Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 8 de 8
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Mol Ther ; 1(2): 165-70, 2000 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-10933927

RESUMEN

Human CXCR4 is the receptor for the CXC chemokine SDF-1alpha and also acts as a coreceptor for T lymphotropic HIV-1 strains. Blocking the surface expression of this receptor via an intrakine approach has recently been shown to efficiently prevent HIV-1 infection of T cells. The CXC-chemokine gene is fused to an endoplasmic reticulum retention signal (KDEL) that retains the newly synthesized chemokine and its receptor within the cell, where both are subsequently degraded. We constructed MoMuLV-based vectors containing the SDF-KDEL construct driven by the "MND" long terminal repeat, using eGFP as a marker gene (MND-SDF-KDEL-IRES-eGFP) and a control vector (MND-X-IRES-eGFP). CEM human T lymphoblastic leukemia cells were transduced with the intrakine vector or the control vector. We detected a marked downregulation of CXCR4 expression in the cells transduced with the intrakine vectors as opposed to the cells transduced with the control vector. However, the eGFP-negative fraction of the cells transduced with the intrakine vector displayed the same CXCR4 downregulation as the eGFP-positive fraction, suggesting an effect in trans. The possibility of this being due to eGFP being silenced while SDF-KDEL was still expressed was excluded by Southern and Northern blot analyses. Upon cultivating the control cells with supernatant of the cells transduced with the intrakine vector, we observed a downregulation of CXCR4 expression on the control cells. Experiments using rhSDF-1alpha showed downregulation by the supernatant to be comparable to that achieved by the exogenous addition of 30 ng/ml SDF-1alpha. To assess the bioactivity of the secreted substance in the supernatant, a chemotaxis assay was performed. The transmigration observed was, once again, within the range of that achieved by the addition of 30 ng/ml SDF-1alpha. We conclude that the intrakine SDF-KDEL, apart from acting within the cell, is also in part secreted and causes the downregulation of the receptor by acting like a secreted chemokine.


Asunto(s)
Quimiocinas/genética , Quimiocinas/fisiología , Receptores CXCR4/metabolismo , Activación Transcripcional , Northern Blotting , Southern Blotting , Línea Celular , Quimiocina CXCL12 , Quimiocinas/metabolismo , Quimiocinas CXC/metabolismo , Quimiotaxis , Relación Dosis-Respuesta a Droga , Regulación hacia Abajo , Citometría de Flujo , Vectores Genéticos , Proteínas Fluorescentes Verdes , Humanos , Células Jurkat , Proteínas Luminiscentes/metabolismo , Receptores CXCR4/genética , Retroviridae/metabolismo , Linfocitos T/metabolismo , Transducción Genética
2.
Blood ; 96(4): 1317-26, 2000 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-10942373

RESUMEN

Cell vaccines engineered to express immunomodulators have shown feasibility in eliminating leukemia in murine models. Vectors for efficient gene delivery to primary human leukemia cells are required to translate this approach to clinical trials. In this study, second-generation lentiviral vectors derived from human immunodeficiency virus 1 were evaluated, with the cytomegalovirus (CMV) promoter driving expression of granulocyte-macrophage-colony-stimulating factor (GM-CSF) and CD80 in separate vectors or in a bicistronic vector. The vectors were pseudotyped with vesicular stomatitis virus G glycoprotein and concentrated to high titers (10(8)-10(9) infective particles/mL). Human acute lymphoblastic leukemia (ALL), acute myeloid leukemia (AML), and chronic myeloid leukemia cell lines transduced with the monocistronic pHR-CD80 vector or the bicistronic pHR-GM/CD vector became 75% to 95% CD80 positive (CD80(+)). More important, transduction of primary human ALL and AML blasts with high-titer lentiviral vectors was consistently successful (40%-95% CD80(+)). The average amount of GM-CSF secretion by the leukemia cell lines transduced with the pHR-GM-CSF monocistronic vector was 2182.9 pg/10(6) cells per 24 hours. Secretion was markedly lower with the bicistronic pHR-GM/CD vector (average, 225.7 pg/10(6) cells per 24 hours). Lower amounts of CMV-driven messenger RNA were detected with the bicistronic vector, which may account for its poor expression of GM-CSF. Primary ALL cells transduced to express CD80 stimulated T-cell proliferation in an autologous mixed lymphocyte reaction. This stimulation was specifically blocked with monoclonal antibodies reactive against CD80 or by recombinant cytotoxic T-lymphocyte antigen 4-immunoglobulin fusion protein. These results show the feasibility of efficiently transducing primary leukemia cells with lentiviral vectors to express immunomodulators to elicit antileukemic immune responses. (Blood. 2000;96:1317-1326)


Asunto(s)
Antígeno B7-1/genética , Terapia Genética , Vectores Genéticos , Factor Estimulante de Colonias de Granulocitos y Macrófagos/genética , Lentivirus , Leucemia Mieloide/genética , Leucemia Mieloide/terapia , Leucemia-Linfoma Linfoblástico de Células Precursoras/genética , Leucemia-Linfoma Linfoblástico de Células Precursoras/terapia , Enfermedad Aguda , Antígeno B7-1/inmunología , Antígeno B7-1/uso terapéutico , Citotoxicidad Inmunológica/genética , Regulación Neoplásica de la Expresión Génica , Técnicas de Transferencia de Gen , Factor Estimulante de Colonias de Granulocitos y Macrófagos/inmunología , Factor Estimulante de Colonias de Granulocitos y Macrófagos/uso terapéutico , Humanos , Inmunoterapia , Leucemia Mieloide/inmunología , Leucemia-Linfoma Linfoblástico de Células Precursoras/inmunología , Células Tumorales Cultivadas
3.
J Virol ; 71(12): 9466-74, 1997 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-9371608

RESUMEN

Gene expression from the Moloney murine leukemia retrovirus (Mo-MuLV) is highly restricted in embryonic carcinoma (EC) and embryonic stem (ES) cells. We compared levels of expression in PA317 fibroblasts, F9 (EC) cells, and CCE (ES) cells by Mo-MuLV-based vectors and vectors based on our previously reported MND backbone, which has alterations to address three viral elements implicated as repressors of expression by Mo-MuLV: the enhancer, the primer binding site, and the negative-control region. Expression was evaluated with three reporter genes, the chloramphenicol acetyltransferase (CAT) gene, whose expression was measured by enzymatic assay and by Northern blotting; a truncated nerve growth factor receptor (tNGFR), whose expression was measured by fluorescence-activated cell sorting (FACS) as a cell surface protein; and the enhanced green fluorescent protein (EGFP), whose expression was measured intracellularly by flow cytometry. We found significantly higher levels of CAT activity (5- to 300-fold) and greater quantities of vector-specific transcripts in ES and EC cells transduced with the modified MND-CAT-SN vector than in those transduced with L-CAT-SN. Northern blot analysis indicated that long terminal repeat transcripts from MND-CAT-SN are >80 times more abundant than the L-CAT-SN transcripts. FACS analysis of tNGFR expression from a pair of vectors, L-tNGFR-SN and MND-tNGFR-SN, indicated that only 1.04% of the CCE cells containing the L-tNGFR-SN vector expressed the cell surface reporter, while the MND-tNGFR-SN vector drove expression in 99.54% of the CCE cells. Of the F9 cells containing the L-tNGFR-SN vector, 13.32% expressed tNGFR, while 99.89% of the F9 cells transduced with MND-tNGFR-SN showed expression. Essentially identical results were produced with an analogous pair of vectors encoding EGFP. In unselected pools of F9 cells 48 h posttransduction, the L-EGFP-SN vector drove expression in only 5% of the population while the MND-EGFP-SN vector drove expression in 88% of the cells. After more than 3 weeks in culture without selection, the proportion of cells showing expression from L-EGFP-SN decreased slightly to 3% while expression from the MND-EGFP-SN vector persisted in 80% of the cells. Interestingly, in the few ES and EC cells which did show expression from the L-tNGFR-SN or L-EGFP-SN vectors, the magnitude of reporter expression was similar to that from the MND-tNGFR-SN or MND-EGFP-SN vector in nearly all cells, suggesting that the MND vectors are far less susceptible to position-dependent variegation of expression than are the Mo-MuLV-based vectors. Therefore, the modified retroviral vector, MND, achieves higher net levels of expression due to a greater frequency of expression, which may be useful for the expression of exogenous genes in EC and ES cells.


Asunto(s)
Expresión Génica , Vectores Genéticos , Virus de la Leucemia Murina de Moloney , Células 3T3 , Animales , Northern Blotting , Southern Blotting , Carcinoma Embrionario , Línea Celular , Cloranfenicol O-Acetiltransferasa/genética , Cloranfenicol O-Acetiltransferasa/metabolismo , Metilación de ADN , Genes Reporteros , Proteínas Fluorescentes Verdes , Proteínas Luminiscentes/genética , Ratones , Provirus , Receptores de Factor de Crecimiento Nervioso/genética , Retroviridae , Células Madre , Células Tumorales Cultivadas , Integración Viral
4.
J Virol ; 71(3): 1776-83, 1997 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-9032306

RESUMEN

We tested the ability of a recombinant adeno-associated virus (rAAV) vector to express and integrate exogenous DNA into human hematopoietic cells in the absence of selection. We developed an rAAV vector, AAV-tNGFR, carrying a truncated rat nerve growth factor receptor (tNGFR) cDNA as a cell surface reporter under the control of the Moloney murine leukemia virus (MoMuLV) long terminal repeat. An analogous MoMuLV-based retroviral vector (L-tNGFR) was used in parallel, and gene transfer and expression in human hematopoietic cells were assessed by flow cytometry and DNA analyses. Following gene transfer into K562 cells with AAV-tNGFR at a multiplicity of infection (MOI) of 13 infectious units (IU), 26 to 38% of cells expressed tNGFR on the surface early after transduction, but the proportion of tNGFR expressing cells steadily declined to 3.0 to 3.5% over 1 month of culture. At an MOI of 130 IU, nearly all cells expressed tNGFR immediately posttransduction, but the proportion of cells expressing tNGFR declined to 62% over 2 months of culture. The decline in the proportion of AAV-tNGFR-expressing cells was associated with ongoing losses of vector genomes. In contrast, K562 cells transduced with the retroviral vector L-tNGFR expressed tNGFR in a constant fraction. Integration analyses on clones showed that integration occurred at different sites. Integration frequencies were estimated at about 49% at an MOI of 130 and 2% at an MOI of 1.3. Transduction of primary human CD34+ progenitor cells by AAV-tNGFR was less efficient than with K562 cells and showed a declining percentage of cells expressing tNGFR over 2 weeks of culture. Thus, purified rAAV caused very high gene transfer and expression in human hematopoietic cells early after transduction, which steadily declined during cell passage in the absence of selection. Although the efficiency of integration was low, overall integration was markedly improved at a high MOI. While prolonged episomal persistence may be adequate for gene therapy of nondividing cells, a very high MOI or improvements in basic aspects of AAV-based vectors may be necessary to improve integration frequency in the rapidly dividing hematopoietic cell population.


Asunto(s)
Dependovirus/genética , Técnicas de Transferencia de Gen , Vectores Genéticos , Receptores de Factor de Crecimiento Nervioso/genética , Integración Viral , Animales , Antígenos CD34 , Línea Celular , Células Cultivadas , ADN/análisis , Expresión Génica , Genes Reporteros , Genoma Viral , Humanos , Ratas , Recombinación Genética , Selección Genética , Células Tumorales Cultivadas
5.
Stem Cells ; 15(6): 443-54, 1997.
Artículo en Inglés | MEDLINE | ID: mdl-9402657

RESUMEN

Human hematopoiesis can be supported in beige/nude/ XID (bnx) mice by coinjection of human bone marrow stromal cells engineered to secrete human interleukin 3 (HuIL-3). The major limitation is a total absence of human B cell development in the mice, which could be due to supraphysiological levels of HuIL-3 in the circulation. In an effort to obtain human B lymphoid, as well as T lymphoid and myeloid cell development in the mice, CD34+ cells were coinjected with human marrow stromal cells engineered to secrete human IL-2, IL-7, stem cell factor or FLT3 ligand, +/- IL-3. No single factor other than IL-3 supported sustained human hematopoiesis in the mice, although cytokines were expressed for four to six months post-transplantation. Production of both HuIL-3 and IL-7 in the mice supported extrathymic development of human T lymphocytes, but no B cells, myeloid cells, or clonogenic progenitors were detected. Human B cells were not produced from CD34+ cells in the bnx mice under any condition tested. Another limitation to the bnx/Hu system is a lack of maturation of human red blood cells, although BFU-E are maintained. Stromal cells secreting human erythropoietin and IL-3 were cotransplanted into mice with HuCD34+ cells and an increase in hematocrit from 40%-45% to 80%-85% resulted, with production of human and murine red blood cells. Unfortunately, all mice (n = 9) suffered strokes, displayed paralysis and died within three weeks. The bnx/Hu cotransplantation model provides an interesting system in which to study human hematopoietic cell differentiation under the influence of various cytokines.


Asunto(s)
Células de la Médula Ósea/metabolismo , Citocinas/farmacología , Hematopoyesis , Animales , Antígenos CD34/inmunología , Linfocitos B/citología , Células de la Médula Ósea/citología , Citocinas/biosíntesis , Citocinas/genética , Trasplante de Células Madre Hematopoyéticas , Humanos , Ratones , Ratones Desnudos , Modelos Biológicos , Proteínas Recombinantes/farmacología , Células del Estroma/metabolismo , Células del Estroma/trasplante , Linfocitos T/citología , Linfocitos T/metabolismo , Factores de Tiempo , Quimera por Trasplante , Trasplante Heterólogo
6.
Hum Gene Ther ; 7(13): 1595-603, 1996 Aug 20.
Artículo en Inglés | MEDLINE | ID: mdl-8864760

RESUMEN

Three dogs with deficiency of the lysosomal enzyme alpha-L-iduronidase were treated by gene replacement therapy targeted at muscle. Direct intramuscular injections of plasmid encoding the alpha-L-iduronidase gene cDNA resulted in no detectable enzyme production, but may have resulted in immunologic sensitization to iduronidase protein, which the dogs lack totally. Myoblasts were grown from skeletal muscle biopsies and transduced with a retroviral vector containing the canine gene under control of the muscle creatine kinase enhancer. Several hundred-fold overexpression of enzyme production occurred in cultured cells; however, following reintroduction of the cultured cells into dogs, enzyme production declined rapidly. Concurrent with the falling enzyme levels, there was production of specific immunoglobulin G (IgG) antibody against iduronidase that was further associated with cellular infiltration of the myoblast injection sites. Most inflammatory cells were lymphocytes and plasma cells, suggesting local humoral and cellular immune responses to the enzyme-producing muscle cells. PCR analysis of tissues collected 2-22 weeks after the final treatment showed the persistence of Neo and canine alpha-L-iduronidase sequences in a progressively decreasing percentage of myoblasts. Results from this study in a canine model of mucopolysaccharidosis I underscore the fact that immunologic reactions to cells producing desirable, normal, but foreign, proteins may be as much an impediment to gene therapy as reactions to the viral vectors used to introduce the foreign gene.


Asunto(s)
Terapia Genética , Iduronidasa/inmunología , Iduronidasa/metabolismo , Mucopolisacaridosis I/terapia , Animales , Células Cultivadas , ADN Complementario/genética , Modelos Animales de Enfermedad , Perros , Femenino , Regulación de la Expresión Génica/genética , Vectores Genéticos/genética , Histocitoquímica , Iduronidasa/genética , Inmunoglobulina G/inmunología , Inmunoglobulina G/metabolismo , Terapia de Inmunosupresión , Leucocitos/enzimología , Mucopolisacaridosis I/genética , Músculos/citología , Músculos/metabolismo , Plásmidos/genética , Retroviridae/genética , Transfección/genética , Trasplante Autólogo/efectos adversos , Trasplante Autólogo/inmunología
7.
Blood ; 87(12): 5232-41, 1996 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-8652838

RESUMEN

NBXFO hybridoma cells produced both the membrane and secreted isoforms of macrophage colony-stimulating factor (M-CSF). Murine bone marrow cells stimulated by the secreted form of M-CSF (sM-CSF) became Mac1+, Mac2+, Mac3+, and F4/80+ macrophages that inhibited the growth of NBXFO cells, but not L1210 or P815 tumor cells. In cytotoxicity studies, M-CSF activated macrophages and freshly isolated macrophages killed NBXFO cells in the presence of polymyxin B, eliminating the possibility that contaminating lipopolysaccharide (LPS) was responsible for the delivery of the cytotoxic signal. Retroviral-mediated transfection of T9 glioma cells with the gene for the membrane isoform of M-CSF (mM-CSF), but not for the secreted isoform of M-CSF, transferred the ability of macrophages to kill these transfected T9 cells in a mM-CSF dose-dependent manner. Macrophage-mediated killing of the mM-CSF transfected clone was blocked by using a 100-fold excess of recombinant M-CSF. Catalase, superoxide dismutase, and the nitric oxide inhibitor, N-omega-nitro-arginine methyl ester (NAME), did not effect macrophage cytotoxicity against the mM-CSF transfectant T9 clones. T9 parental cells when cultured in the presence of an equal number of the mM-CSF transfectant cells were not killed, indicating specific target cell cytotoxicity by the macrophages. Electron microscopy showed that macrophages were capable of phagocytosizing mM-CSF bearing T9 tumor cells and NBXFO hybridoma cells; this suggested a possible mechanism of this cytotoxicity. This study indicates that mM-CSF provides the necessary binding and triggering molecules through which macrophages can initiate direct tumor cell cytotoxicity.


Asunto(s)
Hibridomas , Factor Estimulante de Colonias de Macrófagos/fisiología , Macrófagos/inmunología , Proteínas de Neoplasias/fisiología , Animales , Células de la Médula Ósea , Citotoxicidad Inmunológica , Vectores Genéticos/genética , Hibridomas/metabolismo , Activación de Macrófagos/fisiología , Factor Estimulante de Colonias de Macrófagos/genética , Factor Estimulante de Colonias de Macrófagos/farmacología , Macrófagos/efectos de los fármacos , Masculino , Ratones , Ratones Endogámicos DBA , Fagocitosis , Ratas , Ratas Sprague-Dawley , Retroviridae/genética , Bazo/citología , Transfección
8.
J Immunol ; 144(5): 1614-24, 1990 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-2106551

RESUMEN

The ability to express the growth hormone IL-2 upon stimulation gives T lymphocytes one of their major effector functions in the immune system. IL-2 is apparently synthesized only by T cells, and only by a subset of T cells which constitutes a "helper" class. It remains unknown how and when the IL-2-producing lineage becomes distinct from other functional effector lineages. We have therefore examined immature T cell precursors to determine when IL-2 inducibility is acquired in relation to other maturation events, such as expression of an Ag-binding TCR, which is suspected to play an influential role in the determination of subclass commitment. In mature T cells, IL-2 is inducible via agonists of the phosphoinositide pathway, a network of signaling mediators shared by a wide variety of metazoan cell types. The universality of this activation pathway makes it seem less likely, a priori, to be a target of developmental change than the intrinsic susceptibility to induction of the IL-2 locus. However, our results presented here refute this expectation. In this report, we show that both TCR+ cells and pre-T cells too immature to express TCR can be induced to express IL-2 at high levels. The induction requirements for IL-2 expression, however, are different in TCR- and TCR+ cells. Even by using Ca2+ ionophore and phorbol ester to bypass the requirement for the TCR in cell activation, the TCR- cells also require the presence of the polypeptide hormone IL-1. By contrast, TCR+ mature cells not only can express IL-2 without IL-1, but also show no response to IL-1 when Ca2+ ionophore and phorbol ester are present. IL-1-dependent IL-2 producers appear in the thymus of repopulating radiation chimeras before "mature" (TCR+) T cells, whereas IL-1-independent IL-2 production is found only afterward. Thus, IL-2 inducibility per se apparently precedes TCR expression and all TCR-associated fate determination events. However, developmental alteration of signal transduction pathways may play a vital regulatory role in the later allocation of particular functional responses to appropriate lineages of T cells.


Asunto(s)
Interleucina-2/genética , Receptores de Antígenos de Linfocitos T/genética , Linfocitos T/fisiología , Animales , Antígenos de Diferenciación/análisis , Antígenos de Diferenciación de Linfocitos T/análisis , Antígenos CD4/análisis , Antígenos CD8 , Diferenciación Celular , Expresión Génica , Interleucina-1/farmacología , Ratones , ARN Mensajero/genética , Quimera por Radiación , Transducción de Señal , Timo/citología , Factores de Tiempo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...