Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 36
Filtrar
Más filtros

Bases de datos
Tipo del documento
Intervalo de año de publicación
1.
Mol Cell ; 73(1): 166-182.e7, 2019 01 03.
Artículo en Inglés | MEDLINE | ID: mdl-30609389

RESUMEN

Subcellular localization is a main determinant of protein function; however, a global view of cellular proteome organization remains relatively unexplored. We have developed a robust mass spectrometry-based analysis pipeline to generate a proteome-wide view of subcellular localization for proteins mapping to 12,418 individual genes across five cell lines. Based on more than 83,000 unique classifications and correlation profiling, we investigate the effect of alternative splicing and protein domains on localization, complex member co-localization, cell-type-specific localization, as well as protein relocalization after growth factor inhibition. Our analysis provides information about the cellular architecture and complexity of the spatial organization of the proteome; we show that the majority of proteins have a single main subcellular location, that alternative splicing rarely affects subcellular location, and that cell types are best distinguished by expression of proteins exposed to the surrounding environment. The resource is freely accessible via www.subcellbarcode.org.


Asunto(s)
Cromatografía Liquida , Espectrometría de Masas , Proteínas/metabolismo , Proteoma , Proteómica/métodos , Fracciones Subcelulares/metabolismo , Biomarcadores/metabolismo , Fraccionamiento Celular , Biología Computacional , Receptores ErbB/antagonistas & inhibidores , Receptores ErbB/metabolismo , Gefitinib/farmacología , Humanos , Focalización Isoeléctrica , Células MCF-7 , Inhibidores de Proteínas Quinasas/farmacología , Transporte de Proteínas , Proteínas/antagonistas & inhibidores , Proteínas/clasificación , Proteínas/genética , Reproducibilidad de los Resultados , Fracciones Subcelulares/clasificación , Fracciones Subcelulares/efectos de los fármacos
2.
Mol Cell ; 59(6): 984-97, 2015 Sep 17.
Artículo en Inglés | MEDLINE | ID: mdl-26321255

RESUMEN

Transcriptionally active and inactive chromatin domains tend to segregate into separate sub-nuclear compartments to maintain stable expression patterns. However, here we uncovered an inter-chromosomal network connecting active loci enriched in circadian genes to repressed lamina-associated domains (LADs). The interactome is regulated by PARP1 and its co-factor CTCF. They not only mediate chromatin fiber interactions but also promote the recruitment of circadian genes to the lamina. Synchronization of the circadian rhythm by serum shock induces oscillations in PARP1-CTCF interactions, which is accompanied by oscillating recruitment of circadian loci to the lamina, followed by the acquisition of repressive H3K9me2 marks and transcriptional attenuation. Furthermore, depletion of H3K9me2/3, inhibition of PARP activity by olaparib, or downregulation of PARP1 or CTCF expression counteracts both recruitment to the envelope and circadian transcription. PARP1- and CTCF-regulated contacts between circadian loci and the repressive chromatin environment at the lamina therefore mediate circadian transcriptional plasticity.


Asunto(s)
Cromatina/genética , Células Madre Embrionarias Humanas/enzimología , Poli(ADP-Ribosa) Polimerasas/metabolismo , Proteínas Represoras/metabolismo , Transcripción Genética , Proteínas Adaptadoras Transductoras de Señales , Factor de Unión a CCCTC , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Inmunoprecipitación de Cromatina , Ritmo Circadiano , Cuerpos Embrioides/enzimología , Epistasis Genética , Regulación de la Expresión Génica , Redes Reguladoras de Genes , Células HCT116 , Humanos , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Lámina Nuclear/metabolismo , Poli(ADP-Ribosa) Polimerasa-1 , Unión Proteica , ARN Largo no Codificante/genética , ARN Largo no Codificante/metabolismo
3.
Nature ; 517(7535): 489-92, 2015 Jan 22.
Artículo en Inglés | MEDLINE | ID: mdl-25363767

RESUMEN

Next-generation sequencing of human tumours has refined our understanding of the mutational processes operative in cancer initiation and progression, yet major questions remain regarding the factors that induce driver mutations and the processes that shape mutation selection during tumorigenesis. Here we performed whole-exome sequencing on adenomas from three mouse models of non-small-cell lung cancer, which were induced either by exposure to carcinogens (methyl-nitrosourea (MNU) and urethane) or by genetic activation of Kras (Kras(LA2)). Although the MNU-induced tumours carried exactly the same initiating mutation in Kras as seen in the Kras(LA2) model (G12D), MNU tumours had an average of 192 non-synonymous, somatic single-nucleotide variants, compared with only six in tumours from the Kras(LA2) model. By contrast, the Kras(LA2) tumours exhibited a significantly higher level of aneuploidy and copy number alterations compared with the carcinogen-induced tumours, suggesting that carcinogen-induced and genetically engineered models lead to tumour development through different routes. The wild-type allele of Kras has been shown to act as a tumour suppressor in mouse models of non-small-cell lung cancer. We demonstrate that urethane-induced tumours from wild-type mice carry mostly (94%) Kras Q61R mutations, whereas those from Kras heterozygous animals carry mostly (92%) Kras Q61L mutations, indicating a major role for germline Kras status in mutation selection during initiation. The exome-wide mutation spectra in carcinogen-induced tumours overwhelmingly display signatures of the initiating carcinogen, while adenocarcinomas acquire additional C > T mutations at CpG sites. These data provide a basis for understanding results from human tumour genome sequencing, which has identified two broad categories of tumours based on the relative frequency of single-nucleotide variations and copy number alterations, and underline the importance of carcinogen models for understanding the complex mutation spectra seen in human cancers.


Asunto(s)
Transformación Celular Neoplásica/inducido químicamente , Transformación Celular Neoplásica/genética , Genes ras/genética , Neoplasias Pulmonares/inducido químicamente , Neoplasias Pulmonares/genética , Mutación/genética , Proteína Oncogénica p21(ras)/genética , Proteínas Proto-Oncogénicas p21(ras)/genética , Adenocarcinoma/inducido químicamente , Adenocarcinoma/genética , Animales , Carcinógenos/toxicidad , Carcinoma de Pulmón de Células no Pequeñas/inducido químicamente , Carcinoma de Pulmón de Células no Pequeñas/genética , Variaciones en el Número de Copia de ADN/genética , Progresión de la Enfermedad , Femenino , Inestabilidad Genómica/genética , Mutación de Línea Germinal/genética , Humanos , Masculino , Metilnitrosourea/toxicidad , Ratones , Modelos Genéticos , Mutación Puntual/genética , Uretano/toxicidad
4.
Int J Cancer ; 146(4): 1125-1138, 2020 02 15.
Artículo en Inglés | MEDLINE | ID: mdl-31219615

RESUMEN

Increased expression of GLI1, the main Hedgehog signalling pathway effector, is related to unfavourable prognosis and progressive disease of certain breast cancer subtypes. We used conditional transgenic mice induced to overexpress GLI1 in the mammary epithelium either alone or in combination with deletion of one Trp53 allele to address the role of elevated GLI1 expression in breast tumour initiation and progression. Induced GLI1 expression facilitates mammary gland tumour formation and this was further increased upon heterozygous deletion of Trp53. The GLI1-induced primary tumours were of different murine molecular subtypes, including Normal-likeEx , Class8Ex , Claudin-LowEx and Erbb2-likeEx . The gene expression profiles of some of the tumours correlated well with the PAM50 subtypes for human breast cancer. Whole-exome sequencing revealed somatic mutation profiles with only little overlap between the primary tumours. Orthotopically serially transplanted GLI1-induced tumours maintained the main morphological characteristics of the primary tumours for ≥10 generations. Independent of Trp53 status and molecular subtype, the serially transplanted GLI1-induced tumours were able to grow both in the absence of transgenic GLI1 expression and in the presence of the GLI1 inhibitor GANT61. These data suggest that elevated GLI1 expression has a determinant role in tumour initiation; however, additional genetic events are required for tumour progression.


Asunto(s)
Neoplasias Mamarias Experimentales/genética , Neoplasias Mamarias Experimentales/patología , Proteína con Dedos de Zinc GLI1/genética , Animales , Femenino , Expresión Génica , Heterogeneidad Genética , Humanos , Inmunohistoquímica , Masculino , Ratones , Ratones Transgénicos , Trasplante de Neoplasias , Proteína con Dedos de Zinc GLI1/biosíntesis
5.
Proc Natl Acad Sci U S A ; 113(38): E5618-27, 2016 09 20.
Artículo en Inglés | MEDLINE | ID: mdl-27608497

RESUMEN

Vascular pericytes, an important cellular component in the tumor microenvironment, are often associated with tumor vasculatures, and their functions in cancer invasion and metastasis are poorly understood. Here we show that PDGF-BB induces pericyte-fibroblast transition (PFT), which significantly contributes to tumor invasion and metastasis. Gain- and loss-of-function experiments demonstrate that PDGF-BB-PDGFRß signaling promotes PFT both in vitro and in in vivo tumors. Genome-wide expression analysis indicates that PDGF-BB-activated pericytes acquire mesenchymal progenitor features. Pharmacological inhibition and genetic deletion of PDGFRß ablate the PDGF-BB-induced PFT. Genetic tracing of pericytes with two independent mouse strains, TN-AP-CreERT2:R26R-tdTomato and NG2-CreERT2:R26R-tdTomato, shows that PFT cells gain stromal fibroblast and myofibroblast markers in tumors. Importantly, coimplantation of PFT cells with less-invasive tumor cells in mice markedly promotes tumor dissemination and invasion, leading to an increased number of circulating tumor cells and metastasis. Our findings reveal a mechanism of vascular pericytes in PDGF-BB-promoted cancer invasion and metastasis by inducing PFT, and thus targeting PFT may offer a new treatment option of cancer metastasis.


Asunto(s)
Carcinoma de Células Renales/genética , Pericitos/metabolismo , Proteínas Proto-Oncogénicas c-sis/genética , Receptor beta de Factor de Crecimiento Derivado de Plaquetas/genética , Animales , Becaplermina , Carcinoma de Células Renales/metabolismo , Carcinoma de Células Renales/patología , Línea Celular Tumoral , Fibroblastos/metabolismo , Fibroblastos/patología , Humanos , Ratones , Ratones Noqueados , Metástasis de la Neoplasia , Neovascularización Patológica/genética , Neovascularización Patológica/patología , Pericitos/patología , Proteínas Proto-Oncogénicas c-sis/metabolismo , Receptor beta de Factor de Crecimiento Derivado de Plaquetas/antagonistas & inhibidores , Microambiente Tumoral/genética , Ensayos Antitumor por Modelo de Xenoinjerto
6.
Mol Cell ; 40(5): 762-73, 2010 Dec 10.
Artículo en Inglés | MEDLINE | ID: mdl-21145484

RESUMEN

The miR-17-92 microRNA cluster is often activated in cancer cells, but the identity of its targets remains elusive. Using SILAC and quantitative mass spectrometry, we examined the effects of activation of the miR-17-92 cluster on global protein expression in neuroblastoma (NB) cells. Our results reveal cooperation between individual miR-17-92 miRNAs and implicate miR-17-92 in multiple hallmarks of cancer, including proliferation and cell adhesion. Most importantly, we show that miR-17-92 is a potent inhibitor of TGF-ß signaling. By functioning both upstream and downstream of pSMAD2, miR-17-92 activation triggers downregulation of multiple key effectors along the TGF-ß signaling cascade as well as direct inhibition of TGF-ß-responsive genes.


Asunto(s)
MicroARNs/genética , Neuroblastoma/metabolismo , Factor de Crecimiento Transformador beta/metabolismo , Animales , Adhesión Celular , Línea Celular , Proliferación Celular , Ratones , Ratones Desnudos , MicroARNs/metabolismo , Neuroblastoma/genética , Proteína Smad2/genética , Proteína Smad2/metabolismo , Factor de Crecimiento Transformador beta/genética , Trasplante Heterólogo
7.
Acta Orthop ; 88(1): 35-40, 2017 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-27892748

RESUMEN

Background and purpose - Porous tantalum cups have been introduced as an alternative to various reinforcement rings in revision hip surgery. We hypothesized that porous tantalum cups would be superior to Müller acetabular roof reinforcement rings (MARRs) in revision hip surgery with re-revision for aseptic loosening as the primary outcome measure. Patients and methods - 207 hips operated with either a porous tantalum cup (TM cup, n = 111) or a MARR (n = 96) at index procedure were identified in our local arthroplasty register. Acetabular defects were classified according to Paprosky. There were 96 men and 111 women with a median age of 71 (35-95) years, presenting acetabular defect size type I in 39 cases, IIA in 22, IIB in 27, IIC in 43, IIIA in 32, and IIIB in 37 cases. Analysis of medical records identified all patients with subsequent re-revision and reasons for re-revisions. Kaplan-Meier survival functions were used to estimate implant survival. Results - With re-revision for aseptic loosening as the endpoint, the 6-year unadjusted cumulative survival was 97% (95% CI: 94-100) for TM cups and 96% (CI: 92-100) for MARR (p = 0.6). Using re-revision for any reason as the endpoint, 6-year survival was 87% (CI: 81-94) for TM cups and 95% (CI: 90-99) for MARR (p = 0.06). The main reason for re-revision in the TM group was dislocation (n = 10), followed by loosening (n = 3), whereas the main reason for re-revision in the MARR group was aseptic loosening (n = 8). Duration of the index procedure and perioperative blood loss were lower in the TM group. Interpretation - Both TM and MARR lead to good 6-year results in acetabular revision surgery. The methods differ in their respective failure mechanisms. We conclude that TM cups are a valuable treatment option in acetabular revision surgery, but the reasons underlying dislocations after the use of TM cups must be analyzed further.


Asunto(s)
Acetábulo/cirugía , Artroplastia de Reemplazo de Cadera/instrumentación , Prótesis de Cadera , Sistema de Registros , Tantalio , Adulto , Anciano , Anciano de 80 o más Años , Femenino , Estudios de Seguimiento , Humanos , Masculino , Persona de Mediana Edad , Porosidad , Complicaciones Posoperatorias , Diseño de Prótesis , Reoperación , Estudios Retrospectivos , Resultado del Tratamiento
8.
Proc Natl Acad Sci U S A ; 110(18): 7312-7, 2013 Apr 30.
Artículo en Inglés | MEDLINE | ID: mdl-23589885

RESUMEN

Hypoxia, or low oxygen tension, is a major regulator of tumor development and aggressiveness. However, how cancer cells adapt to hypoxia and communicate with their surrounding microenvironment during tumor development remain important questions. Here, we show that secreted vesicles with exosome characteristics mediate hypoxia-dependent intercellular signaling of the highly malignant brain tumor glioblastoma multiforme (GBM). In vitro hypoxia experiments with glioma cells and studies with patient materials reveal the enrichment in exosomes of hypoxia-regulated mRNAs and proteins (e.g., matrix metalloproteinases, IL-8, PDGFs, caveolin 1, and lysyl oxidase), several of which were associated with poor glioma patient prognosis. We show that exosomes derived from GBM cells grown at hypoxic compared with normoxic conditions are potent inducers of angiogenesis ex vivo and in vitro through phenotypic modulation of endothelial cells. Interestingly, endothelial cells were programmed by GBM cell-derived hypoxic exosomes to secrete several potent growth factors and cytokines and to stimulate pericyte PI3K/AKT signaling activation and migration. Moreover, exosomes derived from hypoxic compared with normoxic conditions showed increased autocrine, promigratory activation of GBM cells. These findings were correlated with significantly enhanced induction by hypoxic compared with normoxic exosomes of tumor vascularization, pericyte vessel coverage, GBM cell proliferation, as well as decreased tumor hypoxia in a mouse xenograft model. We conclude that the proteome and mRNA profiles of exosome vesicles closely reflect the oxygenation status of donor glioma cells and patient tumors, and that the exosomal pathway constitutes a potentially targetable driver of hypoxia-dependent intercellular signaling during tumor development.


Asunto(s)
Vasos Sanguíneos/patología , Neoplasias Encefálicas/irrigación sanguínea , Neoplasias Encefálicas/patología , Transformación Celular Neoplásica/patología , Exosomas/metabolismo , Glioma/irrigación sanguínea , Glioma/patología , Animales , Comunicación Autocrina , Neoplasias Encefálicas/genética , Hipoxia de la Célula/genética , Línea Celular Tumoral , Transformación Celular Neoplásica/genética , Células Endoteliales/metabolismo , Células Endoteliales/patología , Femenino , Regulación Neoplásica de la Expresión Génica , Glioma/genética , Humanos , Ratones , Ratones SCID , Proteínas de Neoplasias/metabolismo , Neovascularización Patológica/genética , Neovascularización Patológica/patología , Comunicación Paracrina , Pericitos/metabolismo , Pericitos/patología , Proteoma/metabolismo , Transducción de Señal/genética , Donantes de Tejidos , Transcriptoma/genética , Ensayos Antitumor por Modelo de Xenoinjerto
9.
BMC Cancer ; 15: 311, 2015 Apr 25.
Artículo en Inglés | MEDLINE | ID: mdl-25907971

RESUMEN

BACKGROUND: Deregulated Notch signaling is linked to a variety of tumors and it is therefore important to learn more about the frequency and distribution of Notch mutations in a tumor context. METHODS: In this report, we use data from the recently developed Cancer Cell Line Encyclopedia to assess the frequency and distribution of Notch mutations in a large panel of cancer cell lines in silico. RESULTS: Our results show that the mutation frequency of Notch receptor and ligand genes is at par with that for established oncogenes and higher than for a set of house-keeping genes. Mutations were found across all four Notch receptor genes, but with notable differences between protein domains, mutations were for example more prevalent in the regions encoding the LNR and PEST domains in the Notch intracellular domain. Furthermore, an in silico estimation of functional impact showed that deleterious mutations cluster to the ligand-binding and the intracellular domains of NOTCH1. For most cell line groups, the mutation frequency of Notch genes is higher than in associated primary tumors. CONCLUSIONS: Our results shed new light on the spectrum of Notch mutations after in vitro culturing of tumor cells. The higher mutation frequency in tumor cell lines indicates that Notch mutations are associated with a growth advantage in vitro, and thus may be considered to be driver mutations in a tumor cell line context.


Asunto(s)
Frecuencia de los Genes , Receptores Notch/genética , Línea Celular Tumoral , Variaciones en el Número de Copia de ADN , Análisis Mutacional de ADN , Genes Esenciales , Humanos , Mutación Missense
10.
Cancer Cell ; 10(5): 413-23, 2006 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-17097563

RESUMEN

In neuroblastoma specimens, HIF-2alpha but not HIF-1alpha is strongly expressed in well-vascularized areas. In vitro, HIF-2alpha protein was stabilized at 5% O2 (resembling end capillary oxygen conditions) and, in contrast to the low HIF-1alpha activity at this oxygen level, actively transcribed genes like VEGF. Under hypoxia (1% O2), HIF-1alpha was transiently stabilized and primarily mediated acute responses, whereas HIF-2alpha protein gradually accumulated and governed prolonged hypoxic gene activation. Knockdown of HIF-2alpha reduced growth of neuroblastoma tumors in athymic mice. Furthermore, high HIF-2alpha protein levels were correlated with advanced clinical stage and high VEGF expression and predicted poor prognosis in a clinical neuroblastoma material. Our results demonstrate the relevance of HIF-2alpha in neuroblastoma progression and have general tumor biological implications.


Asunto(s)
Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Regulación Neoplásica de la Expresión Génica , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Neuroblastoma/metabolismo , Animales , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Niño , Femenino , Perfilación de la Expresión Génica , Humanos , Hipoxia , Subunidad alfa del Factor 1 Inducible por Hipoxia/genética , Ratones , Trasplante de Neoplasias , Neuroblastoma/genética , Neuroblastoma/patología , Análisis de Secuencia por Matrices de Oligonucleótidos , Oxígeno/metabolismo , Fenotipo , Procolágeno-Prolina Dioxigenasa/genética , Procolágeno-Prolina Dioxigenasa/metabolismo , ARN Mensajero/metabolismo , Activación Transcripcional , Células Tumorales Cultivadas
11.
Nucleic Acids Res ; 39(20): e136, 2011 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-21835775

RESUMEN

While a growing body of evidence implicates regulatory miRNA modules in various aspects of human disease and development, insights into specific miRNA function remain limited. Here, we present an innovative approach to elucidate tissue-specific miRNA functions that goes beyond miRNA target prediction and expression correlation. This approach is based on a multi-level integration of corresponding miRNA and mRNA gene expression levels, miRNA target prediction, transcription factor target prediction and mechanistic models of gene network regulation. Predicted miRNA functions were either validated experimentally or compared to published data. The predicted miRNA functions are accessible in the miRNA bodymap, an interactive online compendium and mining tool of high-dimensional newly generated and published miRNA expression profiles. The miRNA bodymap enables prioritization of candidate miRNAs based on their expression pattern or functional annotation across tissue or disease subgroup. The miRNA bodymap project provides users with a single one-stop data-mining solution and has great potential to become a community resource.


Asunto(s)
MicroARNs/metabolismo , Programas Informáticos , Animales , Línea Celular Tumoral , Minería de Datos , Perfilación de la Expresión Génica , Regulación de la Expresión Génica , Redes Reguladoras de Genes , Genómica , Humanos , Ratones , Modelos Genéticos , Anotación de Secuencia Molecular , ARN Mensajero/metabolismo , Ratas , Factores de Transcripción/metabolismo
12.
Proc Natl Acad Sci U S A ; 107(4): 1553-8, 2010 Jan 26.
Artículo en Inglés | MEDLINE | ID: mdl-20080637

RESUMEN

MYCN, a proto-oncogene normally expressed in the migrating neural crest, is in its amplified state a key factor in the genesis of human neuroblastoma (NB). However, the mechanisms underlying MYCN-mediated NB progression are poorly understood. Here, we present a MYCN-induced miRNA signature in human NB involving the activation and transrepression of several miRNA genes from paralogous clusters. Several family members derived from the miR-17 approximately 92 cluster, including miR-18a and miR-19a, were among the up-regulated miRNAs. Expression analysis of these miRNAs in NB tumors confirmed increased levels in MYCN-amplified samples. Specifically, we show that miR-18a and miR-19a target and repress the expression of estrogen receptor-alpha (ESR1), a ligand-inducible transcription factor implicated in neuronal differentiation. Immunohistochemical staining demonstrated ESR1 expression in human fetal sympathetic ganglia, suggesting a role for ESR1 during sympathetic nervous system development. Concordantly, lentiviral restoration of ESR1 in NB cells resulted in growth arrest and neuronal differentiation. Moreover, lentiviral-mediated inhibition of miR-18a in NB cells led to severe growth retardation, outgrowth of varicosity-containing neurites, and induction of neuronal sympathetic differentiation markers. Bioinformatic analyses of microarray data from NB tumors revealed that high ESR1 expression correlates with increased event-free survival in NB patients and favorable disease outcome. Thus, MYCN amplification may disrupt estrogen signaling sensitivity in primitive sympathetic cells through deregulation of ESR1, thereby preventing the normal induction of neuroblast differentiation. Collectively, our findings demonstrate the molecular consequences of abnormal miRNA transcription in a MYCN-driven tumor and offer unique insights into the pathology underlying MYCN-amplified NB.


Asunto(s)
Diferenciación Celular , Receptor alfa de Estrógeno/metabolismo , Regulación de la Expresión Génica , MicroARNs/genética , Neuroblastoma/metabolismo , Neuroblastoma/patología , Proteínas Nucleares/metabolismo , Proteínas Oncogénicas/metabolismo , Regiones no Traducidas 3' , Línea Celular , Línea Celular Tumoral , Proliferación Celular , Receptor alfa de Estrógeno/genética , Humanos , Proteína Proto-Oncogénica N-Myc , Neuroblastoma/genética , Neuronas/citología , Neuronas/metabolismo , Proteínas Nucleares/genética , Proteínas Oncogénicas/genética , Proto-Oncogenes Mas , Transducción de Señal , Transcripción Genética
13.
Breast Cancer Res ; 14(4): R113, 2012 Jul 27.
Artículo en Inglés | MEDLINE | ID: mdl-22839103

RESUMEN

INTRODUCTION: Gene expression data derived from clinical cancer specimens provide an opportunity to characterize cancer-specific transcriptional programs. Here, we present an analysis delineating a correlation-based gene expression landscape of breast cancer that identifies modules with strong associations to breast cancer-specific and general tumor biology. METHODS: Modules of highly connected genes were extracted from a gene co-expression network that was constructed based on Pearson correlation, and module activities were then calculated using a pathway activity score. Functional annotations of modules were experimentally validated with an siRNA cell spot microarray system using the KPL-4 breast cancer cell line, and by using gene expression data from functional studies. Modules were derived using gene expression data representing 1,608 breast cancer samples and validated in data sets representing 971 independent breast cancer samples as well as 1,231 samples from other cancer forms. RESULTS: The initial co-expression network analysis resulted in the characterization of eight tightly regulated gene modules. Cell cycle genes were divided into two transcriptional programs, and experimental validation using an siRNA screen showed different functional roles for these programs during proliferation. The division of the two programs was found to act as a marker for tumor protein p53 (TP53) gene status in luminal breast cancer, with the two programs being separated only in luminal tumors with functional p53 (encoded by TP53). Moreover, a module containing fibroblast and stroma-related genes was highly expressed in fibroblasts, but was also up-regulated by overexpression of epithelial-mesenchymal transition factors such as transforming growth factor beta 1 (TGF-beta1) and Snail in immortalized human mammary epithelial cells. Strikingly, the stroma transcriptional program related to less malignant tumors for luminal disease and aggressive lymph node positive disease among basal-like tumors. CONCLUSIONS: We have derived a robust gene expression landscape of breast cancer that reflects known subtypes as well as heterogeneity within these subtypes. By applying the modules to TP53-mutated samples we shed light on the biological consequences of non-functional p53 in otherwise low-proliferating luminal breast cancer. Furthermore, as in the case of the stroma module, we show that the biological and clinical interpretation of a set of co-regulated genes is subtype-dependent.


Asunto(s)
Neoplasias de la Mama/genética , Transición Epitelial-Mesenquimal/genética , Regulación Neoplásica de la Expresión Génica , Proteína p53 Supresora de Tumor/genética , Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , Ciclo Celular/genética , Análisis por Conglomerados , Conjuntos de Datos como Asunto , Femenino , Perfilación de la Expresión Génica , Redes Reguladoras de Genes , Humanos , Células del Estroma/metabolismo , Transcriptoma
14.
Proc Natl Acad Sci U S A ; 106(39): 16805-10, 2009 Sep 29.
Artículo en Inglés | MEDLINE | ID: mdl-19805377

RESUMEN

High hypoxia-inducible factor-2alpha (HIF-2alpha) protein levels predict poor outcome in neuroblastoma, and hypoxia dedifferentiates cultured neuroblastoma cells toward a neural crest-like phenotype. Here, we identify HIF-2alpha as a marker of normoxic neural crest-like neuroblastoma tumor-initiating/stem cells (TICs) isolated from patient bone marrows. Knockdown of HIF-2alpha reduced VEGF expression and induced partial sympathetic neuronal differentiation when these TICs were grown in vitro under stem cell-promoting conditions. Xenograft tumors of HIF-2alpha-silenced cells were widely necrotic, poorly vascularized, and resembled the bulk of tumor cells in clinical neuroblastomas by expressing additional sympathetic neuronal markers, whereas control tumors were immature, well-vascularized, and stroma-rich. Thus, HIF-2alpha maintains an undifferentiated state of neuroblastoma TICs. Because low differentiation is associated with poor outcome and angiogenesis is crucial for tumor growth, HIF-2alpha is an attractive target for neuroblastoma therapy.


Asunto(s)
Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Diferenciación Celular , Cresta Neural/metabolismo , Neuroblastoma/metabolismo , Animales , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Hipoxia de la Célula , Línea Celular Tumoral , Regulación hacia Abajo , Femenino , Humanos , Ratones , Ratones Desnudos , Factores de Crecimiento Endotelial Vascular/genética , Factores de Crecimiento Endotelial Vascular/metabolismo
15.
J Proteome Res ; 10(4): 1645-56, 2011 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-21235201

RESUMEN

Nontransient hypoxia is strongly associated with malignant lesions, resulting in aggressive behavior and resistance to treatment. We present an analysis of mRNA and protein expression changes in neuroblastoma cell lines occurring upon the transition from normoxia to hypoxia. The correlation between mRNA and protein level changes was poor, although some known hypoxia-driven genes and proteins correlated well. We present previously undescribed membrane proteins expressed under hypoxic conditions that are candidates for evaluation as biomarkers.


Asunto(s)
Biomarcadores/química , Membrana Celular/química , Hipoxia/metabolismo , Proteínas de la Membrana/química , Biomarcadores/metabolismo , Línea Celular Tumoral , Humanos , Espectrometría de Masas/métodos , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Análisis por Micromatrices , Neoplasias/química , Neoplasias/metabolismo , Neoplasias/patología , ARN Mensajero/metabolismo , Electroforesis Bidimensional Diferencial en Gel/métodos
16.
Blood ; 114(4): 844-59, 2009 Jul 23.
Artículo en Inglés | MEDLINE | ID: mdl-19454749

RESUMEN

Ischemia exists in many diseased tissues, including arthritic joints, atherosclerotic plaques, and malignant tumors. Macrophages accumulate in these sites and up-regulate hypoxia-inducible transcription factors (HIFs) 1 and 2 in response to the hypoxia present. Here we show that the gene expression profile in primary human and murine macrophages changes markedly when they are exposed to hypoxia for 18 hours. For example, they were seen to up-regulate the cell surface receptors, CXCR4 and GLUT1, and the potent, tumor-promoting cytokines, vascular endothelial growth factor A, interleukin (IL)-1beta and IL-8, adrenomedullin, CXCR4, and angiopoietin-2. Hypoxia also stimulated their expression and/or phosphorylation of various proteins in the nuclear factor-kappaB (NF-kappaB) signaling pathway. We then used both genetic and pharmacologic methods to manipulate the levels of HIFs-1alpha and 2alpha or NF-kappaB in primary macrophages to elucidate their role in the hypoxic induction of many of these key genes. These studies showed that both HIF-1 and -2, but not NF-kappaB, are important transcriptional effectors regulating the responses of macrophages to such a period of hypoxia. Further studies using experimental mouse models are now warranted to investigate the role of such macrophage responses in the progression of various diseased tissues, such as malignant tumors.


Asunto(s)
Factor 1 Inducible por Hipoxia/fisiología , Macrófagos/fisiología , Factores de Transcripción/fisiología , Animales , Técnicas de Cultivo de Célula , Hipoxia de la Célula/genética , Células Cultivadas , Femenino , Perfilación de la Expresión Génica , Regulación de la Expresión Génica , Humanos , Factor 1 Inducible por Hipoxia/genética , Factor 1 Inducible por Hipoxia/metabolismo , Macrófagos/metabolismo , Ratones , Ratones Transgénicos , FN-kappa B/fisiología , Análisis de Secuencia por Matrices de Oligonucleótidos , Factores de Transcripción/genética , Factores de Transcripción/metabolismo
17.
Proc Natl Acad Sci U S A ; 105(37): 14094-9, 2008 Sep 16.
Artículo en Inglés | MEDLINE | ID: mdl-18780787

RESUMEN

The childhood cancer neuroblastoma arises in the developing sympathetic nervous system and is a genotypically and phenotypically heterogeneous disease. Prognostic markers of poor survival probability include amplification of the MYCN oncogene and an undifferentiated morphology. Whereas these features discriminate high- from low-risk patients with precision, identification of poor outcome low- and intermediate-risk patients is more challenging. In this study, we analyze two large neuroblastoma microarray datasets using a priori-defined gene expression signatures. We show that differential overexpression of Myc transcriptional targets and low expression of genes involved in sympathetic neuronal differentiation predicts relapse and death from disease. This was evident not only for high-risk patients but was also robust in identifying groups of poor prognosis patients who were otherwise judged to be at low- or intermediate-risk for adverse outcome. These data suggest that pathway-specific gene expression profiling might be useful in the clinic to adjust treatment strategies for children with neuroblastoma.


Asunto(s)
Diferenciación Celular , Neuroblastoma/metabolismo , Neuroblastoma/patología , Proteínas Proto-Oncogénicas c-myc/metabolismo , Transducción de Señal , Estudios de Seguimiento , Amplificación de Genes , Perfilación de la Expresión Génica , Humanos , Estadificación de Neoplasias , Neuroblastoma/genética , Neuroblastoma/terapia , Proteínas Proto-Oncogénicas c-myc/genética , Factores de Riesgo , Resultado del Tratamiento , Regulación hacia Arriba
18.
Exp Cell Res ; 315(11): 1924-36, 2009 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-19254710

RESUMEN

Adaptation to low oxygen conditions is essential for maintaining homeostasis and viability in oxygen-consuming multi-cellular tissues, including solid tumors. Central in these processes are the hypoxia-inducible transcription factors, HIF-1 and HIF-2, controlling genes involved in e.g. glucose metabolism and neovascularization. Tumor hypoxia and HIF expression have also been associated with a dedifferentiated phenotype and increased aggressiveness. In this report we show that the MAX interactor-1 (MXI1) gene is directly regulated by HIF proteins in neuroblastoma and breast cancer cells. HIF-binding and transactivation were detected within MXI1 gene regulatory sequences in the vicinity of the MXI1-0 promoter, leading to rapid induction of the alternate MXI1-0 isoform followed by a long-term induction of both the MXI1-0 and MXI1 isoforms. Importantly, knock-down of MXI1 had limited effect on MYC/MYCN activity under hypoxia, an observation that might be related to the different functional attributes of the two MXI1 isoforms.


Asunto(s)
Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/biosíntesis , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Neuroblastoma/genética , Neuroblastoma/metabolismo , Proteínas Supresoras de Tumor/biosíntesis , Proteínas Supresoras de Tumor/genética , Secuencia de Bases , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/antagonistas & inhibidores , Neoplasias de la Mama/genética , Neoplasias de la Mama/metabolismo , Hipoxia de la Célula/genética , Hipoxia de la Célula/fisiología , Línea Celular Tumoral , Femenino , Regulación Neoplásica de la Expresión Génica , Genes Supresores de Tumor , Genes myc , Humanos , Subunidad alfa del Factor 1 Inducible por Hipoxia/antagonistas & inhibidores , Subunidad alfa del Factor 1 Inducible por Hipoxia/genética , Análisis de Secuencia por Matrices de Oligonucleótidos , Regiones Promotoras Genéticas , ARN Interferente Pequeño/genética , Transfección , Proteínas Supresoras de Tumor/antagonistas & inhibidores , Regulación hacia Arriba
19.
Elife ; 92020 07 06.
Artículo en Inglés | MEDLINE | ID: mdl-32628111

RESUMEN

Inhibition of WEE1 kinase by AZD1775 has shown promising results in clinical cancer trials, but markers predicting AZD1775 response are lacking. Here we analysed AZD1775 response in a panel of human breast cancer (BC) cell lines by global proteome/transcriptome profiling and identified two groups of basal-like BC (BLBCs): 'PTEN low' BLBCs were highly sensitive to AZD1775 and failed to recover following removal of AZD1775, while 'PTEN high' BLBCs recovered. AZD1775 induced phosphorylation of DNA-PK, protecting cells from replication-associated DNA damage and promoting cellular recovery. Deletion of DNA-PK or PTEN, or inhibition of DNA-PK sensitized recovering BLBCs to AZD1775 by abrogating replication arrest, allowing replication despite DNA damage. This was linked to reduced CHK1 activation, increased cyclin E levels and apoptosis. In conclusion, we identified PTEN and DNA-PK as essential regulators of replication checkpoint arrest in response to AZD1775 and defined PTEN as a promising biomarker for efficient WEE1 cancer therapy.


Asunto(s)
Antineoplásicos/farmacología , Neoplasias de la Mama/tratamiento farmacológico , Proteínas de Ciclo Celular/genética , Proteína Quinasa Activada por ADN/genética , Fosfohidrolasa PTEN/genética , Proteínas Tirosina Quinasas/genética , Pirazoles/farmacología , Pirimidinonas/farmacología , Proteínas de Ciclo Celular/metabolismo , Línea Celular Tumoral , Proteína Quinasa Activada por ADN/metabolismo , Femenino , Perfilación de la Expresión Génica , Humanos , Fosfohidrolasa PTEN/metabolismo , Proteínas Tirosina Quinasas/metabolismo , Proteoma
20.
Nat Commun ; 11(1): 394, 2020 01 20.
Artículo en Inglés | MEDLINE | ID: mdl-31959748

RESUMEN

Ionising radiation (IR) is a recognised carcinogen responsible for cancer development in patients previously treated using radiotherapy, and in individuals exposed as a result of accidents at nuclear energy plants. However, the mutational signatures induced by distinct types and doses of radiation are unknown. Here, we analyse the genetic architecture of mammary tumours, lymphomas and sarcomas induced by high (56Fe-ions) or low (gamma) energy radiation in mice carrying Trp53 loss of function alleles. In mammary tumours, high-energy radiation is associated with induction of focal structural variants, leading to genomic instability and Met amplification. Gamma-radiation is linked to large-scale structural variants and a point mutation signature associated with oxidative stress. The genomic architecture of carcinomas, sarcomas and lymphomas arising in the same animals are significantly different. Our study illustrates the complex interactions between radiation quality, germline Trp53 deficiency and tissue/cell of origin in shaping the genomic landscape of IR-induced tumours.


Asunto(s)
Carcinogénesis/efectos de la radiación , Inestabilidad Genómica/efectos de la radiación , Neoplasias Inducidas por Radiación/genética , Traumatismos Experimentales por Radiación/genética , Proteína p53 Supresora de Tumor/genética , Animales , Carcinogénesis/genética , Daño del ADN/efectos de la radiación , Análisis Mutacional de ADN , Relación Dosis-Respuesta en la Radiación , Femenino , Amplificación de Genes/efectos de la radiación , Mutación de Línea Germinal , Humanos , Masculino , Ratones , Ratones Noqueados , Neoplasias Inducidas por Radiación/patología , Mutación Puntual/efectos de la radiación , Proteínas Proto-Oncogénicas c-met/genética , Traumatismos Experimentales por Radiación/patología , Secuenciación Completa del Genoma
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA