Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 57
Filtrar
1.
Cancer Res ; 82(7): 1193-1207, 2022 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-35101866

RESUMEN

Noncoding cis-regulatory variants have gained interest as cancer drivers, yet progress in understanding their significance is hindered by the numerous challenges and limitations of variant prioritization. To overcome these limitations, we focused on active cis-regulatory elements (aCRE) to design a customized panel for the deep sequencing of 56 neuroblastoma tumor and normal DNA sample pairs. To search for driver mutations, aCREs were defined by reanalysis of H3K27ac chromatin immunoprecipitation sequencing peaks in 25 neuroblastoma cell lines. These regulatory genomic regions were tested for an excess of somatic mutations and assessed for statistical significance using a global approach that accounted for chromatin accessibility and replication timing. Additional validation was provided by whole genome sequence analysis of 151 neuroblastomas. Analysis of HiC data determined the presence of candidate target genes interacting with mutated regions. An excess of somatic mutations in aCREs of diverse genes were identified, including IPO7, HAND2, and ARID3A. CRISPR-Cas9 editing was utilized to assess the functional consequences of mutations in the IPO7-aCRE. Patients with noncoding mutations in aCREs showed inferior overall and event-free survival independent of age at diagnosis, stage, risk stratification, and MYCN status. Expression of aCRE-interacting genes correlated strongly with negative prognostic markers and low survival rates. Moreover, a convergence between the biological functions of aCRE target genes and transcription factors with mutated binding motifs was associated with embryonic development and immune system response. Overall, this strategy enabled the identification of somatic mutations in regulatory elements that collectively promote neuroblastoma tumorigenesis. SIGNIFICANCE: Assessment of noncoding cis-regulatory variants and long-range interaction data highlight the combined effect of somatic mutations in regulatory elements in driving neuroblastoma.


Asunto(s)
Regulación Neoplásica de la Expresión Génica , Neuroblastoma , Proteínas de Unión al ADN/genética , Desarrollo Embrionario , Humanos , Sistema Inmunológico/patología , Mutación , Neuroblastoma/patología , Factores de Transcripción/genética , Factores de Transcripción/metabolismo
2.
Nat Commun ; 12(1): 1749, 2021 03 19.
Artículo en Inglés | MEDLINE | ID: mdl-33741928

RESUMEN

Sonic hedgehog medulloblastoma encompasses a clinically and molecularly diverse group of cancers of the developing central nervous system. Here, we use unbiased sequencing of the transcriptome across a large cohort of 250 tumors to reveal differences among molecular subtypes of the disease, and demonstrate the previously unappreciated importance of non-coding RNA transcripts. We identify alterations within the cAMP dependent pathway (GNAS, PRKAR1A) which converge on GLI2 activity and show that 18% of tumors have a genetic event that directly targets the abundance and/or stability of MYCN. Furthermore, we discover an extensive network of fusions in focally amplified regions encompassing GLI2, and several loss-of-function fusions in tumor suppressor genes PTCH1, SUFU and NCOR1. Molecular convergence on a subset of genes by nucleotide variants, copy number aberrations, and gene fusions highlight the key roles of specific pathways in the pathogenesis of Sonic hedgehog medulloblastoma and open up opportunities for therapeutic intervention.


Asunto(s)
Neoplasias Cerebelosas/genética , Regulación Neoplásica de la Expresión Génica , Proteínas Hedgehog/genética , Meduloblastoma/genética , Transcriptoma , Adolescente , Adulto , Niño , Preescolar , Femenino , Redes Reguladoras de Genes , Variación Genética , Humanos , Lactante , Masculino , Persona de Mediana Edad , Transducción de Señal/genética , Adulto Joven
3.
J Pediatr Hematol Oncol ; 43(2): e180-e183, 2021 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-31876779

RESUMEN

WT1-mutant Wilms tumors exhibit a high rate of concomitant CTNNB1 mutations, associated with activated Wnt signaling. Here, we show by laser and manual microdissection of different histologic cell types from 6 WT1-mutant tumor samples that 1 patient's tumor can contain up to 4 distinct mutations in CTNNB1 and/or WTX. Consecutive sections may also harbor different CTNNB1 mutations. The variability of activating CTNNB1 mutations demonstrates the multifocal nature of WT1-mutant Wilms tumors. As multiple independent tumors can occur in patients with constitutional WT1 mutations, they need to be surveyed more closely for tumor development.


Asunto(s)
Evolución Clonal , Neoplasias Renales/patología , Mutación , Proteínas WT1/genética , Tumor de Wilms/patología , beta Catenina/genética , Progresión de la Enfermedad , Humanos , Neoplasias Renales/genética , Pronóstico , Tumor de Wilms/genética
4.
Cancer Res ; 80(3): 382-393, 2020 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-31784426

RESUMEN

The contribution of coding mutations to oncogenesis has been largely clarified, whereas little is known about somatic mutations in noncoding DNA and their role in driving tumors remains controversial. Here, we used an alternative approach to interpret the functional significance of noncoding somatic mutations in promoting tumorigenesis. Noncoding somatic mutations of 151 neuroblastomas were integrated with ENCODE data to locate somatic mutations in regulatory elements specifically active in neuroblastoma cells, nonspecifically active in neuroblastoma cells, and nonactive. Within these types of elements, transcription factors (TF) were identified whose binding sites were enriched or depleted in mutations. For these TFs, a gene expression signature was built to assess their implication in neuroblastoma. DNA- and RNA-sequencing data were integrated to assess the effects of those mutations on mRNA levels. The pathogenicity of mutations was significantly higher in transcription factor binding site (TFBS) of regulatory elements specifically active in neuroblastoma cells, as compared with the others. Within these elements, there were 18 over-represented TFs involved mainly in cell-cycle phase transitions and 15 under-represented TFs primarily regulating cell differentiation. A gene expression signature based on over-represented TFs correlated with poor survival and unfavorable prognostic markers. Moreover, recurrent mutations in TFBS of over-represented TFs such as EZH2 affected MCF2L and ADP-ribosylhydrolase like 1 expression, among the others. We propose a novel approach to study the involvement of regulatory variants in neuroblastoma that could be extended to other cancers and provide further evidence that alterations of gene expression may have relevant effects in neuroblastoma development. SIGNIFICANCE: These findings propose a novel approach to study regulatory variants in neuroblastoma and suggest that noncoding somatic mutations have relevant implications in neuroblastoma development.


Asunto(s)
Biomarcadores de Tumor/metabolismo , Carcinogénesis/patología , ADN de Neoplasias/metabolismo , Regulación Neoplásica de la Expresión Génica , Mutación , Neuroblastoma/patología , Factores de Transcripción/metabolismo , Sitios de Unión , Biomarcadores de Tumor/genética , Carcinogénesis/genética , Carcinogénesis/metabolismo , ADN de Neoplasias/genética , Humanos , Neuroblastoma/genética , Neuroblastoma/metabolismo , Unión Proteica , Secuencias Reguladoras de Ácidos Nucleicos , Factores de Transcripción/genética , Secuenciación Completa del Genoma
5.
Mol Oncol ; 13(9): 1959-1975, 2019 09.
Artículo en Inglés | MEDLINE | ID: mdl-31293052

RESUMEN

We have previously reported the expression of parathyroid hormone-like hormone (PTHLH) in well-differentiated, Schwannian stroma-rich neuroblastic tumors. The aim of this study was to functionally assess the role of PTHLH and its receptor, PTH1R, in neuroblastoma. Stable knockdown of PTHLH and PTH1R was conducted in neuroblastoma cell lines to investigate the succeeding phenotype induced both in vitro and in vivo. Downregulation of PTHLH reduced MYCN expression and subsequently induced cell cycle arrest, senescence, and migration and invasion impairment in a MYCN-amplified, TP53-mutated neuroblastoma cell line. These phenotypes were associated with reduced tumorigenicity in a murine model. We also show that PTHLH expression is not under the control of the calcium-sensing receptor in neuroblastoma. Conversely, its production is stimulated by epidermal growth factor receptor (EGFR). Accordingly, irreversible EGFR inhibition with canertinib abolished PTHLH expression. The oncogenic role of PTHLH appeared to be a consequence of its intracrine function, as downregulation of its receptor, PTH1R, increased anchorage-independent growth and induced a more undifferentiated, invasive phenotype. Respectively, high PTH1R mRNA expression was found in MYCN nonamplified primary tumors and also significantly associated with other prognostic factors of good outcome. This study provides the first evidence of the dual role of PTHLH in the behavior of neuroblastomas. Moreover, the identification of EGFR as a transcriptional regulator of PTHLH in neuroblastoma provides a novel therapeutic opportunity to promote a less aggressive tumor phenotype through irreversible inhibition of EGFR tyrosine kinase activity.


Asunto(s)
Regulación Neoplásica de la Expresión Génica , Neuroblastoma/metabolismo , Proteína Relacionada con la Hormona Paratiroidea/metabolismo , Receptor de Hormona Paratiroídea Tipo 1/biosíntesis , Animales , Línea Celular Tumoral , Femenino , Células HEK293 , Humanos , Ratones , Ratones Desnudos , Mutación , Neuroblastoma/genética , Neuroblastoma/patología , Proteína Relacionada con la Hormona Paratiroidea/genética , Receptor de Hormona Paratiroídea Tipo 1/genética , Proteína p53 Supresora de Tumor/genética , Proteína p53 Supresora de Tumor/metabolismo
6.
Int J Cancer ; 143(11): 2828-2837, 2018 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-30132831

RESUMEN

A previous genome-wide association study (GWAS) identified common variation at the BARD1 locus as being highly associated with susceptibility to high-risk neuroblastoma, but the mechanisms underlying this association have been not extensively investigated. Here, we performed a fine mapping analysis of BARD1 locus (2q35) using GWAS data from 556 high-risk neuroblastoma patients and 2,575 controls of European-American ancestry, and identified two independent genome-wide neuroblastoma-associated loci. Functional single-nucleotide polymorphism (SNP) prioritization identified two causative variants that independently contributed to neuroblastoma risk, and each replicated robustly in multiple independent cohorts comprising 445 high-risk cases and 3,170 controls (rs17489363: combined p = 1.07 × 10-31 , OR:1.79, 95% CI:1.62-1.98 and rs1048108: combined p = 7.27 × 10-14 , OR:0.65, 95% CI:0.58-0.73). Particularly, the T risk allele of rs17489363 in the canonical promoter region of full-length BARD1 altered binding site of the transcription factor HSF1 and correlated with low expression of full-length BARD1 mRNA and protein. Low-level expression of full-length BARD1 associated with advanced neuroblastoma. In human neuroblastoma cells, attenuating full-length BARD1 increased proliferation and invasion capacity. In conclusion, we have identified two potentially causative SNPs at the BARD1 locus associated with predisposition to high-risk neuroblastoma, and have shown that full-length BARD1 may act as tumor suppressor.


Asunto(s)
Predisposición Genética a la Enfermedad/genética , Neuroblastoma/genética , Polimorfismo de Nucleótido Simple/genética , Proteínas Supresoras de Tumor/genética , Ubiquitina-Proteína Ligasas/genética , Alelos , Estudios de Casos y Controles , Línea Celular Tumoral , Proliferación Celular/genética , Genes Supresores de Tumor/fisiología , Estudio de Asociación del Genoma Completo/métodos , Genotipo , Células HEK293 , Humanos , Lactante , Invasividad Neoplásica/genética , Regiones Promotoras Genéticas/genética , ARN Mensajero/genética
7.
Nat Med ; 24(8): 1204-1215, 2018 08.
Artículo en Inglés | MEDLINE | ID: mdl-29967352

RESUMEN

The failure to develop effective therapies for pediatric glioblastoma (pGBM) and diffuse intrinsic pontine glioma (DIPG) is in part due to their intrinsic heterogeneity. We aimed to quantitatively assess the extent to which this was present in these tumors through subclonal genomic analyses and to determine whether distinct tumor subpopulations may interact to promote tumorigenesis by generating subclonal patient-derived models in vitro and in vivo. Analysis of 142 sequenced tumors revealed multiple tumor subclones, spatially and temporally coexisting in a stable manner as observed by multiple sampling strategies. We isolated genotypically and phenotypically distinct subpopulations that we propose cooperate to enhance tumorigenicity and resistance to therapy. Inactivating mutations in the H4K20 histone methyltransferase KMT5B (SUV420H1), present in <1% of cells, abrogate DNA repair and confer increased invasion and migration on neighboring cells, in vitro and in vivo, through chemokine signaling and modulation of integrins. These data indicate that even rare tumor subpopulations may exert profound effects on tumorigenesis as a whole and may represent a new avenue for therapeutic development. Unraveling the mechanisms of subclonal diversity and communication in pGBM and DIPG will be an important step toward overcoming barriers to effective treatments.


Asunto(s)
Neoplasias del Tronco Encefálico/patología , Glioblastoma/patología , Animales , Neoplasias del Tronco Encefálico/genética , Carcinogénesis/patología , Separación Celular , Niño , Células Clonales , Genotipo , Glioblastoma/genética , Humanos , Ratones Desnudos , Fenotipo , Células Tumorales Cultivadas
9.
Front Oncol ; 8: 127, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29755954

RESUMEN

BACKGROUND AND OBJECTIVE: Diffuse intrinsic pontine glioma (DIPG) is a lethal brainstem tumor in children. Dendritic cells (DCs) have T-cell stimulatory capacity and, therefore, potential antitumor activity for disease control. DCs vaccines have been shown to reactivate tumor-specific T cells in both clinical and preclinical settings. We designed a phase Ib immunotherapy (IT) clinical trial with the use of autologous dendritic cells (ADCs) pulsed with an allogeneic tumors cell-lines lysate in patients with newly diagnosed DIPG after irradiation (radiation therapy). METHODS: Nine patients with newly diagnosed DIPG met enrollment criteria. Autologous dendritic cell vaccines (ADCV) were prepared from monocytes obtained by leukapheresis. Five ADCV doses were administered intradermally during induction phase. In the absence of tumor progression, patients received three boosts of tumor lysate every 3 months during the maintenance phase. RESULTS: Vaccine fabrication was feasible in all patients included in the study. Non-specific KLH (9/9 patients) and specific (8/9 patients) antitumor response was identified by immunologic studies in peripheral blood mononuclear cells (PBMC). Immunological responses were also confirmed in the T lymphocytes isolated from the cerebrospinal fluid (CSF) of two patients. Vaccine administration resulted safe in all patients treated with this schema. CONCLUSION: These preliminary results demonstrate that ADCV preparation is feasible, safe, and generate a DIPG-specific immune response detected in PBMC and CSF. This strategy shows a promising backbone for future schemas of combination IT.

10.
Cancer Med ; 7(4): 1359-1368, 2018 04.
Artículo en Inglés | MEDLINE | ID: mdl-29542868

RESUMEN

Wilms tumors (WT) with WT1 mutations do not respond well to preoperative chemotherapy by volume reduction, suggesting resistance to chemotherapy. The histologic pattern of this tumor subtype indicates an intrinsic mesenchymal differentiation potential. Currently, it is unknown whether cytotoxic treatments can induce a terminal differentiation state as a direct comparison of untreated and chemotherapy-treated tumor samples has not been reported so far. We conducted gene expression profiling of 11 chemotherapy and seven untreated WT1-mutant Wilms tumors and analyzed up- and down-regulated genes with bioinformatic methods. Cell culture experiments were performed from primary Wilms tumors and genetic alterations in WT1 and CTNNB1 analyzed. Chemotherapy induced MYF6 165-fold and several MYL and MYH genes more than 20-fold and repressed many genes from cell cycle process networks. Viable tumor cells could be cultivated when patients received less than 8 weeks of chemotherapy but not in two cases with longer treatments. In one case, viable cells could be extracted from a lung metastasis occurring after 6 months of intensive chemotherapy and radiation. Comparison of primary tumor and metastasis cells from the same patient revealed up-regulation of RELN and TBX2, TBX4 and TBX5 genes and down-regulation of several HOXD genes. Our analyses demonstrate that >8 weeks of chemotherapy can induce terminal myogenic differentiation in WT1-mutant tumors, but this is not associated with volume reduction. The time needed for all tumor cells to achieve the terminal differentiation state needs to be evaluated. In contrast, prolonged treatments can result in genetic alterations leading to resistance.


Asunto(s)
Músculo Esquelético/metabolismo , Músculo Esquelético/patología , Mutación , Proteínas WT1/genética , Tumor de Wilms/genética , Tumor de Wilms/patología , Protocolos de Quimioterapia Combinada Antineoplásica/efectos adversos , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Biomarcadores de Tumor , Ciclo Celular/genética , Línea Celular Tumoral , Perfilación de la Expresión Génica , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Neoplasias Pulmonares/secundario , Desarrollo de Músculos/genética , Proteína Reelina , Transcripción Genética , Tumor de Wilms/tratamiento farmacológico
11.
Genome Med ; 10(1): 25, 2018 03 28.
Artículo en Inglés | MEDLINE | ID: mdl-29592813

RESUMEN

While tumor genome sequencing has become widely available in clinical and research settings, the interpretation of tumor somatic variants remains an important bottleneck. Here we present the Cancer Genome Interpreter, a versatile platform that automates the interpretation of newly sequenced cancer genomes, annotating the potential of alterations detected in tumors to act as drivers and their possible effect on treatment response. The results are organized in different levels of evidence according to current knowledge, which we envision can support a broad range of oncology use cases. The resource is publicly available at http://www.cancergenomeinterpreter.org .


Asunto(s)
Genoma Humano , Anotación de Secuencia Molecular , Neoplasias/genética , Programas Informáticos , Antineoplásicos/uso terapéutico , Biomarcadores de Tumor/metabolismo , Bases de Datos Genéticas , Genes Relacionados con las Neoplasias , Humanos , Mutación/genética , Neoplasias/tratamiento farmacológico
12.
Biosens Bioelectron ; 107: 62-68, 2018 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-29438908

RESUMEN

In this work, we propose for the first time the use of anodic aluminum oxide (AAO) nanoporous membranes for in situ monitoring of parathyroid hormone-like hormone (PTHLH) secretion in cultured human cells. The biosensing system is based on the nanochannels blockage upon immunocomplex formation, which is electrically monitored through the voltammetric oxidation of Prussian blue nanoparticles (PBNPs). Models evaluated include a neuroblastoma cell line (SK-N-AS) and immortalized keratinocytes (HaCaT) as a control of high PTHLH production. The effect of total number of seeded cells and incubation time on the secreted PTHLH levels is assessed, finding that secreted PTHLH levels range from approximately 60 to 400 ng/mL. Moreover, our methodology is also applied to analyse PTHLH production following PTHLH gene knockdown upon transient cell transfection with a specific silencing RNA (siRNA). Given that inhibition of PTHLH secretion reduces cell proliferation, survival and invasiveness in a number of tumors, our system provides a powerful tool for the preclinical evaluation of therapies that regulate PTHLH production. This nanoporous membrane - based sensing technology might be useful to monitor the active secretion of other proteins as well, thus contributing to characterize their regulation and function.


Asunto(s)
Técnicas Biosensibles/instrumentación , Membranas Artificiales , Nanoporos/ultraestructura , Neuroblastoma/metabolismo , Proteína Relacionada con la Hormona Paratiroidea/metabolismo , Línea Celular , Línea Celular Tumoral , Diseño de Equipo , Humanos , Proteína Relacionada con la Hormona Paratiroidea/análisis
13.
Clin Cancer Res ; 24(6): 1355-1363, 2018 03 15.
Artículo en Inglés | MEDLINE | ID: mdl-29351917

RESUMEN

Purpose: The classification of medulloblastoma into WNT, SHH, group 3, and group 4 subgroups has become of critical importance for patient risk stratification and subgroup-tailored clinical trials. Here, we aimed to develop a simplified, clinically applicable classification approach that can be implemented in the majority of centers treating patients with medulloblastoma.Experimental Design: We analyzed 1,577 samples comprising previously published DNA methylation microarray data (913 medulloblastomas, 457 non-medulloblastoma tumors, 85 normal tissues), and 122 frozen and formalin-fixed paraffin-embedded medulloblastoma samples. Biomarkers were identified applying stringent selection filters and Linear Discriminant Analysis (LDA) method, and validated using DNA methylation microarray data, bisulfite pyrosequencing, and direct-bisulfite sequencing.Results: Using a LDA-based approach, we developed and validated a prediction method (EpiWNT-SHH classifier) based on six epigenetic biomarkers that allowed for rapid classification of medulloblastoma into the clinically relevant subgroups WNT, SHH, and non-WNT/non-SHH with excellent concordance (>99%) with current gold-standard methods, DNA methylation microarray, and gene signature profiling analysis. The EpiWNT-SHH classifier showed high prediction capacity using both frozen and formalin-fixed material, as well as diverse DNA methylation detection methods. Similarly, we developed a classifier specific for group 3 and group 4 tumors, based on five biomarkers (EpiG3-G4) with good discriminatory capacity, allowing for correct assignment of more than 92% of tumors. EpiWNT-SHH and EpiG3-G4 methylation profiles remained stable across tumor primary, metastasis, and relapse samples.Conclusions: The EpiWNT-SHH and EpiG3-G4 classifiers represent a new simplified approach for accurate, rapid, and cost-effective molecular classification of single medulloblastoma DNA samples, using clinically applicable DNA methylation detection methods. Clin Cancer Res; 24(6); 1355-63. ©2018 AACR.


Asunto(s)
Biomarcadores de Tumor , Neoplasias Cerebelosas/diagnóstico , Neoplasias Cerebelosas/genética , Estudios de Asociación Genética , Predisposición Genética a la Enfermedad , Meduloblastoma/diagnóstico , Meduloblastoma/genética , Biopsia , Islas de CpG , Metilación de ADN , Epigénesis Genética , Epigenómica/métodos , Femenino , Perfilación de la Expresión Génica , Humanos , Masculino , Reproducibilidad de los Resultados
14.
Oncotarget ; 8(52): 89775-89792, 2017 Oct 27.
Artículo en Inglés | MEDLINE | ID: mdl-29163787

RESUMEN

Pediatric tumors arise upon oncogenic transformation of stem/progenitor cells during embryonic development. Given this scenario, the existence of non-tumorigenic stem cells included within the aberrant tumoral niche, with a potential role in tumor biology, is an intriguing and unstudied possibility. Here, we describe the presence and function of non-tumorigenic neural crest-derived progenitor cells in aggressive neuroblastoma (NB) tumors. These cells differentiate into neural crest typical mesectodermal derivatives, giving rise to tumor stroma and promoting proliferation and tumor aggressiveness. Furthermore, an analysis of gene expression profiles in stage 4/M NB revealed a neural crest stem cell (NCSC) gene signature that was associated to stromal phenotype and high probability of relapse. Thus, this NCSC gene expression signature could be used in prognosis to improve stratification of stage 4/M NB tumors. Our results might facilitate the design of new therapies by targeting NCSCs and their contribution to tumor stroma.

15.
Cancer Cell ; 32(4): 520-537.e5, 2017 10 09.
Artículo en Inglés | MEDLINE | ID: mdl-28966033

RESUMEN

We collated data from 157 unpublished cases of pediatric high-grade glioma and diffuse intrinsic pontine glioma and 20 publicly available datasets in an integrated analysis of >1,000 cases. We identified co-segregating mutations in histone-mutant subgroups including loss of FBXW7 in H3.3G34R/V, TOP3A rearrangements in H3.3K27M, and BCOR mutations in H3.1K27M. Histone wild-type subgroups are refined by the presence of key oncogenic events or methylation profiles more closely resembling lower-grade tumors. Genomic aberrations increase with age, highlighting the infant population as biologically and clinically distinct. Uncommon pathway dysregulation is seen in small subsets of tumors, further defining the molecular diversity of the disease, opening up avenues for biological study and providing a basis for functionally defined future treatment stratification.


Asunto(s)
Neoplasias del Tronco Encefálico/genética , Glioma/genética , Histonas/genética , Mutación , Adolescente , Neoplasias del Tronco Encefálico/patología , Proteínas de Ciclo Celular/genética , Niño , Preescolar , ADN-Topoisomerasas de Tipo I/genética , Exoma , Proteínas F-Box/genética , Proteína 7 que Contiene Repeticiones F-Box-WD , Femenino , Dosificación de Gen , Glioma/patología , Humanos , Lactante , Recién Nacido , Masculino , Proteínas Proto-Oncogénicas/genética , Proteínas Represoras/genética , Ubiquitina-Proteína Ligasas/genética , Adulto Joven
16.
Sci Rep ; 7(1): 11762, 2017 09 18.
Artículo en Inglés | MEDLINE | ID: mdl-28924151

RESUMEN

This work provides a comprehensive CpG methylation landscape of the different layers of the human eye that unveils the gene networks associated with their biological functions and how these are disrupted in common visual disorders. Herein, we firstly determined the role of CpG methylation in the regulation of ocular tissue-specification and described hypermethylation of retinal transcription factors (i.e., PAX6, RAX, SIX6) in a tissue-dependent manner. Second, we have characterized the DNA methylome of visual disorders linked to internal and external environmental factors. Main conclusions allow certifying that crucial pathways related to Wnt-MAPK signaling pathways or neuroinflammation are epigenetically controlled in the fibrotic disorders involved in retinal detachment, but results also reinforced the contribution of neurovascularization (ETS1, HES5, PRDM16) in diabetic retinopathy. Finally, we had studied the methylome in the most frequent intraocular tumors in adults and children (uveal melanoma and retinoblastoma, respectively). We observed that hypermethylation of tumor suppressor genes is a frequent event in ocular tumors, but also unmethylation is associated with tumorogenesis. Interestingly, unmethylation of the proto-oncogen RAB31 was a predictor of metastasis risk in uveal melanoma. Loss of methylation of the oncogenic mir-17-92 cluster was detected in primary tissues but also in blood from patients.


Asunto(s)
Metilación de ADN , ADN de Neoplasias , Retinopatía Diabética , Epigénesis Genética , Neoplasias del Ojo/metabolismo , Proteínas del Ojo , Ojo , Regulación Neoplásica de la Expresión Génica , Sistema de Señalización de MAP Quinasas , Proteínas de Neoplasias , Neovascularización Retiniana , Adulto , Niño , Preescolar , ADN de Neoplasias/genética , ADN de Neoplasias/metabolismo , Retinopatía Diabética/genética , Retinopatía Diabética/metabolismo , Retinopatía Diabética/patología , Ojo/crecimiento & desarrollo , Ojo/patología , Neoplasias del Ojo/genética , Neoplasias del Ojo/patología , Proteínas del Ojo/biosíntesis , Proteínas del Ojo/genética , Femenino , Humanos , Masculino , Proteínas de Neoplasias/biosíntesis , Proteínas de Neoplasias/genética , Neovascularización Retiniana/genética , Neovascularización Retiniana/metabolismo , Neovascularización Retiniana/patología
17.
J Clin Oncol ; 35(25): 2934-2941, 2017 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-28727518

RESUMEN

Purpose BRAF V600E is a potentially highly targetable mutation detected in a subset of pediatric low-grade gliomas (PLGGs). Its biologic and clinical effect within this diverse group of tumors remains unknown. Patients and Methods A combined clinical and genetic institutional study of patients with PLGGs with long-term follow-up was performed (N = 510). Clinical and treatment data of patients with BRAF V600E mutated PLGG (n = 99) were compared with a large international independent cohort of patients with BRAF V600E mutated-PLGG (n = 180). Results BRAF V600E mutation was detected in 69 of 405 patients (17%) with PLGG across a broad spectrum of histologies and sites, including midline locations, which are not often routinely biopsied in clinical practice. Patients with BRAF V600E PLGG exhibited poor outcomes after chemotherapy and radiation therapies that resulted in a 10-year progression-free survival of 27% (95% CI, 12.1% to 41.9%) and 60.2% (95% CI, 53.3% to 67.1%) for BRAF V600E and wild-type PLGG, respectively ( P < .001). Additional multivariable clinical and molecular stratification revealed that the extent of resection and CDKN2A deletion contributed independently to poor outcome in BRAF V600E PLGG. A similar independent role for CDKN2A and resection on outcome were observed in the independent cohort. Quantitative imaging analysis revealed progressive disease and a lack of response to conventional chemotherapy in most patients with BRAF V600E PLGG. Conclusion BRAF V600E PLGG constitutes a distinct entity with poor prognosis when treated with current adjuvant therapy.


Asunto(s)
Neoplasias Encefálicas/enzimología , Glioma/enzimología , Proteínas Proto-Oncogénicas B-raf/genética , Adolescente , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/patología , Neoplasias Encefálicas/terapia , Neoplasias del Tronco Encefálico/enzimología , Neoplasias del Tronco Encefálico/genética , Neoplasias del Tronco Encefálico/patología , Neoplasias del Tronco Encefálico/terapia , Niño , Preescolar , Estudios de Cohortes , Diencéfalo/enzimología , Diencéfalo/patología , Femenino , Glioma/genética , Glioma/patología , Glioma/terapia , Humanos , Lactante , Masculino , Mutación , Clasificación del Tumor , Pronóstico
18.
J Control Release ; 255: 108-119, 2017 06 10.
Artículo en Inglés | MEDLINE | ID: mdl-28412222

RESUMEN

Neuroblastoma is a pediatric solid tumor with high expression of the tumor associated antigen disialoganglioside GD2. Despite initial response to induction therapy, nearly 50% of high-risk neuroblastomas recur because of chemoresistance. Here we encapsulated the topoisomerase-I inhibitor SN-38 in polymeric nanoparticles (NPs) surface-decorated with the anti-GD2 mouse mAb 3F8 at a mean density of seven antibody molecules per NP. The accumulation of drug-loaded NPs targeted with 3F8 versus with control antibody was monitored by microdialysis in patient-derived GD2-expressing neuroblastoma xenografts. We showed that the extent of tumor penetration by SN-38 was significantly higher in mice receiving the targeted nano-drug delivery system when compared to non-targeted system or free drug. This selective penetration of the tumor extracellular fluid translated into a strong anti-tumor effect prolonging survival of mice bearing GD2-high neuroblastomas in vivo.


Asunto(s)
Anticuerpos Monoclonales/administración & dosificación , Antineoplásicos Fitogénicos/administración & dosificación , Camptotecina/análogos & derivados , Líquido Extracelular/metabolismo , Inmunoglobulina G/administración & dosificación , N-Acetilgalactosaminiltransferasas/antagonistas & inhibidores , Nanopartículas/administración & dosificación , Neuroblastoma/metabolismo , Animales , Anticuerpos Monoclonales/química , Anticuerpos Monoclonales de Origen Murino , Antígenos de Neoplasias/genética , Antígenos de Neoplasias/metabolismo , Antineoplásicos Fitogénicos/química , Antineoplásicos Fitogénicos/farmacocinética , Camptotecina/administración & dosificación , Camptotecina/química , Camptotecina/farmacocinética , Línea Celular Tumoral , Preescolar , Liberación de Fármacos , Regulación Neoplásica de la Expresión Génica , Humanos , Inmunoglobulina G/química , Irinotecán , Masculino , Ratones Desnudos , N-Acetilgalactosaminiltransferasas/genética , N-Acetilgalactosaminiltransferasas/inmunología , N-Acetilgalactosaminiltransferasas/metabolismo , Nanopartículas/química , Neuroblastoma/tratamiento farmacológico , Distribución Tisular , Ensayos Antitumor por Modelo de Xenoinjerto
19.
Nat Med ; 23(4): 483-492, 2017 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-28263309

RESUMEN

Diffuse intrinsic pontine glioma (DIPG) is an aggressive brain tumor that is located in the pons and primarily affects children. Nearly 80% of DIPGs harbor mutations in histone H3 genes, wherein lysine 27 is substituted with methionine (H3K27M). H3K27M has been shown to inhibit polycomb repressive complex 2 (PRC2), a multiprotein complex responsible for the methylation of H3 at lysine 27 (H3K27me), by binding to its catalytic subunit EZH2. Although DIPGs with the H3K27M mutation show global loss of H3K27me3, several genes retain H3K27me3. Here we describe a mouse model of DIPG in which H3K27M potentiates tumorigenesis. Using this model and primary patient-derived DIPG cell lines, we show that H3K27M-expressing tumors require PRC2 for proliferation. Furthermore, we demonstrate that small-molecule EZH2 inhibitors abolish tumor cell growth through a mechanism that is dependent on the induction of the tumor-suppressor protein p16INK4A. Genome-wide enrichment analyses show that the genes that retain H3K27me3 in H3K27M cells are strong polycomb targets. Furthermore, we find a highly significant overlap between genes that retain H3K27me3 in the DIPG mouse model and in human primary DIPGs expressing H3K27M. Taken together, these results show that residual PRC2 activity is required for the proliferation of H3K27M-expressing DIPGs, and that inhibition of EZH2 is a potential therapeutic strategy for the treatment of these tumors.


Asunto(s)
Neoplasias del Tronco Encefálico/genética , Proliferación Celular/genética , Proteína Potenciadora del Homólogo Zeste 2/genética , Glioma/genética , Histonas/genética , Animales , Benzamidas/farmacología , Compuestos de Bifenilo , Neoplasias Encefálicas/genética , Sistemas CRISPR-Cas , Línea Celular Tumoral , Inmunoprecipitación de Cromatina , Cromatografía Liquida , Inhibidor p16 de la Quinasa Dependiente de Ciclina/efectos de los fármacos , Inhibidor p16 de la Quinasa Dependiente de Ciclina/genética , Modelos Animales de Enfermedad , Proteína Potenciadora del Homólogo Zeste 2/antagonistas & inhibidores , Técnicas de Inactivación de Genes , Glioblastoma/genética , Humanos , Inmunohistoquímica , Hibridación Fluorescente in Situ , Indazoles/farmacología , Ratones , Ratones SCID , Terapia Molecular Dirigida , Morfolinas , Mutación , Trasplante de Neoplasias , Células-Madre Neurales , Complejo Represivo Polycomb 2/genética , Piridonas/farmacología , Espectrometría de Masas en Tándem , Proteína p14ARF Supresora de Tumor/efectos de los fármacos , Proteína p14ARF Supresora de Tumor/genética
20.
Mol Carcinog ; 56(4): 1281-1289, 2017 04.
Artículo en Inglés | MEDLINE | ID: mdl-27862333

RESUMEN

We have previously reported that the calcium-sensing receptor (CaSR) is expressed in benign, differentiated neuroblastic tumors, and epigenetically silenced in undifferentiated, malignant cases. Furthermore, cinacalcet, an allosteric activator of the CaSR, reduces neuroblastoma tumor growth in preclinical models. However, to identify patients that might benefit from this treatment, a complete understanding of mechanisms governing CaSR expression in these tumors would be required. We have now analyzed two polymorphisms in the promoter region of the CASR gene (rs7652589 and rs1501899) by allelic discrimination in neuroblastoma patients and cell lines. Association of genotypes and haplotypes with CaSR mRNA levels and CASR promoter P2 methylation status was determined. Data presented show that minor alleles rs7652589 and rs1501899, present either in homo- or heterozygosis, were correlated with reduced CaSR mRNA levels in matching primary tumors and this association was independent of CASR promoter P2 hypermethylation. Haplotype AA was independently associated with reduced CaSR expression after adjusting by promoter P2 methylation status. These polymorphisms were identified in some ganglioneuromas in which CaSR expression is low despite exhibiting a high degree of differentiation. Furthermore, homozygous variants rs7652589 and rs1501899 were detected in SH-SY5Y cells, which are devoid of CaSR expression in the absence of hypermethylation of CASR promoter P2. In summary, minor alleles rs7652589 and rs1501899 are associated with reduced CaSR expression in neuroblastic tumors and neuroblastoma cell lines in which the CASR gene promoter P2 is not hypermethylated. Therefore, they potentially represent an additional mechanism of CASR transcriptional regulation in this group of developmental malignancies. © 2016 Wiley Periodicals, Inc.


Asunto(s)
Regulación hacia Abajo , Regulación Neoplásica de la Expresión Génica , Neuroblastoma/genética , Neoplasias del Sistema Nervioso Periférico/genética , Polimorfismo de Nucleótido Simple , Receptores Sensibles al Calcio/genética , Línea Celular Tumoral , Metilación de ADN , Supervivencia sin Enfermedad , Variación Genética , Haplotipos , Humanos , Lactante , Neuroblastoma/epidemiología , Neoplasias del Sistema Nervioso Periférico/epidemiología , Regiones Promotoras Genéticas
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA