Sequência de DNA , Homologia de Sequência de Aminoácidos , Vírus Formadores de Foco no Baço/metabolismo , Vírus Formadores de Foco no Baço/patogenicidade , Esplenomegalia/virologia , Infecções Tumorais por Vírus/virologia
10.
J Virol ; 72(3): 2272-9, 1998 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-9499086

RESUMO

We previously reported (N. Watanabe, M. Nishi, Y. Ikawa, and H. Amanuma, J. Virol. 65:132-137, 1991) that the mutant Friend spleen focus-forming virus (F-SFFV(MS)), which encodes a mutant gp55 membrane glycoprotein with an ecotropic env gp70 sequence, was nonpathogenic. Here we injected the F-SFFV(MS)-Friend murine leukemia virus (F-MuLV) clone 57 complex into newborn DBA/2 mice. We obtained four groups of pathogenic variant F-SFFV complexes, each showing a different degree of pathogenicity in adult mice and a different gp55 profile. Of these, group 1 variant F-SFFV was particularly interesting, because it was the most frequently obtained and because it produced doublet bands of gp55 (59 and 57 kDa), neither of which reacted with the nonecotropic gp70-specific monoclonal antibody, and because its DNA intermediate did not hybridize with the nonecotropic env-specific probe. Cloning and DNA sequence analysis of the env region of one isolate of the group 1 variant F-SFFV revealed that this virus consisted of two distinct F-SFFV genomes; one (clone 117) differed from the other (clone 118) due to the presence of a 39-bp in-frame deletion. Reconstitution to full-length F-SFFV genomes and a pathogenicity assay showed that each reconstituted F-SFFV was pathogenic, with clone 117 showing a higher degree of pathogenicity than clone 118. Both reconstituted F-SFFVs caused activation of the mouse erythropoietin receptor in the factor-independent cell proliferation assay, although much less efficiently than the wild-type polycythemia-inducing isolate F-SFFVp. Clone 118 produced a gp55 of 59 kDa, while clone 117 produced one of 57 kDa. Clone 118 had a substitution by the F-MuLV clone 57 gp70 sequence, indicating that it was derived from the F-SFFV(MS) env gene by a homologous recombination with the F-MuLV clone 57 env gene. The site of the 39-bp deletion in clone 117 corresponded to the portion of the clone 118 sequence which was unique to the ecotropic env genes. These results indicated the importance for the biological activity of gp55 of the sequences in the gp70 differential region, which are contained in both polytropic and ecotropic env genes.


Assuntos
Proteínas Oncogênicas de Retroviridae/genética , Vírus Formadores de Foco no Baço/genética , Proteínas do Envelope Viral/genética , Células 3T3 , Sequência de Aminoácidos , Animais , Animais Recém-Nascidos , Sequência de Bases , Clonagem Molecular , DNA Viral , Produtos do Gene env/genética , Vírus da Leucemia Murina , Camundongos , Camundongos Endogâmicos DBA , Dados de Sequência Molecular , Receptores da Eritropoetina/metabolismo , Homologia de Sequência do Ácido Nucleico , Vírus Formadores de Foco no Baço/isolamento & purificação , Vírus Formadores de Foco no Baço/patogenicidade , Proteínas do Envelope Viral/química , Proteínas do Envelope Viral/metabolismo
11.
Virology ; 252(1): 46-53, 1998 Dec 05.
Artigo em Inglês | MEDLINE | ID: mdl-9875316

RESUMO

The membrane glycoprotein encoded by the env gene of either the polycythemia- or anemia-inducing spleen focus-forming virus (SFFVp or SFFVa, respectively) is responsible for the induction of erythroleukemia in mice. It has been shown that the SFFVp glycoprotein, gp55, interacts with the erythropoietin receptor (EPO-R) and promotes EPO-independent proliferation of an EPO-R-expressing hematopoietic cell line, Ba/F3 (Li et al., Nature 343:762, 1990). We show here that when residues within the transmembrane (TM) sequence of an SFFVp gp55 are altered based on the sequences of the anemia-inducing gp55s by a methionine-to-isoleucine (M-I) substitution, a di-leucine deletion (dLL), or both, the resulting mutants display an attenuated phenotype that resembles an SFFVa: they induce milder erythroproliferative disease without polycythemia in vivo and are unable to promote EPO-independent cell proliferation in vitro. The dLL mutation directly interferes with EPO-R binding by decreasing the affinity of gp55 for the receptor. On the other hand, the M-I mutation hampers the full mitogenic activation of EPO-R while having no effect on receptor binding and asserts a dominant negative effect over the wild-type SFFVp gp55. Two other sequence changes within the TM sequence did not affect the biological activities of the SFFVp gp55. These results indicate that the TM sequence of the SFFV env glycoprotein plays a prominent role in SFFV-induced erythroleukemogenesis through its influence on the mitogenic activation of EPO-R.


Assuntos
Leucemia Eritroblástica Aguda/virologia , Glicoproteínas de Membrana/química , Vírus Formadores de Foco no Baço/patogenicidade , Proteínas do Envelope Viral/química , Células 3T3 , Sequência de Aminoácidos , Animais , Glicoproteínas de Membrana/genética , Camundongos , Dados de Sequência Molecular , Mutagênese Sítio-Dirigida , Vírus Formadores de Foco no Baço/genética , Relação Estrutura-Atividade , Proteínas do Envelope Viral/genética
12.
Leukemia ; 11 Suppl 3: 152-4, 1997 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-9209327

RESUMO

Friend leukemia virus complex (FLV) consists of replication-defective, Friend spleen focus-forming virus (F-SFFV) and replication-competent, Friend murine leukemia virus (F-MuLV). We produced transgenic mice possessing F-SFFV gp55 gene and clarified that the gp55 glycoprotein encoded by F-SFFV env-related gene is, by itself, responsible for the initiation of erythroleukemia. The occurrence of erythroleukemia, however, is sporadic in these mice. Erythroleukemia cell lines established from these mice possessed mutations in the p53 allele. One had a temperature-sensitive mutant p53 allele, p53Val-135 and showed induction of apoptosis by expressing a wild-type p53 protein at 32 degrees C. Superinfection of the mice with Moloney murine leukemia virus (Mo-MuLV) conferred 100% induction of erythroleukemia, mutating p53 gene or activating Spfi-1 gene by insertional events. Activation of the JAK/STAT pathway, which is involved in cytokine signaling, was investigated in the gp55 signaling mediated by the erythropoietin receptor. JAK1 and STAT5 were constitutively tyrosine-phosphorylated but the DNA binding activity of STAT5 was not induced.


Assuntos
Vírus da Leucemia Murina de Friend/patogenicidade , Genes env , Leucemia Eritroblástica Aguda/fisiopatologia , Proteínas do Leite , Infecções por Retroviridae/fisiopatologia , Vírus Formadores de Foco no Baço/patogenicidade , Infecções Tumorais por Vírus/fisiopatologia , Animais , Linhagem Celular , Citocinas/fisiologia , Proteínas de Ligação a DNA/metabolismo , Eritropoetina/farmacologia , Éxons , Vírus da Leucemia Murina de Friend/genética , Janus Quinase 1 , Leucemia Eritroblástica Aguda/genética , Leucemia Eritroblástica Aguda/virologia , Camundongos , Camundongos Transgênicos , Vírus da Leucemia Murina de Moloney/patogenicidade , Mutação , Proteínas Tirosina Quinases/metabolismo , Receptores da Eritropoetina/fisiologia , Infecções por Retroviridae/genética , Fator de Transcrição STAT4 , Fator de Transcrição STAT5 , Transdução de Sinais , Vírus Formadores de Foco no Baço/genética , Transativadores/metabolismo , Proteína Supressora de Tumor p53/genética , Infecções Tumorais por Vírus/genética
13.
J Virol ; 69(12): 7606-11, 1995 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-7494268

RESUMO

Friend spleen focus-forming virus (F-SFFV) causes acute erythroleukemia in mice and encodes in its defective env gene an Env-like membrane glycoprotein (gp55). The F-SFFV env gene has three characteristic structures compared with that of ecotropic murine leukemia viruses (MuLVs): substitution by the polytropic MuLV env sequence, a 585-bp deletion, and a 1-bp insertion. All of these characteristic structures are essential for the leukemogenic potential of gp55 of polycythemia-inducing isolates of F-SFFV (F-SFFVp). The 1-bp insertion causes changes of six amino acids and truncation by 34 amino acids at the C terminus. In this study, we constructed 12 mutant F-SFFV genomes starting from the wild-type F-SFFVp and examined the effect of the C-terminal truncation and the six altered amino acids on the pathogenic activity of gp55. The results indicated that at least 18 to 24 amino acids must be deleted from the C terminus for the env product to be pathogenically active. We also found that the six altered amino acids contributed significantly to the pathogenic activity of gp55. Analyses of the cellular processing of these mutant gp55s supported a correlation between the pathogenic activity of gp55 and its efficiency in overall cellular processing.


Assuntos
Genes env , Leucemia Eritroblástica Aguda/virologia , Leucemia Experimental/virologia , Vírus Formadores de Foco no Baço/genética , Vírus Formadores de Foco no Baço/patogenicidade , Proteínas do Envelope Viral/biossíntese , Células 3T3 , Sequência de Aminoácidos , Animais , Sequência de Bases , Elementos de DNA Transponíveis , Glucosamina/metabolismo , Vírus da Leucemia Murina/genética , Leucemia Eritroblástica Aguda/fisiopatologia , Leucemia Experimental/fisiopatologia , Camundongos , Dados de Sequência Molecular , Mutagênese Insercional , Mutagênese Sítio-Dirigida , Oligodesoxirribonucleotídeos , Proteínas Recombinantes/biossíntese , Proteínas Recombinantes/metabolismo , Deleção de Sequência , Transfecção , Proteínas do Envelope Viral/metabolismo , Virulência/genética
14.
Baillieres Clin Haematol ; 8(1): 225-47, 1995 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-7663048

RESUMO

The Friend spleen focus-forming virus has been a valuable tool for understanding the molecular events involved in the multiple stages of leukaemia. As summarized in Figure 3, the primary effect of SFFV, which occurs within days, is to cause a polyclonal proliferation of erythroid precursor cells that can proliferate in the absence of their normal regulator erythropoietin. This is the direct result of the unique envelope glycoprotein encoded by SFFV, which is transported to the cell surface and apparently interacts with the EpoR or another component of the multimeric EpoR complex, resulting in the constitutive activation of the Epo signal transduction pathway. Within this proliferating population of erythroid cells is a rare cell that has undergone several genetic changes due to the integration of the viral genome in specific sites in the mouse DNA. This leads to the activation of a gene encoding the PU.1 transcription factor, whose high expression in erythroid cells may be the cause of the block in differentiation that is characteristic of SFFV-transformed erythroid cells. SFFV integration can also lead to the inactivation of the p53 tumour supressor gene, giving these cells a growth advantage in the mouse. The disease induced by SFFV in mice is very similar to polycythaemia vera in humans (Golde et al, 1981). The major clinical feature of polycythaemia vera is the continuous expansion of the number of mature red blood cells in the presence of low serum Epo levels. Also, BFU-E and CFU-E from these patients can form in the absence of Epo like the analogous cells from SFFV-infected mice (Casadevall et al, 1982). It is possible that haematopoietic cells from individuals suffering from this disease express a protein similar to the envelope glycoprotein of SFFV that can interact with the EpoR and lead to its constitutive activation. Alternatively, these patients may contain a mutant EpoR gene that is constitutively activated like the mutant EpoR described earlier. As we understand more fully how the SFFV envelope protein constitutively activates te EpoR complex, we can begin to design therapies to counteract its action that can then be applied to treating patients with polycythaemia vera or other human diseases associated with uncontrolled erythropoiesis.


Assuntos
Vírus Defeituosos/patogenicidade , Vírus Auxiliares/fisiologia , Leucemia Eritroblástica Aguda/virologia , Leucemia Experimental/virologia , Infecções por Retroviridae/virologia , Vírus Formadores de Foco no Baço/patogenicidade , Infecções Tumorais por Vírus/virologia , Animais , Transformação Celular Neoplásica , Transformação Celular Viral , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/fisiologia , Vírus Defeituosos/genética , Vírus Defeituosos/fisiologia , Células Precursoras Eritroides/patologia , Células Precursoras Eritroides/virologia , Eritropoese , Eritropoetina/fisiologia , Vírus da Leucemia Murina de Friend/genética , Vírus da Leucemia Murina de Friend/fisiologia , Genes env , Genoma Viral , Vírus Auxiliares/genética , Hiperplasia , Camundongos , Mutagênese Insercional , Receptores da Eritropoetina/fisiologia , Proteínas Oncogênicas de Retroviridae , Transdução de Sinais , Vírus Formadores de Foco no Baço/genética , Vírus Formadores de Foco no Baço/fisiologia , Proteínas do Envelope Viral/genética , Proteínas do Envelope Viral/fisiologia , Replicação Viral
15.
Trends Microbiol ; 3(2): 51-7, 1995 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-7728385

RESUMO

Since its discovery in 1957, Friend viral erythroleukemia has been the major model for understanding host genetic barriers to retroviral diseases and has facilitated the discovery of many polymorphic leukemia-control genes of mice. Some of these genes limit helper-virus replication, target-cell (erythroblast) pools or immune responses. At least one host gene appears to block the viral oncoprotein.


Assuntos
Vírus da Leucemia Murina de Friend/patogenicidade , Vírus Formadores de Foco no Baço/patogenicidade , Proteínas do Envelope Viral/fisiologia , Sequência de Aminoácidos , Animais , Vírus da Leucemia Murina de Friend/genética , Humanos , Leucemia Experimental/virologia , Camundongos , Dados de Sequência Molecular , Infecções por Retroviridae/virologia , Vírus Formadores de Foco no Baço/genética , Proteínas do Envelope Viral/genética
16.
J Virol ; 69(2): 856-63, 1995 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-7815553

RESUMO

Although the Friend virus-encoded membrane glycoprotein (gp55) activates erythropoietin receptors (EpoR) to cause erythroblastosis only in certain inbred strains of mice but not in other species, mutant viruses can overcome aspects of mouse resistance. Thus, mice homozygous for the resistance allele of the Fv-2 gene are unaffected by gp55 but are susceptible to mutant glycoproteins that have partial deletions in their ecotropic domains. These and other results have suggested that proteins coded for by polymorphic Fv-2 alleles might directly or indirectly interact with EpoR and that changes in gp55 can overcome this defense. A new viral mutant with an exceptionally large deletion in its ecotropic domain is now also shown to overcome Fv-2rr resistance. In all cases, the glycoproteins that activate EpoR are processed to cell surfaces as disulfide-bonded dimers. To initiate analysis of nonmurine resistances, we expressed human EpoR and mouse EpoR in the interleukin 3-dependent mouse cell line BaF3 and compared the abilities of Friend virus-encoded glycoproteins to convert these cells to growth factor independence. Human EpoR was activated in these cells by erythropoietin but was resistant to gp55. However, human EpoR was efficiently activated in these cells by the same viral mutants that overcome Fv-2rr resistance in mice. By construction and analysis of human-mouse EpoR chimeras, we obtained evidence that the cytosolic domain of human EpoR contributes to its resistance to gp55 and that this resistance is mediated by accessory cellular factors. Aspects of host resistance in both murine and nonmurine species are targeted specifically against the ecotropic domain of gp55.


Assuntos
Leucemia Eritroblástica Aguda/etiologia , Receptores da Eritropoetina/fisiologia , Vírus Formadores de Foco no Baço/genética , Proteínas do Envelope Viral/fisiologia , Animais , Produtos do Gene env/metabolismo , Camundongos , Camundongos Endogâmicos DBA , Mutação , Vírus Formadores de Foco no Baço/patogenicidade , Proteínas do Envelope Viral/genética
17.
J Virol ; 67(6): 3665-70, 1993 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-8497076

RESUMO

Friend murine leukemia virus (F-MuLV), an erythroleukemogenic replication-competent retrovirus, induces leukemia in its host after a long latency. However, the early effects of infection may determine the pathway that eventually leads to malignant transformation. To determine how F-MuLV affects host cell proliferation soon after infection, BALB/c mice were inoculated with virus and then were assayed for susceptibility to appropriately pseudotyped spleen focus-forming virus (SFFV) as an indicator of erythropoietic activity. Twelve-week-old mice exposed to F-MuLV for 9 days were more susceptible (by a factor of 30) to superinfection by SFFV than were nonviremic mice. To test whether increased susceptibility was the result of increased hematopoietic activity, hematopoietic progenitors from the spleens of F-MuLV-infected mice were enumerated with a clonal culture assay. Nine days after inoculation with F-MuLV, the numbers of colony-forming progenitors increased by a factor of 4. Morphological analysis of the cultured colonies showed that erythroid, granulocytic, monocytic, and mixed granulocytic-monocytic progenitors all had increased. Thus, F-MuLV more rapidly induced a generalized increase in hematopoiesis than has previously been reported. The splenic hyperplasia induced by F-MuLV soon after infection may explain its ability to accelerate leukemogenesis in mice also infected by the polytropic Friend mink cell focus-forming virus.


Assuntos
Hematopoese , Leucemia Experimental/fisiopatologia , Baço/microbiologia , Fatores Etários , Animais , Células Clonais , Eritrócitos , Feminino , Vírus da Leucemia Murina de Friend , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos , Baço/crescimento & desenvolvimento , Vírus Formadores de Foco no Baço/patogenicidade , Superinfecção , Viremia
18.
J Gen Virol ; 74 ( Pt 4): 699-705, 1993 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-8468555

RESUMO

The product of the envelope gene (gp55) of Friend spleen focus-forming virus is responsible for the acute form of erythroleukaemia caused by this virus. In order to investigate the role that the four known N-linked carbohydrate side-chains of gp55 play in pathogenesis, we have inactivated the four N-glycosylation signals by mutating the asparagine residues of these four sites into serine. When glycosylation sites 1 and/or 2 were altered, the viruses remained fully pathogenic. However, mutation at either of glycosylation sites 3 or 4 rendered the virus apathogenic, independent of mutations at other sites. Furthermore, when site 3 was changed, a new product appeared which seemed to have acquired a carbohydrate chain at a position normally not glycosylated, presumably at position Asn378.


Assuntos
Produtos do Gene env/metabolismo , Vírus Formadores de Foco no Baço/patogenicidade , Animais , Sequência de Bases , Produtos do Gene env/química , Glicoproteínas/fisiologia , Glicosilação , Leucemia Eritroblástica Aguda/microbiologia , Leucemia Experimental/microbiologia , Camundongos , Dados de Sequência Molecular , Peso Molecular , Mutagênese Sítio-Dirigida , Oligodesoxirribonucleotídeos/química , Vírus Formadores de Foco no Baço/química , Relação Estrutura-Atividade
19.
J Virol ; 67(3): 1322-7, 1993 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-8437218

RESUMO

Recent evidence suggests that interactions between spleen focus-forming virus (SFFV) env products and the erythropoietin receptor (EpoR) are responsible for viral pathogenicity. Infection of factor-dependent cell lines expressing epoR (the cloned gene for EpoR) with SFFVP is mitogenic, generating cell lines that are no longer dependent on added growth factor, and an immunoprecipitable complex between EpoR and immature env protein in the endoplasmic reticulum has been identified. The dependence of these in vitro activities on env protein processing and their relationship to pathogenicity of SFFV were explored by using glycosylation site mutants of SFFV env. Mutants carrying Asn-->Asp mutations at each of the two consensus signals for N-linked glycosylation in the N-terminal domain of SFFVAP-L env (gs1 and gs2), the gs1-2- double mutant, and the gs0 quadruple mutant (mutated at all four signals utilized for N-linked glycosylation in SFFVAP-L env) were made. The primary translation products (gp52) of single-site mutant envs were processed into more highly glycosylated forms, and the corresponding viruses induced splenomegaly in susceptible mice, whereas the gs1-2- and gs0 proteins were not processed, and these viruses were not pathogenic. Unprocessed env proteins of both pathogenic and nonpathogenic mutants coprecipitated with EpoR. In the BaF3 cell assay for epoR-dependent mitogenicity, the pathogenic single mutants induced factor-independent growth efficiently whereas the nonpathogenic gs1-2- and gs0 mutants did not. These data demonstrate that the ability of gp52 to form complexes with EpoR in the endoplasmic reticulum is not sufficient for either mitogenicity in cell culture or induction of splenomegaly in mice while supporting the hypothesis that pathogenicity and mitogenicity of SFFV both result from an interaction between EpoR and SFFV env protein.


Assuntos
Transformação Celular Viral , Genes env/genética , Processamento de Proteína Pós-Traducional , Receptores da Eritropoetina/metabolismo , Vírus Formadores de Foco no Baço/patogenicidade , Proteínas do Envelope Viral/metabolismo , Células 3T3 , Sequência de Aminoácidos , Animais , Sequência de Bases , Divisão Celular , Sequência Consenso , Glicosilação , Camundongos , Dados de Sequência Molecular , Mutagênese Sítio-Dirigida , Transfecção , Proteínas do Envelope Viral/genética
20.
J Virol ; 66(9): 5696-702, 1992 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-1501302

RESUMO

Retroviral interference is manifested in chronically infected cells as a decrease in susceptibility to superinfection by virions using the same cellular receptor. The pattern of interference reflects the cellular receptor specificity of the chronically infecting retrovirus and is mediated by the viral envelope glycoprotein, which is postulated to bind competitively all cellular receptors available for viral attachment. We established retroviral interference in mice by infecting them with Friend murine leukemia virus and them measured susceptibility to superinfection by challenging the mice with the erythroproliferative spleen focus-forming virus. Infection of approximately 10% of nucleated splenocytes rendered mice 1% as susceptible to superinfection as untreated controls. The magnitude of this effect was the same in mice incapable of producing neutralizing antibodies or genetically deficient for T cells. The results indicated that retroviral interference in vivo was established rapidly with infection of a fraction of the host cell population and that the decrease in susceptibility to superinfection occurred without a detectable contribution by immunologic factors.


Assuntos
Vírus da Leucemia Murina de Friend/fisiologia , Leucemia Experimental/prevenção & controle , Vírus Formadores de Foco no Baço/fisiologia , Interferência Viral/fisiologia , Animais , Feminino , Citometria de Fluxo , Masculino , Camundongos , Camundongos Endogâmicos , Camundongos Nus , Formação de Roseta , Baço/citologia , Vírus Formadores de Foco no Baço/patogenicidade , Linfócitos T/microbiologia , Virulência
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...