Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
1.
Stem Cells ; 28(5): 928-38, 2010 May.
Artículo en Inglés | MEDLINE | ID: mdl-20235097

RESUMEN

The mammary gland represents a unique model system to study gene functions in adult stem cells. Mammary stem cells (MaSCs) can regenerate a functional epithelium on transplantation into cleared fat pads. We studied the consequences of distinct genetic modifications of MaSCs on their repopulation and differentiation ability. The reconstitution of ductal trees was used as a stem cell selection procedure and the nearly quantitative lentiviral infection efficiency of the primary mammary epithelial cells (MECs) rendered the enrichment of MaSCs before their transplantation unnecessary. The repopulation frequency of transduced MaSCs was nearly 100% in immunodeficient recipients and the resulting transgenic ducts homogeneously expressed the virally encoded fluorescent marker proteins. Transplantation of a mixture of MECs, expressing different fluorescent proteins, resulted in a distinct pattern of ductal outgrowths originating from a small number of individually transduced MaSCs. We used genetically modified MECs to define multiple functions of Stat5 during mammary gland development and differentiation. Stat5-downregulation in MaSCs did not affect primary ductal outgrowth, but impaired side branching and the emergence of mature alveolar cells from luminal progenitors during pregnancy. Conversely, the expression of a constitutively active variant of Stat5 (cS5-F) caused epithelial hyperproliferation, thickening of the ducts and precocious, functional alveoli formation in virgin mice. Expression of cS5-F also prevented involution and caused the formation of estrogen and progesterone receptor positive (ER(+)PR(+)) adenocarcinomas. The tumors expressed activated Stat5 and Stat3 and contained a small fraction of CD44(+) cells, possibly indicative of cancer stem cells.


Asunto(s)
Adenocarcinoma/etiología , Adenocarcinoma/metabolismo , Linaje de la Célula/genética , Células Epiteliales/metabolismo , Neoplasias Mamarias Animales/etiología , Neoplasias Mamarias Animales/metabolismo , Factor de Transcripción STAT5/fisiología , Células Madre/metabolismo , Adenocarcinoma/patología , Animales , Diferenciación Celular/genética , Células Cultivadas , Células Epiteliales/citología , Femenino , Regulación Neoplásica de la Expresión Génica/genética , Glándulas Mamarias Animales/citología , Glándulas Mamarias Animales/crecimiento & desarrollo , Glándulas Mamarias Animales/metabolismo , Neoplasias Mamarias Animales/patología , Ratones , Ratones Endogámicos BALB C , Células Madre Neoplásicas/metabolismo , Células Madre Neoplásicas/patología , Factor de Transcripción STAT5/genética , Células Madre/citología , Células Tumorales Cultivadas
2.
Blood ; 111(9): 4532-41, 2008 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-18326819

RESUMEN

Mef2c is a MADS (MCM1-agamous-deficient serum response factor) transcription factor best known for its role in muscle and cardiovascular development. A causal role of up-regulated MEF2C expression in myelomonocytic acute myeloid leukemia (AML) has recently been demonstrated. Due to the pronounced monocytic component observed in Mef2c-induced AML, this study was designed to assess the importance of Mef2c in normal myeloid differentiation. Analysis of bone marrow (BM) cells manipulated to constitutively express Mef2c demonstrated increased monopoiesis at the expense of granulopoiesis, whereas BM isolated from Mef2c(Delta/-) mice showed reduced levels of monocytic differentiation in response to cytokines. Mechanistic studies showed that loss of Mef2c expression correlated with reduced levels of transcripts encoding c-Jun, but not PU.1, C/EBPalpha, or JunB transcription factors. Inhibiting Jun expression by short-interfering RNA impaired Mef2c-mediated inhibition of granulocyte development. Moreover, retroviral expression of c-Jun in BM cells promoted monocytic differentiation. The ability of Mef2c to modulate cell-fate decisions between monocyte and granulocyte differentiation, coupled with its functional sensitivity to extracellular stimuli, demonstrate an important role in immunity--and, consistent with findings of other myeloid transcription factors, a target of oncogenic lesions in AML.


Asunto(s)
Células Mieloides/citología , Factores Reguladores Miogénicos/fisiología , Proteínas Proto-Oncogénicas c-jun/fisiología , Animales , Células de la Médula Ósea , Diferenciación Celular , Granulocitos/citología , Hematopoyesis , Factores de Transcripción MEF2 , Ratones , Ratones Mutantes , Monocitos/citología , Factores de Transcripción/fisiología
3.
Mol Ther ; 16(4): 698-706, 2008 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-18362927

RESUMEN

Functional gene analysis requires the possibility of overexpression, as well as downregulation of one, or ideally several, potentially interacting genes. Lentiviral vectors are well suited for this purpose as they ensure stable expression of complementary DNAs (cDNAs), as well as short-hairpin RNAs (shRNAs), and can efficiently transduce a wide spectrum of cell targets when packaged within the coat proteins of other viruses. Here we introduce a multicolor panel of novel lentiviral "gene ontology" (LeGO) vectors designed according to the "building blocks" principle. Using a wide spectrum of different fluorescent markers, including drug-selectable enhanced green fluorescent protein (eGFP)- and dTomato-blasticidin-S resistance fusion proteins, LeGO vectors allow simultaneous analysis of multiple genes and shRNAs of interest within single, easily identifiable cells. Furthermore, each functional module is flanked by unique cloning sites, ensuring flexibility and individual optimization. The efficacy of these vectors for analyzing multiple genes in a single cell was demonstrated in several different cell types, including hematopoietic, endothelial, and neural stem and progenitor cells, as well as hepatocytes. LeGO vectors thus represent a valuable tool for investigating gene networks using conditional ectopic expression and knock-down approaches simultaneously.


Asunto(s)
Aminohidrolasas/genética , Vectores Genéticos , Proteínas Fluorescentes Verdes/genética , Lentivirus/genética , Proteínas Recombinantes de Fusión/biosíntesis , Transducción Genética , Aminohidrolasas/biosíntesis , Animales , Células Cultivadas , Cricetinae , Cricetulus , Proteínas Fluorescentes Verdes/biosíntesis , Humanos , Ratones , Regiones Promotoras Genéticas , Proteínas Recombinantes de Fusión/genética
4.
Nat Protoc ; 7(5): 839-49, 2012 Apr 05.
Artículo en Inglés | MEDLINE | ID: mdl-22481527

RESUMEN

Cells transduced with lentiviral vectors are individually marked by a highly characteristic pattern of insertion sites inherited by all their progeny. We have recently extended this principle of clonal cell marking by introducing the method of RGB marking, which makes use of the simultaneous transduction of target cells with three lentiviral gene ontology (LeGO) vectors encoding red, green or blue fluorescent proteins. In accordance with the additive color model, individual RGB-marked cells display a large variety of unique and highly specific colors. Color codes remain stable after cell division and can thus be used for clonal tracking in vivo and in vitro. Our protocol for efficient RGB marking is based on established methods of lentiviral vector production (3-4 d) and titration (3 d). The final RGB-marking step requires concurrent transduction with the three RGB vectors at equalized multiplicities of infection (1-12 h). The initial efficiency of RGB marking can be assessed after 2-4 d by flow cytometry and/or fluorescence microscopy.


Asunto(s)
Rastreo Celular/métodos , Proteínas Luminiscentes/análisis , Citometría de Flujo , Vectores Genéticos , Proteínas Fluorescentes Verdes/análisis , Proteínas Fluorescentes Verdes/genética , Células HEK293 , Humanos , Lentivirus/genética , Proteínas Luminiscentes/genética , Microscopía Fluorescente , Transfección , Proteína Fluorescente Roja
5.
PLoS One ; 7(8): e43468, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22927971

RESUMEN

Glioblastomas are highly aggressive brain tumors of adults with poor clinical outcome. Despite a broad range of new and more specific treatment strategies, therapy of glioblastomas remains challenging and tumors relapse in all cases. Recent work demonstrated that the posttranslational hypusine modification of the eukaryotic initiation factor 5A (eIF-5A) is a crucial regulator of cell proliferation, differentiation and an important factor in tumor formation, progression and maintenance. Here we report that eIF-5A as well as the hypusine-forming enzymes deoxyhypusine synthase (DHS) and deoxyhypusine hydroxylase (DOHH) are highly overexpressed in glioblastoma patient samples. Importantly, targeting eIF-5A and its hypusine modification with GC7, a specific DHS-inhibitor, showed a strong antiproliferative effect in glioblastoma cell lines in vitro, while normal human astrocytes were not affected. Furthermore, we identified p53 dependent premature senescence, a permanent cell cycle arrest, as the primary outcome in U87-MG cells after treatment with GC7. Strikingly, combined treatment with clinically relevant alkylating agents and GC7 had an additive antiproliferative effect in glioblastoma cell lines. In addition, stable knockdown of eIF-5A and DHS by short hairpin RNA (shRNA) could mimic the antiproliferative effects of GC7. These findings suggest that pharmacological inhibition of eIF-5A may represent a novel concept to treat glioblastomas and may help to substantially improve the clinical course of this tumor entity.


Asunto(s)
Regulación Neoplásica de la Expresión Génica , Glioblastoma/genética , Lisina/análogos & derivados , Oxigenasas de Función Mixta/genética , Terapia Molecular Dirigida , Oxidorreductasas actuantes sobre Donantes de Grupo CH-NH/genética , Factores de Iniciación de Péptidos/genética , Proteínas de Unión al ARN/genética , Adulto , Anciano , Anciano de 80 o más Años , Antineoplásicos Alquilantes/farmacología , Apoptosis/efectos de los fármacos , Carmustina/farmacología , Ciclo Celular/efectos de los fármacos , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Senescencia Celular/efectos de los fármacos , Dacarbazina/análogos & derivados , Dacarbazina/farmacología , Femenino , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Técnicas de Silenciamiento del Gen , Glioblastoma/tratamiento farmacológico , Glioblastoma/enzimología , Glioblastoma/patología , Guanina/análogos & derivados , Guanina/farmacología , Humanos , Lisina/biosíntesis , Masculino , Oxigenasas de Función Mixta/metabolismo , Clasificación del Tumor , Oxidorreductasas actuantes sobre Donantes de Grupo CH-NH/antagonistas & inhibidores , Oxidorreductasas actuantes sobre Donantes de Grupo CH-NH/deficiencia , Oxidorreductasas actuantes sobre Donantes de Grupo CH-NH/metabolismo , Factores de Iniciación de Péptidos/deficiencia , Temozolomida , Factor 5A Eucariótico de Iniciación de Traducción
6.
Cancer Lett ; 317(1): 106-13, 2012 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-22108531

RESUMEN

Skeletal metastases are a frequent complication of prostate, breast and lung cancer, and the interactions of tumor cells with bone-forming osteoblasts and bone-resorbing osteoclasts have been suggested to play critical roles in disease progression. We have previously shown that treatment of primary murine osteoblasts with conditioned medium of the human osteolytic prostate cancer cell line PC-3 results in a rapid induction of chemokine expression, thereby providing further evidence for a molecular crosstalk between bone and tumor cells. The aim of our current study was to identify PC-3-derived molecules mediating this effect. Using Affymetrix Gene Chip hybridization followed by qRT-PCR we were able to confirm that the expression of chemokine-encoding genes is markedly induced in human primary osteoblasts following incubation with PC-3-conditioned medium. Since this induction was significantly affected upon alteration of p65-levels in PC-3 cells, we performed a second genome-wide expression analysis to identify p65-regulated cytokines, which were then tested for their ability to induce chemokine expression. Here we observed that interleukin-1ß (IL-1B) did not only increase the expression of chemokines in osteoblasts, but also the phosphorylation of p65 and thereby its own expression. Since immunohistochemistry on bone biopsy sections from prostate cancer metastases demonstrated IL-1B expression in both, tumor cells and osteoblasts, our data suggest that IL-1B is one of the relevant cytokines involved in the skeletal complications of cancer metastases.


Asunto(s)
Neoplasias Óseas/metabolismo , Quimiocinas/metabolismo , Interleucina-1beta/metabolismo , Osteoblastos/metabolismo , Comunicación Paracrina , Neoplasias de la Próstata/metabolismo , Factor de Transcripción ReIA/metabolismo , Neoplasias Óseas/genética , Neoplasias Óseas/inmunología , Neoplasias Óseas/secundario , Línea Celular Tumoral , Quimiocinas/genética , Medios de Cultivo Condicionados/metabolismo , Perfilación de la Expresión Génica/métodos , Humanos , Inmunohistoquímica , Masculino , Análisis de Secuencia por Matrices de Oligonucleótidos , Osteoblastos/inmunología , Fosforilación , Neoplasias de la Próstata/genética , Neoplasias de la Próstata/inmunología , Neoplasias de la Próstata/patología , Interferencia de ARN , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Factor de Transcripción ReIA/genética , Transfección , Regulación hacia Arriba
7.
J Mol Med (Berl) ; 89(11): 1113-24, 2011 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-21698427

RESUMEN

Suicide gene therapy is a promising concept in oncology. We have recently introduced a novel suicide gene, TK.007, which was shown to excel established herpes simplex virus thymidine kinase (HSVtk) variants when used for donor-lymphocyte modification in adoptive immunotherapy models. Here, the potential of TK.007 in killing cancer cells was studied. Initially, we transduced tumour cell lines derived from different neoplasias (glioblastoma, melanoma, lung cancer, colon cancer) with lentiviral LeGO vectors encoding TK.007 or the splice-corrected (sc)HSVtk together with an eGFP/Neo-marker. Based on direct in vitro comparison, we found that TK.007 facilitates more efficient tumour cell killing at significantly lower ganciclovir doses in all tumour cell lines tested. Also, using different readout systems, we found a significantly stronger bystander effect of TK.007 as compared to scHSVtk. Importantly, in vitro data were confirmed in vivo using a subcutaneous G62 glioblastoma model in NOD/SCID mice. In mice transplanted with scHSVtk-positive tumours, treatment with low (10 mg/kg) or standard (50 mg/kg) ganciclovir doses resulted only in short-term growth inhibition or transient tumour remission, respectively. In striking contrast, in the TK.007 group, all animals achieved continuous complete remission after both standard and low-dose ganciclovir. Finally, a substantial bystander effect for TK.007 was also confirmed with the G62 model in vivo, where significantly prolonged survival for mice bearing tumours containing only 10% or 50% TK.007-expressing cells was observed. In summary, our data indicate strongly improved anti-tumour activity of TK.007 as compared to conventional HSVtk. We therefore suppose that TK.007 is an excellent candidate for cancer suicide gene therapy.


Asunto(s)
Efecto Espectador , Genes Transgénicos Suicidas , Terapia Genética/métodos , Neoplasias/terapia , Simplexvirus/enzimología , Timidina Quinasa/metabolismo , Proteínas Virales/metabolismo , Animales , Antivirales/farmacología , Línea Celular Tumoral , Relación Dosis-Respuesta a Droga , Ganciclovir/farmacología , Humanos , Ratones , Ratones Endogámicos NOD , Ratones SCID , Trasplante de Neoplasias , Neoplasias/etnología , Neoplasias/genética , Neoplasias/patología , Simplexvirus/genética , Timidina Quinasa/genética , Trasplante Heterólogo , Proteínas Virales/genética , Ensayos Antitumor por Modelo de Xenoinjerto
8.
Nat Med ; 17(4): 504-9, 2011 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-21441917

RESUMEN

We simultaneously transduced cells with three lentiviral gene ontology (LeGO) vectors encoding red, green or blue fluorescent proteins. Individual cells were thereby marked by different combinations of inserted vectors, resulting in the generation of numerous mixed colors, a principle we named red-green-blue (RGB) marking. We show that lentiviral vector-mediated RGB marking remained stable after cell division, thus facilitating the analysis of clonal cell fates in vitro and in vivo. Particularly, we provide evidence that RGB marking allows assessment of clonality after regeneration of injured livers by transplanted primary hepatocytes. We also used RGB vectors to mark hematopoietic stem/progenitor cells that generated colored spleen colonies. Finally, based on limiting-dilution and serial transplantation assays with tumor cells, we found that clonal tumor cells retained their specific color-code over extensive periods of time. We conclude that RGB marking represents a useful tool for cell clonality studies in tissue regeneration and pathology.


Asunto(s)
Rastreo Celular/métodos , Células Clonales/citología , Células Clonales/metabolismo , Proteínas Luminiscentes/genética , Proteínas Luminiscentes/metabolismo , Animales , Color , Vectores Genéticos , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Hepatocitos/citología , Hepatocitos/metabolismo , Regeneración Hepática , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos NOD , Ratones SCID , Ratones Transgénicos , Trasplante de Neoplasias , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Transducción Genética , Células Tumorales Cultivadas/metabolismo , Células Tumorales Cultivadas/patología , Proteína Fluorescente Roja
10.
Blood Coagul Fibrinolysis ; 21(5): 464-73, 2010 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-20595824

RESUMEN

Postnatal vasculogenesis has been implicated as an important mechanism for neovascularization via bone marrow-derived endothelial progenitor cells (EPCs) circulating in peripheral blood. In preparation of the utilization of EPCs in clinical protocols, we have generated blood-derived EPCs according to two established protocols by culturing either nonadherent mononuclear cells on fibronectin or adherent mononuclear cells on collagen. To explore the feasibility of these EPCs for their potential clinical use as target cells for genetic transduction to enhance their thromboresistance, newly designed retroviral and lentiviral gene ontology expression vectors were tested. Whereas cell clusters derived from the nonadherent cells demonstrated an only limited proliferative potential, cell colonies derived from collagen-adherent cells expanded more than a million-fold. Characterization of the exponentially growing cells by surface antigen and gene expression profiling revealed a consistently strong expression of characteristic endothelial markers, whereas expression of leukocyte markers was gradually lost. Using a single-step transduction protocol, we were able to achieve gene transfer efficiency of up to 99%. Our results suggest that the generated blood-derived EPC population might be attractive target cells for tissue engineering and gene therapy protocols due to their well defined phenotype, extensive proliferative potential, and efficient genetic transducibility, three important qualities that need to be defined prior to any clinical use.


Asunto(s)
Células Endoteliales/citología , Lentivirus/genética , Células Madre/metabolismo , Transducción Genética , Adulto , Células Cultivadas , Células Endoteliales/metabolismo , Células Endoteliales/virología , Femenino , Genotipo , Humanos , Masculino , Persona de Mediana Edad , Fenotipo , Células Madre/citología , Células Madre/virología
11.
Neoplasia ; 12(12): 1023-30, 2010 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-21170266

RESUMEN

The human immunodeficiency virus (HIV) protease inhibitor saquinavir shows anticancer activity. Although its nitric oxide-modified derivative saquinavir-NO (saq-NO) was less toxic to normal cells, it exerted stronger inhibition of B16 melanoma growth in syngeneic C57BL/6 mice than saquinavir did. Saq-NO has been shown to block proliferation, upregulate p53 expression, and promote differentiation of C6 glioma and B16 cells. The anticancer activity of substances is frequently hampered by cancer cell chemoresistance mechanisms. Therefore, we here investigated the roles of p53 and the ATP-binding cassette (ABC) transporters P-glycoprotein (P-gp), multidrug resistance-associated protein 1 (MRP1), and breast cancer resistance protein 1 (BCRP1) in cancer cell sensitivity to saq-NO to get more information about the potential of saq-NO as anticancer drug. Saq-NO exerted anticancer effects in lower concentrations than saquinavir in a panel of human cancer cell lines. Neither p53 mutation or depletion nor expression of P-gp, MRP1, or BCRP1 affected anticancer activity of saq-NO or saquinavir. Moreover, saq-NO sensitized P-gp-, MRP1-, or BCRP1-expressing cancer cells to chemotherapy. Saq-NO induced enhanced sensitization of P-gp- or MRP1-expressing cancer cells to chemotherapy compared with saquinavir, whereas both substances similarly sensitized BCRP1-expressing cells. Washout kinetics and ABC transporter ATPase activities demonstrated that saq-NO is a substrate of P-gp as well as of MRP1. These data support the further investigation of saq-NO as an anticancer drug, especially in multidrug-resistant tumors.


Asunto(s)
Transportadoras de Casetes de Unión a ATP/metabolismo , Antineoplásicos/farmacología , Proliferación Celular/efectos de los fármacos , Resistencia a Antineoplásicos , Saquinavir/análogos & derivados , Saquinavir/farmacología , Miembro 1 de la Subfamilia B de Casetes de Unión a ATP/metabolismo , Animales , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Resistencia a Múltiples Medicamentos , Citometría de Flujo , Genes p53 , Humanos , Ratones , Proteínas Asociadas a Resistencia a Múltiples Medicamentos/metabolismo
12.
Hum Gene Ther ; 21(8): 929-41, 2010 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-20201626

RESUMEN

Conditional elimination of infused gene-modified alloreactive T cells, using suicide gene activation, has been shown to be an efficient strategy to abrogate severe graft-versus-host disease (GvHD) in the context of adoptive immunotherapy. To overcome shortcomings of the most widely used suicide gene, wild-type (splice-corrected) herpes simplex virus thymidine kinase (scHSVtk), we generated two new variants: the codon-optimized coHSVtk and, by introducing an additional mutation (A168H), the novel TK.007. We transduced human hematopoietic cell lines and primary T cells with retroviral "sort-suicide vectors" encoding combinations of selection markers (tCD34 and OuaSelect) with one of three HSVtk variants. In vitro we observed higher expression levels and sustained long-term expression of TK.007, indicating lower nonspecific toxicity. Also, we noted significantly improved kinetics of ganciclovir (GCV)-mediated killing for TK.007-transduced cells. In an experimental (murine) allogeneic transplantation model, TK.007-transduced T cells mediated severe GvHD, which was readily abrogated by application of GCV (10 mg/kg). Last, we established a modified allotransplantation model that allowed quantitative comparison of the in vivo activities of TK.007 versus scHSVtk. We found that TK.007 mediates both significantly faster and higher absolute killing at low GCV concentrations (10 and 25 mg/kg). In summary, we demonstrate that the novel TK.007 suicide gene combines better killing performance with reduced nonspecific toxicity (as compared with the frequently used splice-corrected wild-type scHSVtk gene), thus representing a promising alternative for suicide gene therapy.


Asunto(s)
Genes Transgénicos Suicidas , Terapia Genética/métodos , Vectores Genéticos , Enfermedad Injerto contra Huésped/terapia , Timidina Quinasa/metabolismo , Animales , Línea Celular , Codón/genética , Codón/metabolismo , Ganciclovir/metabolismo , Enfermedad Injerto contra Huésped/genética , Humanos , Inmunoterapia Adoptiva , Ratones , Ratones Endogámicos C57BL , Retroviridae/genética , Linfocitos T/metabolismo , Linfocitos T/virología , Timidina Quinasa/genética , Transducción Genética
13.
Cancer Res ; 69(2): 416-21, 2009 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-19147553

RESUMEN

Murine double minute 2 (MDM2) negatively regulates the activity of the tumor suppressor protein p53. Nutlin-3 is a MDM2 inhibitor under preclinical investigation as nongenotoxic activator of the p53 pathway for cancer therapy. Here, nutlin-3 was evaluated for its activity alone or in combination with established chemotherapeutic drugs for antitumor action in chemosensitive and chemoresistant neuroblastoma and rhabdomyosarcoma cell lines. Effects of nutlin-3 single treatment were much more pronounced in p53 wild-type cell lines (IC(50)s <3 micromol/L) than in p53-mutated cell lines (IC(50)s >17 micromol/L). In sharp contrast to the expectations, nutlin-3 concentrations that did not affect viability of p53-mutated cell lines strongly increased the efficacy of vincristine in p53-mutated, P-glycoprotein (P-gp)-overexpressing cell lines (decrease in IC(50)s 92- to 3,434-fold). Similar results were obtained for other P-gp substrates. Moreover, nutlin-3 reduced efflux of rhodamine 123 and other fluorescence dyes that are effluxed by P-gp. Investigation of Madin-Darby canine kidney (MDCK) II cells stably transfected with plasmids encoding for P-gp (MDCKII MDR1) or multidrug resistance protein 1 (MRP-1, MDCKII MRP1) revealed that nutlin-3 not only interferes with P-gp but also affects MRP-1-mediated efflux. Kinetic studies and investigation of P-gp-ATPase activity showed that nutlin-3 is likely to act as a P-gp transport substrate. Examination of the nutlin-3 enantiomers nutlin-3a and nutlin-3b revealed that, in contrast to MDM2-inhibitory activity that is limited to nutlin-3a, both enantiomers similarly interfere with P-gp-mediated drug efflux. In conclusion, nutlin-3-induced inhibition of P-gp and MRP-1 was discovered as a novel anticancer mechanism of the substance in this report.


Asunto(s)
Miembro 1 de la Subfamilia B de Casetes de Unión a ATP/antagonistas & inhibidores , Protocolos de Quimioterapia Combinada Antineoplásica/farmacología , Imidazoles/farmacología , Neuroblastoma/tratamiento farmacológico , Piperazinas/farmacología , Rabdomiosarcoma Alveolar/tratamiento farmacológico , Miembro 1 de la Subfamilia B de Casetes de Unión a ATP/biosíntesis , Miembro 1 de la Subfamilia B de Casetes de Unión a ATP/metabolismo , Animales , Línea Celular Tumoral , Resistencia a Antineoplásicos , Sinergismo Farmacológico , Humanos , Imidazoles/administración & dosificación , Imidazoles/farmacocinética , Ratones , Mutación , Neuroblastoma/genética , Neuroblastoma/metabolismo , Piperazinas/administración & dosificación , Piperazinas/farmacocinética , Proteínas Proto-Oncogénicas c-mdm2/antagonistas & inhibidores , Rabdomiosarcoma Alveolar/genética , Rabdomiosarcoma Alveolar/metabolismo , Rodamina 123/farmacocinética , Estereoisomerismo , Proteína p53 Supresora de Tumor/biosíntesis , Proteína p53 Supresora de Tumor/genética , Vincristina/administración & dosificación
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA