Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Gene Ther ; 22(11): 883-92, 2015 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-26125609

RESUMO

Successful application of gene therapy strategies may require stringently regulated transgene expression. Along this line, we describe a doxycycline (Dox)-inducible 'all-in-one' lentiviral vector design using the pTET-T11 (TII) minimal-promoter and a reverse transactivator protein (rtTA2S-M2) driven by the phosphoglycerate kinase promoter allowing for tight regulation of transgene expression (Lv.TII vectors). Vector design was evaluated in human hematopoietic cells in the context of cytidine deaminase (hCDD)-based myeloprotective gene therapy. Upon Dox administration, a rapid (16-24 h) and dose-dependent (>0.04 µg ml(-1) Dox) onset of transgene expression was detected in Lv.TII.CDD gene-modified K562 cells as well as in primary human CD34(+) hematopoietic cells. Importantly, in both cell models low background transgene expression was observed in the absence of Dox. Functionality of Dox-inducible hCDD expression was demonstrated by >10-fold increase in cytosine arabinoside (1-ß-d-arabinofuranosylcytosine, Ara-C) resistance of Lv.TII.CDD-transduced K562 cells. In addition, Lv.TII.CDD-transduced CD34(+)-derived myeloid cells were protected from up to 300 nm Ara-C (control affected from 50 nm onwards). These data clearly demonstrate the suitability of our self-inactivating lentiviral vector to induce robust, tightly regulated transgene expression in human hematopoietic cells with minimal background activity and highlight the potential of our construct in myeloprotective gene therapy strategies.


Assuntos
Terapia Genética/métodos , Células-Tronco Hematopoéticas/efeitos dos fármacos , Células-Tronco Hematopoéticas/fisiologia , Lentivirus/genética , Antimetabólitos Antineoplásicos/toxicidade , Citarabina/toxicidade , Citidina Desaminase/biossíntese , Citidina Desaminase/genética , Doxiciclina/farmacologia , Vetores Genéticos/administração & dosagem , Vetores Genéticos/genética , Células-Tronco Hematopoéticas/virologia , Humanos , Células K562 , Cultura Primária de Células , Regiões Promotoras Genéticas , Transgenes
2.
Leukemia ; 29(7): 1530-42, 2015 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-25652739

RESUMO

Co-transplantation of hematopoietic stem cells with those engineered to express leukemia-reactive T-cell receptors (TCRs) and differentiated ex vivo into precursor T cells (preTs) may reduce the risk of leukemia relapse. As expression of potentially self-(leukemia-) reactive TCRs will lead to negative selection or provoke autoimmunity upon thymic maturation, we investigated a novel concept whereby TCR expression set under the control of an inducible promoter would allow timely controlled TCR expression. After in vivo maturation and gene induction, preTs developed potent anti-leukemia effects. Engineered preTs provided protection even after repeated leukemia challenges by giving rise to effector and central memory cells. Importantly, adoptive transfer of TCR-transduced allogeneic preTs mediated anti-leukemia effect without evoking graft-versus-host disease (GVHD). Earlier transgene induction forced CD8(+) T-cell development was required to obtain a mature T-cell subset of targeted specificity, allowed engineered T cells to efficiently pass positive selection and abrogated the endogenous T-cell repertoire. Later induction favored CD4 differentiation and failed to produce a leukemia-reactive population emphasizing the dominant role of positive selection. Taken together, we provide new functional insights for the employment of TCR-engineered precursor cells as a controllable immunotherapeutic modality with significant anti-leukemia activity.


Assuntos
Doença Enxerto-Hospedeiro/prevenção & controle , Efeito Enxerto vs Leucemia/imunologia , Leucemia Mieloide/imunologia , Células Precursoras de Linfócitos T/imunologia , Receptores de Antígenos de Linfócitos T/genética , Transferência Adotiva , Animais , Citometria de Fluxo , Engenharia Genética , Doença Enxerto-Hospedeiro/mortalidade , Transplante de Células-Tronco Hematopoéticas , Humanos , Leucemia Mieloide/mortalidade , Leucemia Mieloide/terapia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Ovalbumina/genética , Regiões Promotoras Genéticas/genética , Receptores de Antígenos de Linfócitos T/imunologia , Transplante Homólogo
3.
Stem Cells ; 32(2): 436-46, 2014 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-24420904

RESUMO

Fanconi anemia (FA) is a complex genetic disease associated with a defective DNA repair pathway known as the FA pathway. In contrast to many other FA proteins, BRCA2 participates downstream in this pathway and has a critical role in homology-directed recombination (HDR). In our current studies, we have observed an extremely low reprogramming efficiency in cells with a hypomorphic mutation in Brca2 (Brca2(Δ) (27/) (Δ27)), that was associated with increased apoptosis and defective generation of nuclear RAD51 foci during the reprogramming process. Gene complementation facilitated the generation of Brca2(Δ) (27/) (Δ27) induced pluripotent stem cells (iPSCs) with a disease-free FA phenotype. Karyotype analyses and comparative genome hybridization arrays of complemented Brca2(Δ) (27/) (Δ27) iPSCs showed, however, the presence of different genetic alterations in these cells, most of which were not evident in their parental Brca2(Δ) (27/) (Δ27) mouse embryonic fibroblasts. Gene-corrected Brca2(Δ) (27/) (Δ27) iPSCs could be differentiated in vitro toward the hematopoietic lineage, although with a more limited efficacy than WT iPSCs or mouse embryonic stem cells, and did not engraft in irradiated Brca2(Δ) (27/) (Δ27) recipients. Our results are consistent with previous studies proposing that HDR is critical for cell reprogramming and demonstrate that reprogramming defects characteristic of Brca2 mutant cells can be efficiently overcome by gene complementation. Finally, based on analysis of the phenotype, genetic stability, and hematopoietic differentiation potential of gene-corrected Brca2(Δ) (27/) (Δ) (27) iPSCs, achievements and limitations in the application of current reprogramming approaches in hematopoietic stem cell therapy are also discussed.


Assuntos
Proteína BRCA2/genética , Anemia de Fanconi/genética , Terapia Genética , Células-Tronco Hematopoéticas , Células-Tronco Pluripotentes Induzidas/citologia , Animais , Proteína BRCA2/biossíntese , Diferenciação Celular/genética , Células Cultivadas , Reprogramação Celular , Dano ao DNA/genética , Anemia de Fanconi/patologia , Anemia de Fanconi/terapia , Fibroblastos/metabolismo , Células-Tronco Hematopoéticas/metabolismo , Células-Tronco Pluripotentes Induzidas/metabolismo , Camundongos
4.
Leukemia ; 21(10): 2171-80, 2007 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-17673903

RESUMO

Neurotrophins and their receptors play a key role in neurogenesis and survival. However, we and others have recently obtained evidence for a potential involvement of this receptor system in leukemia. To investigate mechanisms underlying the leukemogenic potential of activated neurotrophin receptor signaling, we analyzed in vivo leukemogenesis mediated by deltaTrkA, a mutant of TRKA (tropomyosin-related kinase A) isolated from a patient with acute myeloid leukemia (AML). Retroviral expression of deltaTrkA in myeloid 32D cells induced AML in syngeneic C3H/Hej mice (n=11/11, latency approximately 4 weeks). C57Bl/6J mice transplanted with deltaTrkA-transduced primary lineage negative (Lin-) bone marrow cells died of a transient polyclonal AML (n=7/15, latency of <12 days). Serial transplantation of AML cells did not re-induce this disease but rather acute lymphoblastic leukemia (ALL, latency >78 days). All primary recipients surviving the early AML developed clonal ALL or myeloid leukemia (latency >72 days) that required additional genetic lesions. PI3K and mTOR-raptor were identified as the crucial mediators of leukemic transformation, whereas STAT and MAP kinase signaling pathways were not activated. Thus, our findings reveal potent and unique transforming properties of altered neurotrophin receptor signaling in leukemogenesis, and encourage further analyses of neurotrophin receptors and downstream signaling events in hematological malignancies.


Assuntos
Regulação Leucêmica da Expressão Gênica , Leucemia/metabolismo , Receptor trkA/metabolismo , Receptor trkA/fisiologia , Animais , Células da Medula Óssea/citologia , Transplante de Medula Óssea , Neoplasias Hematológicas/metabolismo , Cariotipagem , Sistema de Sinalização das MAP Quinases , Camundongos , Camundongos Endogâmicos C3H , Camundongos Endogâmicos C57BL , Modelos Biológicos , Fosfatidilinositol 3-Quinases/metabolismo , Receptores Proteína Tirosina Quinases/metabolismo , Transdução de Sinais
5.
Gene Ther ; 13(13): 1037-47, 2006 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-16541120

RESUMO

The failure of pharmacological approaches to cure infection with the human immunodeficiency virus (HIV) has renewed the interest in gene-based therapies. Among the various strategies that are currently explored, the blockade of HIV entry into susceptible T cells and macrophages promises to be the most powerful intervention. For long-term protection of both of these lineages, genetic modification of hematopoietic stem cells (HSCs) would be required. Here, we tested whether HSCs and their progeny can be modified to express therapeutic levels of M87o, a gammaretroviral vector encoding an artificial transmembrane molecule that blocks fusion-mediated uptake of HIV. In serial murine bone marrow transplantations, efficient and multilineage expression of M87o was observed for more than 1 year (range 37-75% of mononuclear cells), without signs of toxicity related to the transmembrane molecule. To allow enrichment of M87o-modified HSCs after transplant, we constructed vectors coexpressing the P140K mutant of O(6)-methylguanine-DNA-methyltransferase (MGMT-P140K). This clinically relevant selection marker mediates a survival advantage in HSCs if exposed to combinations of methylguanine-methyltransferase (MGMT) inhibitors and alkylating agents. A bicistronic vector mediated sufficient expression of both M87o and MGMT to confer a selective survival advantage in the presence of HIV and alkylating agents, respectively. These data encourage further investigations in large animal models and clinical trials.


Assuntos
Vacinas contra a AIDS/administração & dosagem , Terapia Genética/métodos , Inibidores da Fusão de HIV/uso terapêutico , Infecções por HIV/prevenção & controle , HIV-1 , Células-Tronco Hematopoéticas/metabolismo , Vacinas contra a AIDS/genética , Animais , Citometria de Fluxo , Expressão Gênica , Vetores Genéticos/administração & dosagem , Vetores Genéticos/genética , Infecções por HIV/metabolismo , Transplante de Células-Tronco Hematopoéticas/métodos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Modelos Animais , O(6)-Metilguanina-DNA Metiltransferase/análise , Retroviridae/genética , Transdução Genética/métodos
6.
Leukemia ; 16(9): 1655-63, 2002 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-12200677

RESUMO

Many applications of hematopoietic gene therapy require selection for clones with active transgene expression. However, it was unclear whether the clonal progeny of a retrovirally transduced hematopoietic stem cell would be capable of maintaining transgene expression through serial repopulation and multilineage differentiation. Such investigations require simultaneous analyses of clonality, multilineage activity and transgene copy numbers. Using a mouse model, the present study demonstrates that a single hematopoietic stem cell expressing a marker gene from one or two insertions of a simple retroviral vector actively maintains multilineage transgene expression in the vast majority (80-99%) of bone marrow and peripheral blood cells. Gene expression persisted through serial transplantations for at least 97 weeks post gene transfer and was observed in the lymphoid (B, T and NK cells), myeloid (CD11b(+), Gr-1(+)), erythroid (Ter119(+), mature red blood cells) and megakaryocytic (as indicated by platelets) progeny. Therefore, a single immunoselection for hematopoietic stem cells expressing the transgene in vivo was sufficient to establish a completely chimeric hematopoiesis. These observations imply that the retroviral vectors used in this study contain cis-elements that mediate expression through massive clonal expansion and multilineage differentiation, provided the insertion occurred in genetic loci permissive for expression in hematopoietic stem cells.


Assuntos
Antígenos CD34/genética , Linhagem da Célula/fisiologia , Hematopoese/fisiologia , Células-Tronco Hematopoéticas/fisiologia , Retroviridae/genética , Transgenes/fisiologia , Animais , Antígenos CD34/metabolismo , Células da Medula Óssea/metabolismo , Quimera , Ensaio de Unidades Formadoras de Colônias , Feminino , Dosagem de Genes , Inativação Gênica , Técnicas de Transferência de Genes , Proteínas de Fluorescência Verde , Transplante de Células-Tronco Hematopoéticas , Humanos , Proteínas Luminescentes/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Transdução Genética , Transfecção
7.
Cancer Gene Ther ; 8(6): 440-9, 2001 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-11498764

RESUMO

Peripheral blood progenitor cells are a prime target for gene therapy approaches. As recent data point to the relevance of soluble stroma factors for the efficient transduction of progenitor cells, we tested the stroma-conditioned medium (SCM) of the two cell lines FBMD-1 and L88/5 as well as desulfated and O-sulfated heparin (HS dS and HS OS) for their effect on transduction of peripheral blood progenitor cells. We transduced CD34+ cells of nine tumor patients with the retroviral SF-MDR vector containing the human multidrug resistance 1 (MDR1) gene under serum-free conditions on the fibronectin fragment CH-296 with or without SCM. Provirus-specific polymerase chain reaction showed a median 1.6-fold higher integration rate of the transgene into committed progenitor cells for the group with added FBMD-1 SCM (P=.008). This was maintained after 2 (P=.02) and, as a trend, after 5 weeks of stroma-dependent long-term culture. We found a median 1.5-fold increase in rhodamine-123 (Rh-123) exclusion in myeloid lineage-committed progeny cells following transduction in the presence of FBMD-1 SCM (P=.0004). After 2 or 5 weeks of long-term culture, a significantly higher proportion of Rh-123(dull) cells could still be detected in the FBMD-1 SCM transduction group (P=.003 and P=.04, respectively). L88/5 SCM or HS OS or HS dS was not effective as supplement for improving gene transfer. The FBMD-1 stroma cell line appears to secrete a unique moiety, which can increase retroviral transduction of lineage-committed and primitive progenitor cells. The FBMD-1 stroma activity is not attributable to heparan sulfate.


Assuntos
Sangue/metabolismo , Neoplasias/sangue , Retroviridae/genética , Células Estromais/citologia , Células Estromais/metabolismo , Transdução Genética , Adolescente , Adulto , Antígenos CD34/metabolismo , Linhagem da Célula , Células Cultivadas , Meios de Cultura Livres de Soro/farmacologia , Feminino , Fibronectinas/química , Humanos , Masculino , Pessoa de Meia-Idade , Reação em Cadeia da Polimerase , Rodaminas/metabolismo , Células-Tronco/metabolismo , Fatores de Tempo , Transgenes
8.
Gene Ther ; 8(10): 811-7, 2001 May.
Artigo em Inglês | MEDLINE | ID: mdl-11420646

RESUMO

Retroviral vector-mediated expression of the homeoboxgene, HoxB4, in hematopoietic cells has been reported to mediate a benign expansion of gene-modified hematopoietic stem and precursor cells in vivo. In the present study, we used a novel coexpression strategy for coordinated expression of HoxB4 along with a cytoplasmic protein from a retroviral vector. The novel coexpression strategy, based on cotranslational protein separation mediated by the 2A sequence of foot-and-mouth disease virus (FMDV), allows an indirect quantification of HoxB4 expression levels when inserting a reporter such as the enhanced green fluorescent protein (GFP) in the retroviral vector. Presence of the 2A sequence did not interfere with the correct subcellular localization of HoxB4 (nuclear) and GFP (cytoplasmic), nor with the titer of bicistronic vectors, and mediated functional long-term coexpression (at least 1 year) of GFP and HoxB4 after transplantation of transduced mouse bone marrow cells in mice.


Assuntos
Terapia Genética/métodos , Vetores Genéticos/administração & dosagem , Células-Tronco Hematopoéticas/metabolismo , Retroviridae/genética , Transdução Genética/métodos , Animais , Aphthovirus/genética , Western Blotting , Núcleo Celular/metabolismo , Citoplasma/metabolismo , Feminino , Imunofluorescência , Expressão Gênica , Proteínas de Fluorescência Verde , Proteínas Luminescentes/genética , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Poliovirus/genética , Fatores de Tempo
9.
Gene Ther ; 8(3): 239-46, 2001 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-11313796

RESUMO

We describe the functional analysis of a novel retroviral vector, SF91m3, which was designed for improved expression of the in vivo selectable marker, multidrug resistance 1 gene (MDR1), in hematopoietic cells. SF91m3 combines several promising features. The vector backbone lacks viral coding sequences and AUG-start codons 5' of the MDR1 cDNA. A point mutation of a cryptic splice acceptor of the MDR1 cDNA increases the probability of transferring an intact provirus. The titer of a PG13 packaging cell clone containing a single proviral integration is high (>2 x 10(6) particles/ml from frozen stocks of serum-free vector harvests). Human hematopoietic cells transduced with SF91m3 reliably express MDR1 before and after passage through NOD/SCID mice, as shown by quantitative PCR and efflux assays with rhodamine 123 or Hoechst 33342. Finally, SF91m3 mediates resistance to escalated doses of cytotoxic agents, as shown by survival and differentiation of transduced colony-forming cells in the presence of colchicine at 48 ng/ml (>10 x IC(50)). Thus, SF91m3 may represent an interesting candidate for future trials addressing the safety and utility of MDR1 gene transfer; moreover, this study demonstrates that sequence alterations improving post-transcriptional processing of retroviral vectors have a substantial impact for gene expression in hematopoietic cells.


Assuntos
Membro 1 da Subfamília B de Cassetes de Ligação de ATP/genética , Resistência a Múltiplos Medicamentos/genética , Vetores Genéticos , Células-Tronco Hematopoéticas/efeitos dos fármacos , Retroviridae/genética , Animais , Antineoplásicos/farmacologia , Ensaio de Unidades Formadoras de Colônias , Feminino , Técnicas de Transferência de Genes , Transplante de Células-Tronco Hematopoéticas , Humanos , Camundongos , Camundongos Endogâmicos NOD , Camundongos SCID , Processamento Pós-Transcricional do RNA , Transdução Genética
10.
Mol Ther ; 2(6): 609-18, 2000 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-11124062

RESUMO

We have adapted a recently published protocol for retroviral gene transfer into hematopoietic cells [A. J. Schilz et al. (1998) Blood 92: 3163-3171] with respect to clinical requirements such as large-volume vector stock generation, adequate cell source, high cell numbers, and serum-free conditions. We present data on transduction efficacy and expression of the multidrug resistance 1 (MDR1) gene in human CD34(+) cells from mobilized peripheral blood (PB) mediated by a gibbon ape leukemia virus (GALV)-pseudotyped retroviral vector. Using a 1-day cytokine-mediated prestimulation, consisting of human interleukin (IL)-3, IL-6, stem cell factor (SCF), Flt-3 ligand (FL), and thrombopoietin (TPO), followed by a 3-day transduction procedure, we were able to detect up to 51% CD34(+) cells expressing MDR1. Xenotransplantation of transduced cells into NOD/LtSz-scid/scid (NOD/SCID) mice resulted in a mean engraftment level of 23% (0.1 to 87%). As shown by quantitative PCR analysis, a mean of 12.7% (range 0.3 to 55%) of the engrafted human cells in the bone marrow of chimeric mice contained the MDR1 cDNA. Furthermore, enhanced expression of MDR1 above control levels was detected in up to 15% of the engrafted human cell population. Our data suggest that NOD/SCID repopulating cells derived from mobilized PB can be transduced efficiently with existing retroviral vector systems under clinically applicable conditions.


Assuntos
Membro 1 da Subfamília B de Cassetes de Ligação de ATP/genética , Expressão Gênica , Técnicas de Transferência de Genes , Células-Tronco Hematopoéticas/citologia , Retroviridae/genética , Animais , Antígenos CD34/análise , Sequência de Bases , Divisão Celular , Meios de Cultura Livres de Soro , Primers do DNA , Feminino , Células-Tronco Hematopoéticas/imunologia , Camundongos , Camundongos Endogâmicos NOD , Camundongos SCID
11.
Bone Marrow Transplant ; 25 Suppl 2: S118-24, 2000 May.
Artigo em Inglês | MEDLINE | ID: mdl-10933204

RESUMO

Mobilized peripheral blood progenitor cells (PBPC) are an attractive target for the retrovirus-mediated transfer of cytostatic drug resistance genes. We analyzed NOD/SCID mouse repopulating CD34+ PBPC from cancer patients following retroviral Transwell transduction in various cytokine combinations with the FMEV-based (Friend-mink cell focus forming/murine embryonic stem cell virus) hybrid vector SF-MDR carrying the human multidrug resistance-1 (MDR1) gene. Five to 10 weeks following transplantation of 2.0 x 10(6) CD34+ PBPC into NOD/SCID mice we observed medium to high levels of human cell engraftment with up to 33%. The extent of vector-marked human cells was assessed by a quantitative real-time polymerase chain reaction (PCR). SF-MDR gene transfer into long-term in vivo repopulating human hematopoietic cells was optimal in the presence of either IL-3/IL-6/SCF/FL or FL/TPO/SCF resulting in three-fold (12.4% +/- 1.7%) or four-fold (16.5% +/- 6.8%) higher average proportions of gene-marked human cells in NOD/SCID mice as compared to IL-3 alone (P < 0.01). In conclusion, we could optimize the engraftment capacity and the retroviral gene transfer to CD34+ PBPC using cocktails of early acting cytokines in combination with the recombinant fibronectin fragment CH-296. Our data suggest that the NOD/SCID model provides a valid assay to estimate the gene transfer efficiency to repopulating human PBPC that may be achievable in clinical autologous transplantation settings.


Assuntos
Genes MDR , Células-Tronco Hematopoéticas/metabolismo , Transdução Genética , Animais , Sequência de Bases , Primers do DNA/genética , Terapia Genética , Sobrevivência de Enxerto , Transplante de Células-Tronco Hematopoéticas , Humanos , Camundongos , Camundongos Endogâmicos NOD , Camundongos SCID , Retroviridae/genética , Transplante Heterólogo
12.
Cancer Gene Ther ; 7(4): 597-604, 2000 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-10811478

RESUMO

The use of autologous hematopoietic stem cell (HSC) grafts after high-dose chemotherapy protocols may be hampered by contamination of the grafts with tumor cells. Because epithelial cells seem to be the natural hosts of adeno-associated virus 2 (AAV-2), we speculated that epithelial tumor cells in HSC grafts might be selective targets for AAV-2-based vectors. To test this hypothesis, the breast cancer cell lines T47D and MCF-7 were infected with a recombinant AAV-2 vector expressing the green fluorescent protein (GFP) gene; in addition, human CD34+ mobilized peripheral progenitor cells were infected with the same vector. At a multiplicity of infection of 100, only 1.39% +/- 0.51% CD34+ cells expressed the GFP gene whereas, 36.06% +/- 6.53% of the infected T47D cells and 41.52% +/- 3.16% of the infected MCF-7 cells expressed the transduced GFP gene. After further optimizing the transduction procedure by using higher multiplicities of infection (100-500) and preincubation of samples with the tyrosine kinase inhibitor genistein, up to 82.52% and 85.35% GFP+ T47D and MCF-7 cells, respectively, were observed. The GFP fluorescence intensity in transduced mammary tumor cells was up to 3 logs higher than that of transduced CD34+ cells. The differential expression of recombinant AAV-2 vectors in hematopoietic and epithelial tumor cells warrants further research with this vector system, including the use of suicide genes for the purging of autologous HSC grafts.


Assuntos
Neoplasias da Mama/patologia , Dependovirus/genética , Vetores Genéticos , Células-Tronco Hematopoéticas/citologia , Transfecção/métodos , Antígenos CD34 , Células Cultivadas , Feminino , Genes Reporter , Proteínas de Fluorescência Verde , Mobilização de Células-Tronco Hematopoéticas , Células-Tronco Hematopoéticas/virologia , Humanos , Proteínas Luminescentes/análise , Proteínas Luminescentes/genética , Recombinação Genética , Células Tumorais Cultivadas
13.
Blood ; 95(4): 1237-48, 2000 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-10666196

RESUMO

Mobilized peripheral blood progenitor cells (PBPC) are a potential target for the retrovirus-mediated transfer of cytostatic drug-resistance genes. We analyzed nonobese diabetic/severe combined immunodeficient (NOD/SCID) mouse-repopulating CD34+ PBPC from patients with cancer after retroviral transduction in various cytokine combinations with the hybrid vector SF-MDR, which is based on the Friend mink cell focus-forming/murine embryonic stem-cell virus and carries the human multidrug resistance 1 (MDR1) gene. Five to 13 weeks after transplantation of CD34+ PBPC into NOD/SCID mice (n = 84), a cell dose-dependent multilineage engraftment of human leukocytes up to an average of 33% was observed. The SF-MDR provirus was detected in the bone marrow (BM) and in its granulocyte fractions in 96% and 72%, respectively, of chimeric NOD/SCID mice. SF-MDR provirus integration assessed by quantitative real-time polymerase chain reaction (PCR) was optimal in the presence of Flt-3 ligand/thrombopoietin/stem-cell factor, resulting in a 6-fold (24% +/- 5% [mean +/- SE]) higher average proportion of gene-marked human cells in NOD/SCID mice than that achieved with IL-3 alone (P <.01). A population of clearly rhodamine-123(dull) human myeloid progeny cells could be isolated from BM samples from chimeric NOD/SCID mice. On the basis of PCR and rhodamine-123 efflux data, up to 18% +/- 4% of transduced cells were calculated to express the transgene. Our data suggest that the NOD/SCID model provides a valid assay for estimating the gene-transfer efficiency to repopulating human PBPC that may be achievable in clinical autologous transplantation. P-glycoprotein expression sufficient to prevent marrow aplasia in vivo may be obtained with this SF-MDR vector and an optimized transduction protocol. (Blood. 2000;95:1237-1248)


Assuntos
Adipócitos/citologia , Genes MDR , Terapia Genética/métodos , Transplante de Células-Tronco Hematopoéticas , Células-Tronco Hematopoéticas/citologia , Retroviridae , Transfecção/métodos , Transplante Heterólogo/imunologia , Animais , Antígenos CD34 , Células da Medula Óssea/citologia , Linhagem Celular , Vetores Genéticos , Células-Tronco Hematopoéticas/imunologia , Células-Tronco Hematopoéticas/fisiologia , Humanos , Antígenos Comuns de Leucócito/imunologia , Camundongos , Camundongos Endogâmicos NOD , Camundongos SCID , Neoplasias/sangue , Reação em Cadeia da Polimerase , Células Estromais/citologia
14.
Eur J Cancer ; 35(7): 1136-42, 1999 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-10533460

RESUMO

Autologous peripheral blood progenitor cell (PBPC) grafts can be contaminated with tumour cells that potentially give rise to relapse following myeloablative therapy and PBPC transplantation. Adeno-associated virus (AAV)-based vectors produced by a new adenovirus-free technique are a gene delivery system which may be applicable for tumour cell purging. To test for the host range of these vectors, solid tumours of clinical relevance and normal CD34+ PBPC were selected as target cells for an AAV-vector, encoding the green-fluorescent protein (GFP) as the indicator gene. At a multiplicity of infection (MOI) of 100: 79.94% +/- 14.36% (mean +/- SEM) of the connective tissue sarcoma cell line (HS-1) and 64.84% +/- 6.91% of the cervical carcinoma cell line cells (HeLa-RC) expressed GFP while the other cell lines tested (1 ovarian tumour, 1 germ cell tumour, 1 osteosarcoma, 2 small cell lung cancer) ranged between 2.82% and 11.94%. Optimising the transduction protocol by use of higher MOIs of up to 500 and by pretreatment with the tyrosine kinase inhibitor, genistein, resulted in up to 95.97% and 94.10% green-fluorescent HS-1 and HeLa-RC cells, respectively. In contrast, only 1.39% +/- 0.51% of the normal haematopoietic CD34+ progenitor cells expressed GFP at a MOI of 100. The differential infectivity between HS-1 and CD34+ cells was maintained after tumour cell spiking in leucapheresis products. Our observations suggest that AAV-based vectors may prove useful for purging of autologous PBPC grafts from solid tumour cells.


Assuntos
Técnicas de Transferência de Genes , Vetores Genéticos/administração & dosagem , Transplante de Células-Tronco Hematopoéticas/métodos , Neoplasias/terapia , Adenoviridae , Antígenos CD , Células HeLa , Mobilização de Células-Tronco Hematopoéticas/métodos , Humanos , Células Tumorais Cultivadas
15.
Cancer Chemother Pharmacol ; 44(5): 433-8, 1999.
Artigo em Inglês | MEDLINE | ID: mdl-10501919

RESUMO

PURPOSE: In order to assess the effect of the tyrosine kinase inhibitor CGP57148B on lineage-committed and primitive chronic myelogenous leukemia (CML) progenitor cells, peripheral blood progenitor cells (PBPC) mobilized in chronic phase CML were exposed to this compound in vitro. METHODS: Both short-term (/=2 weeks) to CGP57148B were investigated using suspension culture, semisolid (methylcellulose) assay or stroma-dependent long-term culture (LTC). The proportion of bcr/abl-positive progenitors was determined after direct plating [2 weeks in colony-forming cell (CFC) assay] as well as after 2 or 6 weeks LTC (LTC always followed by CFC replates). RESULTS: Incubation of CML PBPC over 48 h in suspension culture with 100 microM CGP57148B reduced the proportion of bcr/abl-positive colonies to 4.4 +/- 4.3% (n = 5) after direct plating, 6.6 +/- 4.2% (n = 5) after 2 weeks LTC and 5 +/- 5.6% (n = 2) after 6 weeks LTC. At this dose, survival of drug-exposed normal PBPC was 53 +/- 4.2%, 51 +/- 2.8% and 54.5 +/- 4.9% (n = 2), respectively. Incubation with CGP57148B at a concentration of 10 microM over 1 week under LTC conditions reduced the number of bcr/abl-positive colonies to 11.8 +/- 6.1% (n = 5) after direct plating, 12 +/- 6.4% (n = 4) after 2 weeks LTC and 14.3 +/- 11.4% (n = 3) after 6 weeks LTC; survival of normal PBPC was 84.5 +/- 2.1%, 93 +/- 4.2% and 86 +/- 1.4% (n = 2), respectively. Following long-term exposure to CGP57148B at a concentration of 1 microM, the proportion of remaining bcr/abl-positive colonies was 35%, 9% and 25% of untreated CML samples after direct plating as well as after 2 and 6 weeks LTC, respectively. The respective values for 10 microM CGP57148B were 10%, 11% and 19%. Long-term exposure of normal progenitors to CGP57148B yielded a survival of 98%, 100% and 93% (1 microM) or 77%, 86% and 80% (10 microM), respectively. CONCLUSION: The results support the use of CGP57148B either for short-term exposure in vitro (e.g. purging) or for continuous treatment of CML in vivo.


Assuntos
Antineoplásicos/toxicidade , Inibidores Enzimáticos/toxicidade , Proteínas de Fusão bcr-abl/metabolismo , Células-Tronco Hematopoéticas/patologia , Leucemia Mielogênica Crônica BCR-ABL Positiva/patologia , Piperazinas/toxicidade , Proteínas Tirosina Quinases/antagonistas & inibidores , Pirimidinas/toxicidade , Benzamidas , Técnicas de Cocultura , Ensaio de Unidades Formadoras de Colônias , Ensaios de Seleção de Medicamentos Antitumorais , Células-Tronco Hematopoéticas/efeitos dos fármacos , Humanos , Mesilato de Imatinib , Leucemia Mielogênica Crônica BCR-ABL Positiva/sangue , Células Estromais/citologia , Células Estromais/patologia , Células Tumorais Cultivadas
16.
Hum Gene Ther ; 10(9): 1443-52, 1999 Jun 10.
Artigo em Inglês | MEDLINE | ID: mdl-10395370

RESUMO

Hematopoietic stem cells (HSCs) are a potential target for the retrovirus-mediated transfer of chemotherapeutic drug resistance genes. For integration of the proviral DNA in the HSC genome cell division is required. In the bone marrow (BM) hematopoiesis occurs in the vicinity of stroma cells. Soluble stroma components were shown to play a permissive role for the proliferation of lineage-committed and primitive hematopoietic progenitors in conjunction with cytokines. We investigated the effect of stroma-conditioned medium (SCM) of the FBMD1 cell line on the gene transfer rate of the human multidrug resistance 1 (MDR1) gene contained in the retroviral SF-MDR vector into human mobilized peripheral blood progenitor cells (PBPCs) from tumor patients (n = 14) during transwell transduction in the presence of the recombinant fibronectin fragment CH-296. Addition of SCM during transduction increased the gene transfer efficiency into myeloid lineage-committed colony-forming cells by an average of 1.5-fold (p = 0.02) as detected by an SF-MDR provirus-specific polymerase chain reaction (PCR). These data were paralleled by significantly (p = 0.04 to p = 0.007) higher proportions of MDR1-expressing myelo-monocytic progeny after transduction in SCM plus interleukin 3 (IL-3), IL-3/Flt3 ligand (FL), IL-3/IL-6/FL, or IL-3/IL-6/stem cell factor (SCF) when compared with transductions without SCM as measured by rhodamine-123 exclusion. A similar trend was observed for SCM employed in combination with IL-3/IL-6/SCF/FL or FL/thrombopoietin (TPO)/SCF during transduction. The latter combination plus SCM yielded the highest proportion, 19.16 +/- 3.10% Rh-123dull cells. The beneficial effect of SCM on transduction efficiency was confirmed in additional four patients' samples, using a serum-free viral supernatant transduction protocol. As soluble BM stroma factors are able to increase the efficiency of retrovirus-mediated gene transfer into committed progenitor cells, beyond that achieved with fibronectin fragment CH-296, their effect on gene transfer into primitive repopulating hematopoietic cells may also prove beneficial.


Assuntos
Membro 1 da Subfamília B de Cassetes de Ligação de ATP/genética , Células da Medula Óssea/metabolismo , Técnicas de Transferência de Genes , Vetores Genéticos , Células-Tronco Hematopoéticas/efeitos dos fármacos , Retroviridae , Antineoplásicos Fitogênicos/farmacologia , Células da Medula Óssea/citologia , Divisão Celular/efeitos dos fármacos , Linhagem Celular , Meios de Cultivo Condicionados , Meios de Cultura Livres de Soro , Células-Tronco Hematopoéticas/citologia , Humanos , Monócitos , Rodamina 123 , Solubilidade , Células Estromais/citologia , Células Estromais/metabolismo , Vincristina/farmacologia
17.
Proc Natl Acad Sci U S A ; 91(11): 5080-4, 1994 May 24.
Artigo em Inglês | MEDLINE | ID: mdl-8197189

RESUMO

Stable nuclear transformation of Volvox carteri was achieved using the cloned V. carteri nitA+ gene (which encodes nitrate reductase) to complement a nitA- mutation. Following bombardment of mutant cells with plasmid-coated gold particles, putative transformants able to utilize nitrate as a nitrogen source were recovered with an efficiency of approximately 2.5 x 10(5). DNA analysis indicated that the plasmid integrated into the genome, often in multiple copies, at sites other than the nitA locus. Cotransformants were recovered with a frequency of 40-80% when cells were cobombarded with a selected and an unselected marker. Thus, V. carteri becomes one of the simplest multicellular organisms that is accessible to detailed molecular studies of genes regulating cellular differentiation and morphogenesis.


Assuntos
Clorófitas/genética , Nitrato Redutases/genética , Transformação Genética , Southern Blotting , Diferenciação Celular/genética , Clorófitas/citologia , Clorófitas/enzimologia , Marcadores Genéticos , Nitrato Redutase , Plasmídeos
18.
Curr Genet ; 25(2): 169-77, 1994 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-8087887

RESUMO

Southern analysis indicated the presence of at least four ubiquitin gene loci in the Volvox carteri genome. Three of these, a polyubiquitin gene described here and a non-segregating ubiquitin gene pair, were assigned to two different linkage groups by RFLP mapping; the non-polymorphic fourth gene locus remained unassigned. The polyubiquitin gene was cloned and its 2,116-bp sequence determined. It contains six exons each interrupted by an intron at Gly35, and it encodes a pentameric polyubiquitin polypeptide consisting of five runs of 76 identical amino-acid residues and a C-terminal extension of one leucine. The five tandem repeats of coding units plus introns exhibit an unusually high degree of overall sequence identity indicating an efficient process of gene homogenization in this region of the V. carteri genome. S1 mapping revealed two closely-spaced transcription starts, 24 and 28 nucleotides downstream from a putative TATA sequence. Preceding the TATA box are two 14-bp conserved heat-shock elements (HSEs) and two octameric sequences closely resembling an yesat HSE. Consistent with a 1.6-kb transcript seen on Northern blots are two polyadenylation signals (TGTAA) located 99 bp and 169 bp downstream from the TGA translational stop. The polyubiquitin gene was transcribed throughout the Volvox life cycle with peaks in the 1.6-kb mRNA levels during pre-cleavage, cleavage, and post-inversion. In contrast, an 0.6-kb monoubiquitin transcript was abundant only at the pre-cleavage stage suggesting a different type of gene control. Heat shock increased the level of polyubiquitin mRNA, whereas the level of monoubiquitin mRNA was down-regulated.


Assuntos
Biopolímeros/genética , Clorófitas/genética , Genes de Plantas , Ubiquitinas/genética , Sequência de Aminoácidos , Sequência de Bases , Clonagem Molecular , Códon , Dados de Sequência Molecular , Poliubiquitina , Sequências Repetitivas de Ácido Nucleico
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...