Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
1.
Genome Res ; 25(9): 1391-400, 2015 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-26163319

RESUMEN

Chromatin immunoprecipitation coupled with next-generation sequencing (ChIP-seq) is a powerful technology to identify the genome-wide locations of transcription factors and other DNA binding proteins. Computational ChIP-seq peak calling infers the location of protein-DNA interactions based on various measures of enrichment of sequence reads. In this work, we introduce an algorithm, Q, that uses an assessment of the quadratic enrichment of reads to center candidate peaks followed by statistical analysis of saturation of candidate peaks by 5' ends of reads. We show that our method not only is substantially faster than several competing methods but also demonstrates statistically significant advantages with respect to reproducibility of results and in its ability to identify peaks with reproducible binding site motifs. We show that Q has superior performance in the delineation of double RNAPII and H3K4me3 peaks surrounding transcription start sites related to a better ability to resolve individual peaks. The method is implemented in C++ and is freely available under an open source license.


Asunto(s)
Inmunoprecipitación de Cromatina , Genómica/métodos , Secuenciación de Nucleótidos de Alto Rendimiento , Algoritmos , Sitios de Unión/genética , Proteínas de Unión al ADN , Humanos , Motivos de Nucleótidos , Unión Proteica , Reproducibilidad de los Resultados , Factores de Transcripción/metabolismo , Sitio de Iniciación de la Transcripción
2.
BMC Genomics ; 17(1): 873, 2016 11 04.
Artículo en Inglés | MEDLINE | ID: mdl-27814676

RESUMEN

BACKGROUND: ChIP-nexus, an extension of the ChIP-exo protocol, can be used to map the borders of protein-bound DNA sequences at nucleotide resolution, requires less input DNA and enables selective PCR duplicate removal using random barcodes. However, the use of random barcodes requires additional preprocessing of the mapping data, which complicates the computational analysis. To date, only a very limited number of software packages are available for the analysis of ChIP-exo data, which have not yet been systematically tested and compared on ChIP-nexus data. RESULTS: Here, we present a comprehensive software package for ChIP-nexus data that exploits the random barcodes for selective removal of PCR duplicates and for quality control. Furthermore, we developed bespoke methods to estimate the width of the protected region resulting from protein-DNA binding and to infer binding positions from ChIP-nexus data. Finally, we applied our peak calling method as well as the two other methods MACE and MACS2 to the available ChIP-nexus data. CONCLUSIONS: The Q-nexus software is efficient and easy to use. Novel statistics about duplication rates in consideration of random barcodes are calculated. Our method for the estimation of the width of the protected region yields unbiased signatures that are highly reproducible for biological replicates and at the same time very specific for the respective factors analyzed. As judged by the irreproducible discovery rate (IDR), our peak calling algorithm shows a substantially better reproducibility. An implementation of Q-nexus is available at http://charite.github.io/Q/ .


Asunto(s)
Inmunoprecipitación de Cromatina , Biología Computacional/métodos , Secuenciación de Nucleótidos de Alto Rendimiento , Programas Informáticos , Algoritmos , Sitios de Unión , Proteínas de Unión al ADN/metabolismo , Motivos de Nucleótidos , Unión Proteica , Reproducibilidad de los Resultados , Factores de Transcripción/metabolismo
3.
Nat Chem Biol ; 10(7): 574-81, 2014 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-24838012

RESUMEN

Ten eleven translocation (Tet) enzymes oxidize the epigenetically important DNA base 5-methylcytosine (mC) stepwise to 5-hydroxymethylcytosine (hmC), 5-formylcytosine and 5-carboxycytosine. It is currently unknown whether Tet-induced oxidation is limited to cytosine-derived nucleobases or whether other nucleobases are oxidized as well. We synthesized isotopologs of all major oxidized pyrimidine and purine bases and performed quantitative MS to show that Tet-induced oxidation is not limited to mC but that thymine is also a substrate that gives 5-hydroxymethyluracil (hmU) in mouse embryonic stem cells (mESCs). Using MS-based isotope tracing, we show that deamination of hmC does not contribute to the steady-state levels of hmU in mESCs. Protein pull-down experiments in combination with peptide tracing identifies hmU as a base that influences binding of chromatin remodeling proteins and transcription factors, suggesting that hmU has a specific function in stem cells besides triggering DNA repair.


Asunto(s)
Proteínas de Unión al ADN/metabolismo , ADN/metabolismo , Células Madre Embrionarias/metabolismo , Pentoxil (Uracilo)/análogos & derivados , Proteínas Proto-Oncogénicas/metabolismo , Timina/metabolismo , 5-Metilcitosina/análogos & derivados , Animales , Secuencia de Bases , Isótopos de Carbono , Ensamble y Desensamble de Cromatina , Cromatografía Liquida , Citosina/análogos & derivados , Citosina/metabolismo , Proteínas de Unión al ADN/genética , Dioxigenasas , Células Madre Embrionarias/citología , Expresión Génica , Ratones , Datos de Secuencia Molecular , Oxidación-Reducción , Pentoxil (Uracilo)/metabolismo , Unión Proteica , Proteínas Proto-Oncogénicas/genética , Espectrometría de Masa por Ionización de Electrospray , Factores de Transcripción/genética , Factores de Transcripción/metabolismo
4.
Angew Chem Int Ed Engl ; 51(26): 6516-20, 2012 Jun 25.
Artículo en Inglés | MEDLINE | ID: mdl-22644704

RESUMEN

Eraserhead: Stem cells seem to erase epigenetic information by decarboxylation of the newly discovered epigenetic base 5-carboxycytosine (caC; see picture). This reaction is likely to involve a nucleophilic attack of the C5-C6 double bond.


Asunto(s)
Citosina/análogos & derivados , Células Madre Embrionarias/química , Animales , Secuencia de Bases , Citosina/química , Citosina/metabolismo , ADN/química , Descarboxilación , Células Madre Embrionarias/metabolismo , Ratones , Isótopos de Nitrógeno/química , Oxidación-Reducción
5.
Nat Commun ; 12(1): 3582, 2021 06 11.
Artículo en Inglés | MEDLINE | ID: mdl-34117224

RESUMEN

In mouse development, long-term silencing by CpG island DNA methylation is specifically targeted to germline genes; however, the molecular mechanisms of this specificity remain unclear. Here, we demonstrate that the transcription factor E2F6, a member of the polycomb repressive complex 1.6 (PRC1.6), is critical to target and initiate epigenetic silencing at germline genes in early embryogenesis. Genome-wide, E2F6 binds preferentially to CpG islands in embryonic cells. E2F6 cooperates with MGA to silence a subgroup of germline genes in mouse embryonic stem cells and in embryos, a function that critically depends on the E2F6 marked box domain. Inactivation of E2f6 leads to a failure to deposit CpG island DNA methylation at these genes during implantation. Furthermore, E2F6 is required to initiate epigenetic silencing in early embryonic cells but becomes dispensable for the maintenance in differentiated cells. Our findings elucidate the mechanisms of epigenetic targeting of germline genes and provide a paradigm for how transient repression signals by DNA-binding factors in early embryonic cells are translated into long-term epigenetic silencing during mouse development.


Asunto(s)
Islas de CpG/genética , Factor de Transcripción E2F6/genética , Factor de Transcripción E2F6/metabolismo , Desarrollo Embrionario/genética , Epigénesis Genética , Células Germinativas/metabolismo , Animales , Sitios de Unión , Sistemas CRISPR-Cas , Diferenciación Celular , Metilación de ADN , Silenciador del Gen , Ratones , Ratones Noqueados , Células Madre Embrionarias de Ratones , Complejo Represivo Polycomb 1/metabolismo , ARN Interferente Pequeño
6.
Pediatr Res ; 67(1): 35-41, 2010 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-19770690

RESUMEN

Hypoxia and other adverse conditions are commonly encountered by rapidly growing cells. The RNA-binding protein RBM3 (RNA-binding motif protein 3), which is transcriptionally induced by low temperature and hypoxia, has recently been implicated in survival of colon cancer cells by mechanisms involving cyclooxygenase-2 (COX-2) signaling. Immunohistochemically, we found strong RBM3 expression in a variety of malignant and proliferating tissues but low expression in resting and terminally differentiated cells. RBM3 expression in fibroblasts and human embryonal kidney (HEK293) cells subjected to serum deprivation or contact inhibition closely paralleled proliferation rates, assessed by real-time RT-PCR and immunoblotting. siRNA-mediated RBM3 knockdown reduced cell viability and finally led to cell death, which did not involve caspase-3-mediated apoptosis, cell cycle arrest, or COX-2 regulation. In contrast, RBM3 over-expression rescued cells from death under serum starvation. This was associated with increased translation rates, as measured by C serine and H phenylalanine incorporation. Together, RBM3 is a critical factor providing cellular survival advantages in an adverse microenvironment presumably by restoring translation efficacy.


Asunto(s)
Muerte Celular/fisiología , Proliferación Celular , Proteínas de Unión al ARN/fisiología , Secuencia de Bases , Línea Celular , Medio de Cultivo Libre de Suero , Cartilla de ADN , Humanos , Inmunohistoquímica , ARN Interferente Pequeño
7.
Oncol Res ; 18(10): 469-80, 2010.
Artículo en Inglés | MEDLINE | ID: mdl-20681406

RESUMEN

Prognosis for patients with glioblastoma multiforme (GBM) is poor. Inhibitors of histone deacetylases (HDACi) like trichostatin A (TSA) are promising alternatives to conventional treatment. Deficient tumor suppressor functions, such as TP53 mutations and p14(ARF)/p16(INK4a) deletions, are characteristic for GBM and can cause resistance to DNA damaging agents such as cisplatin and to HDACi like TSA. The type II tumor suppressor Inhibitor of growth 1 (ING1) is involved in DNA damage response and histone modification. We have previously shown that ING1 is downregulated in GBM and involved in glioma-induced angiogenesis and in cisplatin-induced apoptosis in malignant glioma cells. Hence, the goal of our present study was to investigate whether TSA affects ING1 protein expression and also whether modulating ING1 levels affects TSA-induced apoptosis in malignant glioma cells that contain deficient p53 function and inactive pl4(ARF)/p16(INK4a) signaling. If so, we asked, which apoptotic pathway might be the major mediator beyond this interaction. To test whether ING1 proteins function in TSA-induced apoptosis in GBM, we analyzed TSA effects in LN229 GBM cells, which harbor TP53 mutations and INK4a deletion, following ING1 knockdown by siRNA. Expression of ING1, acetylated core histones H3 and H4, and the proapoptotic proteins caspase 3 and Fas-associated death domain (FADD) was determined by Western blotting. Percentages of apoptotic cells were obtained by flow cytometry. TSA induced the major ING1 isoform p33(ING1b) and increased levels of both histone acetylation and apoptosis in LN229 cells. ING1 knockdown cells revealed marked resistance to TSA-induced apoptosis, impairment of caspase 3 activation, and suppression of FADD. The data suggest that ING1 contributes to TSA-induced apoptosis in GBM cells with deficient p53 and p14(ARF)/p16(INK4a) functions, possibly by regulating FADD/caspase 3 signaling.


Asunto(s)
Apoptosis/efectos de los fármacos , Caspasa 3/fisiología , Glioblastoma/tratamiento farmacológico , Inhibidores de Histona Desacetilasas/farmacología , Ácidos Hidroxámicos/farmacología , Péptidos y Proteínas de Señalización Intracelular/fisiología , Proteínas Nucleares/fisiología , Transducción de Señal/fisiología , Proteína p53 Supresora de Tumor/deficiencia , Proteínas Supresoras de Tumor/fisiología , Acetilación , Línea Celular Tumoral , Proteína de Dominio de Muerte Asociada a Fas/análisis , Glioblastoma/patología , Histonas/metabolismo , Humanos , Proteína Inhibidora del Crecimiento 1 , Péptidos y Proteínas de Señalización Intracelular/análisis , Péptidos y Proteínas de Señalización Intracelular/antagonistas & inhibidores , Proteínas Nucleares/análisis , Proteínas Nucleares/antagonistas & inhibidores , Proteínas Supresoras de Tumor/análisis , Proteínas Supresoras de Tumor/antagonistas & inhibidores
8.
Oncol Res ; 18(2-3): 95-105, 2009.
Artículo en Inglés | MEDLINE | ID: mdl-20066899

RESUMEN

The inhibitor of growth 1 (ING1) homologue ING4 has previously been implicated as a negative regulator of angiogenesis in a murine glioma and a multiple myeloma model. An association between ING1 and angiogenesis has not been reported yet. Our previous studies using tumor samples from patients have shown that ING1 levels are downregulated in glioblastoma multiforme (GBM), one of the most highly vascularized malignancies. Based on this background, the goal of this study was to test the effects of the major ING1 splicing isoforms, p47ING1a and p33ING1b, on pathological angiogenesis induced by human GBM cells. We used a chorioallantoic membrane (CAM) assay to examine whether LN229 human GBM cells can induce angiogenesis and whether alterations in ING1 expression, such as ING1 knockdown by siRNA or ectopic ING1 overexpression using ING1a and ING1b expression constructs, can affect this process. Increased ING1 protein expression significantly suppressed LN229 cell-induced angiogenesis in the CAM assay. While no effects on the proangiogenic factors VEGF or IL-8 were noted, the expression of angiopoietins (Ang) 1 and 4 were increased by the p47ING1a, but not by the p33ING1b isoform. Levels of Ang-2 were not sensitive to altered ING1 levels. Our data are the first to suggest that ING1 proteins suppress neoangiogenesis in GBM. Moreover, our results may support the idea that ING1 proteins regulate the expression of proteins that are critical for angiogenesis in GBM such as the angiopoietins.


Asunto(s)
Angiopoyetinas/genética , Glioblastoma/irrigación sanguínea , Péptidos y Proteínas de Señalización Intracelular/fisiología , Neovascularización Patológica/prevención & control , Proteínas Nucleares/fisiología , Proteínas Supresoras de Tumor/fisiología , Animales , Línea Celular Tumoral , Embrión de Pollo , Membrana Corioalantoides/irrigación sanguínea , Regulación Neoplásica de la Expresión Génica , Glioblastoma/metabolismo , Humanos , Proteína Inhibidora del Crecimiento 1 , Péptidos y Proteínas de Señalización Intracelular/antagonistas & inhibidores , Proteínas Nucleares/antagonistas & inhibidores , ARN Interferente Pequeño/genética , Proteínas Supresoras de Tumor/antagonistas & inhibidores , Factor A de Crecimiento Endotelial Vascular/fisiología
9.
Curr Biol ; 15(11): 1051-7, 2005 Jun 07.
Artículo en Inglés | MEDLINE | ID: mdl-15936277

RESUMEN

E2F transcription factors play a pivotal role in the regulation of cellular proliferation and can be subdivided into activating and repressing family members [1]. Like other E2Fs, E2F6 binds to E2F consensus sites, but in contrast to E2F1-5, it lacks an Rb binding domain and functions as an Rb-independent transcriptional repressor [2, 3, 4 and 5]. Instead, E2F6 has been shown to complex with Polycomb (PcG) group proteins [6 and 7], which have a well-established role in gene silencing. Here, we show that E2F6 plays an unexpected and essential role in the tissue specificity of gene expression. E2F6-deficient mice ubiquitously express the alpha-tubulin 3 and 7 genes, which are expressed strictly testis-specifically in control mice. Like an additional E2F6 target gene, Tex12, that we identified, tubulin 3 and 7 are normally expressed in male germ cells only. The promoters of the alpha-tubulin and Tex12 genes share a perfectly conserved E2F site, which E2F6 binds to. Mechanistically, E2F6-mediated repression involves CpG hypermethylation locking target promoters in an inactive state. Thus, E2F6 is essential for the long-term somatic silencing of certain male-germ-cell-specific genes, but it is dispensable for cell-cycle regulation.


Asunto(s)
Metilación de ADN , Regulación de la Expresión Génica , Testículo/metabolismo , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Tubulina (Proteína)/metabolismo , Animales , Secuencia de Bases , Sitios de Unión , Inmunoprecipitación de Cromatina , Cromatografía de Afinidad , Huella de ADN , Cartilla de ADN , Factor de Transcripción E2F6 , Ensayo de Cambio de Movilidad Electroforética , Masculino , Ratones , Ratones Noqueados , Análisis por Micromatrices , Datos de Secuencia Molecular , Regiones Promotoras Genéticas/genética , Alineación de Secuencia , Análisis de Secuencia de ADN
10.
Nucleic Acids Res ; 33(Database issue): D108-11, 2005 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-15608157

RESUMEN

Small interfering RNAs (siRNAs) have become a standard tool in functional genomics. Once incorporated into the RNA-induced silencing complex (RISC), siRNAs mediate the specific recognition of corresponding target mRNAs and their cleavage. However, only a small fraction of randomly chosen siRNA sequences is able to induce efficient gene silencing. In common laboratory practice, successful RNA interference experiments typically require both, the labour and cost-intensive identification of an active siRNA sequence and the optimization of target cell line-specific procedures for optimal siRNA delivery. To optimize the design and performance of siRNA experiments, we have established the human siRNA database (HuSiDa). The database provides sequences of published functional siRNA molecules targeting human genes and important technical details of the corresponding gene silencing experiments, including the mode of siRNA generation, recipient cell lines, transfection reagents and procedures and direct links to published references (PubMed). The database can be accessed at http://www.human-siRNA-database.net. We used the siRNA sequence information stored in the database for scrutinizing published sequence selection parameters for efficient gene silencing.


Asunto(s)
Bases de Datos de Ácidos Nucleicos , Interferencia de ARN , ARN Interferente Pequeño/química , Secuencia de Bases , Humanos , ARN Interferente Pequeño/metabolismo , Transfección
12.
Target Oncol ; 10(4): 523-33, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-25559288

RESUMEN

After extensive research on radiochemotherapy, 5-year survival rates of children with high risk neuroblastoma still do not exceed 50%, owing to adverse side-effects exemplified by doxorubicin-induced cardiomyopathy. A promising new approach is the combination of conventional therapies with specific modulation of cell signaling pathways promoting therapeutic resistance, such as inhibition of aberrant kinase activity or re-expression of silenced tumor suppressor genes by means of chromatin remodeling. In this regard, we established a system that allows to identify potential drug targets as well as to validate respective candidate inhibitors in high-risk neuroblastoma model cell lines. Cell culture, drug exposure, shRNA-mediated knockdown and phenotype analysis are integrated into an efficient and versatile single well-based protocol. By utilizing this system, we assessed RG108, SGI-1027 and nanaomycin A, three novel DNA methyltransferase inhibitors that have not been tested in neuroblastoma cell lines so far, for their potential of synergistic anti-tumor activity in combination with doxorubicin. We found that, similarly to azacytidine, SGI-1027 and nanaomycin A mediate synergistic growth inhibition with doxorubicin independently of N-Myc status. However, they display high cytotoxicity but lack global DNA demethylation activity. Secondly, we conducted a lentiviral shRNA screen of F-box proteins, key regulators of protein stability, and identified Fbxw11/ß-TrCP2 as well as Fbxo5/Emi1 as potential therapeutic targets in neuroblastoma. These results complement existing studies and underline the reliability and versatility of our single well-based protocol.


Asunto(s)
Metilación de ADN/efectos de los fármacos , Metilasas de Modificación del ADN/antagonistas & inhibidores , Inhibidores Enzimáticos/farmacología , Proteínas F-Box/genética , Neuroblastoma/terapia , Aminoquinolinas/administración & dosificación , Aminoquinolinas/farmacología , Protocolos de Quimioterapia Combinada Antineoplásica/farmacología , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Doxorrubicina/administración & dosificación , Doxorrubicina/farmacología , Sinergismo Farmacológico , Inhibidores Enzimáticos/administración & dosificación , Células HEK293 , Humanos , Terapia Molecular Dirigida , Naftoquinonas/administración & dosificación , Naftoquinonas/farmacología , Neuroblastoma/tratamiento farmacológico , Neuroblastoma/genética , Neuroblastoma/patología , Ftalimidas/administración & dosificación , Ftalimidas/farmacología , Pirimidinas/administración & dosificación , Pirimidinas/farmacología , ARN Interferente Pequeño/administración & dosificación , ARN Interferente Pequeño/genética , Triptófano/administración & dosificación , Triptófano/análogos & derivados , Triptófano/farmacología
13.
PLoS One ; 6(6): e21065, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-21731648

RESUMEN

The INhibitor of Growth tumor suppressors (ING1-ING5) affect aging, apoptosis, DNA repair and tumorigenesis. Plant homeodomains (PHD) of ING proteins bind histones in a methylation-sensitive manner to regulate chromatin structure. ING1 and ING2 contain a polybasic region (PBR) adjacent to their PHDs that binds stress-inducible phosphatidylinositol monophosphate (PtIn-MP) signaling lipids to activate these INGs. ING1 induces apoptosis independently of p53 but other studies suggest proapoptotic interdependence of ING1 and p53 leaving their functional relationship unclear. Here we identify a novel ubiquitin-binding domain (UBD) that overlaps with the PBR of ING1 and shows similarity to previously described UBDs involved in DNA damage responses. The ING1 UBD binds ubiquitin with high affinity (K(d)∼100 nM) and ubiquitin competes with PtIn-MPs for ING1 binding. ING1 expression stabilized wild-type, but not mutant p53 in an MDM2-independent manner and knockdown of endogenous ING1 depressed p53 levels in a transcription-independent manner. ING1 stabilized unmodified and six multimonoubiquitinated forms of wild-type p53 that were also seen upon DNA damage, but not p53 mutants lacking the six known sites of ubiquitination. We also find that ING1 physically interacts with herpesvirus-associated ubiquitin-specific protease (HAUSP), a p53 and MDM2 deubiquitinase (DUB), and knockdown of HAUSP blocks the ability of ING1 to stabilize p53. These data link lipid stress signaling to ubiquitin-mediated proteasomal degradation through the PBR/UBD of ING1 and further indicate that ING1 stabilizes p53 by inhibiting polyubiquitination of multimonoubiquitinated forms via interaction with and colocalization of the HAUSP-deubiquitinase with p53.


Asunto(s)
Péptidos y Proteínas de Señalización Intracelular/metabolismo , Proteínas Nucleares/metabolismo , Poliubiquitina/metabolismo , Proteína p53 Supresora de Tumor/metabolismo , Proteínas Supresoras de Tumor/metabolismo , Ubiquitinación , Línea Celular Tumoral , Humanos , Proteína Inhibidora del Crecimiento 1 , Péptidos y Proteínas de Señalización Intracelular/química , Modelos Biológicos , Proteínas Mutantes/metabolismo , Proteínas Nucleares/química , Fosfolípidos/metabolismo , Complejo de la Endopetidasa Proteasomal/metabolismo , Unión Proteica , Procesamiento Proteico-Postraduccional , Estabilidad Proteica , Estructura Terciaria de Proteína , Proteínas Supresoras de Tumor/química , Ubiquitina Tiolesterasa/metabolismo , Peptidasa Específica de Ubiquitina 7
14.
J Neurooncol ; 86(1): 23-30, 2008 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-17763999

RESUMEN

Impaired tumor suppressor functions, such as deficient p53, are characteristic for glioblastoma multiforme (GBM) and can cause resistance to DNA-damaging agents like cisplatin. We have recently shown that the INhibitor of Growth 1 (ING1) tumor suppressor is down-regulated in malignant gliomas and that the decrease of ING1 expression correlates with histological grade of malignancy, suggesting a role for ING1 in the pathogenesis and progression of malignant gliomas. Based on this background, the purpose of our current study was to examine the potential impact of ING1 protein levels on DNA-damage response in GBM. Using LN229 GBM cells, which express ING1 proteins and harbor mutant TP53, we are the first to show that DNA damage by cisplatin or ionizing radiation differentially induced the two major ING1 splicing isoforms. The p47 ING1a isoform, that promotes deacetylation of histones, thus formation of heterochromatic regions of DNA, which are less susceptible to DNA damage, was preferentially induced by >50-fold. This might represent a response to protect DNA from damage. Also, ING1 knockdown by siRNA accelerated transit of cells through G1 phase, consistent with ING1 serving a tumor suppressor function, and caused cells to enter apoptosis more rapidly in response to cisplatin. Our results indicate that malignant gliomas may down-regulate ING1 to allow more efficient tumor growth and progression. Also, ING1 down-regulation may sensitize GBM cells with deficient p53 to treatment with cisplatin.


Asunto(s)
Antineoplásicos/uso terapéutico , Cisplatino/uso terapéutico , Regulación hacia Abajo/genética , Glioblastoma , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Proteínas Nucleares/metabolismo , Proteína p53 Supresora de Tumor/deficiencia , Proteínas Supresoras de Tumor/metabolismo , Bromodesoxiuridina/metabolismo , Ciclo Celular/efectos de los fármacos , Línea Celular Tumoral , Daño del ADN/efectos de los fármacos , Daño del ADN/efectos de la radiación , Regulación hacia Abajo/efectos de los fármacos , Glioblastoma/tratamiento farmacológico , Glioblastoma/genética , Glioblastoma/fisiopatología , Humanos , Proteína Inhibidora del Crecimiento 1 , ARN Interferente Pequeño/farmacología , Radiación , Factores de Tiempo , Transfección , Proteína p53 Supresora de Tumor/genética
15.
Blood ; 107(6): 2536-9, 2006 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-16263788

RESUMEN

Hodgkin/Reed-Sternberg (HRS) cells of classical Hodgkin lymphoma (cHL) display unique characteristics that discriminate cHL from other B-cell lymphomas and normal B cells. Therefore, comparative gene expression profiling of Hodgkin and non-Hodgkin B cells could lead to the identification of candidate genes that are critical for the pathogenesis of cHL. We performed microarray analysis of Hodgkin and non-Hodgkin cell lines and identified activating transcription factor 3 (ATF3), a member of the cyclic AMP response element binding protein (CREB)/ATF family, as a differentially expressed candidate gene. Extensive analysis of a large panel of cell lines, primary tumor samples, and normal tissues revealed that high expression of ATF3 is found in nearly all cases of cHL and is almost exclusively restricted to it. Selective knock-down of ATF3 by RNA interference suppressed proliferation and strongly reduced viability of Hodgkin cells. Thus, overexpression of ATF3 is a molecular hallmark of cHL that contributes to the malignant growth of HRS cells.


Asunto(s)
Factor de Transcripción Activador 3/genética , Regulación Neoplásica de la Expresión Génica , Enfermedad de Hodgkin/genética , Células de Reed-Sternberg/patología , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Supervivencia Celular , Perfilación de la Expresión Génica , Enfermedad de Hodgkin/patología , Humanos , Análisis de Secuencia por Matrices de Oligonucleótidos , ARN Interferente Pequeño/farmacología , Células de Reed-Sternberg/metabolismo
16.
J Biol Chem ; 281(13): 8675-85, 2006 Mar 31.
Artículo en Inglés | MEDLINE | ID: mdl-16446370

RESUMEN

DNA damage induced by the topoisomerase I inhibitor irinotecan (CPT-11) triggers in p53(WT) colorectal carcinoma cells a long term cell cycle arrest and in p53MUT cells a transient arrest followed by apoptosis (Magrini, R., Bhonde, M. R., Hanski, M. L., Notter, M., Scherübl, H., Boland, C. R., Zeitz, M., and Hanski, C. (2002) Int. J. Cancer 101, 23-31; Bhonde, M. R., Hanski, M. L., Notter, M., Gillissen, B. F., Daniel, P. T., Zeitz, M., and Hanski, C. (2006) Oncogene 25, 165-175). The mechanism of the p53-independent apoptosis still remains largely unclear. Here we used five p53WT and five p53MUT established colon carcinoma cell lines to identify gene expression alterations associated with apoptosis in p53MUT cells after treatment with SN-38, the irinotecan metabolite. After treatment, 16 mitosis-related genes were found to be expressed at least 2-fold stronger in the apoptosis-executing p53MUT cells than in the cell cycle-arrested p53WT cells by oligonucleotide microarray analysis. One of the genes whose strong post-treatment expression was associated with apoptosis was the mitotic checkpoint kinase hMps1 (human ortholog of the yeast monopolar spindle 1 kinase). hMps1 mRNA and protein expression were suppressed by the treatment-induced and by the exogenous adenovirus-coded p53 protein. The direct suppression of hMps1 on RNA level or inhibition of its activity by a dominant-negative hMps1 partly suppressed apoptosis. Together, these data indicate that the high expression of mitotic genes in p53MUT cells after SN-38 treatment contributes to DNA damage-induced apoptosis, whereas their suppression in p53WT cells acts as a safeguard mechanism preventing mitosis initiation and the subsequent apoptosis. hMps1 kinase is one of the mitotic checkpoint proteins whose expression after DNA damage in p53MUT cells activates the checkpoint and contributes to apoptosis.


Asunto(s)
Apoptosis , Proteínas de Ciclo Celular/metabolismo , Daño del ADN , Regulación Neoplásica de la Expresión Génica , Proteínas Serina-Treonina Quinasas/metabolismo , Proteína p53 Supresora de Tumor/metabolismo , Antineoplásicos Fitogénicos/farmacología , Western Blotting , Camptotecina/análogos & derivados , Camptotecina/farmacología , Carcinoma/genética , Carcinoma/metabolismo , Carcinoma/patología , Ciclo Celular/efectos de los fármacos , Proteínas de Ciclo Celular/genética , Línea Celular Tumoral , Neoplasias del Colon/genética , Neoplasias del Colon/metabolismo , Neoplasias del Colon/patología , Citometría de Flujo , Células HCT116 , Células HT29 , Células HeLa , Humanos , Irinotecán , Modelos Biológicos , Proteínas de Neoplasias/deficiencia , Proteínas Serina-Treonina Quinasas/genética , Proteínas Tirosina Quinasas , ARN Mensajero/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Proteína p53 Supresora de Tumor/genética
17.
J Gen Virol ; 85(Pt 1): 179-184, 2004 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-14718633

RESUMEN

Small interfering RNAs (siRNAs) are the mediators of a sequence-specific process of gene silencing called RNA interference (RNAi). Here, we show that synthetic siRNAs against essential gene products of human cytomegalovirus (HCMV) can trigger RNAi in serum-starved, infected primary fibroblasts, as well as in U373 cells, leading to effective inhibition of viral DNA replication. This opens new possibilities for antiviral strategies and for the analysis of viral and cellular genes important to HCMV physiology.


Asunto(s)
Citomegalovirus/fisiología , Interferencia de ARN , ARN Interferente Pequeño/farmacología , Proteínas Virales , Replicación Viral/efectos de los fármacos , Línea Celular , Células Cultivadas , ADN Polimerasa Dirigida por ADN/genética , ADN Polimerasa Dirigida por ADN/metabolismo , Regulación hacia Abajo , Fibroblastos/virología , Regulación Viral de la Expresión Génica , Humanos , ARN Interferente Pequeño/metabolismo , Transfección
18.
J Virol ; 78(21): 11853-64, 2004 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-15479827

RESUMEN

The IE2 86-kDa gene product is an essential regulatory protein of human cytomegalovirus (HCMV) with several functions, including transactivation, negative autoregulation, and cell cycle regulation. In order to understand the physiological significance of each of the IE2 functions, discriminating mutants of IE2 are required that can be tested in a viral background. However, no such mutants of IE2 are available, possibly reflecting structural peculiarities of the large and ill-defined C-terminal domain of IE2. Here, we revisited the C-terminal domain by analyzing IE2 mutants for transactivation, DNA binding, autoregulation, and cell cycle regulation in parallel. We found it to contain an unexpectedly concise core domain (amino acids 450 to 544) that is defined by its absolute sensitivity to any kind of mutation. In contrast, the region adjacent to the core (amino acids 290 to 449) generally tolerates mutations much better. Although it contributes more specific sequence information to distinct IE2 activities, none of the mutations analyzed abolished any particular function. The core is demarcated from the adjacent region by the putative zinc finger region (amino acids 428 to 452). Surprisingly, the deletion of the putative zinc finger region from IE2 revealed that this region is entirely dispensable for any of the IE2 functions tested here in transfection assays. Our work supports the view that the 100 amino acids of the core domain hold the key to most functions of IE2. A systematic, high-density mutational analysis of this region may identify informative mutants discriminating between various IE2 functions that can then be tested in a viral background.


Asunto(s)
ADN/metabolismo , Proteínas Inmediatas-Precoces/química , Transactivadores/química , Dedos de Zinc , Secuencia de Aminoácidos , Ciclo Celular , Humanos , Proteínas Inmediatas-Precoces/fisiología , Datos de Secuencia Molecular , Proteínas Represoras/química , Proteínas Represoras/fisiología , Relación Estructura-Actividad , Transactivadores/fisiología , Activación Transcripcional
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA