Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 30
Filtrar
Más filtros

Banco de datos
Tipo del documento
País de afiliación
Intervalo de año de publicación
1.
Onkologie ; 34(10): 502-8, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-21985848

RESUMEN

BACKGROUND: The development of distant metastasis is associated with poor outcome in patients with colorectal cancer (CRC). The stromal cell-derived factor-1 (SDF-1) and its receptor CXC chemokine receptor 4 (CXCR4) have pivotal roles in the chemotaxis of migrating tumor cells during metastasis. Thus, hampering the SDF-1/CXCR4 cross-talk is a promising strategy to suppress metastasis. METHODS: We investigated the invasive behavior of the lentivirally CXCR4 overexpressing CRC cell lines SW480, SW620 and RKO in chemotaxis and invasion assays toward an SDF-1α gradient. Low endogenous CXCR4 expression levels were determined by quantitative realtime polymerase chain reaction (PCR) and fluorescence-activated cell sorting (FACS) analyses. RESULTS: A lentiviral CXCR4 overexpression and knockdown model was established in these CRC cells. In transwell migration assays, CXCR4 overexpression favored chemotaxis and invasion of cells in all 3 lines depending on an SDF-1α gradient (p < 0.001 vs. untransduced cells). Functional CXCR4 knockdown using lentiviral short hairpin RNA (shRNA) vectors significantly decreased the migration behavior in CRC cell lines (p < 0.001), confirming a CXCR4-specific effect. Pharmacologic inhibition of the SDF-1α/CXCR4 interaction by the bicyclam Plerixafor(TM) at 100 µM significantly abrogated CXCR4-dependent migration and invasion through Matrigel(TM) (SW480, SW620, RKO; p < 0.05). CONCLUSION: Our results indicate that a CXCR4-antagonistic therapy might prevent tumor cell dissemination and metastasis in CRC patients, consequently improving survival.


Asunto(s)
Fármacos Anti-VIH/farmacología , Quimiocina CXCL12/genética , Quimiotaxis/genética , Neoplasias Colorrectales/genética , Regulación Neoplásica de la Expresión Génica/genética , Vectores Genéticos/genética , Compuestos Heterocíclicos/farmacología , Lentivirus/genética , Invasividad Neoplásica/genética , Receptores CXCR4/genética , Bencilaminas , Línea Celular Tumoral , Neoplasias Colorrectales/patología , Ciclamas , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Técnicas de Silenciamiento del Gen , Humanos , Invasividad Neoplásica/patología , Receptor Cross-Talk/efectos de los fármacos , Receptores CXCR4/antagonistas & inhibidores
2.
Strahlenther Onkol ; 186(2): 99-106, 2010 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-20127227

RESUMEN

BACKGROUND AND PURPOSE: The transmembrane protein caveolin-1 (CAV1) is an essential component of caveolae, small membrane invaginations involved in vesicle formation. CAV1 plays a role in signal transduction, tumor suppression and oncogene transformation. Previous studies with CAV1 knockout mice and CAV1 knockdown in pancreatic tumor cells implicated CAV1 in mediating radioresistance. The aim of this work was to test the effect of CAV1 overexpression after irradiation in human cells lacking endogenous CAV1 expression. MATERIAL AND METHODS: Human CAV1 was overexpressed in lymphoblastoid TK6 cells (TK6-wt) using a eukaryotic expression plasmid, pCI-CAV1, or a lentiviral SIN (self-inactivating) vector, HR'SIN-CAV1. CAV1 expression was verified in TK6 cells with Western blot analysis or intracellular FACS (fluorescence-activated cell sorting) staining. The effect of CAV1 on proliferation kinetics after irradiation of TK6 cells was measured with a growth assay. RESULTS: TK6-wt showed no detectable endogenous CAV1 expression. Lentivirally mediated transduction with HR'SIN-CAV1 (TK6-CAV1) resulted in a considerably stronger CAV1 expression in comparison to TK6 cells electroporated with pCI-CAV1. Intracellular FACS analysis showed that 90% of transduced cells expressed CAV1. CAV1 enhanced early proliferation of TK6 cells after irradiation with a dose of 2 Gy, whereas proliferation of unirradiated cells was not affected. CAV1 also protected cells after irradiation with 4 Gy. This radioprotective effect was supported by a reduction of radiation-induced apoptosis. CONCLUSION: A model system for expression of exogenous CAV1 by stable lentiviral transduction of TK6 cells was established. Functional assays demonstrated enhanced early proliferation by CAV1 expression in TK6 cells after irradiation with clinically relevant doses supporting the role of CAV1 as a prosurvival factor.


Asunto(s)
Apoptosis/efectos de la radiación , Caveolina 1/genética , División Celular/efectos de la radiación , Expresión Génica/efectos de la radiación , Apoptosis/genética , Western Blotting , División Celular/genética , Línea Celular Tumoral , Relación Dosis-Respuesta en la Radiación , Citometría de Flujo , Expresión Génica/genética , Vectores Genéticos/genética , Humanos , Lentivirus/genética , Linfocitos/metabolismo , Linfocitos/efectos de la radiación , Plásmidos , Protección Radiológica , Transducción Genética/métodos
3.
Cytotherapy ; 12(1): 107-12, 2010.
Artículo en Inglés | MEDLINE | ID: mdl-19929455

RESUMEN

BACKGROUND AND AIMS: Because of their pluripotency, human CD34(+) peripheral blood progenitor cells (PBPC) are targets of interest for the treatment of many acquired and inherited disorders using gene therapeutic approaches. Unfortunately, most current vector systems lack either sufficient transduction efficiency or an appropriate safety profile. Standard single-stranded recombinant adeno-associated virus 2 (AAV2)-based vectors offer an advantageous safety profile, yet lack the required efficiency in human PBPC. METHODS: A panel of pseudotyped AAV vectors (designated AAV2/x, containing the vector genome of serotype 2 and capsid of serotype x, AAV2/1-AAV2/6) was screened on primary human granulocyte-colony-stimulating factor (G-CSF)-mobilized CD34(+) PBPC to determine their gene transfer efficacy. Additionally, double-stranded self-complementary AAV (dsAAV) were used to determine possible second-strand synthesis limitations. RESULTS: AAV2/6 vectors proved to be the most efficient [12.8% (1.8-25.4%) transgene-expressing PBPC after a single transduction], being significantly more efficient (all P<0.005) than the other vectors [AAV2/2, 2.0% (0.2-7.3%); AAV2/1, 1.3% (0.1-2.9%); others, <; 1% transgene-expressing PBPC]. In addition, the relevance of the single-to-double-strand conversion block in transduction of human PBPC could be shown using pseudotyped dsAAV vectors: for dsAAV2/2 [9.3% (8.3-20.3%); P<0.001] and dsAAV2/6 [37.7% (23.6-61.0%); P<0.001) significantly more PBPC expressed the transgene compared with their single-stranded counterparts; for dsAAV2/1, no significant increase could be observed. CONCLUSIONS: We have shown that clinically relevant transduction efficiency levels using AAV-based vectors in human CD34(+) PBPC are feasible, thereby offering an efficient alternative vector system for gene transfer into this important target cell population.


Asunto(s)
Antígenos CD34/análisis , Dependovirus/genética , Técnicas de Transferencia de Gen , Vectores Genéticos , Células Madre/metabolismo , Adulto , Femenino , Factor Estimulante de Colonias de Granulocitos y Macrófagos/farmacología , Humanos , Masculino , Persona de Mediana Edad , Células Madre/inmunología , Transducción Genética/métodos , Transgenes
4.
Cytotherapy ; 12(3): 338-48, 2010 May.
Artículo en Inglés | MEDLINE | ID: mdl-20370349

RESUMEN

BACKGROUND AIMS: The discovery of unrestricted somatic stem cells (USSC), a non-hematopoietic stem cell population, brought cord blood (CB) to the attention of regenerative medicine for defining more protocols for non-hematopoietic indications. We demonstrate that a reliable and reproducible method for good manufacturing practice (GMP)-conforming generation of USSC is possible that fulfils safety requirements as well as criteria for clinical applications, such as adherence of strict regulations on cell isolation and expansion. METHODS: In order to maintain GMP conformity, the automated cell processing system Sepax (Biosafe) was implemented for mononucleated cell (MNC) separation from fresh CB. After USSC generation, clinical-scale expansion was achieved by multi-layered CellSTACKs (Costar/Corning). Infectious disease markers, pyrogen and endotoxin levels, immunophenotype, potency, genetic stability and sterility of the cell product were evaluated. RESULTS: The MNC isolation and cell cultivation methods used led to safe and reproducible GMP-conforming USSC production while maintaining somatic stem cell character. CONCLUSIONS: Together with implemented in-process controls guaranteeing contamination-free products with adult stem cell character, USSC produced as suggested here may serve as a universal allogeneic stem cell source for future cell treatment and clinical settings.


Asunto(s)
Sangre Fetal/citología , Trasplante de Células Madre , Células Madre , Animales , Biomarcadores/metabolismo , Técnicas de Cultivo de Célula , Linaje de la Célula , Células Cultivadas , Centrifugación por Gradiente de Densidad/instrumentación , Centrifugación por Gradiente de Densidad/métodos , Citometría de Flujo , Humanos , Inmunofenotipificación , Ratones , Ratones Desnudos , Regeneración/fisiología , Trasplante de Células Madre/legislación & jurisprudencia , Trasplante de Células Madre/métodos , Trasplante de Células Madre/normas , Células Madre/citología , Células Madre/fisiología , Trasplante Autólogo , Trasplante Homólogo
5.
Strahlenther Onkol ; 185(8): 517-23, 2009 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-19652935

RESUMEN

BACKGROUND AND PURPOSE: Protection of normal tissue against radiation-induced damage may increase the therapeutic ratio of radiotherapy. A promising strategy for testing this approach is gene therapy-mediated overexpression of the copper-zinc (CuZnSOD) or manganese superoxide dismutase (MnSOD) using recombinant adeno-associated viral (rAAV2) vectors. The purpose of this study was to test the modulating effects of the SOD genes on human primary lung fibroblasts (HPLF) after irradiation. MATERIAL AND METHODS: HPLF were transduced with rAAV2 vectors containing cDNA for the CuZnSOD, MnSOD or a control gene. The cells were irradiated (1-6 Gy), and gene transfer efficiency, apoptosis, protein expression/activity, and radiosensitivity measured by the colony-forming assay determined. RESULTS: After transduction, 90.0% +/- 6.4% of the cells expressed the transgene. A significant fivefold overexpression of both SOD was confirmed by an SOD activity assay (control: 21.1 +/- 12.6, CuZnSOD: 95.1 +/- 17.1, MnSOD: 108.5 +/- 36.0 U SOD/mg protein) and immunohistochemistry. CuZnSOD and MnSOD overexpression resulted in a significant radioprotection of HPLF compared to controls (surviving fraction [SF] ratio SOD/control > 1): CuZnSOD: 1.18-fold (95% confidence interval [CI]: 1.06-1.32; p = 0.005), MnSOD: 1.23-fold (95% CI: 1.07-1.43; p = 0.01). CONCLUSION: Overexpression of CuZnSOD and MnSOD in HPLF mediated an increase in clonogenic survival after irradiation compared to controls. In previous works, a lack of radioprotection in SOD-overexpressing tumor cells was observed. Therefore, the present results suggest that rAAV2 vectors are promising tools for the delivery of radioprotective genes in normal tissue.


Asunto(s)
Supervivencia Celular/genética , Supervivencia Celular/efectos de la radiación , Técnicas de Transferencia de Gen , Tolerancia a Radiación/genética , Superóxido Dismutasa/genética , Apoptosis/genética , Apoptosis/efectos de la radiación , Línea Celular , Ensayo de Unidades Formadoras de Colonias , Dependovirus , Regulación Enzimológica de la Expresión Génica/genética , Humanos , Microscopía Fluorescente
6.
Exp Hematol ; 36(8): 957-64, 2008 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-18495326

RESUMEN

OBJECTIVE: Currently standard recombinant adeno-associated virus serotype 2(rAAV2)-based vectors lack the efficiency for gene transfer into primary human CD34(+) peripheral blood progenitor cells (PBPC). MATERIALS AND METHODS: An advancement in vector development now allows the generation of rAAV capsid mutants that offer higher target cell efficiency and specificity. To increase the gene transfer into hematopoietic progenitor cells, we applied this method for the first time on primary human CD34(+) PBPC cells. RESULTS: On a panel of leukemia cell lines (CML/AML), significantly higher gene transfer efficiency of the rAAV capsid mutants (up to 100% gene transfer) was observed compared to standard rAAV2 vectors. A higher transduction efficiency in the imatinib-resistant cell line LAMA84-R than in their sensitive counterpart LAMA84-S and a pronounced difference in susceptibility for the capsid mutants vs rAAV2 in LAMA84-S were particularly striking. On solid tumor cell lines, on the other hand, rAAV2 was more efficient than the capsid mutants, suggesting an increased specificity of our capsid mutants for hematopoietic progenitor cells. On primary human CD34(+) PBPC significantly higher (up to eightfold; 16% green fluorescent protein-positive) gene transfer could be obtained with the newly generated vectors compared to standard rAAV2 vectors. CONCLUSION: These novel vectors may enable efficient gene transfer using rAAV-based vectors into primary human blood progenitor cells for a future clinical application.


Asunto(s)
Dependovirus/genética , Técnicas de Transferencia de Gen , Vectores Genéticos/genética , Células Madre Hematopoyéticas/metabolismo , Biblioteca de Péptidos , Cápside/metabolismo , Línea Celular , Evolución Molecular Dirigida , Proteínas Fluorescentes Verdes/biosíntesis , Proteínas Fluorescentes Verdes/genética , Células Madre Hematopoyéticas/virología , Humanos , Leucemia/genética , Leucemia/metabolismo , Leucemia/virología , Mutagénesis , Proteínas Recombinantes/biosíntesis , Proteínas Recombinantes/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
7.
Radiat Res ; 169(3): 301-10, 2008 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-18302483

RESUMEN

Tumor radiotherapy with large-field irradiation results in an increase in apoptosis of the radiosensitive hematopoietic stem cells (CD34(+)). The aim of this study was to demonstrate the radioprotective potential of MDR1 overexpression in human CD34(+) cells using a lentiviral self-inactivating vector. Transduced human undifferentiated CD34(+) cells were irradiated with 0-8 Gy and held in liquid culture under myeloid-specific maturation conditions. After 12 days, MDR1 expression was determined by the rhodamine efflux assay. The proportion of MDR1-positive cells in cells from four human donors increased with increasing radiation dose (up to a 14-fold increase at 8 Gy). Determination of expression of myeloid-specific surface marker proteins revealed that myeloid differentiation was not affected by transduction and MDR1 overexpression. Irradiation after myeloid differentiation also led to an increase of MDR1-positive cells with escalating radiation doses (e.g. 12.5-16% from 0-8 Gy). Most importantly, fractionated irradiation (3 x 2 Gy; 24-h intervals) of MDR1-transduced CD34(+) cells resulted in an increase in MDR1-positive cells (e.g. 3-8% from 0-3 x 2 Gy). Our results clearly support a radioprotective effect of lentiviral MDR1 overexpression in human CD34(+) cells. Thus enhancing repopulation by surviving stem cells may increase the radiation tolerance of the hematopoietic system, which will contribute to widening the therapeutic index in radiotherapy.


Asunto(s)
Miembro 1 de la Subfamilia B de Casetes de Unión a ATP/metabolismo , Células Madre Hematopoyéticas/fisiología , Células Madre Hematopoyéticas/efectos de la radiación , Lentivirus/genética , Protección Radiológica/métodos , Tolerancia a Radiación/fisiología , Receptores de Complemento 3b/metabolismo , Miembro 1 de la Subfamilia B de Casetes de Unión a ATP/genética , Supervivencia Celular/efectos de la radiación , Células Cultivadas , Relación Dosis-Respuesta en la Radiación , Vectores Genéticos/genética , Células Madre Hematopoyéticas/citología , Humanos , Dosis de Radiación , Proteínas Recombinantes/metabolismo , Transfección/métodos
8.
Genet Vaccines Ther ; 6: 12, 2008 Sep 12.
Artículo en Inglés | MEDLINE | ID: mdl-18789140

RESUMEN

BACKGROUND: For many promising target cells (e.g.: haematopoeitic progenitors), the susceptibility to standard adeno-associated viral (AAV) vectors is low. Advancements in vector development now allows the generation of target cell-selected AAV capsid mutants. METHODS: To determine its suitability, the method was applied on a chronic myelogenous leukaemia (CML) cell line (K562) to obtain a CML-targeted vector and the resulting vectors tested on leukaemia, non-leukaemia, primary human CML and CD34+ peripheral blood progenitor cells (PBPC); standard AAV2 and a random capsid mutant vector served as controls. RESULTS: Transduction of CML (BV173, EM3, K562 and Lama84) and AML (HL60 and KG1a) cell lines with the capsid mutants resulted in an up to 36-fold increase in CML transduction efficiency (K562: 2-fold, 60% +/- 2% green fluorescent protein (GFP)+ cells; BV173: 9-fold, 37% +/- 2% GFP+ cells; Lama84: 36-fold, 29% +/- 2% GFP+ cells) compared to controls. For AML (KG1a, HL60) and one CML cell line (EM3), no significant transduction (<1% GFP+ cells) was observed for any vector. Although the capsid mutant clone was established on a cell line, proof-of-principle experiments using primary human cells were performed. For CML (3.2-fold, mutant: 1.75% +/- 0.45% GFP+ cells, p = 0.03) and PBPC (3.5-fold, mutant: 4.21% +/- 3.40% GFP+ cells) a moderate increase in gene transfer of the capsid mutant compared to control vectors was observed. CONCLUSION: Using an AAV random peptide library on a CML cell line, we were able to generate a capsid mutant, which transduced CML cell lines and primary human haematopoietic progenitor cells with higher efficiency than standard recombinant AAV vectors.

9.
Onkologie ; 31(3): 91-6, 2008 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-18322411

RESUMEN

BACKGROUND: The median survival time of patients with malignant pleural mesothelioma (MPM) remains poor. Therefore, novel therapeutic options are in high demand, and well characterized model systems for in vitro/vivo screening have to be established. MATERIAL AND METHODS: For this purpose, 3 MPM cell lines (H-Meso-1, MSTO211H, and NCI-H28) were characterized and tested for susceptibility to recombinant adeno-associated virus 2 (rAAV2)-based vectors which have the potential for a loco-regional application. RESULTS: Using multiplex fluorescence in situ hybridization, several recurrent chromosomal aberrations were observed for each of the MPM cell lines. Tumorigenicity of H-Meso-1 and MSTO-211H cells was shown in an intraperitoneal NOD/SCID mouse model, whereas NCI-H28 cells did not yield any tumors. Although all 3 cell lines were readily susceptible to rAAV2 vectors, differences in susceptibility were observed (H-Meso-1 > NCI-H28 > MSTO-211H). Furthermore, the efficacy of a potential suicide gene therapy using an rAAV2 suicide vector-transduced MPM cell line was determined in a proof-of-feasibility in vivo experiment. CONCLUSION: The characterized cell lines described here may serve as a model for in vitro and in vivo preclinical gene therapy for the treatment of MPM using rAAV2 suicide vectors.


Asunto(s)
Genes Transgénicos Suicidas/genética , Terapia Genética/métodos , Mesotelioma/genética , Mesotelioma/terapia , Transducción Genética/métodos , Animales , Línea Celular Tumoral , Ratones , Ratones SCID
10.
Exp Hematol ; 34(8): 1052-9, 2006 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-16863911

RESUMEN

OBJECTIVE: AMD3100 is a new CXCR4 antagonist that induces a rapid release of hematopoietic progenitors from the bone marrow to the peripheral blood. We conducted a clinical study where patients with multiple myeloma and non-Hodgkin's lymphoma were treated with AMD3100 (A) to increase the number of peripheral blood progenitor cells (PBPCs) when given a mobilization regimen of granulocyte colony-stimulating factor (G-CSF, G). Because experimental data suggest that A+G-mobilized PBPCs are functionally different from G-mobilized PBPCs, we were interested in an intraindividual comparison of the gene expression profile of CD34+ cells in the two different settings. METHODS: To this end peripheral blood CD34+ cells of three patients (three G, three A+G samples) were isolated by immunomagnetic followed by flow cytometric sorting to a purity of >99%. Total RNA was purified. Differentially expressed genes were analyzed by using the Affymetrix GeneChip Human Genome U133 Plus2.0 and the software package Micro Array Solutions 1.3 (SAS Institute Inc.). RESULTS: We found a pattern of unanimously higher (81 genes, log2 ratio > 0.5; p < 0.0001) or lower (29 genes, log2 ratio < -0.4; p < 0.0001) expressed genes in the A+G-mobilized vs G-mobilized CD34+ PBPCs. Significant changes of four selected genes noted in the microarray analysis were validated by quantitative real-time polymerase chain reaction. Genes were grouped according to gene function. Only increased expression was found in the categories antiapoptosis (e.g., MPO, HSPA1B), cell cycle (e.g., MS4A3, RRM2), replication/DNA repair (e.g., MPO, HSPA1B), cell motility (e.g., TNFSF4, HMMR), and oxygen transport. Decreased expression occurred in the proapoptosis gene group (e.g., MDA5, BCL10). CXCR4 receptor gene expression itself was significantly 1.5-fold higher in the A+G vs G group. CONCLUSION: We conclude that A+G-mobilized CD34+ PBPCs express significantly higher amounts of genes that potentially promote superior engraftment after myeloablative therapy than G-mobilized CD34+ PBPCs.


Asunto(s)
Perfilación de la Expresión Génica , Factor Estimulante de Colonias de Granulocitos/farmacología , Movilización de Célula Madre Hematopoyética , Células Madre Hematopoyéticas/metabolismo , Compuestos Heterocíclicos/farmacología , Trasplante de Células Madre de Sangre Periférica , Receptores CXCR4/antagonistas & inhibidores , Bencilaminas , Ciclamas , Humanos , Análisis de Secuencia por Matrices de Oligonucleótidos , Receptores CXCR4/fisiología
11.
Radiat Res ; 166(3): 463-73, 2006 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-16953664

RESUMEN

Overexpression of P-glycoprotein (P-gp), the product of the MDR1 (multidrug resistance 1) gene, might complement chemotherapy and radiotherapy in the treatment of tumors. However, for safety and mechanistic reasons, it is important to know whether MDR1 overexpression influences the expression of other genes. Therefore, we analyzed differential gene expression in cells of the human lymphoblast cell line TK6 retrovirally transduced with MDR1 using the GeneChip Human Genome U133 Plus2.0 (Affymetrix). Sixty-one annotated genes showed a significant change in expression (P < 10(-4)) in MDR1-overexpressing cells compared to untransduced cells and cells transduced with a control virus expressing the neomycin phosphotransferase gene. Several genes coding for proteins involved in detoxification and exocytosis showed approximately 1.4- 4-fold increases in transcript levels (e.g. ALDH1A, UNC13). Additionally, pro-apoptosis genes were down-regulated (e.g. twofold for CASP1, 2.5-fold for NALP7) with concomitant increased expression of the potential anti-apoptosis gene AKT3. In functional assays the influence of MDR1 overexpression on apoptosis signaling was further corroborated by showing reduced rates of apoptosis in response to irradiation in TK6 cells transduced with MDR1. In conclusion, the resistant phenotype of MDR1-mediated P-gp-overexpressing cells is associated with differential expression of genes coding for metabolic and apoptosis-related proteins. These results have important implications for understanding the mechanisms by which MDR1 gene therapy can protect normal tissues from radiation- or chemotherapy-induced damage during tumor treatment.


Asunto(s)
Miembro 1 de la Subfamilia B de Casetes de Unión a ATP/metabolismo , Proteínas Reguladoras de la Apoptosis/metabolismo , Apoptosis/efectos de la radiación , Supervivencia Celular/efectos de la radiación , Linfocitos/fisiología , Miembro 1 de la Subfamilia B de Casetes de Unión a ATP/genética , Apoptosis/genética , Línea Celular , Supervivencia Celular/genética , Relación Dosis-Respuesta en la Radiación , Regulación de la Expresión Génica/fisiología , Regulación de la Expresión Génica/efectos de la radiación , Vectores Genéticos/genética , Humanos , Linfocitos/citología , Linfocitos/efectos de la radiación , Dosis de Radiación , Protección Radiológica , Retroviridae/genética , Transfección/métodos
12.
Anticancer Res ; 26(3A): 2169-77, 2006.
Artículo en Inglés | MEDLINE | ID: mdl-16827161

RESUMEN

BACKGROUND: Resistance to imatinib monotherapy frequently emerges in advanced stages of chronic myelogenous leukemia (CML), supporting the rationale for combination drug therapy. In the present study, the activities of the farnesyltransferase inhibitors (FTIs) L744,832 and LB42918, as single agents and in combination with imatinib, were investigated in different imatinib-sensitive and -resistant BCR-ABL-positive CML cells. MATERIALS AND METHODS: Growth inhibition of the cell lines and primary patient cells was assessed by MTT assays and colony-forming cell assays, respectively. Drug interactions were analyzed according to the median-effect method of Chou and Talalay. The determination of apoptotic cell death was performed by annexin V/propidium iodide staining. RESULTS: Combinations of both FTIs with imatinib displayed synergism or sensitization (potentiation) in all the cell lines tested. In primary chronic phase CML cells, additive and synergistic effects were discernible for the combination of imatinib plus L744,832 and imatinib plus LB42918, respectively. Annexin V/propidium iodide staining showed enhancement of imatinib-induced apoptosis with either drug combination, both in imatinib-sensitive and -resistant cells. CONCLUSION: The results indicated the potential of L744,832 and LB42918 as combination agents for CML patients on imatinib treatment.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/farmacología , Inhibidores Enzimáticos/farmacología , Farnesiltransferasa/antagonistas & inhibidores , Proteínas de Fusión bcr-abl/biosíntesis , Leucemia Mielógena Crónica BCR-ABL Positiva/tratamiento farmacológico , Leucemia Mielógena Crónica BCR-ABL Positiva/metabolismo , Miembro 1 de la Subfamilia B de Casetes de Unión a ATP/metabolismo , Apoptosis/efectos de los fármacos , Benzamidas , Procesos de Crecimiento Celular/efectos de los fármacos , Línea Celular Tumoral , Inhibidores Enzimáticos/administración & dosificación , Humanos , Mesilato de Imatinib , Células K562 , Leucemia Mielógena Crónica BCR-ABL Positiva/enzimología , Leucemia Mielógena Crónica BCR-ABL Positiva/patología , Metionina/administración & dosificación , Metionina/análogos & derivados , Metionina/farmacología , Piperazinas/administración & dosificación , Pirimidinas/administración & dosificación
13.
Hum Gene Ther ; 13(2): 233-42, 2002 Jan 20.
Artículo en Inglés | MEDLINE | ID: mdl-11812280

RESUMEN

Myelosuppression is the main side effect of cancer chemotherapy. An improved rate of retroviral vector-mediated gene transfer to hematopoietic stem cells, shown in more recent clinical trials, has created the basis to test the concept of myeloprotective gene therapy. We transplanted clinical-scale human peripheral blood progenitor cell grafts (n = 2) transduced with retroviral vector SF91m3, which contains the human multidrug resistance 1 gene (MDR1), into nonobese diabetic/severe combined immunodeficient (NOD/SCID) mice. Engrafted mice of one cohort were protected from paclitaxel toxicity (p < 0.05) and we noted a similar trend in the second cohort. In paclitaxel-treated mice that had received gene-transduced cells we found a significant increase in gene marking (p < 0.05 - p < 0.01) or P-glycoprotein expression (p < 0.01) compared with their chemotherapy-naive counterparts. This is the first report showing that cytostatic drug resistance gene therapy can mediate chemoprotection of human clinically relevant stem cell populations with marrow engraftment potential.


Asunto(s)
Genes MDR/genética , Trasplante de Células Madre Hematopoyéticas , Paclitaxel/toxicidad , Animales , Animales Modificados Genéticamente , Resistencia a Medicamentos/genética , Técnicas de Transferencia de Gen , Terapia Genética , Humanos , Ratones , Ratones SCID , Células Tumorales Cultivadas
14.
Cancer Gene Ther ; 11(8): 577-84, 2004 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-15280909

RESUMEN

Soft-tissue sarcomas are mesenchymal tumors that respond poorly to systemic chemotherapy. Suicide gene therapy may be an alternative treatment strategy. Here we show a high susceptibility of human sarcoma cell lines for recombinant adeno-associated virus 2 (rAAV-2) suicide vectors: connective tissue sarcoma (HS-1), fibrosarcoma (HT-1080), Ewing sarcoma (RD-ES), Askin tumor (SK-N-MC), rhabdomyosarcoma (A-204) and soft-tissue sarcoma (WSKL-1). Several vectors containing the thymidine kinase (TK) gene under the control of either the cytomegalovirus promoter or the elongation-factor 1 alpha (EF1alpha) promoter were cloned and tested. Higher expression levels of the transgene were observed in the sarcoma lines when using the EF1alpha-suicide gene-containing vectors. A complete eradication of rAAV-2-EF1alpha-TK/eGFP (TK/enhanced green fluorescent protein fusion gene)-transduced tumor cells was shown following exposure to ganciclovir (2.5 microg/ml) in vitro, while at this dose level > 90% of mock-transduced tumor cells survived. Xenotransplantation tumor models (intraperitoneal, subcutaneous) for the human sarcoma cell line HS-1 were established in nonobese diabetic/severe-combined immunodeficient mice. Mice transplanted with rAAV-2-EF1alpha-TK/eGFP-transduced and ganciclovir-exposed tumor cells survived > 5 months while in the nontransduced group all mice had died approximately 1 month after inoculation. These data hold promise for further development of rAAV-2-based suicide gene therapy of sarcomas.


Asunto(s)
Dependovirus/genética , Genes Transgénicos Suicidas/genética , Terapia Genética/métodos , Vectores Genéticos , Sarcoma/terapia , Timidina Quinasa/genética , Animales , Línea Celular Tumoral , Citomegalovirus/genética , Ganciclovir/uso terapéutico , Vectores Genéticos/genética , Humanos , Ratones , Ratones Endogámicos NOD , Ratones SCID , Factor 1 de Elongación Peptídica/genética , Regiones Promotoras Genéticas/genética , Sarcoma/tratamiento farmacológico , Ensayos Antitumor por Modelo de Xenoinjerto
15.
Radiother Oncol ; 72(3): 341-50, 2004 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-15450734

RESUMEN

BACKGROUND AND PURPOSE: The success rate of any therapeutic approach depends on the therapeutic window, which can be increased by either raising the resistance of the normal tissue without protecting the tumor cells or by sensitizing the tumor cells but not the normal cells. Two promising candidate genes for normal tissue protection against radiation-induced damage may be the copper-zinc (CuZnSOD) and manganese superoxide-dismutase genes (MnSOD). The recombinant adeno-associated virus 2 (rAAV-2) offers attractive advantages over other vector systems: low immunogenicity, ability to infect dividing and non-dividing tissues and a low chance of insertional mutagenesis, due to extra-chromosomal localization. We report the production of novel rAAV-2-SOD vectors and the investigation of their modulating effects on HeLa-RC cells after irradiation. MATERIAL AND METHODS: rAAV-2 vectors were cloned containing the human CuZnSOD or MnSOD as transgene and vector stocks were produced. In the initial experiments human cervix carcinoma (HeLa-RC) cells were chosen for their susceptibility to rAAV-2. On day 0, cells were seeded and transduced with the rAAV-2-SOD vectors. On day 3, cells were harvested, irradiated (0.5-8 Gy) and reseeded in different assays (FACS, SOD, MTT and colony assays). RESULTS: Although >70% of all cells expressed SOD and significant amounts of functional SOD protein were detected, no radioprotective effect of SOD was observed after transduction of HeLa-RC cells. CONCLUSIONS: Novel rAAV-2-SOD vectors that could be produced at high titer, were able to efficiently infect cells and express the SOD genes. The absence of a radioprotective effect in HeLa-RC cancer cells indicates an additional safety feature and suggests that rAAV-mediated MnSOD overexpression might contribute to increasing the therapeutic index when applied for normal tissue protection.


Asunto(s)
Dependovirus , Técnicas de Transferencia de Gen , Tolerancia a Radiación/genética , Superóxido Dismutasa/genética , Femenino , Células HeLa , Humanos , Proteínas Recombinantes , Neoplasias del Cuello Uterino/patología , Neoplasias del Cuello Uterino/radioterapia
16.
Clin Cancer Res ; 20(3): 604-16, 2014 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-24255072

RESUMEN

PURPOSE: In colorectal cancer, increased expression of the CXC chemokine receptor 4 (CXCR4) has been shown to provoke metastatic disease due to the interaction with its ligand stromal cell-derived factor-1 (SDF-1). Recently, a second SDF-1 receptor, CXCR7, was found to enhance tumor growth in solid tumors. Albeit signaling cascades via SDF-1/CXCR4 have been intensively studied, the significance of the SDF-1/CXCR7-induced intracellular communication triggering malignancy is still only marginally understood. EXPERIMENTAL DESIGN: In tumor tissue of 52 patients with colorectal cancer, we observed that expression of CXCR7 and CXCR4 increased with tumor stage and tumor size. Asking whether activation of CXCR4 or CXCR7 might result in a similar expression pattern, we performed microarray expression analyses using lentivirally CXCR4- and/or CXCR7-overexpressing SW480 colon cancer cell lines with and without stimulation by SDF-1α. RESULTS: Gene regulation via SDF-1α/CXCR4 and SDF-1α/CXCR7 was completely different and partly antidromic. Differentially regulated genes were assigned by gene ontology to migration, proliferation, and lipid metabolic processes. Expressions of AKR1C3, AXL, C5, IGFBP7, IL24, RRAS, and TNNC1 were confirmed by quantitative real-time PCR. Using the in silico gene set enrichment analysis, we showed that expressions of miR-217 and miR-218 were increased in CXCR4 and reduced in CXCR7 cells after stimulation with SDF-1α. Functionally, exposure to SDF-1α increased invasiveness of CXCR4 and CXCR7 cells, AXL knockdown hampered invasion. Compared with controls, CXCR4 cells showed increased sensitivity against 5-FU, whereas CXCR7 cells were more chemoresistant. CONCLUSIONS: These opposing results for CXCR4- or CXCR7-overexpressing colon carcinoma cells demand an unexpected attention in the clinical application of chemokine receptor antagonists such as plerixafor.


Asunto(s)
Quimiocina CXCL12/metabolismo , Neoplasias del Colon/metabolismo , Resistencia a Antineoplásicos/fisiología , Regulación Neoplásica de la Expresión Génica/fisiología , Receptores CXCR4/biosíntesis , Receptores CXCR/biosíntesis , Adulto , Anciano , Anciano de 80 o más Años , Biomarcadores de Tumor/análisis , Línea Celular Tumoral , Neoplasias del Colon/genética , Neoplasias del Colon/patología , Femenino , Citometría de Flujo , Humanos , Immunoblotting , Inmunohistoquímica , Masculino , Persona de Mediana Edad , Análisis de Secuencia por Matrices de Oligonucleótidos , Reacción en Cadena en Tiempo Real de la Polimerasa , Transfección
17.
Transl Oncol ; 6(2): 124-32, 2013 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-23544165

RESUMEN

BACKGROUND: Signaling through stromal cell-derived factor-1α (SDF-1α), strongly secreted by bone marrow stromal cells and the CXC chemokine receptor 4 (CXCR4) exposed on tumor cells has pivotal roles in proliferation, metastasis, and tumor cell "dormancy." Dormancy is associated with cytostatic drug resistance and is probably a property of tumor stem cells and minimal residual disease. Thus, hampering the SDF-1α/CXCR4 cross talk by a CXCR4 antagonist like Plerixafor (AMD3100) should overcome tumor cell dormancy bymobilization of tumor cells from "sanctuary" niches. Our aim was to elucidate the direct effects exerted by SDF-1α and Plerixafor on proliferation, chemosensitivity, and apoptosis of CXCR4-expressing tumor cells. METHODS: The ability of SDF-1α and Plerixafor to regulate intracellular signaling, proliferation, and invasion was investigated using two colon cancer cell lines (HT-29 and SW480) with either high endogenous or lentiviral expression of CXCR4 compared to their respective low CXCR4-expressing counterparts as a model system. Efficacy of Plerixafor on sensitivity of these cell lines against 5-fluorouracil, irinotecan, or oxaliplatin was determined in a cell viability assay as well as stroma-dependent cytotoxicity and apoptosis assays. RESULTS: SDF-1α increased proliferation, invasion, and ERK signaling of endogenously and lentivirally CXCR4-expressing cells. Exposure to Plerixafor reduced proliferation, invasion, and extracellular signal-regulated kinase 1/2 (ERK1/2) signaling. Combination of chemotherapy with Plerixafor showed an additive effect on chemosensitivity and apoptosis in CXCR4-overexpressing cells. An SDF-1-secreting feeder layer provideda"protective niche" for CXCR4-overexpressing cells resulting in decreased chemosensitivity. CONCLUSION: CXCR4-antagonistic therapy mobilizes and additionally sensitizes tumor cells toward cytoreductive chemotherapy.

18.
Leuk Lymphoma ; 52(3): 483-90, 2011 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-21323526

RESUMEN

Gene transfer into chronic myelogenous leukemia (CML) cells may become of relevance for overcoming therapy resistance. Single-stranded pseudotyped adeno-associated viruses of serotypes 2/1 to 2/6 (ssAAV2/1-ssAAV2/6) were screened on human CML cell lines and primary cells to determine gene transfer efficiency. Additionally, double-stranded self-complementary vectors (dsAAVs) were used to determine possible second-strand synthesis limitations. On human CML cell lines, ssAAV2/2 and ssAAV2/6 were most efficient. On primary cells, ssAAV2/6 proved significantly more efficient (4.1 ± 2.5% GFP(+) cells, p = 0.011) than the other vectors (<1%). The transduction efficiency could be significantly increased (45.5 ± 13.4%) by using dsAAV2/6 vectors (p < 0.001 vs. ssAAV2/6). In these settings, our data suggest conversion of single- to double-stranded DNA and cell binding/entry as rate-limiting steps. Furthermore, gene transfer was observed in both late and earlier CML (progenitor) populations. For the first time, efficient AAV gene transfer into human CML cells could be shown, with the potential for future clinical application.


Asunto(s)
Dependovirus/genética , Técnicas de Transferencia de Gen , Vectores Genéticos/genética , Leucemia Mielógena Crónica BCR-ABL Positiva/genética , Adolescente , Adulto , Anciano , Técnicas de Cultivo de Célula , Células Cultivadas , Eficiencia , Femenino , Terapia Genética/métodos , Humanos , Células K562 , Leucemia Mielógena Crónica BCR-ABL Positiva/patología , Leucemia Mielógena Crónica BCR-ABL Positiva/terapia , Masculino , Seudogenes/genética , Transgenes
19.
Radiat Res ; 176(6): 725-31, 2011 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-21899432

RESUMEN

Gene therapy-mediated overexpression of superoxide dismutases (SOD) appears to be a promising strategy for modulating radiosensitivity based on detoxification of superoxide radicals and suppression of apoptosis. Using recombinant lentiviral-based vectors, the effects of SOD overexpression on both were tested in human lymphoblastoid cells (TK6) that are sensitive to radiation-induced apoptosis. TK6 cells were transduced with vectors containing CuZnSOD, MnSOD or inverted MnSOD (MSODi) cDNA. Gene transfer efficiency, SOD activity, superoxide-radical resistance, apoptosis and clonogenic survival were determined. A six- to eightfold increase in SOD activity was observed after transduction, rendering MnSOD-overexpressing TK6 cells significantly more resistant to paraquat-induced superoxide radical production than controls. Although significant differences in sensitivity to apoptosis were observed for MnSOD, no differences in clonogenic survival after irradiation were detected between any groups. Our data show that efficient cellular SOD overexpression, an increased superoxide radical detoxifying ability and, for MnSOD, decreased apoptosis did not result in increased clonogenic survival after irradiation. This strengthens the hypothesis of differences in the radiation-modulating effects of SOD on normal and malignant cells (protective and nonprotective, respectively), thereby showing its potential to increase the therapeutic index in future clinical SOD-based radioprotection approaches.


Asunto(s)
Apoptosis/efectos de la radiación , Regulación Enzimológica de la Expresión Génica/efectos de la radiación , Linfocitos/citología , Linfocitos/metabolismo , Superóxido Dismutasa/genética , Superóxido Dismutasa/metabolismo , Línea Celular Tumoral , Supervivencia Celular/efectos de la radiación , Clonación Molecular , Vectores Genéticos/genética , Células HEK293 , Humanos , Lentivirus/genética , Linfocitos/efectos de la radiación , Superóxidos/metabolismo
20.
Hum Gene Ther ; 22(6): 697-710, 2011 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-21319998

RESUMEN

Gene transfer of mutant O(6)-methylguanine-DNA-methyltransferase (MGMT(P140K)) into hematopoietic stem cells (HSCs) protects hematopoiesis from alkylating agents and allows efficient in vivo selection of transduced HSCs. However, insertional mutagenesis, high regenerative stress associated with selection, and the genotoxic potential of alkylating drugs represent considerable risk factors for clinical applications of this approach. Therefore, we investigated the long-term effect of MGMT(P140K) gene transfer followed by repetitive, dose-intensive treatment with alkylating agents in a murine serial bone marrow transplant model and assessed clonality of hematopoiesis up to tertiary recipients. The substantial selection pressure resulted in almost completely transduced hematopoiesis in all cohorts. Ligation-mediated PCR and next-generation sequencing identified several repopulating clones carrying vector insertions in distinct genomic regions that were ∼ 9 kb of size (common integration sites). Beside polyclonal reconstitution in the majority of the mice, we also detected monoclonal or oligoclonal repopulation patterns with HSC clones showing vector insertions in the Usp10 or Tubb3 gene. Interestingly, neither Usp10, Tubb3, nor any of the genes located in common integration sites have been linked to clonal expansion in previous preclinical or clinical gene therapy trials. However, a considerable number of these genes are involved in DNA damage response and cell fate decision pathways following cytostatic drug application. Thus, in summary, our study advocates ligation-mediated PCR and next generation sequencing as an effective and reliable method to identify gene products associated with clonal survival in specific experimental settings such as chemoselection using alkylating agents.


Asunto(s)
Metilasas de Modificación del ADN/genética , Enzimas Reparadoras del ADN/genética , Hematopoyesis/genética , Células Madre Hematopoyéticas , Reacción en Cadena de la Polimerasa/métodos , Análisis de Secuencia de ADN/métodos , Proteínas Supresoras de Tumor/genética , Alquilantes/farmacología , Animales , Trasplante de Médula Ósea , Células Clonales , Técnicas de Transferencia de Gen , Vectores Genéticos , Hematopoyesis/efectos de los fármacos , Ratones , Modelos Animales , Mutación
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA