Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 26
Filtrar
Mais filtros








Base de dados
Intervalo de ano de publicação
1.
Mol Med Rep ; 16(3): 3041-3048, 2017 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-28713922

RESUMO

Gene transfer into normal quiescent human B cells is a challenging procedure. The present study aimed to investigate whether it is possible to increase the levels of transgene expression by using various types of promoters to drive the expression of selected genes­of­interest. To produce lentiviral particles, the present study used the 2nd generation psPAX2 packaging vector and the vesicular stomatitis virus ­expressing envelope vector pMD2.G. Subsequently, lentiviral vectors were generated containing various promoters, including cytomegalovirus (CMV), elongation factor­1 alpha (EF1α) and spleen focus­forming virus (SFFV). The present study was unable to induce satisfactory transduction efficiency in quiescent normal B cells; however, infection of normal B cells with Epstein­Barr virus resulted in increased susceptibility to lentiviral transduction. In addition, the SFFV promoter resulted in a higher level of transgene expression compared with CMV or EF1α promoters. As a proof­of concept that this approach allows for stable gene expression in normal B cells, the present study used bicistronic lentiviral vectors with genes encoding fluorescent reporter proteins, as well as X­box binding protein­1 and binding immunoglobulin protein.


Assuntos
Linfócitos B/metabolismo , Técnicas de Transferência de Genes , Regiões Promotoras Genéticas , Adulto , Linfócitos B/virologia , Biomarcadores/metabolismo , Ligante de CD40/metabolismo , Linhagem Celular Tumoral , Proliferação de Células , Técnicas de Cocultura , Células Alimentadoras/metabolismo , Feminino , Fluorescência , Expressão Gênica , Células HEK293 , Herpesvirus Humano 4/fisiologia , Humanos , Sítios Internos de Entrada Ribossomal/genética , Lentivirus/genética , Masculino , Pessoa de Meia-Idade , Vírus Formadores de Foco no Baço/fisiologia , Transdução Genética , Transgenes
2.
J Virol ; 87(17): 9886-94, 2013 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-23824803

RESUMO

Human cytomegalovirus (HCMV) lytic phase gene expression is repressed upon entry into myeloid lineage cells where the virus establishes latency. Lytic infection is not initiated because the tegument-delivered transactivator protein pp71 fails to enter the nucleus and inactivate the Daxx-mediated cellular intrinsic defense that silences the viral genome. When pp71 is expressed de novo in THP-1 monocytes, it localizes to the nucleus, inactivates the Daxx defense, and initiates lytic infection. We speculated that replacing the native viral promoter that drives pp71 expression with one that is highly and constitutively active in myeloid cells would permit pp71 de novo expression upon infection and that this newly expressed pp71 would accumulate in the nucleus, inactivate the intrinsic defense, and initiate the cascade of lytic gene expression. Surprisingly, we found that this promoter was still subject to normal silencing mechanisms in THP-1 monocytes and primary CD34(+) cells, two independent myeloid lineage cells. A second constitutively active heterologous viral promoter located in a different region of the HCMV genome was also silenced in THP-1 and CD34(+) cells. Furthermore, these two independent heterologous viral promoters inserted into three different regions of the HCMV genome in three different viral strains all required prior expression of the viral immediate early proteins for activation in fibroblasts. From this, we conclude that incorporation within the HCMV genome impacts the proclivity of heterologous viral promoters to initiate transcription. These observations have mechanistic implications for the expression of viral genes and transgenes during both lytic infection and latency.


Assuntos
Citomegalovirus/genética , Latência Viral/genética , Animais , Antígenos CD34/metabolismo , Linhagem Celular , Células Cultivadas , Citomegalovirus/fisiologia , Inativação Gênica , Genoma Viral , Interações Hospedeiro-Patógeno , Humanos , Proteínas Imediatamente Precoces/genética , Proteínas Imediatamente Precoces/metabolismo , Camundongos , Monócitos/virologia , Células Mieloides/imunologia , Células Mieloides/virologia , Regiões Promotoras Genéticas , Recombinação Genética , Vírus Formadores de Foco no Baço/genética , Vírus Formadores de Foco no Baço/fisiologia , Sequências Repetidas Terminais , Proteínas Virais/genética , Proteínas Virais/metabolismo , Latência Viral/fisiologia
3.
J Virol ; 82(1): 419-27, 2008 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-17959667

RESUMO

Infection of erythroid progenitor cells by Friend spleen focus-forming virus (SFFV) leads to acute erythroid hyperplasia and eventually to erythroleukemia in susceptible strains of mice. The viral envelope protein, SFFV gp55, forms a complex with the erythropoietin receptor (EpoR) and a short form of the receptor tyrosine kinase Stk (sf-Stk), activating both and inducing Epo-independent proliferation. Recently, we discovered that coexpression of SFFV gp55 and sf-Stk is sufficient to transform NIH 3T3 and primary fibroblasts. In the current study, we demonstrate that sf-Stk and its downstream effectors are critical to this transformation. Unlike SFFV-derived erythroleukemia cells, which depend on PU.1 expression for maintenance of the transformed phenotype, SFFV gp55-sf-Stk-transformed fibroblasts are negative for PU.1. Underscoring the importance of sf-Stk to fibroblast transformation, knockdown of sf-Stk abolished the ability of these cells to form anchorage-independent colonies. Like SFFV-infected erythroid cells, SFFV gp55-sf-Stk-transformed fibroblasts express high levels of phosphorylated MEK, ERK, phosphatidylinositol 3-kinase (PI3K), Gab1/2, Akt, Jun kinase (JNK), and STAT3, but unlike virus-infected erythroid cells they fail to express phosphorylated STATs 1 and 5, which may require involvement of the EpoR. In addition, the p38 mitogen-activated protein kinase (MAPK) stress response is suppressed in the transformed fibroblasts. Inhibition of either JNK or the PI3K pathway decreases both monolayer proliferation and anchorage-independent growth of the transformed fibroblasts as does the putative kinase inhibitor luteolin, but inhibition of p38 MAPK has no effect. Our results indicate that sf-Stk is a molecular endpoint of transformation that could be targeted directly or with agents against its downstream effectors.


Assuntos
Transformação Celular Viral/fisiologia , Fibroblastos/virologia , Receptores Proteína Tirosina Quinases/fisiologia , Vírus Formadores de Foco no Baço/fisiologia , Proteínas do Envelope Viral/fisiologia , Animais , Proliferação de Células , Inativação Gênica , Camundongos , Células NIH 3T3 , Proteínas Quinases/biossíntese
4.
J Virol ; 79(20): 12752-62, 2005 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-16188978

RESUMO

Members of the mitogen-activated protein kinase (MAPK) family, including Jun amino-terminal kinase (JNK) and extracellular signal-related kinase (ERK), play an important role in the proliferation of erythroid cells in response to erythropoietin (Epo). Erythroid cells infected with the Friend spleen focus-forming virus (SFFV) proliferate in the absence of Epo and show constitutive activation of Epo signal transduction pathways. We previously demonstrated that the ERK pathway was constitutively activated in Friend SFFV-infected erythroid cells, and in this study JNK is also shown to be constitutively activated. Pharmacological inhibitors of both the ERK and JNK pathways stopped the proliferation of primary erythroleukemic cells from Friend SFFV-infected mice, with little induction of apoptosis, and furthermore blocked their ability to form Epo-independent colonies. However, only the JNK inhibitor blocked the proliferation of erythroleukemia cell lines derived from these mice. The JNK inhibitor caused significant apoptosis in these cell lines as well as an increase in the fraction of cells in G(2)/M and undergoing endoreduplication. In contrast, the growth of erythroleukemia cell lines derived from Friend murine leukemia virus (MuLV)-infected mice was inhibited by both the MEK and JNK inhibitors. JNK is important for AP1 activity, and we found that JNK inhibitor treatment reduced AP1 DNA-binding activity in primary erythroleukemic splenocytes from Friend SFFV-infected mice and in erythroleukemia cell lines from Friend MuLV-infected mice but did not alter AP1 DNA binding in erythroleukemia cell lines from Friend SFFV-infected mice. These data suggest that JNK plays an important role in cell proliferation and/or the survival of erythroleukemia cells.


Assuntos
Proteínas Quinases JNK Ativadas por Mitógeno/metabolismo , Leucemia Experimental/metabolismo , Infecções por Retroviridae/metabolismo , Transdução de Sinais , Vírus Formadores de Foco no Baço/fisiologia , Infecções Tumorais por Vírus/metabolismo , Animais , Antracenos/farmacologia , Apoptose , Proteínas Quinases Dependentes de Cálcio-Calmodulina/antagonistas & inibidores , Proteínas Quinases Dependentes de Cálcio-Calmodulina/farmacologia , Linhagem Celular Transformada/virologia , Proliferação de Células , Transformação Celular Viral , Células Cultivadas , Eritropoetina , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Flavonoides/farmacologia , Proteínas Quinases JNK Ativadas por Mitógeno/antagonistas & inibidores , MAP Quinase Quinase Quinases/metabolismo , Camundongos , Fator de Transcrição AP-1/metabolismo , Células Tumorais Cultivadas/fisiologia
5.
Hum Gene Ther ; 13(14): 1783-90, 2002 Sep 20.
Artigo em Inglês | MEDLINE | ID: mdl-12396630

RESUMO

Guidelines for testing gene therapy products for ecotropic replication-competent retrovirus (Eco-RCR) have not been delineated as they have for amphotropic viruses. To evaluate biologic assays that can detect these viruses, we compared an S(+)/L(-) assay and a marker rescue assay designed specifically for Eco-RCR detection. Moloney murine leukemia virus (Mo-MuLV) obtained from the American Type Culture Collection was used as the positive control. For marker rescue, NIH 3T3 cells were transduced with a retroviral vector expressing the neomycin phosphotransferase gene (3T3/Neo). Inoculation and passage of test material in 3T3/Neo cells for 3 weeks (amplification) and subsequent testing in the S(+)/L(-) assay or the marker rescue assay increased the level of sensitivity for virus detection greater than 10-fold compared with direct inoculation of D56 S(+)/L(-) cells. When serial dilutions of Mo-MuLV stock were evaluated, six of six cultures had detectable virus by the S(+)/L(-) and marker rescue assays at dilutions of 10(-5) and 10(-6). At the 10(-7) dilution, five of six assays had detectable virus in both assays. The ability to detect virus-infected cells was also evaluated in a modification that substituted cells for supernatant. Fifteen 3T3/Neo cultures inoculated with 10(6) 293 cells containing 100 or 10 Mo-MuLV/3T3 cells were all positive by marker rescue. For dilution with 1 virus-infected cell per 10(6) 293 cells, 10 of 15 cultures were positive. At the 0.1-cell dilution only 2 of 15 cultures were positive. If we hope to detect one infected cell in a test article, the probability of detecting virus if the assay is performed in triplicate is 96.3%. In summary, after 3 weeks of amplification the S(+)/L(-) and marker rescue assays can detect virus with similar sensitivities. We prefer the marker rescue assay because of the more reliable growth features of NIH 3T3 cells compared with the D56 cell line. For laboratories analyzing clinical materials, this report may prove useful in establishing detection assays for Eco-RCR.


Assuntos
Bioensaio , Terapia Genética/normas , Vetores Genéticos/fisiologia , Canamicina Quinase/análise , Retroviridae/isolamento & purificação , Vírus Formadores de Foco no Baço/isolamento & purificação , Replicação Viral , Células 3T3/virologia , Animais , Linhagem Celular/virologia , Vírus Defeituosos/fisiologia , Genes Reporter , Marcadores Genéticos , Humanos , Canamicina Quinase/genética , Rim , Camundongos , Vírus da Leucemia Murina de Moloney/fisiologia , Proteínas Recombinantes de Fusão/análise , Proteínas Recombinantes de Fusão/genética , Retroviridae/fisiologia , Segurança , Vírus do Sarcoma Murino/fisiologia , Sensibilidade e Especificidade , Vírus Formadores de Foco no Baço/fisiologia , Transdução Genética
6.
Oncogene ; 21(8): 1272-84, 2002 Feb 14.
Artigo em Inglês | MEDLINE | ID: mdl-11850847

RESUMO

Cancer is a multi-step, multi-genetic event. Whether oncogenic mutations cooperate with one another to transform cells and how is not well understood. The Friend murine retroviral erythroleukemia model involves mitogenic activation of the erythropoietin receptor (EpoR) by the virus env gene (F-gp55), aberrant over-expression of the transcription factor PU.1, and inactivating mutations in p53. In this report we demonstrate that concurrent expression of F-gp55 and PU.1 in erythroid target cells, in vivo, cooperate to accelerate erythroleukemia induction. Early in the disease, prior to the detection of clonal leukemic cells, activation of the EpoR by F-gp55, but not erythropoietin, resulted in transcriptional upregulation of PU.1 through a trans regulatory mechanism. This could occur in the absence of an integrated provirus within the PU.1 gene locus. The regulation of PU.1 transcription in established erythroleukemia cell lines differed depending upon the level of PU.1 protein present. Our results suggest that the action of F-gp55 contributes to both early and late stages of Friend erythroleukemia and that persistence of F-gp55 expression may be required not only to initiate erythroleukemia but to also maintain erythroleukemia following Friend virus infection.


Assuntos
Regulação Neoplásica da Expressão Gênica , Leucemia Eritroblástica Aguda/genética , Oncogenes/genética , Proteínas Proto-Oncogênicas/genética , Receptores da Eritropoetina/metabolismo , Vírus Formadores de Foco no Baço/genética , Transativadores/genética , Proteínas do Envelope Viral/metabolismo , Integração Viral , Animais , Células Precursoras Eritroides/efeitos dos fármacos , Células Precursoras Eritroides/metabolismo , Células Precursoras Eritroides/patologia , Células Precursoras Eritroides/virologia , Eritropoetina/metabolismo , Eritropoetina/farmacologia , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Humanos , Interleucina-3/farmacologia , Leucemia Eritroblástica Aguda/metabolismo , Leucemia Eritroblástica Aguda/virologia , Camundongos , Regiões Promotoras Genéticas/genética , Proteínas Proto-Oncogênicas/metabolismo , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Transdução de Sinais/efeitos dos fármacos , Vírus Formadores de Foco no Baço/fisiologia , Fatores de Tempo , Transativadores/metabolismo , Transcrição Gênica , Células Tumorais Cultivadas , Regulação para Cima , Proteínas do Envelope Viral/genética
7.
Oncogene ; 19(55): 6472-81, 2000 Dec 18.
Artigo em Inglês | MEDLINE | ID: mdl-11175363

RESUMO

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


Assuntos
Transformação Celular Viral/genética , Regulação Neoplásica da Expressão Gênica , Regulação Viral da Expressão Gênica , Família Multigênica , Oncogenes , Proteínas Proto-Oncogênicas , Retroviridae/genética , Células 3T3 , Alpharetrovirus/genética , Alpharetrovirus/fisiologia , Animais , Vírus da Mieloblastose Aviária/genética , Vírus da Mieloblastose Aviária/fisiologia , Galinhas , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/fisiologia , Fibroblastos/metabolismo , Hematopoese/genética , Humanos , Vírus da Leucemia Murina/genética , Vírus da Leucemia Murina/fisiologia , Camundongos , Mutagênese Insercional , Proteína Proto-Oncogênica c-fli-1 , Provírus/genética , Retroviridae/fisiologia , Vírus Formadores de Foco no Baço/genética , Vírus Formadores de Foco no Baço/fisiologia , Transativadores/genética , Transativadores/fisiologia
8.
Int J Biochem Cell Biol ; 31(10): 1089-109, 1999 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-10582341

RESUMO

The proliferation and differentiation of erythroid cells is a highly regulated process that is controlled primarily at the level of interaction of erythropoietin (Epo) with its specific cell surface receptor (EpoR). However, this process is deregulated in mice infected with the Friend spleen focus-forming virus (SFFV). Unlike normal erythroid cells, erythroid cells from SFFV-infected mice are able to proliferate and differentiate in the absence of Epo, resulting in erythroid hyperplasia and leukemia. Over the past 20 years, studies have been carried out to identify the viral genes responsible for the pathogenicity of SFFV and to understand how expression of these genes leads to the deregulation of erythropoiesis in infected animals. The studies have revealed that SFFV encodes a unique envelope glycoprotein which interacts specifically with the EpoR at the cell surface, resulting in activation of the receptor and subsequent activation of erythroid signal transduction pathways. This leads to the proliferation and differentiation of erythroid precursor cells in the absence of Epo. Although the precise mechanism by which the viral protein activates the EpoR is not yet known, it has been proposed that it causes dimerization of the receptor, resulting in constitutive activation of Epo signal transduction pathways. While interaction of the SFFV envelope glycoprotein with the EpoR leads to Epo-independent erythroid hyperplasia, this is not sufficient to transform these cells. Transformation requires the viral activation of the cellular gene Sfpi-1, whose product is thought to block erythroid cell differentiation. By understanding how SFFV can deregulate erythropoiesis, we may gain insights into the causes and treatment of related diseases in man.


Assuntos
Eritropoese , Leucemia Eritroblástica Aguda/fisiopatologia , Leucemia Eritroblástica Aguda/virologia , Vírus Formadores de Foco no Baço/fisiologia , Animais , Transformação Celular Viral , Células Precursoras Eritroides/patologia , Células Precursoras Eritroides/virologia , Humanos , Hiperplasia , Glicoproteínas de Membrana/genética , Glicoproteínas de Membrana/metabolismo , Glicoproteínas de Membrana/fisiologia , Camundongos , Receptores da Eritropoetina/metabolismo , Proteínas do Envelope Viral/genética , Proteínas do Envelope Viral/metabolismo , Proteínas do Envelope Viral/fisiologia
9.
Arch Virol ; 144(1): 73-87, 1999.
Artigo em Inglês | MEDLINE | ID: mdl-10076510

RESUMO

The genomes of all replication-competent retroviruses contain cis-acting elements that regulate gene expression. However, the identities of many of these elements remain to be characterized. Inserting sequences into the 3' untranslated region of a replication-competent spleen necrosis virus (SNV) vector disrupted its ability to replicate. Inserts varying in sequences and sizes (0.4-kb to 1.6-kb) all resulted in this defect. Genetic compensation experiments and immunostaining revealed that env gene expression was deficient. Northern analysis indicated the presence of spliced viral mRNA of the correct size although at a reduced level compared to a wildtype vector. It is likely that the block in env expression occurs at a post-transcriptional step. These results suggest that the function of a cis-acting element distinct from the constitutive transport element is disrupted by the inserted sequences into the 3' untranslated region of SNV.


Assuntos
Regiões 3' não Traduzidas/genética , Regulação Viral da Expressão Gênica , Genes env , Vírus Formadores de Foco no Baço/fisiologia , Replicação Viral/genética , Elementos de DNA Transponíveis/genética , Vetores Genéticos
10.
Leukemia ; 11 Suppl 3: 160-1, 1997 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-9209329

RESUMO

Friend spleen focus-forming virus (F-SFFV) causes acute erythroleukemia in adult mice. F-SFFV encodes an envelope protein-like membrane glycoprotein called gp55 in its defective env gene. Gp55 is responsible for the early stage of leukemogenesis by F-SFFV by specifically binding to and activating the murine erythropoietin receptor (EPO-R). Gp55 has a polytropic env sequence in its N-terminal portion. This portion probably contains the binding site for the EPO-R. In order to obtain a clue for the structure of the binding site to the EPO-R, we isolated and analyzed many spontaneous revertant F-SFFVs which derived from the non-leukemogenic mutant F-SFFV having an ecotropic env sequence instead of the polytropic env sequence in its gp55 gene.


Assuntos
Genes env , Vírus Formadores de Foco no Baço/fisiologia , Proteínas do Envelope Viral/biossíntese , Animais , Animais Recém-Nascidos , Análise Mutacional de DNA , DNA Viral/metabolismo , Produtos do Gene env/biossíntese , Produtos do Gene env/metabolismo , Leucemia Eritroblástica Aguda/virologia , Camundongos , Camundongos Endogâmicos DBA , Peso Molecular , Policitemia , Receptores da Eritropoetina/metabolismo , Proteínas Recombinantes/biossíntese , Vírus Formadores de Foco no Baço/genética , Esplenomegalia , Proteínas do Envelope Viral/metabolismo
11.
Leukemia ; 11 Suppl 3: 251-4, 1997 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-9209356

RESUMO

The erythroleukemia-inducing Friend spleen focus-forming virus (SFFV) encodes a unique envelope glycoprotein which allows erythroid cells to proliferate and differentiate in the absence of the erythroid hormone erythropoietin (Epo). In an attempt to understand how the virus alters the growth of erythroid cells, studies were carried out to determine if virus infection leads to the constitutive activation of the Jak-Stat pathway, one of the signal transduction pathways activated by Epo. Our data indicates that expression of SFFV in erythroid cells leads to the constitutive activation of the same Stat proteins that are transiently activated by Epo. While constitutive activation of Stat proteins by SFFV is associated with Epo-independent proliferation of splenic erythroid progenitor cells from Fv-2-sensitive mice and Epo-dependent HCD-57 cells, it is not sufficient to induce their differentiation. Although constitutive activation of the same Stat proteins is detected in erythroid cells from SFFV-infected Fv-2-resistant mice, it does not lead to their Epo-independent growth. It is also not required for transformation of erythroid cells by SFFV. Studies are in progress to identify the mechanism by which Stat proteins are phosphorylated in SFFV-infected cells in the absence of Epo. Although it has been shown that Epo activates Stat proteins through Jak2 kinase, our results suggest that the SFFV-induced Stat protein activation is Jak2-independent.


Assuntos
Proteínas de Ligação a DNA/metabolismo , Genes fos , Células-Tronco Hematopoéticas/fisiologia , Leucemia Eritroblástica Aguda/fisiopatologia , Transdução de Sinais/fisiologia , Vírus Formadores de Foco no Baço/fisiologia , Replicação Viral , Animais , Diferenciação Celular , Linhagem Celular , Eritropoese , Eritropoetina/farmacologia , Células-Tronco Hematopoéticas/citologia , Células-Tronco Hematopoéticas/virologia , Leucemia Eritroblástica Aguda/virologia , Camundongos , Proteínas Quinases/metabolismo , Transdução de Sinais/efeitos dos fármacos , Replicação Viral/efeitos dos fármacos
12.
Leukemia ; 11 Suppl 3: 432-4, 1997 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-9209415

RESUMO

Friend spleen focus forming-virus (F-SFFV) induces acute erythroleukemia in susceptible mice. Initiation of the erythroleukemia is due to binding of the env-related glycoprotein gp55 encoded by F-SFFV to the erythropoietin receptor (EPOR). The gp55/EPOR interaction induces prolonged and growth factor independent proliferation in a factor-dependent cell line. In erythropoietin (EPO) signaling, the JAK2/STAT5 pathway was shown to be activated downstream of the EPOR to transmit the signal to the cells. To determine members of the JAK family and the STAT transcription factor family involved in the gp55/EPOR signaling, we examined tyrosine phosphorylation of JAKs and STATs in F-SFFV-infected erythroid or erythroleukemic cells. JAK1 and STAT5 were constitutively tyrosine-phosphorylated but the DNA binding activity of STAT5 was not induced without EPO stimulation in erythroblastoid cells from spleens of F-SFFV-infected mice and erythroleukemia cell lines derived from gp55-transgenic mice. These results indicate that JAK1 is involved in the gp55/EPOR signaling but STAT5 is not playing an essential role in the growth of those erythroid cells.


Assuntos
Proteínas de Ligação a DNA/metabolismo , Vírus da Leucemia Murina de Friend/fisiologia , Proteínas do Leite , Proteínas Tirosina Quinases/metabolismo , Proteínas Proto-Oncogênicas , Receptores da Eritropoetina/metabolismo , Transdução de Sinais , Vírus Formadores de Foco no Baço/fisiologia , Transativadores/metabolismo , Proteínas do Envelope Viral/metabolismo , Animais , Ativação Enzimática , Genes env , Janus Quinase 1 , Janus Quinase 2 , Leucemia Eritroblástica Aguda/metabolismo , Camundongos , Camundongos Endogâmicos DBA , Ligação Proteica , Fator de Transcrição STAT5 , Baço , Fatores de Transcrição
14.
Blood ; 85(6): 1454-62, 1995 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-7888668

RESUMO

The signal transduction mechanism of erythropoietin (Epo), which regulates growth and differentiation of erythroid cells, is still unclear. Recent studies showing the activation by various ligands of a group of proteins called Stat (signal transducers and activators of transcription) proteins raised the possibility that such proteins may also be involved in the Epo signal transduction pathway. In this report, we show that Epo induces factors that specifically bind to the sis-inducible element and the gamma response region of the Fc gamma receptor factor I gene in the Epo-dependent mouse erythroleukemia cell line HCD-57. These factors contain phosphotyrosine and antibodies against Stat1 and Stat3 proteins reacted with them. In HCD-57 cells infected with Friend spleen focus-forming virus, which now grow in an Epo-independent manner, the DNA-binding factors were constitutively activated even in the absence of Epo. These results suggest that the factors induced by Epo contain components identical or related to known Stat proteins. It is also suggested that continuous activation of these DNA-binding factors may be responsible for the ability of spleen focus-forming virus to abrogate the Epo-dependence of HCD-57 cells and cause erythroleukemia in susceptible mice.


Assuntos
DNA/metabolismo , Eritropoetina/farmacologia , Vírus Formadores de Foco no Baço/fisiologia , Animais , Sequência de Bases , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/fisiologia , Leucemia Eritroblástica Aguda/metabolismo , Camundongos , Dados de Sequência Molecular , RNA Mensageiro/análise , Receptores de IgG/genética , Fator de Transcrição STAT1 , Fator de Transcrição STAT3 , Transativadores/genética , Transativadores/fisiologia , Células Tumorais Cultivadas
15.
J Mol Med (Berl) ; 73(3): 113-20, 1995 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-7633947

RESUMO

Ping-pong amplification is an efficient process by which helper-free retrovirions replicate in cocultures of cell lines that package retroviruses into distinct host-range envelopes [11]. Transfection of a retroviral vector DNA into these cocultures results in massive virus production, with potentially endless cross-infection between different types of packaging cells. Because the helper-free virus spreads efficiently throughout the coculture, it is unnecessary to use dominant selectable marker genes, and the retroviral vectors can be simplified and optimized for expressing a single gene of interest. The most efficient ping-pong vector, pSFF, derived from the Friend erythroleukemia virus, has been used for high-level expression of several genes that could not be expressed with commonly employed two-gene retroviral vectors. Contrary to previous claims, problems of vector recombination are not inherent to ping-pong methods. Indeed, the pSFF vector has not formed replication-competent recombinants as shown by stringent assays. Here we review these methods, characterize the ping-pong process using the human erythropoietin gene as a model, and describe a new vector (pSFY) designed for enhanced expression in T lymphocytes. Factors that limit tissue-specific expression are reviewed.


Assuntos
Expressão Gênica , Vetores Genéticos/genética , Retroviridae/genética , Transfecção , Animais , Eritropoetina/genética , Humanos , Camundongos , Vírus da Leucemia Murina de Moloney/genética , Provírus/genética , Provírus/fisiologia , Sequências Repetitivas de Ácido Nucleico/genética , Retroviridae/fisiologia , Vírus Formadores de Foco no Baço/genética , Vírus Formadores de Foco no Baço/fisiologia , Células Tumorais Cultivadas
16.
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
17.
J Biol Chem ; 268(8): 5741-7, 1993 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-8449938

RESUMO

The membrane glycoprotein (gp55) encoded by the env gene of Friend spleen focus-forming virus (SFFV) causes mitogenesis of infected erythroblasts and is inefficiently (3-5%) processed from the rough endoplasmic reticulum (RER) to plasma membranes. Recent evidence suggested that gp55 binds to erythropoietin receptors (EpoR) in the RER, and it was proposed that this intracellular interaction causes mitogenesis (Li, J.-P., D'Andrea, A. D., Lodish, H. F., and Baltimore, D. (1990) Nature 343, 762-764). Other evidence has indicated that the plasma membrane component (gp55P) can also complex with EpoR. To identify the site of functional complexes and to study the factors that control their formation, we analyzed eight SFFV env mutants that contain in-frame deletions or linker insertions. The three nonpathogenic mutants encode gp55s that fail to leave the RER, whereas the five pathogenic mutants encode glycoproteins that occur on cell surfaces. Although BaF3 hematopoietic cells are interleukin 3 (IL-3)-dependent, a cellular derivative BaF3/EpoR that contains EpoR survives and grows in either IL-3 or erythropoietin (Epo). The five pathogenic SFFV env mutants converted BaF3/EpoR but not BaF3 cells to growth factor independence, whereas the nonpathogenic mutants did not eliminate growth factor requirements of any cells. Studies using 125I-Epo and the covalent cross-linking reagent disuccinimidyl suberate provided unambiguous evidence for ternary complexes of 125I-Epo.EpoR.gp55P on surfaces of all factor-independent cells. Size of the cell surface complex was correspondingly reduced in the case of a newly isolated SFFV mutant that encodes a severely truncated (M(r) approximately 41,000) but mitogenically active env glycoprotein. Our results support the hypothesis that activation of EpoR by the SFFV env glycoprotein occurs exclusively on cell surfaces.


Assuntos
Receptores da Eritropoetina/metabolismo , Vírus Formadores de Foco no Baço/genética , Proteínas do Envelope Viral/metabolismo , Células 3T3 , Sequência de Aminoácidos , Animais , Divisão Celular , Linhagem Celular , Membrana Celular/metabolismo , Vírus da Leucemia Murina de Friend/genética , Vírus da Leucemia Murina de Friend/patogenicidade , Genes Virais , Genes env , Camundongos , Mitógenos/metabolismo , Dados de Sequência Molecular , Mutação , Vírus Formadores de Foco no Baço/fisiologia , Proteínas do Envelope Viral/genética , Vírion
18.
Blood ; 80(12): 3070-8, 1992 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-1467516

RESUMO

The erythropoietin receptor (EPO-R) can be activated to signal cell growth by binding either EPO or gp55, the Friend spleen focus-forming virus (SFFV) glycoprotein. EPO binding induces tyrosine kinase activity and rapid tyrosine phosphorylation of several cellular substrates. To test for gp55-induced tyrosine kinase activity, we performed immunoblots on two murine cell lines that stably express EPO-R and gp55. Stimulation of the parental cell line, Ba/F3, with murine interleukin-3 (IL-3) resulted in rapid, dose-dependent tyrosine phosphorylation of a 97-Kd substrate. Stimulation with IL-3 or EPO of the Ba/F3 cells expressing the recombinant EPO-R (Ba/F3-EPO-R) resulted in tyrosine phosphorylation of the same p97 substrate. These latter cells, when transformed to growth factor-independence by the Friend gp55 glycoprotein, exhibited constitutive tyrosine phosphorylation of the 97-Kd substrate. Other growth factor-independent Ba/F3 subclones, transformed with either the oncoprotein, v-abl, or with a constitutively activated EPO-R, also had constitutive phosphorylation of a 97-Kd substrate. In CTLL-2-EPO-R cells, a T-lymphocyte line stably transfected with the EPO-R, the 97-Kd substrate was tyrosine-phosphorylated in response to IL-2 or EPO. The 97-Kd protein was constitutively phosphorylated in CTLL-2-EPO-R-gp55 cells. In conclusion, a 97-Kd protein found in two murine cell lines is tyrosine-phosphorylated in response to multiple growth factors and viral oncoproteins, and appears to be a central phosphoprotein in signal transduction.


Assuntos
Eritropoetina/farmacologia , Fosfoproteínas/metabolismo , Proteínas Tirosina Quinases/metabolismo , Receptores da Eritropoetina/efeitos dos fármacos , Receptores da Eritropoetina/fisiologia , Vírus Formadores de Foco no Baço/fisiologia , Proteínas do Envelope Viral/metabolismo , Animais , Divisão Celular/efeitos dos fármacos , Linhagem Celular , Eletroforese em Gel de Poliacrilamida , Humanos , Interleucina-2/farmacologia , Interleucina-3/farmacologia , Camundongos , Peso Molecular , Fosfoproteínas/isolamento & purificação , Fosforilação , Proteínas Recombinantes/farmacologia , Acetato de Tetradecanoilforbol/farmacologia
19.
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
20.
Int J Hematol ; 54(2): 117-24, 1991 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-1660735

RESUMO

Mice infected with the polycythemia-inducing strain of the Friend virus complex (FVC-P) have been used as a leukemic mouse model. In the present study, purified iron-saturated human lactoferrin (LF) and recombinant murine (rmu) interferon-gamma (IFN-gamma), alone or in combination, were used to influence disease progression in virally infected mice. DBA/2 mice were injected i.v. with FVC-P, and were treated s.c. with 100 micrograms LF at day 7, and/or rmuIFN-gamma at 5 x 10(4) units/day for 3 days beginning at day 6 after viral infection. Mice were assessed for survival, and also 14 days after virus inoculation, the mice were killed and spleen extracts were assessed for spleen focus forming virus (SFFV) titers by spleen focus forming unit (SFFU) assay, SFFV mRNA and genomic DNA expression, and natural killer (NK) cell activity. Treatment with LF or rmuIFN-gamma alone had little or no effect on SFFU numbers or SFFV mRNA or genomic DNA expression. However, dramatically decreased SFFV titers and levels of SFFV mRNA and genomic DNA were observed in mice treated with the combination of LF and rmuIFN-gamma. NK cell activity decreased by FVC-P was returned to normal levels by LF and rmuIFN-gamma. The combined treatment also enhanced the survival rates of FVC-P-infected mice. The results suggest synergistic suppressive effects of LF with rmuIFN-gamma on disease progression in FVC-P-infected mice. This information might be of significance as a potential therapy for patients with leukemia and those infected with retroviruses.


Assuntos
Antivirais/uso terapêutico , Vírus da Leucemia Murina de Friend , Interferon gama/uso terapêutico , Lactoferrina/uso terapêutico , Leucemia Experimental/tratamento farmacológico , Policitemia/tratamento farmacológico , Infecções por Retroviridae/tratamento farmacológico , Animais , Antivirais/farmacologia , Sinergismo Farmacológico , Feminino , Interferon gama/farmacologia , Células Matadoras Naturais/imunologia , Lactoferrina/farmacologia , Leucemia Experimental/imunologia , Leucemia Experimental/microbiologia , Camundongos , Camundongos Endogâmicos DBA , Tamanho do Órgão/efeitos dos fármacos , Policitemia/imunologia , Policitemia/microbiologia , Proteínas Recombinantes , Infecções por Retroviridae/imunologia , Infecções por Retroviridae/microbiologia , Baço/patologia , Vírus Formadores de Foco no Baço/isolamento & purificação , Vírus Formadores de Foco no Baço/fisiologia , Replicação Viral/efeitos dos fármacos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA