Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
1.
Nat Med ; 5(7): 768-73, 1999 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-10395321

RESUMEN

Definitive cure of an animal model of a human disease by gene transfer into hematopoietic stem cells has not yet been accomplished in the absence of spontaneous in vivo selection for transduced cells. Erythropoietic protoporphyria is a genetic disease in which ferrochelatase is defective. Protoporphyrin accumulates in erythrocytes, leaks into the plasma and results in severe skin photosensitivity. Using a mouse model of erythropoietic protoporphyria, we demonstrate here that ex vivo preselection of hematopoietic stem cells transduced with a polycistronic retrovirus expressing both human ferrochelatase and green fluorescent protein results in complete and long-term correction of skin photosensitivity in all transplanted mice.


Asunto(s)
Trasplante de Médula Ósea , Ferroquelatasa/genética , Terapia Genética , Trastornos por Fotosensibilidad/terapia , Porfiria Hepatoeritropoyética/terapia , Animales , Eritrocitos/metabolismo , Ferroquelatasa/biosíntesis , Técnicas de Transferencia de Gen , Proteínas Fluorescentes Verdes , Células Madre Hematopoyéticas , Humanos , Proteínas Luminiscentes/biosíntesis , Proteínas Luminiscentes/genética , Masculino , Ratones , Ratones Endogámicos BALB C , Trastornos por Fotosensibilidad/sangre , Trastornos por Fotosensibilidad/patología , Porfiria Hepatoeritropoyética/sangre , Porfiria Hepatoeritropoyética/genética , Protoporfiria Eritropoyética , Protoporfirinas/sangre , Piel/patología , Trasplante Isogénico
2.
Science ; 294(5550): 2368-71, 2001 Dec 14.
Artículo en Inglés | MEDLINE | ID: mdl-11743206

RESUMEN

Sickle cell disease (SCD) is caused by a single point mutation in the human betaA globin gene that results in the formation of an abnormal hemoglobin [HbS (alpha2betaS2)]. We designed a betaA globin gene variant that prevents HbS polymerization and introduced it into a lentiviral vector we optimized for transfer to hematopoietic stem cells and gene expression in the adult red blood cell lineage. Long-term expression (up to 10 months) was achieved, without preselection, in all transplanted mice with erythroid-specific accumulation of the antisickling protein in up to 52% of total hemoglobin and 99% of circulating red blood cells. In two mouse SCD models, Berkeley and SAD, inhibition of red blood cell dehydration and sickling was achieved with correction of hematological parameters, splenomegaly, and prevention of the characteristic urine concentration defect.


Asunto(s)
Anemia de Células Falciformes/terapia , Terapia Genética , Vectores Genéticos , Globinas/genética , VIH-1/genética , Anemia de Células Falciformes/genética , Animales , Modelos Animales de Enfermedad , Eritrocitos/metabolismo , Expresión Génica , Globinas/metabolismo , Trasplante de Células Madre Hematopoyéticas , Células Madre Hematopoyéticas/metabolismo , Hemoglobina Falciforme/metabolismo , Humanos , Lentivirus/genética , Región de Control de Posición , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Oxihemoglobinas/metabolismo , ARN Mensajero/genética , ARN Mensajero/metabolismo , Talasemia/genética , Talasemia/terapia , Transducción Genética , Transgenes , Globinas beta
3.
Exp Hematol ; 27(5): 817-25, 1999 May.
Artículo en Inglés | MEDLINE | ID: mdl-10340397

RESUMEN

Retroviral transduction of human hematopoietic stem cells is still limited by lack of information about conditions that will maximize stem cell self-renewal divisions in vitro. To address this, we first compared the kinetics of entry into division of single human CD34+CD38- cord blood (CB) cells exposed in vitro to three different flt3-ligand (FL)-containing cytokine combinations. Of the three combinations tested, FL + hyperinterleukin 6 (HIL-6) yielded the least clones and these developed at a slow rate. With either FL + Steel factor (SF) + HIL-6 + thrombopoietin (TPO) or FL + SF + interleukin 3 (IL-3) + IL-6 + granulocyte-colony-stimulating factor (G-CSF), >90% of the cells that formed clones within 6 days undertook their first division within 4 days, although not until after 24 hours. These latter two, more stimulatory, cytokine combinations then were used to assess the effect of duration of cytokine exposure on the efficiency of transducing primitive CB cells with a gibbon ape leukemia virus-pseudotyped murine retroviral vector containing the enhanced green fluorescent protein (GFP) cDNA and the neomycin resistance gene. Fresh lin- CB cells exposed once to medium containing this virus plus cytokines on fibronectin-coated dishes yielded 23% GFP+ CD34+ cells and 52-57% G418-resistant CFC when assessed after 2 days. Prestimulation of the target cells (before exposing them to virus) with either the four or five cytokine combination increased their susceptibility. In both cases, the effect of prestimulation assessed using the same infection protocol was maximal with 2 days of prestimulation and resulted in 47-54% GFP+ CD34+ cells and 67-69% G418-resistant CFC. Repeated daily addition of new virus (up to three times), with assessment of the cells 2 days after the last addition of fresh virus, gave only a marginal improvement in the proportion of transduced CD34+ cells and CFC, but greatly increased the proportion of transduced LTC-IC (from 40% to >99%). Transplantation of lin- CB cells transduced using this latter 6-day protocol into NOD/SCID mice yielded readily detectable GFP+ cells in 10 of 11 mice that were engrafted with human cells. The proportion of the regenerated human cells that were GFP+ ranged from 0.2-72% in individual mice and included both human lymphoid and myeloid cells in all cases. High-level reconstitution with transduced human cells was confirmed by Southern blot analysis. These findings demonstrate that transplantable hematopoietic stem cells in human CB can be reproducibly transduced at high efficiency using a 6-day period of culture in a retrovirus-containing medium with either FL + SF + HIL-6 + TPO or FL + SF + IL-3 + IL-6 + G-CSF in which virus is added on the third, fourth, and fifth day.


Asunto(s)
Antígenos CD , Sangre Fetal/citología , Técnicas de Transferencia de Gen , Proteínas de la Membrana/genética , Retroviridae/genética , ADP-Ribosil Ciclasa , ADP-Ribosil Ciclasa 1 , Animales , Antígenos CD34/análisis , Antígenos de Diferenciación/análisis , División Celular , Sangre Fetal/inmunología , Vectores Genéticos , Humanos , Inmunofenotipificación , Recién Nacido , Glicoproteínas de Membrana , Ratones , Ratones Endogámicos NOD , Ratones SCID , NAD+ Nucleosidasa/análisis , Transducción Genética
4.
Hum Gene Ther ; 8(13): 1595-604, 1997 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-9322092

RESUMEN

Despite improvements in retrovirus-mediated gene transfer to primitive murine hematopoietic cells, high-level reconstitution with provirally marked cells with continued expression of the transferred gene(s) remains a challenge in many situations. We evaluated a physical preselection strategy for isolating transduced cells after their infection with different vectors. The small (240-bp) cDNA coding region for the human CD24 cell-surface antigen was inserted into myeloproliferative sarcoma virus (MPSV) and murine stem cell virus (MSCV)-based retroviral vectors such that expression of CD24 was under the control of the viral long terminal repeat (LTR). After infection of (Ly-5.1) mouse bone marrow (BM), those expressing CD24 were isolated by fluorescence-activated cell sorting (FACS) and a transplant dose estimated to contain approximately 12 +/- 4 long-term competitive repopulating cells (CRU) injected into lethally irradiated congenic Ly-5.2 recipients. Six months later, virtually all recipients showed high-level (> 80%) reconstitution of their BM and thymus with Ly-5.1 (transplant-derived) cells, the majority of which were also transduced (mean of 2.5 or 2.6 proviral copies for the two vectors). All spleen colonies generated in secondary recipients of cells obtained from the BM of the 6-month-old primary mice contained the provirus. However, only in recipients of MSCVCD24-infected marrow was a correspondingly high level of CD24 expression seen: a maximum of 88% for whole BM (all mice positive), 58% for peripheral blood leukocytes (all mice positive), and 21% for thymocytes (11 of 13 mice positive). CD24 was also readily detected on the regenerated Sca-1+Lin- cells present in the primary and secondary recipients when these were studied 6 months post-transplant, but again on more of the Sca-1+Lin- cells in recipients of MSCVCD24-infected cells as compared to recipients of MPSVCD24-infected cells. These results point to the utility of preselection strategies and suggest an approach for the development of better vectors for achieving regulated, lineage-specific or stage-specific gene expression patterns in particular subsets of hematopoietic cells.


Asunto(s)
Antígenos CD/genética , Células de la Médula Ósea/inmunología , Hematopoyesis/genética , Glicoproteínas de Membrana , Transducción Genética , Animales , Antígeno CD24 , Linaje de la Célula/genética , Femenino , Vectores Genéticos , Hematopoyesis/inmunología , Humanos , Masculino , Ratones , Ratones Endogámicos C3H , Ratones Endogámicos C57BL , Retroviridae/genética
5.
Invest Ophthalmol Vis Sci ; 40(9): 2141-6, 1999 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-10440271

RESUMEN

PURPOSE: To determine whether human retinal pigment epithelium (RPE) can be modified by retroviral-mediated gene transfer and to monitor the human RPE cells in the subretinal space of living rabbits with scanning laser ophthalmoscopy (SLO). METHODS: Cultured human fetal retinal pigment epithelium (HFRPE) was exposed to green fluorescent protein (GFP)-transducing retroviral vectors, Moloney murine leukemia virus, and lentivirus. The cultured cells were followed by fluorescence microscopy. Suspensions of GFP-expressing HFRPE were transplanted into the subretinal space of pigmented rabbits, and the transplant sites were examined by SLO for fluorescence, including fluorescein and indocyanine green angiography. The rabbits were euthanatized at different times after transplantation, and the retinas were studied histologically. RESULTS: Retroviral gene transfer can introduce a foreign gene such as GFP into cultured HFRPE. Gene expression is maintained in cultured RPE for at least 3 months. The lentiviral vector transduced both nondividing and dividing cells; the Moloney vector only transduced the latter. GFP-expressing cells can be followed in the living retina. Their changes reflect the rejection response followed histologically. CONCLUSIONS: Cultured HFRPE could be transduced to express GFP for long periods of time by retroviral gene transfer. GFP allowed retinal transplants and gene expression to be monitored in vivo. These results provide a model for potential ex vivo gene therapy in the subretinal space.


Asunto(s)
Trasplante de Tejido Fetal , Técnicas de Transferencia de Gen , Proteínas Luminiscentes/metabolismo , Epitelio Pigmentado Ocular/trasplante , Retina/cirugía , Retroviridae/genética , Animales , Células Cultivadas , Virus Defectuosos , Proteínas Fluorescentes Verdes , Humanos , Proteínas Luminiscentes/genética , Microscopía Fluorescente , Oftalmoscopía , Epitelio Pigmentado Ocular/metabolismo , Epitelio Pigmentado Ocular/virología , Conejos , Retina/citología , Retina/metabolismo , Retroviridae/metabolismo
6.
Ann N Y Acad Sci ; 850: 151-62, 1998 Jun 30.
Artículo en Inglés | MEDLINE | ID: mdl-9668537

RESUMEN

We are focusing on the development of complex retroviral vectors containing human beta-globin gene and beta-LCR for the gene therapy of sickle cell disease and beta-thalassemias. First generation vectors containing mutated splice-sites to insure stability of proviral transfer enabled long-term reconstitution in 10/12 transplanted mice for a least 8 months with high expression levels in 2 out of 3 mice analyzed (5% and 20% murine beta). Transfer and expression were also achieved in secondary recipients (range: 3-11% murine beta). Position independent expression was not observed. In an effort to increase the efficiency of gene transfer and obtain complete reconstitution of recipient mice with exclusively transduced cells while enriching for proviral integration into active chromatin regions, we have incorporated a cassette expressing CD24 or the green fluorescent protein (GFP). Stable transfer to murine bone marrow cells allowed efficient FACS-sorting of pure populations of transduced cells. A family of vectors based on these principles and containing segments of gamma- or delta-globin genes were also designed for systematic analysis of their anti-sickling properties.


Asunto(s)
Anemia de Células Falciformes/terapia , Terapia Genética/métodos , Globinas/genética , Talasemia beta/terapia , Anemia de Células Falciformes/genética , Animales , Mapeo Cromosómico , Técnicas de Transferencia de Gen , Humanos , Región de Control de Posición , Ratones , Regiones Promotoras Genéticas , Retroviridae , Talasemia beta/genética
7.
Blood ; 88(8): 2852-8, 1996 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-8874181

RESUMEN

Recent assessment of the long-term repopulating activity of defined subsets of hematopoietic cells has offered new insights into the characteristics of the transplantable stem cells of this system; however, as yet, there is very little known about mechanisms that regulate their self-renewal in vivo. We have now exploited the ability to quantitate these cells using the competitive repopulating unit (CRU) assay to identify the role of both intrinsic (ontological) and extrinsic (transplanted dose-related) variables that may contribute to the regulation of CRU recovery in vivo. Ly5.1 donor cells derived from day-14.5 fetal liver (FL) or the bone marrow (BM) of adult mice injected 4 days previously with 5-fluorouracil were transplanted at doses estimated to contain 10, 100, or 1,000 long-term CRU into irradiated congenic Ly5.2 adult recipient mice. Eight to 12 months after transplantation, there was a complete recovery of BM cellularity and in vitro clonogenic progenitor numbers and a nearly full recovery of day-12 colony-forming unit-spleen numbers irrespective of the number or origin of cells initially transplanted. In contrast, regeneration of Ly5.1+ donor-derived CRU was incomplete in all cases and was dependent on both the origin and dose of the transplant, with FL being markedly superior to that of adult BM. As a result, the final recovery of the adult marrow CRU compartment ranged from 15% to 62% and from 1% to 18% of the normal value in recipients of FL and adult BM transplantation, respectively, with an accompanying maximum CRU amplification of 150-fold for recipients of FL cells and 15-fold for recipients of adult BM cells. Interestingly, the extent of CRU expansion from either source was inversely related to the number of CRU transplanted. These data suggest that recovery of mature blood cell production in vivo may activate negative feedback regulatory mechanisms to prematurely limit stem cell self-renewal ability. Proviral integration analysis of mice receiving retrovirally transduced BM cells confirmed regeneration of totipotent lymphomyeloid repopulating cells and provided evidence for a greater than 300-fold clonal amplification of a single transduced stem cell. These results highlight the differential regenerative capacities of CRU from fetal and adult sources that likely reflect intrinsic, genetically defined determinants of CRU expansion but whose contribution to the magnitude of stem cell amplification ultimately obtained in vivo is also strongly influenced by the initial number of CRU transplanted. Such findings set the stage for attempts to enhance CRU regeneration by administration of agents that may enable full expression of regenerative potential or through the expression of intracellular gene products that may alter intrinsic regenerative capacity.


Asunto(s)
Trasplante de Médula Ósea , Trasplante de Tejido Fetal , Trasplante de Células Madre Hematopoyéticas , Células Madre Hematopoyéticas/citología , Animales , Antígenos Ly/genética , Biomarcadores , Recuento de Células , Retroalimentación , Femenino , Genes Reporteros , Vectores Genéticos/aislamiento & purificación , Supervivencia de Injerto , Hematopoyesis , Humanos , Hígado/citología , Hígado/embriología , Masculino , Ratones , Ratones Endogámicos C3H , Ratones Endogámicos C57BL , Provirus/aislamiento & purificación , Quimera por Radiación , Retroviridae/aislamiento & purificación
8.
Blood ; 84(9): 2868-77, 1994 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-7949162

RESUMEN

We have investigated the use of a cell surface antigen as a dominant selectable marker to facilitate the detection and selection of retrovirally infected target cells. The small coding region of the human cell surface antigen CD24 (approximately 240 bp) was introduced into a myeloproliferative sarcoma virus (MPSV)-based retroviral vector, which was then used to infect day 4 5-fluorouracil (5-FU)-treated murine bone marrow cells. Within 48 hours of termination of the infection procedure CD24-expressing cells were selected by fluorescent-activated cell sorting (FACS) with an antibody directed against the CD24 antigen. Functional analysis of these cells showed that they included not only in vitro clonogenic progenitors and day 12 colony-forming unit-spleen but also cells capable of competitive long-term hematopoietic repopulation. Double-antibody labeling studies performed on recipients of retrovirally transduced marrow cells showed that some granulocytes, macrophages, erythrocytes, and, to a lesser extent, B and T lymphocytes still expressed the transduced CD24 gene at high levels 4 months later. No gross abnormalities in hematopoiesis were detected in mice repopulated with CD24-expressing cells. Our results show that the use of the CD24 cell surface antigen as a retrovirally encoded marker enables the rapid, efficient, and nontoxic selection in vitro of infected primary cells, facilitates tracking and phenotyping of their progeny, and should provide a unique tool to identify elements that regulate the expression of transduced genes in the most primitive hematopoietic cells.


Asunto(s)
Antígenos CD/genética , Células Madre Hematopoyéticas , Glicoproteínas de Membrana , Animales , Biomarcadores , Células de la Médula Ósea , Antígeno CD24 , Separación Celular , Femenino , Citometría de Flujo , Genes , Masculino , Ratones , Ratones Endogámicos C3H , Ratones Endogámicos C57BL , Retroviridae , Transfección
9.
Proc Natl Acad Sci U S A ; 97(10): 5411-5, 2000 May 09.
Artículo en Inglés | MEDLINE | ID: mdl-10792053

RESUMEN

Transcriptional silencing of genes transferred into hematopoietic stem cells poses one of the most significant challenges to the success of gene therapy. If the transferred gene is not completely silenced, a progressive decline in gene expression as the mice age often is encountered. These phenomena were observed to various degrees in mouse transplant experiments using retroviral vectors containing a human beta-globin gene, even when cis-linked to locus control region derivatives. Here, we have investigated whether ex vivo preselection of retrovirally transduced stem cells on the basis of expression of the green fluorescent protein driven by the CpG island phosphoglycerate kinase promoter can ensure subsequent long-term expression of a cis-linked beta-globin gene in the erythroid lineage of transplanted mice. We observed that 100% of mice (n = 7) engrafted with preselected cells concurrently expressed human beta-globin and the green fluorescent protein in 20-95% of their RBC for up to 9.5 mo posttransplantation, the longest time point assessed. This expression pattern was successfully transferred to secondary transplant recipients. In the presence of beta-locus control region hypersensitive site 2 alone, human beta-globin mRNA expression levels ranged from 0.15% to 20% with human beta-globin chains detected by HPLC. Neither the proportion of positive blood cells nor the average expression levels declined with time in transplanted recipients. Although suboptimal expression levels and heterocellular position effects persisted, in vivo stem cell gene silencing and age-dependent extinction of expression were avoided. These findings support the further investigation of this type of vector for the gene therapy of human hemoglobinopathies.


Asunto(s)
Eritrocitos/metabolismo , Silenciador del Gen , Globinas/genética , Trasplante de Células Madre Hematopoyéticas , Células Madre Hematopoyéticas/fisiología , Transcripción Genética , Animales , Células de la Médula Ósea/citología , Vectores Genéticos , Globinas/análisis , Proteínas Fluorescentes Verdes , Células Madre Hematopoyéticas/citología , Humanos , Región de Control de Posición , Proteínas Luminiscentes/genética , Ratones , Ratones Endogámicos C3H , Ratones Endogámicos C57BL , Ratones Endogámicos , Fosfoglicerato Quinasa/genética , Regiones Promotoras Genéticas , ARN Mensajero/genética , Proteínas Recombinantes de Fusión/biosíntesis , Retroviridae , Factores de Tiempo , Transfección
10.
Proc Natl Acad Sci U S A ; 92(26): 12075-9, 1995 Dec 19.
Artículo en Inglés | MEDLINE | ID: mdl-8618847

RESUMEN

The gene transfer efficiency of human hematopoietic stem cells is still inadequate for efficient gene therapy of most disorders. To overcome this problem, a selectable retroviral vector system for gene therapy has been developed for gene therapy of Gaucher disease. We constructed a bicistronic retroviral vector containing the human glucocerebrosidase (GC) cDNA and the human small cell surface antigen CD24 (243 bp). Expression of both cDNAs was controlled by the long terminal repeat enhancer/promoter of the Molony murine leukemia virus. The CD24 selectable marker was placed downstream of the GC cDNA and its translation was enhanced by inclusion of the long 5' untranslated region of encephalomyocarditis virus internal ribosomal entry site. Virus-producing GP+envAM12 cells were created by multiple supernatant transductions to create vector producer cells. The vector LGEC has a high titer and can drive expression of GC and the cell surface antigen CD24 simultaneously in transduced NIH 3T3 cells and Gaucher skin fibroblasts. These transduced cells have been successfully separated from untransduced cells by fluorescence-activated cell sorting, based on cell surface expression of CD24. Transduced and sorted NIH 3T3 cells showed higher GC enzyme activity than the unsorted population, demonstrating coordinated expression of both genes. Fibroblasts from Gaucher patients were transduced and sorted for CD24 expression, and GC enzyme activity was measured. The transduced sorted Gaucher fibroblasts had a marked increase in enzyme activity (149%) compared with virgin Gaucher fibroblasts (17% of normal GC enzyme activity). Efficient transduction of CD34+ hematopoietic progenitors (20-40%) was accomplished and fluorescence-activated cell sorted CD24(+)-expressing progenitors generated colonies, all of which (100%) were vector positive. The sorted, CD24-expressing progenitors generated erythroid burst-forming units, colony-forming units (CFU)-granulocyte, CFU-macrophage, CFU-granulocyte/macrophage, and CFU-mix hematopoietic colonies, demonstrating their ability to differentiate into these myeloid lineages in vitro. The transduced, sorted progenitors raised the GC enzyme levels in their progeny cells manyfold compared with untransduced CD34+ progenitors. Collectively, this demonstrates the development of high titer, selectable bicistronic vectors that allow isolation of transduced hematopoietic progenitors and cells that have been metabolically corrected.


Asunto(s)
Antígenos CD34/inmunología , Antígenos CD/genética , Enfermedad de Gaucher/genética , Enfermedad de Gaucher/inmunología , Genes , Glucosilceramidasa/genética , Células Madre Hematopoyéticas/inmunología , Glicoproteínas de Membrana , Células 3T3 , Animales , Antígenos CD/biosíntesis , Secuencia de Bases , Antígeno CD24 , Ensayo de Unidades Formadoras de Colonias , Cartilla de ADN , Elementos de Facilitación Genéticos , Enfermedad de Gaucher/terapia , Expresión Génica , Terapia Genética , Vectores Genéticos , Glucosilceramidasa/biosíntesis , Células Madre Hematopoyéticas/fisiología , Humanos , Ratones , Datos de Secuencia Molecular , Virus de la Leucemia Murina de Moloney , Reacción en Cadena de la Polimerasa , Regiones Promotoras Genéticas , Secuencias Repetitivas de Ácidos Nucleicos , Transducción Genética
11.
Blood ; 87(5): 1754-62, 1996 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-8634421

RESUMEN

Corrective gene transfer for therapeutic intervention in metabolic and hematopoietic disorders has been hampered by the relatively inefficient transduction of human hematopoietic stem cells. To overcome this, a bicistronic recombinant retrovirus has been generated that delivers both a therapeutic glucocerebrosidase (GC) cDNA for the treatment of Gaucher disease, and a small murine cell surface antigen (heat-stable antigen [HSA]) as a selectable marker. An amphotropic retroviral-producing cell clone was created, and filtered supernatant was used to transduce NIH 3T3 cells. Sorting of transduced cells by flow cytometry enabled separation into populations based on cell surface fluorescence intensity derived from the expressed HSA. Significant increases in GC enzyme activity were seen for the transduced and especially the transduced and sorted cells. Similarly, increases in GC specific activity were seen in transduced and sorted skin fibroblasts from a patient with Gaucher disease. To streamline future transfer and sorting protocols for hematopoietic cells, transformed B-cell lines from Gaucher patients were created. Type I B cells were transduced and sorted, and large increases in GC specific activity occurred with concomitant increases in integrated retroviral copy numbers. In addition, toward the goal of using this selectable approach for corrective gene transfer to bone marrow stem cells, CD34+ cells were isolated from normal BM donors, transduced, and sorted based on cell surface expression of HSA. Proviral DNA was detected in approximately 40% of clonogenic progenitor colonies derived from unsorted, transduced CD34+ cells, demonstrating the high titer of the vector. However, after sorting, 100% of the colonies had the corrective GC cDNA, demonstrating the efficiency of this selective system for human hematopoietic progenitors. It is expected that strategies based on this approach will allow sorting of transduced cells of many types before implantation of transduced cells to animals or patients. This vector system may also be used to simplify manipulations and studies on retroviral-mediated gene delivery in vitro and for in vivo models.


Asunto(s)
Separación Celular/métodos , Citometría de Flujo , Enfermedad de Gaucher/enzimología , Terapia Genética , Vectores Genéticos/genética , Glucosilceramidasa/genética , Células Madre Hematopoyéticas/citología , Glicoproteínas de Membrana , Virus de la Leucemia Murina de Moloney/genética , Retroviridae/genética , Células 3T3 , Animales , Antígenos CD/genética , Antígenos CD34 , Linfocitos B , Antígeno CD24 , Línea Celular Transformada , Células Cultivadas , ADN Complementario/genética , Fibroblastos/enzimología , Enfermedad de Gaucher/genética , Enfermedad de Gaucher/patología , Enfermedad de Gaucher/terapia , Glucosilceramidasa/deficiencia , Células Madre Hematopoyéticas/enzimología , Humanos , Ratones , Selección Genética , Transfección
12.
Science ; 302(5644): 415-9, 2003 Oct 17.
Artículo en Inglés | MEDLINE | ID: mdl-14564000

RESUMEN

We have previously shown correction of X-linked severe combined immunodeficiency [SCID-X1, also known as gamma chain (gamma(c)) deficiency] in 9 out of 10 patients by retrovirus-mediated gamma(c) gene transfer into autologous CD34 bone marrow cells. However, almost 3 years after gene therapy, uncontrolled exponential clonal proliferation of mature T cells (with gammadelta+ or alphabeta+ T cell receptors) has occurred in the two youngest patients. Both patients' clones showed retrovirus vector integration in proximity to the LMO2 proto-oncogene promoter, leading to aberrant transcription and expression of LMO2. Thus, retrovirus vector insertion can trigger deregulated premalignant cell proliferation with unexpected frequency, most likely driven by retrovirus enhancer activity on the LMO2 gene promoter.


Asunto(s)
Proteínas de Unión al ADN/genética , Terapia Genética/efectos adversos , Vectores Genéticos , Leucemia-Linfoma de Células T del Adulto/etiología , Metaloproteínas/genética , Retroviridae/genética , Inmunodeficiencia Combinada Grave/terapia , Linfocitos T/fisiología , Proteínas Adaptadoras Transductoras de Señales , Ensayos Clínicos como Asunto , Células Clonales/fisiología , Regulación de la Expresión Génica , Técnicas de Transferencia de Gen , Trasplante de Células Madre Hematopoyéticas , Células Madre Hematopoyéticas/fisiología , Humanos , Lactante , Proteínas con Dominio LIM , Mutagénesis Insercional , Regiones Promotoras Genéticas , Proto-Oncogenes Mas , Proteínas Proto-Oncogénicas , Proto-Oncogenes , Receptores de Interleucina-2/genética , Retroviridae/fisiología , Transcripción Genética , Integración Viral , Replicación Viral
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA