Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 40
Filtrar
Más filtros

Banco de datos
Tipo del documento
País de afiliación
Intervalo de año de publicación
1.
Nat Med ; 4(1): 58-64, 1998 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-9427607

RESUMEN

Janus kinase-3 (JAK3) deficiency has recently been identified as a cause of severe combined immunodeficiency (SCID) in humans. We used a mouse model of Jak3-deficient SCID to test a gene therapy approach for treatment of this disease. Transfer of a Jak3 retroviral vector to repopulating hematopoietic stem cells resulted in increased numbers of T and B lymphocytes, reversal of hypogammaglobulinemia, restoration of T-cell activation upon stimulation with mitogens, and development of an antigen-specific immune response after immunization. Analysis for vector copy number in lymphoid and myeloid populations showed a large in vivo selective advantage for Jak3-expressing lymphoid cells. These results show that gene replacement is a feasible treatment strategy for this disease and that naturally occurring in vivo selection of corrected cells is an important advantage of this approach.


Asunto(s)
Linfocitos B/inmunología , Terapia Genética/métodos , Proteínas Tirosina Quinasas/deficiencia , Proteínas Tirosina Quinasas/genética , Inmunodeficiencia Combinada Grave/inmunología , Inmunodeficiencia Combinada Grave/terapia , Linfocitos T/inmunología , Agammaglobulinemia/etiología , Agammaglobulinemia/terapia , Animales , Formación de Anticuerpos , Células de la Médula Ósea/citología , Trasplante de Médula Ósea/métodos , Técnicas de Transferencia de Gen , Vectores Genéticos , Humanos , Inmunoglobulina A/sangre , Inmunoglobulina G/sangre , Inmunoglobulina M/sangre , Janus Quinasa 3 , Activación de Linfocitos , Ratones , Ratones Endogámicos C57BL , Ratones SCID , Reacción en Cadena de la Polimerasa , Proteínas Tirosina Quinasas/biosíntesis , Retroviridae , Inmunodeficiencia Combinada Grave/enzimología , Bazo/inmunología
2.
Nat Med ; 4(10): 1136-43, 1998 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-9771746

RESUMEN

One of the main impediments to effective gene therapy of blood disorders is the resistance of human hematopoietic stem cells to stable genetic modification. We show here that a small minority of retrovirally transduced stem cells can be selectively enriched in vivo, which might be a way to circumvent this obstacle. We constructed two retroviral vectors containing an antifolate-resistant dihydrofolate reductase cDNA transcriptionally linked to a reporter gene. Mice were transplanted with transduced bone marrow cells and then treated with an antifolate-based regimen that kills unmodified stem cells. Drug treatment significantly increased the percentage of vector-expressing peripheral blood erythrocytes, platelets, granulocytes, and T and B lymphocytes. Secondary transplant experiments demonstrated that selection occurred at the level of hematopoietic stem cells. This system for in vivo stem-cell selection provides a means to increase the number of genetically modified cells after transplant, and may circumvent an substantial obstacle to successful gene therapy for human blood diseases.


Asunto(s)
Vectores Genéticos , Trasplante de Células Madre Hematopoyéticas , Retroviridae/genética , Selección Genética , Tetrahidrofolato Deshidrogenasa/genética , Animales , Células Sanguíneas/enzimología , Resistencia a Medicamentos , Femenino , Antagonistas del Ácido Fólico/farmacología , Genes Reporteros , Terapia Genética/métodos , Ratones , Ratones Endogámicos C57BL , Tioinosina/análogos & derivados , Tioinosina/farmacología , Tionucleótidos/farmacología , Transformación Genética , Trimetrexato/farmacología
3.
Nat Med ; 7(9): 1028-34, 2001 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-11533706

RESUMEN

Stem cells from bone marrow, skeletal muscle and possibly other tissues can be identified by the 'side-population' (SP) phenotype. Although it has been assumed that expression of ABC transporters is responsible for this phenotype, the specific molecules involved have not been defined. Here we show that expression of the Bcrp1 (also known as Abcg2 murine/ABCG2 human) gene is a conserved feature of stem cells from a wide variety of sources. Bcrp1 mRNA was expressed at high levels in primitive murine hematopoietic stem cells, and was sharply downregulated with differentiation. Enforced expression of the ABCG2 cDNA directly conferred the SP phenotype to bone-marrow cells and caused a reduction in maturing progeny both in vitro and in transplantation-based assays. These results show that expression of the Bcrp1/ABCG2 gene is an important determinant of the SP phenotype, and that it might serve as a marker for stem cells from various sources.


Asunto(s)
Transportadoras de Casetes de Unión a ATP/metabolismo , Proteínas Inflamatorias de Macrófagos , Glicoproteínas de Membrana , Proteínas Asociadas a Resistencia a Múltiples Medicamentos , Proteínas de Neoplasias , Células Madre/fisiología , Miembro 1 de la Subfamilia B de Casetes de Unión a ATP/genética , Miembro 1 de la Subfamilia B de Casetes de Unión a ATP/metabolismo , Transportador de Casetes de Unión a ATP, Subfamilia G, Miembro 2 , Transportadoras de Casetes de Unión a ATP/genética , Animales , Antígenos CD/metabolismo , Antígenos CD34/metabolismo , Biomarcadores , Células de la Médula Ósea/fisiología , Células Cultivadas , Quimiocinas CC , Citocinas/metabolismo , Femenino , Humanos , Masculino , Ratones , Ratones Endogámicos , Ratones Mutantes , Proteínas Ribosómicas/metabolismo , Células Madre/citología , Tetraspanina 29 , Trasplantes
4.
Science ; 257(5066): 99-103, 1992 Jul 03.
Artículo en Inglés | MEDLINE | ID: mdl-1352414

RESUMEN

Experiments were performed to determine if retroviral-mediated transfer of the human multidrug resistance 1 gene (MDR1) into murine bone marrow cells would confer drug resistance to the cells and whether the MDR1 gene could be used as a dominant selectable marker in vivo. When mice transplanted with bone marrow cells containing a transferred MDR1 gene were treated with the cytotoxic drug taxol, a substantial enrichment for transduced bone marrow cells was observed. This demonstration of positive selection establishes the ability to amplify clones of transduced hematopoietic cells in vivo and suggests possible applications in human therapy.


Asunto(s)
Alcaloides/farmacología , Antineoplásicos Fitogénicos/farmacología , Trasplante de Médula Ósea/fisiología , Médula Ósea/fisiología , Resistencia a Medicamentos/genética , Células Madre Hematopoyéticas/citología , Transfección , Secuencia de Bases , ADN/genética , ADN/aislamiento & purificación , Eritrocitos/fisiología , Vectores Genéticos , Células Madre Hematopoyéticas/efectos de los fármacos , Humanos , Datos de Secuencia Molecular , Oligodesoxirribonucleótidos , Paclitaxel , Reacción en Cadena de la Polimerasa/métodos , Provirus/genética , Retroviridae/genética
6.
Leukemia ; 19(9): 1590-6, 2005 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-16001089

RESUMEN

Selective inhibition of the BCR/ABL tyrosine kinase by imatinib has become a first-line therapy for chronic myelogenous leukemia (CML). However, BCR/ABL-positive progenitors often persist despite treatment, and relapse associated with resistance to imatinib has been described in many patients with advanced disease. Drug efflux by P-glycoprotein (P-gp), as well as point mutations in BCR/ABL oncoprotein, has been implicated in the mechanism of resistance to imatinib. In this study, we established a murine transplantation model of CML-like myeloproliferative disease using Mdr1a/1b-null mice and analyzed the effects of loss of P-gp on resistance to imatinib. We found that mice transplanted with Mdr1a/1b-null bone marrow (BM) that had been transduced with a BCR/ABL retroviral vector displayed similar responses to imatinib, compared with those transplanted with BCR/ABL-transduced wild-type BM. In the absence of P-gp, the incidence and latency of disease in secondary recipients was not changed in imatinib-treated mice, relative to wild-type controls. Furthermore, K562 cells engineered to overexpress P-gp remained sensitive to imatinib-induced growth inhibition and cell death. Together, our findings suggest that P-gp expression in hematopoietic stem cells does not significantly contribute to imatinib resistance in CML.


Asunto(s)
Miembro 1 de la Subfamilia B de Casetes de Unión a ATP/biosíntesis , Modelos Animales de Enfermedad , Células Madre Hematopoyéticas/metabolismo , Leucemia Mielógena Crónica BCR-ABL Positiva/tratamiento farmacológico , Leucemia Mielógena Crónica BCR-ABL Positiva/metabolismo , Piperazinas/farmacología , Pirimidinas/farmacología , Miembro 1 de la Subfamilia B de Casetes de Unión a ATP/genética , Animales , Benzamidas , Trasplante de Médula Ósea/métodos , Supervivencia Celular/efectos de los fármacos , Trasplante de Células , Resistencia a Antineoplásicos , Humanos , Mesilato de Imatinib , Células K562 , Ratones , Ratones Noqueados , Análisis de Supervivencia
7.
Leukemia ; 10(3): 439-46, 1996 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-8642859

RESUMEN

Resistance to methotrexate (MTX) in some sublines of mammalian cells is reported to be due to one of the following amino acid substitutions in dihydrofolate reductase (DHFR) that lower inhibition by MTX: Gly15 to Trp, Leu22 to Arg or Phe or Phe31 to Trp or Ser. We have produced variants of human DHFR (hDHFR) with these substitutions by directed mutagenesis. Recombinant hDHFR variants expressed in Escherichia coli have greatly decreased inhibition by MTX, but decreased catalytic efficiency, and in one case decreased stability. When a retroviral vector encoding wild-type (wt) hDHFR or one of these variants was introduced into murine fibroblasts or bone marrow progenitors, modest protection from MTX was conferred, even by wt. Relapsed pediatric patients with acute lymphoblastic leukemia who have received multiple courses of high-dose MTX seem most likely to develop such MTX resistance. cDNA was reverse transcribed from blast mRNA from 17 of these patients. However, upon amplification and sequencing of DHFR cDNA, no resistance mutation was found. The explanation for this probably lies in the need for considerable gene amplification to offset lowered catalytic efficiency, and the need for two-base changes for most substitutions, both of which are probably infrequent events.


Asunto(s)
Antimetabolitos Antineoplásicos/uso terapéutico , Genes , Metotrexato/uso terapéutico , Mutación , Leucemia-Linfoma Linfoblástico de Células Precursoras/genética , Tetrahidrofolato Deshidrogenasa/genética , Adolescente , Animales , Secuencia de Bases , Niño , Preescolar , Resistencia a Antineoplásicos , Femenino , Vectores Genéticos , Humanos , Lactante , Masculino , Ratones , Ratones Endogámicos C57BL , Datos de Secuencia Molecular , Leucemia-Linfoma Linfoblástico de Células Precursoras/tratamiento farmacológico , Leucemia-Linfoma Linfoblástico de Células Precursoras/enzimología , Recurrencia , Retroviridae/genética , Tetrahidrofolato Deshidrogenasa/aislamiento & purificación
8.
Clin Cancer Res ; 1(11): 1359-68, 1995 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-9815932

RESUMEN

The chloroethylnitrosoureas (CENUs) are important antineoplastic drugs for which clinical utility has been restricted by the development of severe delayed myelosuppression in most patients. To investigate the potential of DNA repair proteins to reduce bone marrow sensitivity to the CENUs, we transferred the Escherichia coli ada gene, which encodes a Mr 39,000 O6-alkylguanine-DNA alkyltransferase (ATase), into murine bone marrow cells by the use of a high-titer ecotropic retrovirus. The ada-encoded ATase is resistant to O6-benzylguanine (O6-BG), a potent inhibitor of the mammalian ATases, thus affording the bone marrow an additional level of protection against CENUs. In methylcellulose cultures, ada-infected hematopoietic progenitor cells were twice as resistant as uninfected cells to the toxic effects of 1, 3-bis(2-chloroethyl)-1-nitrosourea (BCNU) following treatment with O6-BG. Although showing no obvious protective effects against leukopenia, overexpression of the bacterial ATase activity reduced the severity of anemia and thrombocytopenia in mice treated with O6-BG and BCNU. These effects, which were maximal at a BCNU dose of 12.5 mg/kg, were associated with improved survival when BCNU was given at this dose. At lower BCNU doses cytotoxicity was limited in both transduced and control mice, and at higher doses the protective effect was saturated due to cytotoxicity. These results suggest that ada gene therapy may be a feasible approach to amelioration of delayed myelosuppression following O6-BG plus CENU combination chemotherapy.


Asunto(s)
Antineoplásicos Alquilantes/toxicidad , Proteínas Bacterianas/genética , Médula Ósea/efectos de los fármacos , Proteínas de Escherichia coli , Técnicas de Transferencia de Gen , O(6)-Metilguanina-ADN Metiltransferasa/genética , Animales , Médula Ósea/enzimología , Médula Ósea/virología , Carmustina/toxicidad , Inhibidores Enzimáticos/toxicidad , Etilnitrosourea/toxicidad , Estudios de Factibilidad , Guanina/análogos & derivados , Guanina/toxicidad , Ratones , Ratones Endogámicos CBA , O(6)-Metilguanina-ADN Metiltransferasa/metabolismo , Retroviridae/genética , Factores de Transcripción
9.
Hum Gene Ther ; 11(17): 2353-64, 2000 Nov 20.
Artículo en Inglés | MEDLINE | ID: mdl-11096440

RESUMEN

Janus kinase 3 (JAK3) is an essential component of cytokine receptor signal transduction pathways required for normal lymphocyte development and function. JAK3 deficiency in both mice and humans results in severe combined immunodeficiency (SCID) and increased susceptibility to opportunistic infections. We have previously shown that JAK3 gene transfer into irradiated recipients could restore immune function. However, since this toxic conditioning would be undesirable for infants in a clinical application, we have tested whether immune function could be restored in nonmyeloablated JAK3-deficient (-/-) mice. Murine JAK3 retroviral vectors were transduced into hematopoietic stem cells from the livers of newborn JAK3(-/-) mice. These cells were then injected intraperitoneally into nonirradiated JAK3(-/-) neonates. Transduced cells were detectable in these mice at time points 4 to 6 months after injection and resulted in significant correction of T and B lymphocyte numbers and circulating immunoglobulin (Ig) levels. After immune challenge with a dose of influenza A virus that was lethal to nonmanipulated JAK3(-/-) mice, mice injected with transduced cells showed development of circulating virus-specific IgG and enhanced survival. This work shows that the large selective advantage for JAK3-corrected lymphoid cells may be sufficient to overcome the need for myeloablative conditioning in JAK3 gene therapy protocols.


Asunto(s)
Terapia Genética/métodos , Células Madre Hematopoyéticas/fisiología , Linfocitos/fisiología , Proteínas Tirosina Quinasas/deficiencia , Proteínas Tirosina Quinasas/genética , Inmunodeficiencia Combinada Grave/terapia , Animales , Animales Recién Nacidos , Formación de Anticuerpos , Trasplante de Células , Vectores Genéticos , Virus de la Influenza A/patogenicidad , Janus Quinasa 3 , Hígado/citología , Ratones , Ratones Mutantes , Células Mieloides/fisiología , Infecciones por Orthomyxoviridae/inmunología , Fenotipo , Retroviridae/genética , Selección Genética , Inmunodeficiencia Combinada Grave/inmunología , Inmunodeficiencia Combinada Grave/patología , Tasa de Supervivencia
10.
Hum Gene Ther ; 8(15): 1773-83, 1997 Oct 10.
Artículo en Inglés | MEDLINE | ID: mdl-9358027

RESUMEN

Chemoresistance gene transfer is an experimental method to protect hematopoietic cells from the toxicity of anticancer drugs. Because multiple drugs are usually given together in cancer therapy, this strategy will ultimately require vectors expressing multiple chemoresistance genes. For this reason, we designed a bicistronic retroviral vector (HaMID) containing a modified human multidrug resistance-1 cDNA and a mutant human dihydrofolate reductase cDNA bearing a leucine to tyrosine substitution at codon 22 (L22Y). To determine if this vector would confer dual drug resistance to hematopoietic cells, recombinant retrovirus was used to transduce the human CEM T lymphoblastic cell line as well as primary murine myeloid progenitors. Growth suppression assays, using polyclonal transduced CEM cells, demonstrated increased resistance to taxol (13-fold), trimetrexate (8.9-fold), vinblastine (5.6-fold), methotrexate (2.5-fold), and etoposide (1.5-fold) when used as single agents. HaMID-transduced cells also grew at a logarithmic rate in the simultaneous presence of 25 nM taxol and 100 nM trimetrexate while control cells were entirely growth inhibited by this drug combination. Similarly, HaMID-transduced murine myeloid progenitors acquired increased resistance to taxol (2.9-fold) and trimetrexate (140-fold), and were able to form colonies in the simultaneous presence of both drugs. Our results suggest that retroviral transfer of HaMID into primary hematopoietic cells should reduce the myelosuppression associated with the combined use of antifolates and P-glycoprotein-effluxed drugs.


Asunto(s)
Miembro 1 de la Subfamilia B de Casetes de Unión a ATP/genética , Antineoplásicos Fitogénicos/efectos adversos , Resistencia a Múltiples Medicamentos , Antagonistas del Ácido Fólico/efectos adversos , Vectores Genéticos , Células Madre Hematopoyéticas/efectos de los fármacos , Retroviridae/genética , Tetrahidrofolato Deshidrogenasa/genética , Miembro 1 de la Subfamilia B de Casetes de Unión a ATP/biosíntesis , Células Cultivadas , Clonación Molecular , Etopósido/efectos adversos , Expresión Génica , Células Madre Hematopoyéticas/citología , Células Madre Hematopoyéticas/metabolismo , Humanos , Metotrexato/efectos adversos , Paclitaxel/efectos adversos , Provirus , Transformación Genética , Trimetrexato/efectos adversos , Células Tumorales Cultivadas , Vinblastina/efectos adversos
11.
Hum Gene Ther ; 8(13): 1531-43, 1997 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-9322086

RESUMEN

Class 1 aldehyde dehydrogenases (ALDH-1) function as drug resistance gene products by catalyzing the irreversible conversion of aldophosphamide, an active metabolite of cyclophosphamide, to an inert compound. Because the dose-limiting toxicity of cyclophosphamide is myelosuppression, retrovirus-mediated transfer of ALDH-1 to bone marrow cells has been proposed as a protective strategy. Here we show that expression of ALDH-1 vectors was problematic due to low levels of ALDH-1 mRNA accumulation. A number of vectors containing several different ALDH-1 cDNAs were introduced into a variety of different cell lines either by transfection or transduction. Detectable ALDH-1 protein and enzyme activity was only seen in one transfected cell clone. Cells transduced with ALDH-1 retroviral vectors had no detectable protein expression and very low levels of ALDH-1 mRNA. Analogous vectors containing other drug resistance cDNAs led to much higher levels of steady-state mRNA. The mRNA half-life from ALDH-1 vectors was less than 2 hr suggesting that vector-derived mRNAs were destabilized by ALDH-1 coding sequences. These results suggest that methods which increase the stability of ALDH-1 mRNAs will be important for increased drug resistance in retrovirally transduced hematopoietic cells.


Asunto(s)
Aldehído Deshidrogenasa/genética , Vectores Genéticos , ARN Mensajero/genética , Retroviridae/genética , Animales , ADN Complementario , Resistencia a Medicamentos/genética , Semivida , Ratones , ARN Mensajero/metabolismo , Tetrahidrofolato Deshidrogenasa/genética , Transducción Genética , Transfección , Células Tumorales Cultivadas
12.
Hum Gene Ther ; 8(17): 2069-77, 1997 Nov 20.
Artículo en Inglés | MEDLINE | ID: mdl-9414255

RESUMEN

Retroviral transduction of antifolate-resistant variants of human dihydrofolate reductase (hDHFR) into cells can increase their resistance to the cytotoxic effects of these drugs. We evaluated the ability of wild-type hDHFR and 20 mutant enzymes (13 with single-amino acid substitutions, 7 with two substitutions) to prevent growth inhibition in antifolate-treated CCRF-CEM cells. The wild-type enzyme and all of the variants significantly protected transduced cells from trimetrexate (TMTX)-induced growth inhibition. However, only half of the variants conferred more protection than does the wild-type enzyme. For the variants tested, the observed protective effect was higher for TMTX than for methotrexate (< or =7.5-fold increased resistance), piritrexim (< or =16-fold), and edatrexate (negligible). Transduction of the variants L22Y-F31S and L22Y-F31R led to the greatest protection against TMTX (approximately 200-fold). Protection from loss of cell viability was similar to protection from growth inhibition. The protection associated with a particular mutant hDHFR did not result from the level of expression: Efficient protection resulted from low affinity of the variant for antifolates, reasonable catalytic activity, and good thermal stability. Clones isolated from a polyclonal population of transduced cells varied by as much as 30-fold in their resistance to TMTX, the resistance differences depending on hDHFR expression levels.


Asunto(s)
Antagonistas del Ácido Fólico/farmacología , Tetrahidrofolato Deshidrogenasa/genética , Aminopterina/análogos & derivados , Aminopterina/antagonistas & inhibidores , Animales , Supervivencia Celular , Resistencia a Antineoplásicos/genética , Expresión Génica , Variación Genética , Inhibidores de Crecimiento/antagonistas & inhibidores , Humanos , Cinética , Metotrexato/antagonistas & inhibidores , Pirimidinas/antagonistas & inhibidores , Conejos , Tetrahidrofolato Deshidrogenasa/farmacología , Timidina/metabolismo , Transfección , Trimetrexato/antagonistas & inhibidores
13.
Hum Gene Ther ; 5(7): 891-911, 1994 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-7526902

RESUMEN

Patients with metastatic breast cancer will receive 4-5 cycles of induction chemotherapy on one of the ongoing Medicine Branch protocols. Patients achieving at least a partial response, and who do not have evidence of bone marrow involvement and who do not have metastatic bone disease, will undergo PBSC and bone marrow harvest when hematologic recovery has occurred. Patients who have not achieved a PR, but who are responding to therapy, may be treated with additional cycles of therapy in an attempt to achieve a PR. Such patients will be eligible for transplant if a PR is obtained. 70% of the bone marrow and PBSC will be cryopreserved. The CD34+ subpopulation from the remaining 30% of the bone marrow and PBSC harvest will be obtained using an anti-CD34+ antibody and immunoabsorption column. The bone marrow and peripheral blood CD34 cells will be transduced with a retroviral vector expressing the human MDR-1 cDNA. Patients with positive bone scans or histologic evidence of bone marrow involvement will be excluded from the gene transfer component of the protocol. The MDR-1 transduced CD34 cells will be reinfused along with the non-transduced bone marrow and PBSC into patients following high dose ICE chemotherapy. Serial peripheral blood and bone marrow samples will be obtained to study hematopoietic reconstitution with MDR-1 transduced cells. Patients with residual or progressive disease after ABMT will be treated with taxol or vinblastine. In these relapsed patients, peripheral blood and bone marrow samples will be obtained to study whether chemotherapy amplifies the proportion of hematopoietic cells containing the MDR-1 provirus. We will monitor the nadir blood counts of each patient receiving salvage chemotherapy for evidence of myeloprotection and correlate this data with changes in the mean proviral copy number. Sites of relapsed tumor will be biopsied to test for the presence of the MDR-1 provirus.


Asunto(s)
Miembro 1 de la Subfamilia B de Casetes de Unión a ATP/genética , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Neoplasias de la Mama/tratamiento farmacológico , Vectores Genéticos , Trasplante de Células Madre Hematopoyéticas , Células Madre Hematopoyéticas/metabolismo , Pancitopenia/terapia , Retroviridae , Miembro 1 de la Subfamilia B de Casetes de Unión a ATP/biosíntesis , Adolescente , Adulto , Protocolos de Quimioterapia Combinada Antineoplásica/efectos adversos , Trasplante de Médula Ósea , Carboplatino/administración & dosificación , Protocolos Clínicos , Resistencia a Medicamentos/genética , Etopósido/administración & dosificación , Factores de Crecimiento de Célula Hematopoyética/farmacología , Células Madre Hematopoyéticas/efectos de los fármacos , Humanos , Ifosfamida/administración & dosificación , Consentimiento Informado , Interleucina-3/farmacología , Interleucina-6/farmacología , Mesna/administración & dosificación , Persona de Mediana Edad , Metástasis de la Neoplasia , Paclitaxel/administración & dosificación , Paclitaxel/farmacología , Pancitopenia/inducido químicamente , Proteínas Recombinantes de Fusión/biosíntesis , Proteínas Recombinantes/farmacología , Factor de Células Madre , Trasplante Autólogo , Vinblastina/administración & dosificación , Vinblastina/farmacología
14.
Cancer Gene Ther ; 5(4): 225-35, 1998.
Artículo en Inglés | MEDLINE | ID: mdl-9694074

RESUMEN

Expression of certain variants of dihydrofolate reductase (DHFR) in mammalian cells protects them from methotrexate. Retroviral transfer of the gene for such a variant DHFR into hematopoietic cells might permit selection of modified cells in vivo by antifolate administration or alleviate antifolate-induced myelosuppression in patients receiving antifolate therapy. We examined protection of cells of the human lymphoblastoid line, CCRF-CEM, transduced with variants of mouse DHFR. In transduced cells selected with G418 but not with antifolate, the variant that had arginine substituted for leucine 22 did not protect against either methotrexate or trimetrexate; however, four other variants did offer protection, with the best having leucine 22 changed to tyrosine. Polyclonal cultures transduced with the different variants express DHFR at about the same level, but clones within each polyclonal population differ in DHFR expression levels and in resistance. These differences in expression were shown to reflect different integration sites for proviral DNA. Exposure to trimetrexate selects highly resistant clones, with high expression due to both high copy number and integration sites that are favorable for expression. Differences in the resistance of cultures expressing different variants at the same level are due to differences in the catalytic activity of the expressed DHFR, its affinity for antifolates, and its stability.


Asunto(s)
Antagonistas del Ácido Fólico/farmacología , Técnicas de Transferencia de Gen , Leucemia-Linfoma Linfoblástico de Células Precursoras/tratamiento farmacológico , Leucemia-Linfoma Linfoblástico de Células Precursoras/enzimología , Tetrahidrofolato Deshidrogenasa/genética , Animales , Clonación Molecular , Resistencia a Antineoplásicos , Estabilidad de Enzimas/genética , Escherichia coli/genética , Dosificación de Gen , Regulación Neoplásica de la Expresión Génica , Humanos , Metotrexato/farmacología , Ratones , Mutación , Leucemia-Linfoma Linfoblástico de Células Precursoras/genética , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Retroviridae/genética , Tetrahidrofolato Deshidrogenasa/metabolismo , Trimetrexato/farmacología , Células Tumorales Cultivadas
15.
Biochem Pharmacol ; 60(3): 325-38, 2000 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-10856427

RESUMEN

Tumors resistant to chemotherapeutic oxazaphosphorines such as cyclophosphamide often overexpress aldehyde dehydrogenase (ALDH), some isozymes of which catalyze the oxidization of aldophosphamide, an intermediate of cyclophosphamide activation, with formation of inert carboxyphosphamide. Since resistance to oxazaphosphorines can be produced in mammalian cells by transfecting them with the gene for human ALDH isozyme 3 (hALDH3), it seems possible that patients receiving therapy for solid tumors with cyclophosphamide might be protected from myelosuppression by their prior transplantation with autologous bone marrow that has been transduced with a retroviral vector causing overexpression of hALDH3. We investigated whether retroviral introduction of hALDH3 into a human leukemia cell line confers resistance to oxazaphosphorines. This was examined in the polyclonal transduced population, that is, without selecting out high expression clones. hALDH3 activity was 0.016 IU/mg protein in the transduced cells (compared with 2x10(-5) IU/mg in untransduced cells), but there was no detectable resistance to aldophosphamide-generating compounds (mafosfamide or 4-hydroperoxycyclophosphamide). The lack of protection was due, in part, to low catalytic activity of hALDH3 towards aldophosphamide, since, with NAD as cofactor, the catalytic efficiency of homogeneous, recombinant hALDH3 for aldophosphamide oxidation was shown to be about seven times lower than that of recombinant hALDH1. The two polymorphic forms of hALDH3 had identical kinetics with either benzaldehyde or aldophosphamide as substrate. Results of initial velocity measurements were consistent with an ordered sequential mechanism for ALDH1 but not for hALDH3; a kinetic mechanism for the latter is proposed, and the corresponding rate equation is presented.


Asunto(s)
Aldehído Deshidrogenasa/metabolismo , Antineoplásicos/metabolismo , Mostazas de Fosforamida/metabolismo , Aldehído Deshidrogenasa/química , Aldehído Deshidrogenasa/genética , Familia de Aldehído Deshidrogenasa 1 , Aldehídos/metabolismo , Antineoplásicos/farmacología , Benzaldehídos/metabolismo , Clonación Molecular , Ciclofosfamida/análogos & derivados , Ciclofosfamida/farmacología , ADN Complementario/análisis , Inhibidores Enzimáticos/farmacología , Células Madre Hematopoyéticas/efectos de los fármacos , Humanos , Isoenzimas/química , Isoenzimas/genética , Isoenzimas/metabolismo , Cinética , Oxidación-Reducción , Mostazas de Fosforamida/farmacología , Proteínas Recombinantes/química , Retinal-Deshidrogenasa , Análisis de Secuencia , Especificidad por Sustrato , Transfección , Células Tumorales Cultivadas
16.
Ann N Y Acad Sci ; 872: 125-40; discussion 140-1, 1999 Apr 30.
Artículo en Inglés | MEDLINE | ID: mdl-10372117

RESUMEN

Ex vivo expansion of hematopoietic stem cells would be useful for bone marrow transplantation and gene therapy applications. Toward this goal, we have investigated whether retrovirally-transduced murine stem cells could be expanded in culture with hematopoietic cytokines. Bone marrow cells were transduced with retroviral vectors expressing either the human multidrug resistance 1 gene (HaMDR1), a variant of human dihydrofolate reductase (HaDHFR), or both MDR1 and DHFR in an internal ribosomal entry site (IRES)-containing bicistronic vector (HaMID). Cells were then expanded for 15 days in cultures stimulated with interleukin (IL)-3, IL-6, and stem cell factor. When very low marrow volumes were injected into lethally irradiated recipient mice, long-term reconstitution with 100% donor cells was seen in all mice injected with HaMDR1- or HaMID-transduced cells. By contrast, engraftment with HaDHFR- or mock-transduced cells ranged from partial to undetectable despite injection of significantly larger marrow volumes. In addition, mice transplanted with expanded HaMDR1- or HaMID-transduced stem cells developed a myeloproliferative disorder that was characterized by an increase in abnormal peripheral blood leukocytes. These results show that MDR1-transduced stem cells can be expanded in vitro with hematopoietic cytokines, but indicate that an increased stem cell division frequency can lead to stem cell damage.


Asunto(s)
Miembro 1 de la Subfamilia B de Casetes de Unión a ATP/genética , Células de la Médula Ósea/citología , Trasplante de Médula Ósea , Células Madre Hematopoyéticas/citología , Células Madre Hematopoyéticas/fisiología , Retroviridae , Transfección/métodos , Miembro 1 de la Subfamilia B de Casetes de Unión a ATP/biosíntesis , Miembro 1 de la Subfamilia B de Casetes de Unión a ATP/sangre , Animales , Animales Recién Nacidos , Diferenciación Celular , División Celular , Células Cultivadas , Humanos , Leucocitos/citología , Leucocitos/fisiología , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos , Modelos Biológicos
17.
Hum Gene Ther ; 4(2): 205-22, 1993 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-8098622
19.
Tissue Antigens ; 68(4): 317-24, 2006 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-17026467

RESUMEN

Human prominin-1 (CD133 or AC133) is an important cell surface marker used to isolate primitive hematopoietic stem cells. The commercially available antibody to human prominin-1 does not recognize rhesus prominin-1. Therefore, we isolated, cloned and characterized the complementary DNA (cDNA) of rhesus prominin-1 gene and determined its coding potential. Following the nomenclature of prominin family of genes, we named this cDNA as rhesus prominin-1.s1. The amino acid sequence data of the putative rhesus prominin-1.s1 could be used in designing antigenic peptides to raise antibodies for use in isolation of pure populations of rhesus prominin-1(+) hematopoietic cells. To the best of our knowledge, there has been no previously published report about the isolation of a prominin-1 cDNA from rhesus monkey (Macaca mulatta).


Asunto(s)
Antígenos CD/biosíntesis , Antígenos CD/aislamiento & purificación , Clonación Molecular , ADN Complementario/biosíntesis , ADN Complementario/aislamiento & purificación , Glicoproteínas/biosíntesis , Glicoproteínas/aislamiento & purificación , Células Madre Hematopoyéticas/inmunología , Péptidos/aislamiento & purificación , Antígeno AC133 , Secuencia de Aminoácidos , Animales , Antígenos CD/genética , Secuencia de Bases , Marcadores Genéticos , Glicoproteínas/genética , Células Madre Hematopoyéticas/metabolismo , Humanos , Macaca mulatta , Ratones , Datos de Secuencia Molecular , Péptidos/genética , Ratas
20.
Invest New Drugs ; 14(1): 87-99, 1996.
Artículo en Inglés | MEDLINE | ID: mdl-8880396

RESUMEN

An increased understanding of the molecular mechanisms of cancer and the ability to introduce exogenous genes into mammalian cells has led to the development of oncologic treatment strategies based upon gene transfer. Preclinical animal models have suggested a variety of approaches which are now being tested in pediatric trials. Studies using marker genes to trace cell origin have already generated important information regarding autologous bone marrow transplantation for pediatric cancers. A variety of therapeutic genes are also being clinically tested. Trials are underway to determine if introduction of immunostimulatory genes into cancer cells can be used to enhance host antitumor immunity. Treatment of primary brain tumors with insertion of drug sensitization genes is a promising new therapy that is also being clinically evaluated. Other strategies such as insertion of drug resistance genes into hematopoietic cells, anti-oncogene therapy, and tumor suppressor gene replacement are being tested in adults and may find use in pediatric cancer treatment. Although gene transfer offers promising new approaches for the therapy of pediatric cancer, many technical problems remain which limit efficacy and widespread use. Further basic research in the molecular biology of cancer and in vector development will be required to realize the full potential of gene therapy strategies.


Asunto(s)
Terapia Genética , Neoplasias/terapia , Niño , Técnicas de Transferencia de Gen , Vectores Genéticos , Humanos , Neoplasias/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA