Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 28
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Reprod Fertil Dev ; 29(3): 521-543, 2017 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-28442062

RESUMO

Restricted availability of retinoic acid (RA) in the testicular milieu regulates transcriptional activity of c-kit (KIT, CD117), which aids in the determination of spermatogonial stem-cell differentiation. The effect of RA on c-kit has been reported previously, but its mode of genomic action remains unresolved. We studied the molecular machinery guiding RA responsiveness to the c-kit gene using spermatogonial stem-cell line C18-4 and primary spermatogonial cells. A novel retinoic acid response element (RARE) positioned at -989 nucleotides upstream of the transcription start site (TSS) was identified, providing a binding site for a dimeric RA receptor (i.e. retinoic acid receptor gamma (RARγ) and retinoic X receptor). RA treatment influenced c-kit promoter activity, along with endogenous c-kit expression in C18-4 cells. A comprehensive promoter deletion assay using the pGL3B reporter system characterised the region spanning -271bp and -1011bp upstream of the TSS, which function as minimal promoter and maximal promoter, respectively. In silico analysis predicted that the region -1011 to +58bp comprised the distal enhancer RARE and activators such as spleen focus forming virus proviral integration oncogene (SPFI1) (PU.1), specificity protein 1 (SP1) and four E26 transformation-specific (ETS) tandem binding sites at the proximal region. Gel retardation and chromatin immunoprecipitation (ChIP) assays showed binding for RARγ, PU.1 and SP1 to the predicted consensus binding sequences, whereas GABPα occupied only two out of four ETS binding sites within the c-kit promoter region. We propose that for RA response, an enhanceosome is orchestrated through scaffolding of a CREB-binding protein (CBP)/p300 molecule between RARE and elements in the proximal promoter region, controlling germ-line expression of the c-kit gene. This study outlines the fundamental role played by RARγ, along with other non-RAR transcription factors (PU.1, SP1 and GABPα), in the regulation of c-kit expression in spermatogonial stem cells in response to RA.


Assuntos
Células-Tronco Germinativas Adultas/efeitos dos fármacos , Proteínas Proto-Oncogênicas c-kit/genética , Tretinoína/farmacologia , Células-Tronco Germinativas Adultas/metabolismo , Animais , Sítios de Ligação , Linhagem Celular , Expressão Gênica/efeitos dos fármacos , Humanos , Regiões Promotoras Genéticas , Proteínas Proto-Oncogênicas c-ets/genética , Proteínas Proto-Oncogênicas c-ets/metabolismo , Proteínas Proto-Oncogênicas c-kit/metabolismo , Receptores do Ácido Retinoico/genética , Receptores do Ácido Retinoico/metabolismo , Vírus Formadores de Foco no Baço/genética , Vírus Formadores de Foco no Baço/metabolismo
2.
Stem Cell Reports ; 6(6): 873-884, 2016 06 14.
Artigo em Inglês | MEDLINE | ID: mdl-27161365

RESUMO

We previously reported the generation of integration-free induced pluripotent stem cells from adult peripheral blood (PB) with an improved episomal vector (EV) system, which uses the spleen focus-forming virus U3 promoter and an extra factor BCL-XL (B). Here we show an ∼100-fold increase in efficiency by optimizing the vector combination. The two most critical factors are: (1) equimolar expression of OCT4 (O) and SOX2 (S), by using a 2A linker; (2) a higher and gradual increase in the MYC (M) to KLF4 (K) ratio during the course of reprogramming, by using two individual vectors to express M and K instead of one. The combination of EV plasmids (OS + M + K + B) is comparable with Sendai virus in reprogramming efficiency but at a fraction of the cost. The generated iPSCs are indistinguishable from those from our previous approach in pluripotency and phenotype. This improvement lays the foundation for broad applications of episomal vectors in PB reprogramming.


Assuntos
Reprogramação Celular , Engenharia Genética/métodos , Vetores Genéticos/metabolismo , Células-Tronco Pluripotentes Induzidas/metabolismo , Leucócitos Mononucleares/metabolismo , Plasmídeos/metabolismo , Adulto , Biomarcadores/metabolismo , Diferenciação Celular , Expressão Gênica , Engenharia Genética/economia , Vetores Genéticos/química , Humanos , Células-Tronco Pluripotentes Induzidas/citologia , Fator 4 Semelhante a Kruppel , Fatores de Transcrição Kruppel-Like/genética , Fatores de Transcrição Kruppel-Like/metabolismo , Leucócitos Mononucleares/citologia , Fator 3 de Transcrição de Octâmero/genética , Fator 3 de Transcrição de Octâmero/metabolismo , Plasmídeos/química , Cultura Primária de Células , Regiões Promotoras Genéticas , Proteínas Proto-Oncogênicas c-myc/genética , Proteínas Proto-Oncogênicas c-myc/metabolismo , Fatores de Transcrição SOXB1/genética , Fatores de Transcrição SOXB1/metabolismo , Vírus Formadores de Foco no Baço/genética , Vírus Formadores de Foco no Baço/metabolismo , Proteína bcl-X/genética , Proteína bcl-X/metabolismo
3.
Proc Natl Acad Sci U S A ; 112(29): E3893-900, 2015 Jul 21.
Artigo em Inglês | MEDLINE | ID: mdl-26150503

RESUMO

Substantial advances have been made in the past two decades in the management of osteoporosis. However, none of the current medications can eliminate the risk of fracture and rejuvenate the skeleton. To this end, we recently reported that transplantation of hematopoietic stem/progenitor cells (HSCs) or Sca1(+) cells engineered to overexpress FGF2 results in a significant increase in lamellar bone matrix formation at the endosteum; but this increase was attended by the development of secondary hyperparathyroidism and severe osteomalacia. Here we switch the therapeutic gene to PDGFB, another potent mitogen for mesenchymal stem cells (MSCs) but potentially safer than FGF2. We found that modest overexpression of PDGFB using a relatively weak phosphoglycerate kinase (PGK) promoter completely avoided osteomalacia and secondary hyperparathyroidism, and simultaneously increased trabecular bone formation and trabecular connectivity, and decreased cortical porosity. These effects led to a 45% increase in the bone strength. Transplantation of PGK-PDGFB-transduced Sca1(+) cells increased MSC proliferation, raising the possibility that PDGF-BB enhances expansion of MSC in the vicinity of the hematopoietic niche where the osteogenic milieu propels the differentiation of MSCs toward an osteogenic destination. Our therapy should have potential clinical applications for patients undergoing HSC transplantation, who are at high risk for osteoporosis and bone fractures after total body irradiation preconditioning. It could eventually have wider application once the therapy can be applied without the preconditioning.


Assuntos
Osso e Ossos/fisiopatologia , Terapia Genética , Transplante de Células-Tronco Hematopoéticas , Proteínas Proto-Oncogênicas c-sis/genética , Proteínas Proto-Oncogênicas c-sis/uso terapêutico , Fosfatase Alcalina/sangue , Animais , Antígenos Ly/metabolismo , Peso Corporal , Remodelação Óssea , Diferenciação Celular , Proliferação de Células , Hiperparatireoidismo/complicações , Hiperparatireoidismo/metabolismo , Hiperparatireoidismo/fisiopatologia , Hiperparatireoidismo/terapia , Antígeno Ki-67/metabolismo , Lentivirus/metabolismo , Proteínas de Membrana/metabolismo , Células-Tronco Mesenquimais/citologia , Camundongos , Modelos Biológicos , Neovascularização Fisiológica , Osteoblastos/metabolismo , Osteoblastos/patologia , Osteocalcina/sangue , Osteogênese , Osteomalacia/complicações , Osteomalacia/fisiopatologia , Fosfoglicerato Quinase/genética , Fosfoglicerato Quinase/metabolismo , Regiões Promotoras Genéticas/genética , Vírus Formadores de Foco no Baço/metabolismo , Transdução Genética , Transgenes , Suporte de Carga
4.
Exp Hematol ; 42(9): 793-803.e1, 2014 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-24746876

RESUMO

Mesenchymal stromal cells (MSCs) are a promising treatment modality for a variety of diseases. Strategies to investigate the fate of MSCs in vivo are important to unravel their therapeutic mechanisms. However, currently available techniques are hampered by their low sensitivity. We therefore aimed to optimize in vivo bioluminescence imaging of MSCs. We compared MSCs transduced with firefly luciferase (Fluc) and transmembrane-bound Gaussia luciferase driven by the human cytomegalovirus, spleen focus-forming virus (SFFV), and elongation factor 1-α (EF1α) promoters. Although cytomegalovirus-transmembrane-bound Gaussia luciferase-transduced MSCs showed the highest light intensity in vitro, the signal was almost undetectable in vivo. Spleen focus-forming virus-Fluc-transduced MSCs revealed a bright signal in vivo, but transgene expression was silenced upon in vitro stimulation with interferon (IFN)-γ. Therefore, the SFFV promoter was replaced by the EF1α promoter. Light emission of Fluc under the control of EF1α was similar to SFFV-Fluc. Although EF1α-Fluc light emission was decreased tenfold in the presence of IFN-γ when compared with unstimulated MSCs, the bioluminescent signal could still be detected and was clearly distinguishable from untransduced MSCs. Furthermore, stimulation of MSCs with tumor necrosis factor-α hardly affected transgene expression in EF1α-Fluc-transduced MSCs. Thus, the use of the EF1α promoter partially overcomes silencing and allows in vivo bioluminescence imaging of IFN-γ-stimulated MSCs.


Assuntos
Antivirais/farmacologia , Genes Reporter , Interferon gama/farmacologia , Luciferases de Vaga-Lume/biossíntese , Medições Luminescentes/métodos , Células-Tronco Mesenquimais/metabolismo , Fator 1 de Elongação de Peptídeos/genética , Regiões Promotoras Genéticas , Animais , Citomegalovirus/genética , Citomegalovirus/metabolismo , Humanos , Luciferases de Vaga-Lume/genética , Masculino , Células-Tronco Mesenquimais/citologia , Camundongos , Camundongos Endogâmicos BALB C , Vírus Formadores de Foco no Baço/genética , Vírus Formadores de Foco no Baço/metabolismo , Transdução Genética
5.
Mol Ther ; 19(1): 122-32, 2011 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-20978475

RESUMO

X-linked chronic granulomatous disease (X-CGD) is a primary immunodeficiency caused by mutations in the CYBB gene encoding the phagocyte nicotinamide adenine dinucleotide phosphate (NADPH)-oxidase catalytic subunit gp91(phox). A recent clinical trial for X-CGD using a spleen focus-forming virus (SFFV)-based γ-retroviral vector has demonstrated clear therapeutic benefits in several patients although complicated by enhancer-mediated mutagenesis and diminution of effectiveness over time due to silencing of the viral long terminal repeat (LTR). To improve safety and efficacy, we have designed a lentiviral vector that directs transgene expression primarily in myeloid cells. To this end, we created a synthetic chimeric promoter that contains binding sites for myeloid transcription factors CAAT box enhancer-binding family proteins (C/EBPs) and PU.1, which are highly expressed during granulocytic differentiation. As predicted, the chimeric promoter regulated higher reporter gene expression in myeloid than in nonmyeloid cells, and in human hematopoietic progenitors upon granulocytic differentiation. In a murine model of stem cell gene therapy for X-CGD, the chimeric vector resulted in high levels of gp91(phox) expression in committed myeloid cells and granulocytes, and restored normal NADPH-oxidase activity. These findings were recapitulated in human neutrophils derived from transduced X-CGD CD34(+) cells in vivo, and suggest that the chimeric promoter will have utility for gene therapy of myeloid lineage disorders such as CGD.


Assuntos
Catepsina G/genética , Doença Granulomatosa Crônica/genética , Doença Granulomatosa Crônica/terapia , Células Mieloides/fisiologia , Proteínas Proto-Oncogênicas c-fes/genética , Proteínas Recombinantes de Fusão/genética , Transgenes , Animais , Sequência de Bases , Sítios de Ligação , Proteínas Estimuladoras de Ligação a CCAAT/metabolismo , Diferenciação Celular/genética , Linhagem Celular Tumoral , Variações do Número de Cópias de DNA , Elementos Facilitadores Genéticos , Regulação da Expressão Gênica , Genes Ligados ao Cromossomo X , Terapia Genética/métodos , Vetores Genéticos/administração & dosagem , Vetores Genéticos/genética , Vetores Genéticos/metabolismo , Granulócitos/metabolismo , Doença Granulomatosa Crônica/enzimologia , Doença Granulomatosa Crônica/metabolismo , Células-Tronco Hematopoéticas/metabolismo , Humanos , Lentivirus/genética , Lentivirus/metabolismo , Camundongos , Dados de Sequência Molecular , Mutagênese/genética , Células Mieloides/metabolismo , NADPH Oxidases/genética , NADPH Oxidases/metabolismo , Regiões Promotoras Genéticas , Proteínas Proto-Oncogênicas/metabolismo , Receptores Imunológicos/genética , Receptores Imunológicos/metabolismo , Proteínas Recombinantes de Fusão/biossíntese , Proteínas Recombinantes de Fusão/metabolismo , Retroviridae/genética , Retroviridae/metabolismo , Vírus Formadores de Foco no Baço/genética , Vírus Formadores de Foco no Baço/metabolismo , Células-Tronco/metabolismo , Sequências Repetidas Terminais , Transativadores/metabolismo
6.
Hum Gene Ther ; 21(5): 623-30, 2010 May.
Artigo em Inglês | MEDLINE | ID: mdl-20001454

RESUMO

Fanconi anemia (FA) is an inherited genetic disease characterized mainly by bone marrow failure and cancer predisposition. Although gene therapy may constitute a good therapeutic option for many patients with FA, none of the clinical trials so far developed has improved the clinical status of these patients. We have proposed strategies for the genetic correction of bone marrow grafts from patients with FA, using lentiviral vectors (LVs). Here we investigate the relevance of the expression of FANCA to confer a therapeutic effect in cells from patients with FA-A, the most frequent complementation group in FA. Our data show that relatively weak promoters such as the vav or phosphoglycerate kinase (PGK) promoter confer, per copy of FANCA, physiological levels of FANCA mRNA in lymphoblastoid cell lines, whereas the cytomegalovirus and, more significantly, spleen focus-forming virus (SFFV) promoters mediated the expression of supraphysiological levels of FANCA mRNA. Insertion of the woodchuck hepatitis virus posttranscriptional regulatory element (WPRE) or a mutated WPRE into the 3' region of PGK-FANCA LVs significantly increased FANCA mRNA levels. At the protein level, however, all tested vectors conferred, per copy of FANCA, similar and physiological levels of the protein, except SFFV LVs, which again conferred supraphysiological levels of FANCA. In spite of their different activity, all tested vectors mediated a similar phenotypic correction in FA-A lymphoblastoid cell lines and also in hematopoietic progenitors from patients with FA-A. On the basis of the efficacy and safety properties of PGK LVs, a PGK LV carrying FANCA and a mutant WPRE is proposed as an optimized vector for the gene therapy of patients with FA-A.


Assuntos
Anemia de Fanconi/terapia , Terapia Genética , Vetores Genéticos/genética , Linhagem Celular , Anemia de Fanconi/genética , Anemia de Fanconi/metabolismo , Humanos , Mutação , Proteínas/genética , Proteínas/metabolismo , Proteínas Proto-Oncogênicas c-vav , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Vírus Formadores de Foco no Baço/genética , Vírus Formadores de Foco no Baço/metabolismo
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.
J Virol ; 75(17): 7893-903, 2001 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-11483734

RESUMO

The Friend spleen focus-forming virus (SFFV) encodes a unique envelope glycoprotein, gp55, which allows erythroid cells to proliferate and differentiate in the absence of erythropoietin (Epo). SFFV gp55 has been shown to interact with the Epo receptor complex, causing constitutive activation of various signal-transducing molecules. When injected into adult mice, SFFV induces a rapid erythroleukemia, with susceptibility being determined by the host gene Fv-2, which was recently shown to be identical to the gene encoding the receptor tyrosine kinase Stk/Ron. Susceptible, but not resistant, mice encode not only full-length Stk but also a truncated form of the kinase, sf-Stk, which may mediate the biological effects of SFFV infection. To determine whether expression of SFFV gp55 leads to the activation of sf-Stk, we expressed sf-Stk, with or without SFFV gp55, in hematopoietic cells expressing the Epo receptor. Our data indicate that sf-Stk interacts with SFFV gp55 as well as gp55(P), the biologically active form of the viral glycoprotein, forming disulfide-linked complexes. This covalent interaction, as well as noncovalent interactions with SFFV gp55, results in constitutive tyrosine phosphorylation of sf-Stk and its association with multiple tyrosine-phosphorylated signal-transducing molecules. In contrast, neither Epo stimulation in the absence of SFFV gp55 expression nor expression of a mutant of SFFV that cannot interact with sf-Stk was able to induce tyrosine phosphorylation of sf-Stk or its association with any signal-transducing molecules. Covalent interaction of sf-Stk with SFFV gp55 and constitutive tyrosine phosphorylation of sf-Stk can also be detected in an erythroleukemia cell line derived from an SFFV-infected mouse. Our results suggest that SFFV gp55 may mediate its biological effects in vivo by interacting with and activating a truncated form of the receptor tyrosine kinase Stk.


Assuntos
Receptores Proteína Tirosina Quinases/química , Receptores Proteína Tirosina Quinases/metabolismo , Receptores de Superfície Celular/química , Receptores de Superfície Celular/metabolismo , Vírus Formadores de Foco no Baço/metabolismo , Proteínas do Envelope Viral/metabolismo , Animais , Leucemia Experimental/virologia , Camundongos , Mutação , Fosforilação , Plasmídeos/genética , Receptores da Eritropoetina/metabolismo , Infecções por Retroviridae/virologia , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transdução de Sinais , Vírus Formadores de Foco no Baço/genética , Células Tumorais Cultivadas , Infecções Tumorais por Vírus/virologia , Tirosina/metabolismo , Proteínas do Envelope Viral/genética
9.
Dev Dyn ; 219(4): 505-13, 2000 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-11084650

RESUMO

In amniote embryos, cells from a rostral portion of the primitive streak migrate anterolaterally and establish the heart field mesoderm, from which two cardiac cell lineages, cardiomyocytes and endocardial endothelial cells, differentiate. The endoderm underlying the heart field has been postulated as the source of several paracrine factors that may serve to induce each of these cell types. However, it has been unclear how these signal molecules, which are expressed broadly in the endoderm, instruct individual cells of the heart field mesoderm to enter either the cardiomyocyte lineage or the endocardial cell lineage. To clarify lineage relationships of these two cardiac cell types, the fate of chicken primitive streak cells was traced for the first time in ovo. By using replication-defective retroviral-mediated gene transfer, we demonstrate that cells in the rostral half of Hamburger and Hamilton (HH) stage 3 primitive streak generate a daughter population that proliferates and migrates into the heart field, differentiating into either endocardial or myocardial cells, but not both cell types. The results suggest that the rostral portion of the primitive streak at HH stage 3 consists of at least two distinct subpopulations, entering either the cardiomyocyte lineage or the endocardial cell lineage. Thus, in ovo these two cell lineages of the heart are already segregated within the primitive streak, significantly before their migration to the heart field. When the precardiomyocytes and pre-endocardial cells arrive at the heart field, each mesodermal cell subpopulation may be permissive to paracrine signal(s) from underlying endoderm to initiate their terminal differentiation into either muscle or endothelial cells.


Assuntos
Diferenciação Celular , Gástrula/citologia , Coração/embriologia , Animais , Linhagem da Célula , Embrião de Galinha , Indução Embrionária , Endocárdio/citologia , Endocárdio/embriologia , Gástrula/fisiologia , Técnicas de Transferência de Genes , Morfogênese , Miocárdio/citologia , Vírus Formadores de Foco no Baço/genética , Vírus Formadores de Foco no Baço/metabolismo , beta-Galactosidase/análise , beta-Galactosidase/genética
10.
J Virol ; 74(18): 8444-51, 2000 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-10954544

RESUMO

Interaction of erythropoietin (Epo) with its cell surface receptor activates signal transduction pathways which result in the proliferation and differentiation of erythroid cells. Infection of erythroid cells with the Friend spleen focus-forming virus (SFFV) leads to the interaction of the viral envelope glycoprotein with the Epo receptor and renders these cells Epo independent. We previously reported that SFFV induces Epo independence by constitutively activating components of several Epo signal transduction pathways, including the Jak-Stat and the Raf-1/mitogen-activated protein kinase (MAPK) pathways. To further evaluate the mechanism by which SFFV activates the Raf-1/MAPK pathway, we investigated the effects of SFFV on upstream components of this pathway, and our results indicate that SFFV activates Shc and Grb2 and that this leads to Ras activation. While studies with a dominant-negative Ras indicated that Ras was required for Epo-induced proliferation of normal erythroid cells, the Epo-independent growth of SFFV-infected cells can still occur in the absence of Ras, although at reduced levels. In contrast, protein kinase C (PKC) was shown to be required for the Epo-independent proliferation of SFFV-infected cells. Further studies indicated that PKC, which is thought to be involved in the activation of both Raf-1 and MAPK, was required only for the activation of MAPK, not Raf-1, in SFFV-infected cells. Our results indicate that Ras and PKC define two distinct signals converging on MAPK in both Epo-stimulated and SFFV-infected erythroid cells and that activation of only PKC is sufficient for the Epo-independent proliferation of SFFV-infected cells.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal , Proteínas Adaptadoras de Transporte Vesicular , Eritroblastos/metabolismo , Eritropoetina/metabolismo , Guanosina Trifosfato/metabolismo , Proteína Quinase C/metabolismo , Vírus Formadores de Foco no Baço/metabolismo , Sulfonamidas , Proteínas ras/metabolismo , Western Blotting , Divisão Celular , Inibidores Enzimáticos/farmacologia , Receptores ErbB/metabolismo , Eritroblastos/virologia , Proteína Adaptadora GRB2 , Humanos , Indóis/farmacologia , Isoquinolinas/farmacologia , Maleimidas/farmacologia , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Fosforilação , Ligação Proteica , Proteína Quinase C/antagonistas & inibidores , Proteínas/metabolismo , Proteínas Proto-Oncogênicas c-raf/metabolismo , Proteínas Adaptadoras da Sinalização Shc , Transdução de Sinais , Proteína 1 de Transformação que Contém Domínio 2 de Homologia de Src , Estaurosporina/farmacologia , Transfecção , Células Tumorais Cultivadas
11.
Virology ; 246(2): 232-40, 1998 Jul 05.
Artigo em Inglês | MEDLINE | ID: mdl-9657942

RESUMO

A murine erythroleukemogenic retrovirus, Friend spleen focus-forming virus, encodes an envelope protein-like membrane glycoprotein (gp55) in its defective env gene which is responsible for activation of the erythropoietin receptor (EpoR) and the abnormally rapid proliferation of erythroid precursor cells. The S34 mutant gp55, which possesses an additional cytoplasmic domain, is nonpathogenic and its processing to the cell surface is severely reduced compared to that of the wild-type gp55. In this study, we found that the S34 mutant gp55 neither binds to nor activates the EpoR. The S34 mutant gp55 formed disulfide-bonded homodimers in the rough endoplasmic reticulum (RER) membrane much less efficiently than the wild-type gp55, which is consistent with the proposal that homodimer formation is a prerequisite for gp55 to be exported from the RER. We found that the wild-type gp55 that is bound to EpoR in the RER consists of a large number of monomers and a small number of dimers, suggesting that monomers of the S34 mutant gp55 have lost the ability to bind to the EpoR. The 1-bp insertion present in the wild-type gp55 gene, causing a loss of the cytoplasmic domain, is essential for pathogenicity in that it renders the encoded protein capable of both binding to the EpoR and transport to the cell surface.


Assuntos
Glicoproteínas de Membrana/metabolismo , Mutação , Receptores da Eritropoetina/metabolismo , Vírus Formadores de Foco no Baço/metabolismo , Proteínas do Envelope Viral/metabolismo , Animais , Linhagem Celular , Dimerização , Dissulfetos , Eritropoetina/farmacologia , Humanos , Glicoproteínas de Membrana/genética , Camundongos , Mitógenos/farmacologia , Receptores da Eritropoetina/genética , Vírus Formadores de Foco no Baço/genética , Proteínas do Envelope Viral/genética
12.
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
13.
J Virol ; 72(5): 3742-50, 1998 May.
Artigo em Inglês | MEDLINE | ID: mdl-9557656

RESUMO

The Friend spleen focus-forming virus (SFFV) env gene encodes a 409-amino-acid glycoprotein with an apparent Mr of 55,000 (gp55) that binds to erythropoietin receptors (EpoR) to stimulate erythroblastosis. We reported previously the in vivo selection during serial passages in mice of several evolutionary intermediates that culminated in the formation of a novel SFFV (M. E. Hoatlin, E. Gomez-Lucia, F. Lilly, J. H. Beckstead, and D. Kabat, J. Virol. 72:3602-3609, 1998). A mouse injected with a retroviral vector in the presence of a nonpathogenic helper virus developed long-latency erythroblastosis, and subsequent viral passages resulted in more pathogenic isolates. The viruses taken from these mice converted an erythropoietin-dependent cell line (BaF3/EpoR) into factor-independent derivatives. Western blot analysis of cell extracts with an antiserum that broadly reacts with murine retroviral envelope glycoproteins suggested that the spleen from the initial mouse with mild erythoblastosis contained an array of viral components that were capable of activating EpoR. DNA sequence analysis of the viral genomes cloned from different factor-independent cell clones revealed env genes with open reading frames encoding 644, 449, and 187 amino acids. All three env genes contained 3' regions identical to that of SFFV, including a 6-bp duplication and a single-base insertion that have been shown previously to be critical for pathogenesis. However, the three env gene sequences did not contain any polytropic sequences and were divergent in their 5' regions, suggesting that they had originated by recombination and partial deletions of endogenously inherited MuLV env sequences. These results suggest that the requirements for EpoR activation by SFFV-related viruses are dependent on sequences at the 3' end of the env gene and not on the polytropic regions or on the 585-base deletions that are common among the classical strains of SFFV. Moreover, sequence analysis of the different recombinants and deletion mutants revealed that short direct and indirect repeat sequences frequently flanked the deletions that had occurred, suggesting a reverse transcriptase template jumping mechanism for this rapid retroviral diversification.


Assuntos
Produtos do Gene env/genética , Receptores da Eritropoetina/metabolismo , Vírus Formadores de Foco no Baço/genética , Sequência de Aminoácidos , Animais , Sequência de Bases , Linhagem Celular , DNA Viral , Feminino , Produtos do Gene env/metabolismo , Camundongos , Dados de Sequência Molecular , Deleção de Sequência , Homologia de Sequência de Aminoácidos , Vírus Formadores de Foco no Baço/classificação , Vírus Formadores de Foco no Baço/metabolismo
14.
Mol Cell Biol ; 18(3): 1172-80, 1998 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-9488432

RESUMO

Abnormal erythropoietin (EPO)-independent cell growth is induced after infection of erythroid progenitor cells with a polycythemic strain of Friend virus (FVp). Binding of its Env-related glycoprotein (gp55) to the EPO receptor (EPOR) mimics the activation of the EPOR with EPO. We investigated the gp55-EPOR signaling in erythroblastoid cells from mice infected with FVp and in cells of FVp-induced or gp55-transgenic-mouse-derived erythroleukemia cell lines, comparing it with the EPO-EPOR signaling in EPO-responsive erythroblastoid cells. While the Janus protein tyrosine kinase JAK2 and the transcription factor STAT5 became tyrosine phosphorylated with the EPO stimulation in EPO-responsive erythroblastoid cells from anemic mice, JAK1 and STAT5 were constitutively tyrosine phosphorylated in all of these FVp gp55-induced erythroblastoid or erythroleukemic cells. Moreover, this constitutively tyrosine-phosphorylated STAT5 was unable to bind to its specific DNA sequences and did not translocate to the nucleus. Nuclear translocation and DNA binding of this STAT5 species required EPO stimulation. These findings clearly indicate that the FVp gp55-EPOR signaling is distinct from the EPO-EPOR signaling and suggest that STAT5 may not play an essential role in the transmission of the cell growth signals in FVp gp55-induced erythroleukemia cells.


Assuntos
Proteínas de Ligação a DNA/metabolismo , Células Precursoras Eritroides/metabolismo , Eritropoetina/metabolismo , Proteínas do Leite , Proteínas Tirosina Quinases/metabolismo , Proteínas Proto-Oncogênicas , Receptores da Eritropoetina/metabolismo , Vírus Formadores de Foco no Baço/metabolismo , Transativadores/metabolismo , Proteínas do Envelope Viral/metabolismo , Animais , Núcleo Celular/metabolismo , Células Cultivadas , DNA/metabolismo , Ativação Enzimática , Células Precursoras Eritroides/citologia , Eritropoetina/farmacologia , Feminino , Humanos , Janus Quinase 1 , Janus Quinase 2 , Camundongos , Camundongos Endogâmicos DBA , Fosforilação , Receptores da Eritropoetina/genética , Proteínas Recombinantes/metabolismo , Proteínas Recombinantes/farmacologia , Fator de Transcrição STAT5 , Vírus Formadores de Foco no Baço/genética , Células Tumorais Cultivadas , Tirosina/metabolismo , Proteínas do Envelope Viral/genética
15.
In Vivo ; 10(2): 191-9, 1996.
Artigo em Inglês | MEDLINE | ID: mdl-8744800

RESUMO

Mice infected with Friend Leukemia Virus (FLV) rapidly develop erythroleukemia and severe immune deficiency which resembles human AIDS. We have reported that mice infected with a lethal dose of FLV can be 100% cured by 150 cGy total body irradiation (TBI). This curative effect was associated with restoration of cellular immunity which was compromised by the virus. This restoration may result from activation of the IFN-gamma system and IL-2 production. Our research work further demonstrated that no spleen focus-forming virus (SFFV) specific mRNAs, no 6.0kb SFFV fragments and SFFV envelope glycoproteins were detectable in FLV-infected mice treated with low dose TBI. Predicated on our report, del Regato has initiated clinical trials to treat AIDS patients with low dose TBI. The preliminary results are encouraging and the study is continuing. We have also studied the effects of low dose TBI on the expression of the P53 gene. The results show loss or inactivation of P53 tumor suppressor genes in FLV-infected mice, but P53 expression was restored in FLV-infected mice treated by low dose TBI. It is intriguing to speculate that in the curative effect of low dose TBI on mice infected with retrovirus, the P53 tumor suppressor gene may play an important role. It would be of interest to see if this type of treatment, which was well tolerated by mice, would be beneficial in other types of virally induced disease, including AIDS.


Assuntos
Síndrome de Imunodeficiência Adquirida Murina/radioterapia , Irradiação Corporal Total , Animais , Vírus da Leucemia Murina de Friend , Genes p53 , Imunidade Celular , Interferon gama/metabolismo , Interleucina-2/metabolismo , Cariotipagem , Células Matadoras Naturais/metabolismo , Camundongos , Camundongos Endogâmicos , Síndrome de Imunodeficiência Adquirida Murina/imunologia , Síndrome de Imunodeficiência Adquirida Murina/metabolismo , Tamanho do Órgão , RNA Mensageiro/metabolismo , Vírus Formadores de Foco no Baço/metabolismo , Linfócitos T/metabolismo , Fator de Necrose Tumoral alfa/metabolismo
16.
J Virol ; 69(3): 1714-19, 1995 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-7853508

RESUMO

The leukemogenic membrane glycoprotein gp55, encoded by Friend spleen focus-forming virus (SFFV), induces erythroid cell proliferation through its interaction with the erythropoietin receptor (EPO-R). There are two forms of gp55 in SFFV-infected cells: an intracellular form (more than 95% of the total protein), which is localized within the endoplasmic reticulum (ER) membranes, and a cell surface form (about 3 to 5%). Because both forms of the viral proteins bind to EPO-R, it is not clear whether the viral protein induces mitogenesis intracellularly or at the cell surface. To address this question, we constructed an EPO-R mutant that contained a 6-amino-acid (DEKKMP) C-terminus ER retention signal. Biochemical and functional analyses with this mutant indicated that it was completely retained in the ER and not expressed at the cell surface. Further analysis showed that the mutant, like the wild-type EPO-R, interacted with SFFV gp55. However, this apparent intracellular interaction between the two proteins failed to induce growth factor-independent proliferation of Ba/F3 cells. Furthermore, spontaneous variants of the ER-retained EPO-R selected on the basis of their ability to induce cell proliferation when coexpressed with gp55 were exclusively expressed at the cell surface. Thus, our results support the hypothesis that the mitogenic activation of the EPO-R by gp55 requires the interaction of the two proteins at the cell surface.


Assuntos
Vírus da Leucemia Murina de Friend/metabolismo , Receptores da Eritropoetina/metabolismo , Vírus Formadores de Foco no Baço/metabolismo , Proteínas do Envelope Viral/metabolismo , Células 3T3 , Sequência de Aminoácidos , Animais , Sequência de Bases , Compartimento Celular , Ciclo Celular , Membrana Celular/metabolismo , Primers do DNA/química , Retículo Endoplasmático/metabolismo , Técnicas In Vitro , Camundongos , Dados de Sequência Molecular
17.
Glycoconj J ; 11(2): 133-9, 1994 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-7804003

RESUMO

Normal rat kidney cells, non-productively infected with the anaemia-inducing variant of Friend spleen focus-forming virus (F-SFFVA), were metabolically labelled with [2-3H]mannose. The primary translation product of the viral envelope gene (env), representing a glycoprotein with an apparent molecular M(r) of 55,000 (gp55), was isolated from cell lysates by immunoaffinity chromatography and purified by preparative SDS/PAGE. Radiolabelled oligosaccharides, released from tryptic glycopeptides by treatment with endo-beta-N-acetylglucosaminidase H, were characterized chromatographically, by enzymic digestion and by acetolysis. The results revealed that F-SFFVA gp55 obtained from this source carried predominantly oligomannose type sugar chains with five to nine mannoses. As a characteristic feature, glycans with seven to nine mannoses contained, in part, an additional glucose residue. Although the amount of glucosylated species found was higher in F-SFFVA gp55 (about 25% of total endo-H-sensitive oligosaccharides) than in gp55 of the corresponding polycythaemia-inducing variant (F-SFFVP, 16.3%), the overall glycosylation pattern of the F-SFFVA env product closely resembled that of F-SFFVP gp55 [Strube et al. (1988) J Biol Chem 263:3762-71]. Hence, our results demonstrate that the different intracellular processing and transport of the primary F-SFFVA env product cannot be attributed to aberrant trimming of its oligomannose type glycans.


Assuntos
Vírus da Leucemia Murina de Friend/metabolismo , Oligossacarídeos/química , Vírus Formadores de Foco no Baço/metabolismo , Proteínas do Envelope Viral/metabolismo , Animais , Aspergillus oryzae/enzimologia , Sequência de Carboidratos , Linhagem Celular , Fabaceae/enzimologia , Vírus da Leucemia Murina de Friend/genética , Glicosilação , Rim , Manose/metabolismo , Manosidases , Dados de Sequência Molecular , Oligossacarídeos/biossíntese , Oligossacarídeos/isolamento & purificação , Plantas Medicinais , Processamento de Proteína Pós-Traducional , Ratos , Vírus Formadores de Foco no Baço/genética , Trítio , Proteínas do Envelope Viral/biossíntese , alfa-Manosidase
18.
Biochem Biophys Res Commun ; 192(3): 1131-8, 1993 May 14.
Artigo em Inglês | MEDLINE | ID: mdl-8507186

RESUMO

Direct interactions of Friend spleen focus-forming virus glycoprotein gp55 with either erythropoietin receptor (EpoR) or interleukin (IL)2 receptor beta chain (IL2R) (but not with IL3 receptor) have been reported to induce factor-independent prolonged proliferation of erythroid or lymphoid cells. In order to clarify the molecular mechanism by which EpoR-gp55 complex transmits an aberrant growth signal in the absence of erythropoietin, various chimeric receptors constituted with IL2R, EpoR or IL3 receptor were constructed. It was found that coexpression of gp55 and the chimeric receptors containing the cytoplasmic domains of EpoR and the extracellular domains of IL3 (or IL2) receptor in IL3-dependent Ba/F3 cells results in factor-independent growth. Since gp55 in cell membrane has only a two amino acid tail in the cytoplasmic domains and thus cannot interact with EpoR in cytoplasm, our data suggest that gp55 does not bind EpoR directly but interacts with EpoR through third membrane component(s).


Assuntos
Eritropoetina/farmacologia , Vírus da Leucemia Murina de Friend/metabolismo , Produtos do Gene env/metabolismo , Receptores da Eritropoetina/metabolismo , Vírus Formadores de Foco no Baço/metabolismo , Proteínas do Envelope Viral/metabolismo , Animais , Linfócitos B , Divisão Celular/efeitos dos fármacos , Linhagem Celular , Vetores Genéticos , Humanos , Interleucina-2/farmacologia , Cinética , Camundongos , Plasmídeos , Receptores da Eritropoetina/genética , Receptores de Interleucina-2/genética , Receptores de Interleucina-2/metabolismo , Receptores de Interleucina-3/genética , Receptores de Interleucina-3/metabolismo , Proteínas Recombinantes de Fusão/metabolismo , Transfecção
20.
Mol Cell Biol ; 12(7): 2949-57, 1992 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-1320192

RESUMO

The erythropoietin receptor (EPO-R), a member of the cytokine receptor superfamily, can be activated by binding either erythropoietin (EPO) or gp55, the Friend spleen focus-forming virus glycoprotein. The highly specific interaction between gp55 and EPO-R triggers cell proliferation and thereby causes the first stage of Friend virus-induced erythroleukemia. We have generated functional chimeric receptors containing regions of the EPO-R and the interleukin-3 receptor (AIC2A polypeptide), a related cytokine receptor which does not interact with gp55. All chimeric receptors were expressed at similar levels, had similar binding affinities for EPO, and conferred EPO-dependent cell growth. Only those chimeric receptors which contained the EPO-R transmembrane region were activated by gp55. These results demonstrate that the transmembrane region of the EPO-R is critical for activation by gp55. In addition, analysis of a soluble, secreted EPO-R and cysteine point mutants of the EPO-R show that the extracytoplasmic region of the EPO-R specifically interacts with gp55.


Assuntos
Leucemia Experimental/metabolismo , Receptores de Superfície Celular/metabolismo , Vírus Formadores de Foco no Baço/metabolismo , Proteínas do Envelope Viral/metabolismo , Sítios de Ligação/genética , Células Cultivadas , Análise Mutacional de DNA , Conformação Proteica , Receptores de Superfície Celular/genética , Receptores da Eritropoetina , Receptores de Interleucina-3/metabolismo , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/metabolismo , Transdução de Sinais , Transfecção
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...