Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Oncogene ; 34(26): 3357-68, 2015 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-25174395

RESUMEN

Neuroblastoma, a childhood cancer that originates from neural crest-derived cells, is the most common deadly solid tumor of infancy. Amplification of the MYCN oncogene, which occurs in approximately 20-25% of human neuroblastomas, is the most prominent genetic marker of high-stage disease. The availability of valid preclinical in vivo models is a prerequisite to develop novel targeted therapies. We here report on the generation of transgenic mice with Cre-conditional induction of MYCN in dopamine ß-hydroxylase-expressing cells, termed LSL-MYCN;Dbh-iCre. These mice develop neuroblastic tumors with an incidence of >75%, regardless of strain background. Molecular profiling of tumors revealed upregulation of the MYCN-dependent miR-17-92 cluster as well as expression of neuroblastoma marker genes, including tyrosine hydroxylase and the neural cell adhesion molecule 1. Gene set enrichment analyses demonstrated significant correlation with MYC-associated expression patterns. Array comparative genome hybridization showed that chromosomal aberrations in LSL-MYCN;Dbh-iCre tumors were syntenic to those observed in human neuroblastomas. Treatment of a cell line established from a tumor derived from a LSL-MYCN;Dbh-iCre mouse with JQ1 or MLN8237 reduced cell viability and demonstrated oncogene addiction to MYCN. Here we report establishment of the first Cre-conditional human MYCN-driven mouse model for neuroblastoma that closely recapitulates the human disease with respect to tumor localization, histology, marker expression and genomic make up. This mouse model is a valuable tool for further functional studies and to assess the effect of targeted therapies.


Asunto(s)
Evaluación Preclínica de Medicamentos/métodos , Integrasas/genética , Neuroblastoma/patología , Proteínas Nucleares/genética , Proteínas Oncogénicas/genética , Regiones Promotoras Genéticas , Transgenes , Animales , Modelos Animales de Enfermedad , Femenino , Perfilación de la Expresión Génica , Humanos , Ratones , Ratones Endogámicos C57BL , Ratones Desnudos , Ratones Transgénicos , Análisis por Micromatrices , Proteína Proto-Oncogénica N-Myc , Neuroblastoma/genética , Células Tumorales Cultivadas
2.
Klin Padiatr ; 225(6): 303-8, 2013 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-24166094

RESUMEN

Treatment for neuroblastoma, the most common extracranial childhood tumor, spans a broad range of aggressiveness that mirrors the risk profiles of disease subtypes, with high-risk neuroblastoma still presenting a clinical challenge. Currently, most patients with relapsed neuro-blastoma die of disease and present a major challenge for treatment. New therapeutic options are urgently needed to improve patient survival. Activating mutations in the gene encoding the anaplastic lymphoma kinase (ALK) remain the most frequent druggable mutations identified in neuroblastomas to date. Preclinical data support an oncogene addiction of neuroblastoma cells to mutated ALK and demonstrate that ALK inhibitory therapy strongly combats tumor models. Most recently, pediatric phase I testing has been completed for the first approved ALK inhibitor, Crizotinib, showing very encouraging antitumoral results in neuroblastoma patients. Subsequently, an international phase I study with the second generation ALK inhibitor, LDK-378, will be launched that makes ALK inhibitory therapy also available to pediatric patients in Germany.


Asunto(s)
Sistemas de Liberación de Medicamentos , Neuroblastoma/tratamiento farmacológico , Inhibidores de Proteínas Quinasas/uso terapéutico , Proteínas Tirosina Quinasas Receptoras/antagonistas & inhibidores , Quinasa de Linfoma Anaplásico , Transformación Celular Neoplásica/efectos de los fármacos , Transformación Celular Neoplásica/genética , Niño , Ensayos Clínicos Fase I como Asunto , Crizotinib , Análisis Mutacional de ADN , Aprobación de Drogas , Alemania , Humanos , Estadificación de Neoplasias , Neuroblastoma/genética , Neuroblastoma/patología , Inhibidores de Proteínas Quinasas/efectos adversos , Pirazoles/efectos adversos , Pirazoles/uso terapéutico , Piridinas/efectos adversos , Piridinas/uso terapéutico , Pirimidinas/efectos adversos , Pirimidinas/uso terapéutico , Proteínas Tirosina Quinasas Receptoras/genética , Sulfonas/efectos adversos , Sulfonas/uso terapéutico
3.
Oncogene ; 32(24): 2927-36, 2013 Jun 13.
Artículo en Inglés | MEDLINE | ID: mdl-22797059

RESUMEN

MicroRNAs (miRNAs) contribute to the pathogenesis of many forms of cancer, including the pediatric cancer neuroblastoma, but the underlying mechanisms leading to altered miRNA expression are often unknown. Here, a novel integrated approach for analyzing DNA methylation coupled with miRNA and mRNA expression data sets identified 67 epigenetically regulated miRNA in neuroblastoma. A large proportion (42%) of these miRNAs was associated with poor patient survival when underexpressed in tumors. Moreover, we demonstrate that this panel of epigenetically silenced miRNAs targets a large set of genes that are overexpressed in tumors from patients with poor survival in a highly redundant manner. The genes targeted by the epigenetically regulated miRNAs are enriched for a number of biological processes, including regulation of cell differentiation. Functional studies involving ectopic overexpression of several of the epigenetically silenced miRNAs had a negative impact on neuroblastoma cell viability, providing further support to the concept that inactivation of these miRNAs is important for neuroblastoma disease pathogenesis. One locus, miR-340, induced either differentiation or apoptosis in a cell context dependent manner, indicating a tumor suppressive function for this miRNA. Intriguingly, it was determined that miR-340 is upregulated by demethylation of an upstream genomic region that occurs during the process of neuroblastoma cell differentiation induced by all-trans retinoic acid (ATRA). Further biological studies of miR-340 revealed that it directly represses the SOX2 transcription factor by targeting of its 3'-untranslated region, explaining the mechanism by which SOX2 is downregulated by ATRA. Although SOX2 contributes to the maintenance of stem cells in an undifferentiated state, we demonstrate that miR-340-mediated downregulation of SOX2 is not required for ATRA induced differentiation to occur. In summary, our results exemplify the dynamic nature of the miRNA epigenome and identify a remarkable network of miRNA/mRNA interactions that significantly contribute to neuroblastoma disease pathogenesis.


Asunto(s)
Epigénesis Genética/genética , Redes Reguladoras de Genes/genética , MicroARNs/genética , Neuroblastoma/etiología , Neuroblastoma/genética , Regiones no Traducidas 3'/genética , Línea Celular Tumoral , Biología Computacional , Metilación de ADN/efectos de los fármacos , Metilación de ADN/genética , Epigénesis Genética/efectos de los fármacos , Redes Reguladoras de Genes/efectos de los fármacos , Genómica , Humanos , Neuroblastoma/patología , Factores de Transcripción SOXB1/genética , Análisis de Supervivencia , Tretinoina/farmacología
4.
Oncogene ; 32(8): 1059-65, 2013 Feb 21.
Artículo en Inglés | MEDLINE | ID: mdl-22484425

RESUMEN

Neuroblastoma is an embryonal tumor with a heterogeneous clinical course. The tumor is presumed to be derived from the neural crest, but the cells of origin remain to be determined. To date, few recurrent genetic changes contributing to neuroblastoma formation, such as amplification of the MYCN oncogene and activating mutations of the ALK oncogene, have been identified. The possibility to model neuroblastoma in mice allows investigation of the cell of origin hypothesis in further detail. Here we present the evidence that murine neural crest progenitor cells can give rise to neuroblastoma upon transformation with MYCN or ALK(F1174L). For this purpose we used JoMa1, a multipotent neural crest progenitor cell line, which is kept in a viable and undifferentiated state by a tamoxifen-activated c-Myc transgene (c-MycER(T)). Expression of MYCN or ALK(F1174L), one of the oncogenic ALK variants identified in primary neuroblastomas, enabled these cells to grow independently of c-MycER(T) activity in vitro and caused formation of neuroblastoma-like tumors in vivo in contrast to parental JoMa1 cells and JoMa1 cells-expressing TrkA or GFP. Tumorigenicity was enhanced upon serial transplantation of tumor-derived cells, and tumor cells remained susceptible to the MYC-inhibitor, NBT-272, indicating that cell growth depended on functional MYCN. Our findings support neural crest progenitor cells as the precursor cells of neuroblastoma, and indicate that neuroblastomas arise as their malignant progeny.


Asunto(s)
Células Madre Neoplásicas/patología , Cresta Neural/patología , Neuroblastoma/genética , Neuroblastoma/patología , Proteínas Nucleares/genética , Proteínas Oncogénicas/genética , Proteínas Tirosina Quinasas Receptoras/genética , Células Madre/patología , Quinasa de Linfoma Anaplásico , Animales , Transformación Celular Neoplásica/genética , Transformación Celular Neoplásica/metabolismo , Transformación Celular Neoplásica/patología , Humanos , Ratones , Ratones Desnudos , Ratones Transgénicos , Proteína Proto-Oncogénica N-Myc , Células Madre Neoplásicas/metabolismo , Cresta Neural/metabolismo , Neuroblastoma/metabolismo , Proteínas Nucleares/biosíntesis , Proteínas Nucleares/metabolismo , Proteínas Oncogénicas/biosíntesis , Proteínas Oncogénicas/metabolismo , Proteínas Tirosina Quinasas Receptoras/biosíntesis , Proteínas Tirosina Quinasas Receptoras/metabolismo , Células Madre/metabolismo , Transfección , Trasplante Heterólogo
5.
Br J Cancer ; 107(8): 1409-17, 2012 Oct 09.
Artículo en Inglés | MEDLINE | ID: mdl-23047593

RESUMEN

BACKGROUND: Using mRNA expression-derived signatures as predictors of individual patient outcome has been a goal ever since the introduction of microarrays. Here, we addressed whether analyses of tumour mRNA at the exon level can improve on the predictive power and classification accuracy of gene-based expression profiles using neuroblastoma as a model. METHODS: In a patient cohort comprising 113 primary neuroblastoma specimens expression profiling using exon-level analyses was performed to define predictive signatures using various machine-learning techniques. Alternative transcript use was calculated from relative exon expression. Validation of alternative transcripts was achieved using qPCR- and cell-based approaches. RESULTS: Both predictors derived from the gene or the exon levels resulted in prediction accuracies >80% for both event-free and overall survival and proved as independent prognostic markers in multivariate analyses. Alternative transcript use was most prominently linked to the amplification status of the MYCN oncogene, expression of the TrkA/NTRK1 neurotrophin receptor and survival. CONCLUSION: As exon level-based prediction yields comparable, but not significantly better, prediction accuracy than gene expression-based predictors, gene-based assays seem to be sufficiently precise for predicting outcome of neuroblastoma patients. However, exon-level analyses provide added knowledge by identifying alternative transcript use, which should deepen the understanding of neuroblastoma biology.


Asunto(s)
Exones/genética , Neuroblastoma/genética , Proteínas Nucleares/genética , Proteínas Oncogénicas/genética , Receptor trkA/genética , Línea Celular Tumoral , Preescolar , Perfilación de la Expresión Génica , Humanos , Lactante , Proteína Proto-Oncogénica N-Myc , Neuroblastoma/mortalidad , Pronóstico , ARN Mensajero , Factores de Riesgo , Análisis de Supervivencia
6.
Leukemia ; 26(9): 2039-51, 2012 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-22699452

RESUMEN

Lysine (K)-specific demethylase 1A (LSD1/KDM1A) has been identified as a potential therapeutic target in solid cancers and more recently in acute myeloid leukemia. However, the potential side effects of a LSD1-inhibitory therapy remain elusive. Here, we show, with a newly established conditional in vivo knockdown model, that LSD1 represents a central regulator of hematopoietic stem and progenitor cells. LSD1 knockdown (LSD1-kd) expanded progenitor numbers by enhancing their proliferative behavior. LSD1-kd led to an extensive expansion of granulomonocytic, erythroid and megakaryocytic progenitors. In contrast, terminal granulopoiesis, erythropoiesis and platelet production were severely inhibited. The only exception was monopoiesis, which was promoted by LSD1 deficiency. Importantly, we showed that peripheral blood granulocytopenia, monocytosis, anemia and thrombocytopenia were reversible after LSD1-kd termination. Extramedullary splenic hematopoiesis contributed to the phenotypic reversion, and progenitor populations remained expanded. LSD1-kd was associated with the upregulation of key hematopoietic genes, including Gfi1b, Hoxa9 and Meis1, which are known regulators of the HSC/progenitor compartment. We also demonstrated that LSD1-kd abrogated Gfi1b-negative autoregulation by crossing LSD1-kd with Gfi1b:GFP mice. Taken together, our findings distinguish LSD1 as a critical regulator of hematopoiesis and point to severe, but reversible, side effects of a LSD1-targeted therapy.


Asunto(s)
Diferenciación Celular , Proliferación Celular , Hematopoyesis/fisiología , Oxidorreductasas N-Desmetilantes/fisiología , Células Madre/citología , Animales , Western Blotting , Eritropoyesis/fisiología , Femenino , Citometría de Flujo , Granulocitos/citología , Granulocitos/metabolismo , Histona Demetilasas , Humanos , Integrasas/metabolismo , Masculino , Megacariocitos/citología , Megacariocitos/metabolismo , Ratones , Ratones Transgénicos , Oxidorreductasas N-Desmetilantes/antagonistas & inhibidores , Proteínas Proto-Oncogénicas/metabolismo , Proteínas Represoras/metabolismo , Células Madre/metabolismo
7.
Oncogene ; 31(12): 1571-81, 2012 Mar 22.
Artículo en Inglés | MEDLINE | ID: mdl-21860421

RESUMEN

Neuroblastoma tumors frequently show loss of heterozygosity of chromosome 11q with a shortest region of overlap in the 11q23 region. These deletions are thought to cause inactivation of tumor suppressor genes leading to haploinsufficiency. Alternatively, micro-deletions could lead to gene fusion products that are tumor driving. To identify such events we analyzed a series of neuroblastomas by comparative genomic hybridization and single-nucleotide polymorphism arrays and integrated these data with Affymetrix mRNA profiling data with the bioinformatic tool R2 (http://r2.amc.nl). We identified three neuroblastoma samples with small interstitial deletions at 11q23, upstream of the forkhead-box R1 transcription factor (FOXR1). Genes at the proximal side of the deletion were fused to FOXR1, resulting in fusion transcripts of MLL-FOXR1 and PAFAH1B2-FOXR1. FOXR1 expression has only been detected in early embryogenesis. Affymetrix microarray analysis showed high FOXR1 mRNA expression exclusively in the neuroblastomas with micro-deletions and rare cases of other tumor types, including osteosarcoma cell line HOS. RNAi silencing of FOXR1 strongly inhibited proliferation of HOS cells and triggered apoptosis. Expression profiling of these cells and reporter assays suggested that FOXR1 is a negative regulator of fork-head box factor-mediated transcription. The neural crest stem cell line JoMa1 proliferates in culture conditional to activity of a MYC-ER transgene. Over-expression of the wild-type FOXR1 could functionally replace MYC and drive proliferation of JoMa1. We conclude that FOXR1 is recurrently activated in neuroblastoma by intrachromosomal deletion/fusion events, resulting in overexpression of fusion transcripts. Forkhead-box transcription factors have not been previously implicated in neuroblastoma pathogenesis. Furthermore, this is the first identification of intrachromosomal fusion genes in neuroblastoma.


Asunto(s)
Cromosomas Humanos Par 11 , Neuroblastoma/genética , Recombinación Genética , Animales , Línea Celular Tumoral , Hibridación Genómica Comparativa , Regulación Neoplásica de la Expresión Génica , Silenciador del Gen , Haploinsuficiencia , Humanos , Pérdida de Heterocigocidad , Ratones , Fusión de Oncogenes , Polimorfismo de Nucleótido Simple , Eliminación de Secuencia
8.
Oncogene ; 29(9): 1394-404, 2010 Mar 04.
Artículo en Inglés | MEDLINE | ID: mdl-19946337

RESUMEN

Increased activity of MYC protein-family members is a common feature in many cancers. Using neuroblastoma as a tumor model, we established a microRNA (miRNA) signature for activated MYCN/c-MYC signaling in two independent primary neuroblastoma tumor cohorts and provide evidence that c-MYC and MYCN have overlapping functions. On the basis of an integrated approach including miRNA and messenger RNA (mRNA) gene expression data we show that miRNA activation contributes to widespread mRNA repression, both in c-MYC- and MYCN-activated tumors. c-MYC/MYCN-induced miRNA activation was shown to be dependent on c-MYC/MYCN promoter binding as evidenced by chromatin immunoprecipitation. Finally, we show that pathways, repressed through c-MYC/MYCN miRNA activation, are highly correlated to tumor aggressiveness and are conserved across different tumor entities suggesting that c-MYC/MYCN activate a core set of miRNAs for cooperative repression of common transcriptional programs related to disease aggressiveness. Our results uncover a widespread correlation between miRNA activation and c-MYC/MYCN-mediated coding gene expression modulation and further substantiate the overlapping functions of c-MYC and MYCN in the process of tumorigenesis.


Asunto(s)
Regulación Neoplásica de la Expresión Génica/fisiología , Redes Reguladoras de Genes/efectos de los fármacos , Genes myc/fisiología , MicroARNs/farmacología , Neuroblastoma/genética , Proteínas Nucleares/farmacología , Proteínas Oncogénicas/farmacología , Regiones Promotoras Genéticas/efectos de los fármacos , Línea Celular Tumoral , Redes Reguladoras de Genes/fisiología , Silenciador del Gen/fisiología , Genes myc/genética , Humanos , MicroARNs/biosíntesis , Proteína Proto-Oncogénica N-Myc , Neuroblastoma/terapia , ARN Interferente Pequeño/farmacología , Factores de Transcripción/fisiología , Resultado del Tratamiento
9.
Oncogene ; 28(19): 2015-23, 2009 May 14.
Artículo en Inglés | MEDLINE | ID: mdl-19363525

RESUMEN

Expression of Trk receptors is an important prognostic factor in neuroblastoma (NB) and other cancers. TrkB and its ligand brain-derived neurotrophic factor (BDNF) are preferentially expressed in NB with poor prognosis, conferring invasive and metastatic potential to the tumor cells as well as enhancing therapy resistance. Galectin-1 (Gal-1) has emerged as an interesting cancer target, as it is involved in modulating cell proliferation, cell death and cell migration, all of which are linked to cancer initiation and progression. We previously identified Gal-1 mRNA to be upregulated in patients with aggressive, relapsing NB and found that Gal-1 protein was upregulated in human SY5Y NB cells on activation of ectopically expressed TrkB (SY5Y-TrkB), but not TrkA (SY5Y-TrkA). Here, we report that Gal-1 mRNA levels positively correlated with TrkB expression and anticorrelated with TrkA expression in a cohort of 102 primary NB. Immunohistochemical analyses of 92 primary NB specimens revealed high Gal-1 expression in stromal septae and in neuroblasts. BDNF-mediated activation of TrkB enhanced invasiveness and migration in vitro, which could be impaired by transient transfection using Gal-1-specific siRNA or a neutralizing antibody directed against Gal-1. The addition of recombinant Gal-1 (rGal-1) in the absence of BDNF partially restored migration and invasive capacity. Using the Trk inhibitor K252a, we could show that the upregulation of Gal-1 protein strictly depended on activated TrkB. Our data suggest that targeting Gal-1 might be a promising strategy for the treatment of aggressive NB.


Asunto(s)
Galectina 1/metabolismo , Neuroblastoma/fisiopatología , Receptor trkB/metabolismo , Factor Neurotrófico Derivado del Encéfalo/metabolismo , Línea Celular Tumoral , Movimiento Celular , Regulación hacia Abajo , Galectina 1/genética , Regulación Neoplásica de la Expresión Génica , Humanos , Receptor trkA/metabolismo
10.
J Pathol ; 216(4): 428-39, 2008 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-18855347

RESUMEN

Synovial sarcomas account for 5-10% of all soft tissue sarcomas and the majority of synovial sarcomas display characteristic t(X;18) translocations that result in enhanced transcription of the insulin-like growth factor-2 (IGF-2) gene. IGF-2 is an essential fetal mitogen involved in the pathogenesis of different tumours, leading to cellular proliferation and inhibition of apoptosis. Here we asked whether activation of IGF signalling is of functional importance in synovial sarcomas. We screened human synovial sarcomas for expression of IGF-2 and the phosphorylated IGF-1 receptor (IGF-1R), which mainly mediates the proliferative and anti-apoptotic effects of IGF-2. Since both the phosphatidylinositol 3'-kinase (PI3K)-AKT pathway and the MAPK signalling cascade are known to be involved in the transmission of IGF-1R signals, expression of phosphorylated (p)-AKT and p-p44/42 MAPK was additionally assessed. All tumours expressed IGF-2 and 78% showed an activated IGF-1R. All tumours were found to express p-AKT and 92% showed expression of activated p44/42 MAPK. To analyse the functional and potential therapeutic relevance of IGF-1R signalling, synovial sarcoma cell lines were treated with the IGF-1R inhibitor NVP-AEW541. Growth was impaired by the IGF-1R antagonist, which was consistently accompanied by a dose-dependent reduction of phosphorylation of AKT and p44/42 MAPK. Functionally, inhibition of the receptor led to increased apoptosis and diminished mitotic activity. Concurrent exposure of selected cells to NVP-AEW541 and conventional chemotherapeutic agents resulted in positive interactions. Finally, synovial sarcoma cell migration was found to be dependent on signals transmitted by the IGF-1R. In summary, our data show that the IGF-1R might represent a promising therapeutic target in synovial sarcomas.


Asunto(s)
Receptor IGF Tipo 1/fisiología , Sarcoma Sinovial/metabolismo , Transducción de Señal/fisiología , Adulto , Antineoplásicos/farmacología , Apoptosis , Western Blotting/métodos , Línea Celular Tumoral , Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Femenino , Técnica del Anticuerpo Fluorescente , Humanos , Factor II del Crecimiento Similar a la Insulina/genética , Factor II del Crecimiento Similar a la Insulina/metabolismo , Sistema de Señalización de MAP Quinasas/fisiología , Masculino , Persona de Mediana Edad , Fosfatidilinositol 3-Quinasas/metabolismo , Fosforilación , Proteínas Proto-Oncogénicas c-akt/metabolismo , Pirimidinas/farmacología , Pirroles/farmacología , ARN Mensajero/análisis , Receptor IGF Tipo 1/antagonistas & inhibidores , Receptor IGF Tipo 1/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa/métodos , Sarcoma Sinovial/patología , Transducción de Señal/efectos de los fármacos
11.
Klin Padiatr ; 215(6): 298-302, 2003.
Artículo en Inglés | MEDLINE | ID: mdl-14677092

RESUMEN

BACKGROUND: Experimental studies for the treatment of relapsed neuroblastoma include the use of hyperthermia in combination with chemotherapeutic drugs. Cytotoxic effects of alkylants and platinum compounds on tumor cells can be enhanced by hyperthermia in various in vitro models. However, the underlying molecular mechanisms are still largely unknown. METHOD: In this study, we used microarray-analysis as a new biological approach to gain insight into the pharmacogenomics and possible target genes of thermochemotherapy. As a model, LAN 1 neuroblastoma cells were treated for 1 h with low doses of cisplatin alone, with simultaneous heating to 42 degrees C or with hyperthermia alone. Gene expression was analyzed at five time points 0 to 24 h after treatment using U95Av2 oligonucleotide arrays (Affymetrix Inc). Significant changes of gene expression levels were calculated by similarity metrices and Pearson correlation. RESULTS: Only a few genes (n = 23) demonstrated altered expression following treatment of LAN 1 cells with cisplatin alone. Hyperthermia alone resulted in significant expression changes of 136 genes in comparison to untreated control cells. Combination therapy of cisplatin and hyperthermia resulted in expression changes of 251 genes, interestingly including 131 genes with unchanged expression under treatment with either cisplatin or hyperthermia alone. Significant changes of expression levels could be annotated to genes involved in heat shock response, protein degradation and apoptosis. These results are now being validated on mRNA- and protein levels by RT-PCR and Western Blot analysis. CONCLUSION: Microarray-Analysis is a suitable new approach for the identification of target genes, which might play an important role for the synergistic effect of hyperthermia and chemotherapy in tumor cells.


Asunto(s)
Antineoplásicos/farmacología , Cisplatino/farmacología , Perfilación de la Expresión Génica , Hipertermia Inducida , Neuroblastoma/genética , Neuroblastoma/terapia , Análisis de Secuencia por Matrices de Oligonucleótidos , Antineoplásicos/administración & dosificación , Apoptosis , Western Blotting , Calorimetría , Muerte Celular , Línea Celular , Cisplatino/administración & dosificación , Terapia Combinada , Regulación Neoplásica de la Expresión Génica , Proteínas de Choque Térmico , Humanos , Neuroblastoma/tratamiento farmacológico , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Factores de Tiempo , Células Tumorales Cultivadas
12.
Acta Crystallogr C ; 57(Pt 11): 1288-9, 2001 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-11706251

RESUMEN

The metallation reaction between dibutylmagnesium and 2,6-diisopropyl-N-(trimethylsilyl)aniline gives the unusual monomeric three-coordinate complex (diethyl ether-kappa O)bis[2,6-diisopropyl-N-(trimethylsilyl)anilido-kappa N]magnesium(II), [Mg(C(15)H(26)NSi)(2)(C(4)H(10)O)] or [Mg[(Me(3)Si)(2,6-(i)Pr(2)C(6)H(3))N](2)(Et(2)O)]. This low-coordinate species has a distorted trigonal-planar coordination environment, with an additional short Mg-C(ipso) contact of 2.799 (2) A.

15.
Arch Environ Health ; 13(5): 662-3, 1966 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-5925641
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...