Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 22
Filtrar
Más filtros

Banco de datos
Tipo del documento
País de afiliación
Intervalo de año de publicación
1.
Lab Invest ; 100(10): 1330-1344, 2020 10.
Artículo en Inglés | MEDLINE | ID: mdl-32404931

RESUMEN

Glioblastomas (GBM) can be classified into three major transcriptional subgroups (proneural, mesenchymal, classical), underlying different molecular alterations, prognosis, and response to therapy. However, transcriptional analysis is not routinely feasible and assessment of a simplified method for glioblastoma subclassification is required. We propose an integrated molecular and immunohistochemical approach aimed at identifying GBM subtypes in routine paraffin-embedded material. RNA-sequencing analysis was performed on representative samples (n = 51) by means of a "glioblastoma transcriptional subtypes (GliTS) redux" custom gene signature including a restricted number (n = 90) of upregulated genes validated on the TCGA dataset. With this dataset, immunohistochemical profiles, based on expression of a restricted panel of gene classifiers, were integrated by a machine-learning approach to generate a GliTS based on protein quantification that allowed an efficient GliTS assignment when applied to an extended cohort (n = 197). GliTS redux maintained high levels of correspondence with the original GliTS classification using the TCGA dataset. The machine-learning approach designed an immunohistochemical (IHC)-based classification, whose concordance was 79.5% with the transcriptional- based classification, and reached 90% for the mesenchymal subgroup. Distribution and survival of GliTS were in line with reported data, with the mesenchymal subgroup given the worst prognosis. Notably, the algorithm allowed the identification of cases with comparable probability to be assigned to different GliTS, thus falling within overlapping regions and reflecting an extreme heterogeneous phenotype that mirrors the underlying genetic and biological tumor heterogeneity. Indeed, while mesenchymal and classical subgroups were well segregated, the proneural types frequently showed a mixed proneural/classical phenotype, predicted as proneural by the algorithm, but with comparable probability of being assigned to the classical subtype. These cases, characterized by concomitant high expression of EGFR and proneural biomarkers, showed lower survival. Collectively, these data indicate that a restricted panel of highly sensitive immunohistochemical markers can efficiently predict GliTS with high accuracy and significant association with different clinical outcomes.


Asunto(s)
Neoplasias Encefálicas/clasificación , Neoplasias Encefálicas/metabolismo , Perfilación de la Expresión Génica , Glioblastoma/clasificación , Glioblastoma/metabolismo , Anciano , Algoritmos , Biomarcadores de Tumor/genética , Biomarcadores de Tumor/metabolismo , Neoplasias Encefálicas/genética , Análisis por Conglomerados , Estudios de Cohortes , Femenino , Perfilación de la Expresión Génica/métodos , Perfilación de la Expresión Génica/estadística & datos numéricos , Regulación Neoplásica de la Expresión Génica , Glioblastoma/genética , Humanos , Inmunohistoquímica , Aprendizaje Automático , Masculino , Persona de Mediana Edad , RNA-Seq
2.
Stem Cells ; 35(11): 2218-2228, 2017 11.
Artículo en Inglés | MEDLINE | ID: mdl-28895245

RESUMEN

Glioblastoma (GBM) is a lethal tumor that displays remarkable genetic heterogeneity. It is also known that GBM contains a cell hierarchy driven by GBM stem-like cells (GSCs), responsible for tumor generation, therapeutic resistance, and relapse. An important and still open issue is whether phylogenetically related GSCs can be found in matched primary and recurrent GBMs, and reflect tumor genetic evolution under therapeutic pressure. To address this, we analyzed the mutational profile of GSCs isolated from either human primary GBMs (primary GSCs) or their matched tumors recurring after surgery and chemoradiotherapy (recurrent GSCs). We found that recurrent GSCs can accumulate temozolomide-related mutations over primary GSCs, following both linear and branched patterns. In the latter case, primary and recurrent GSCs share a common set of lesions, but also harbor distinctive mutations indicating that primary and recurrent GSCs derive from a putative common ancestor GSC by divergent genetic evolution. Interestingly, TP53 mutations distinctive of recurrent GSCs were detectable at low frequency in the corresponding primary tumors and likely marked pre-existent subclones that evolved under therapeutic pressure and expanded in the relapsing tumor. Consistently, recurrent GSCs displayed in vitro greater therapeutic resistance than primary GSCs. Overall, these data indicate that (a) phylogenetically related GSCs are found in matched primary and recurrent GBMs and (b) recurrent GSCs likely pre-exist in the untreated primary tumor and are both mutagenized and positively selected by chemoradiotherapy. Stem Cells 2017;35:2218-2228.


Asunto(s)
Dosificación de Gen/genética , Glioblastoma/genética , Células Madre Neoplásicas/metabolismo , Adulto , Animales , Evolución Molecular , Femenino , Glioblastoma/patología , Humanos , Masculino , Ratones , Persona de Mediana Edad , Células Madre Neoplásicas/patología
3.
STAR Protoc ; 4(4): 102705, 2023 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-37971942

RESUMEN

Cultures enriched in glioblastoma stem-like cells (GSCs) are prominent in vitro models to investigate molecular determinants and therapeutic targets of glioblastoma; however, conventional GSC derivation protocols fail to preserve GSC heterogeneity. Here, we present a protocol for the propagation of heterogeneous GSC cultures starting from cell resuspensions containing the entire tumor mass. We describe steps for isolation of GSCs and their maintenance and expansion in culture. We then detail procedures for preliminary analysis to be performed on freshly isolated material. For complete details on the use and execution of this protocol, please refer to De Bacco et al.1.


Asunto(s)
Glioblastoma , Humanos , Glioblastoma/patología
4.
Mol Oncol ; 17(7): 1280-1301, 2023 07.
Artículo en Inglés | MEDLINE | ID: mdl-36862005

RESUMEN

In colorectal cancer, the mechanisms underlying tumor aggressiveness require further elucidation. Taking advantage of a large panel of human metastatic colorectal cancer xenografts and matched stem-like cell cultures (m-colospheres), here we show that the overexpression of microRNA 483-3p (miRNA-483-3p; also known as MIR-483-3p), encoded by a frequently amplified gene locus, confers an aggressive phenotype. In m-colospheres, endogenous or ectopic miRNA-483-3p overexpression increased proliferative response, invasiveness, stem cell frequency, and resistance to differentiation. Transcriptomic analyses and functional validation found that miRNA-483-3p directly targets NDRG1, known as a metastasis suppressor involved in EGFR family downregulation. Mechanistically, miRNA-483-3p overexpression induced the signaling pathway triggered by ERBB3, including AKT and GSK3ß, and led to the activation of transcription factors regulating epithelial-mesenchymal transition (EMT). Consistently, treatment with selective anti-ERBB3 antibodies counteracted the invasive growth of miRNA-483-3p-overexpressing m-colospheres. In human colorectal tumors, miRNA-483-3p expression inversely correlated with NDRG1 and directly correlated with EMT transcription factor expression and poor prognosis. These results unveil a previously unrecognized link between miRNA-483-3p, NDRG1, and ERBB3-AKT signaling that can directly support colorectal cancer invasion and is amenable to therapeutic targeting.


Asunto(s)
Neoplasias del Colon , Neoplasias Colorrectales , MicroARNs , Neoplasias del Recto , Humanos , Proteínas Proto-Oncogénicas c-akt/metabolismo , Regulación hacia Abajo/genética , Línea Celular Tumoral , MicroARNs/genética , MicroARNs/metabolismo , Neoplasias Colorrectales/patología , Neoplasias del Colon/genética , Factores de Transcripción/metabolismo , Neoplasias del Recto/genética , Transición Epitelial-Mesenquimal/genética , Regulación Neoplásica de la Expresión Génica , Movimiento Celular/genética , Invasividad Neoplásica/genética
5.
Clin Cancer Res ; 29(7): 1252-1266, 2023 04 03.
Artículo en Inglés | MEDLINE | ID: mdl-36648487

RESUMEN

PURPOSE: Current glioma diagnostic guidelines call for molecular profiling to stratify patients into prognostic and treatment subgroups. In case the tumor tissue is inaccessible, cerebrospinal fluid (CSF) has been proposed as a reliable tumor DNA source for liquid biopsy. We prospectively investigated the use of CSF for molecular characterization of newly diagnosed gliomas. EXPERIMENTAL DESIGN: We recruited two cohorts of newly diagnosed patients with glioma, one (n = 45) providing CSF collected in proximity of the tumor, the other (n = 39) CSF collected by lumbar puncture (LP). Both cohorts provided tumor tissues by surgery concomitant with CSF sampling. DNA samples retrieved from CSF and matched tumors were systematically characterized and compared by comprehensive (NGS, next-generation sequencing) or targeted (ddPCR, droplet digital PCR) methodologies. Conventional and molecular diagnosis outcomes were compared. RESULTS: We report that tumor DNA is abundant in CSF close to the tumor, but scanty and mostly below NGS sensitivity threshold in CSF from LP. Indeed, tumor DNA is mostly released by cells invading liquoral spaces, generating a gradient that attenuates by departing from the tumor. Nevertheless, in >60% of LP CSF samples, tumor DNA is sufficient to assess a selected panel of genetic alterations (IDH and TERT promoter mutations, EGFR amplification, CDKN2A/B deletion: ITEC protocol) and MGMT methylation that, combined with imaging, enable tissue-agnostic identification of main glioma molecular subtypes. CONCLUSIONS: This study shows potentialities and limitations of CSF liquid biopsy in achieving molecular characterization of gliomas at first clinical presentation and proposes a protocol to maximize diagnostic information retrievable from CSF DNA.


Asunto(s)
Neoplasias Encefálicas , Glioma , Humanos , Glioma/diagnóstico , Glioma/genética , Glioma/patología , Mutación , Pronóstico , Biopsia Líquida , ADN de Neoplasias , Neoplasias Encefálicas/diagnóstico , Neoplasias Encefálicas/genética , Biomarcadores de Tumor/genética
6.
Cell Rep ; 42(8): 112816, 2023 08 29.
Artículo en Inglés | MEDLINE | ID: mdl-37505981

RESUMEN

Glioblastoma (GBM) is known as an intractable, highly heterogeneous tumor encompassing multiple subclones, each supported by a distinct glioblastoma stem cell (GSC). The contribution of GSC genetic and transcriptional heterogeneity to tumor subclonal properties is debated. In this study, we describe the systematic derivation, propagation, and characterization of multiple distinct GSCs from single, treatment-naive GBMs (GSC families). The tumorigenic potential of each GSC better correlates with its transcriptional profile than its genetic make-up, with classical GSCs being inherently more aggressive and mesenchymal more dependent on exogenous growth factors across multiple GBMs. These GSCs can segregate and recapitulate different histopathological aspects of the same GBM, as shown in a paradigmatic tumor with two histopathologically distinct components, including a conventional GBM and a more aggressive primitive neuronal component. This study provides a resource for investigating how GSCs with distinct genetic and/or phenotypic features contribute to individual GBM heterogeneity and malignant escalation.


Asunto(s)
Neoplasias Encefálicas , Glioblastoma , Humanos , Glioblastoma/patología , Neoplasias Encefálicas/metabolismo , Amplificación de Genes , Células Madre Neoplásicas/metabolismo , Carcinogénesis/patología , Línea Celular Tumoral
7.
Oncologist ; 16(10): 1440-50, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-21948653

RESUMEN

Meningiomas are the most frequent intracranial tumors. Surgery can be curative, but recurrences are possible. We performed gene expression analyses and loss of heterozygosity (LOH) studies looking for new markers predicting the recurrence risk. We analyzed expression profiles of 23 meningiomas (10 grade I, 10 grade II, and 3 grade III) and validated the data using quantitative polymerase chain reaction (qPCR). We performed LOH analysis on 40 meningiomas, investigating chromosomal regions on 1p, 9p, 10q, 14q, and 22q. We found 233 and 268 probe sets to be significantly down- and upregulated, respectively, in grade II or III meningiomas. Genes downregulated in high-grade meningiomas were overrepresented on chromosomes 1, 6, 9, 10, and 14. Based on functional enrichment analysis, we selected LIM domain and actin binding 1 (LIMA1), tissue inhibitor of metalloproteinases 3 (TIMP3), cyclin-dependent kinases regulatory subunit 2 (CKS2), leptin receptor (LEPR), and baculoviral inhibitor of apoptosis repeat-containing 5 (BIRC5) for validation using qPCR and confirmed their differential expression in the two groups of tumors. We calculated ΔCt values of CKS2 and LEPR and found that their differential expression (C-L index) was significantly higher in grade I than in grade II or III meningiomas (p < .0001). Interestingly, the C-L index of nine grade I meningiomas from patients who relapsed in <5 years was significantly lower than in grade I meningiomas from patients who did not relapse. These findings indicate that the C-L index may be relevant to define the progression risk in meningioma patients, helping guide their clinical management. A prospective analysis on a larger number of cases is warranted.


Asunto(s)
Biomarcadores de Tumor/genética , Proteínas Portadoras/genética , Proteínas de Ciclo Celular/genética , Neoplasias Meníngeas/genética , Meningioma/genética , Recurrencia Local de Neoplasia/genética , Proteínas Quinasas/genética , Receptores de Leptina/genética , Biomarcadores de Tumor/metabolismo , Quinasas CDC2-CDC28 , Proteínas Portadoras/biosíntesis , Proteínas de Ciclo Celular/biosíntesis , Progresión de la Enfermedad , Femenino , Expresión Génica , Humanos , Inmunohistoquímica , Pérdida de Heterocigocidad , Masculino , Neoplasias Meníngeas/metabolismo , Neoplasias Meníngeas/patología , Meningioma/metabolismo , Meningioma/patología , Análisis por Micromatrices , Clasificación del Tumor , Recurrencia Local de Neoplasia/patología , Análisis de Secuencia por Matrices de Oligonucleótidos , Proteínas Quinasas/biosíntesis , Reacción en Cadena en Tiempo Real de la Polimerasa , Receptores de Leptina/biosíntesis
8.
Cell Rep ; 36(4): 109455, 2021 07 27.
Artículo en Inglés | MEDLINE | ID: mdl-34320350

RESUMEN

In glioblastoma (GBM), the most frequent and lethal brain tumor, therapies suppressing recurrently altered signaling pathways failed to extend survival. However, in patient subsets, specific genetic lesions can confer sensitivity to targeted agents. By exploiting an integrated model based on patient-derived stem-like cells, faithfully recapitulating the original GBMs in vitro and in vivo, here, we identify a human GBM subset (∼9% of all GBMs) characterized by ERBB3 overexpression and nuclear accumulation. ERBB3 overexpression is driven by inheritable promoter methylation or post-transcriptional silencing of the oncosuppressor miR-205 and sustains the malignant phenotype. Overexpressed ERBB3 behaves as a specific signaling platform for fibroblast growth factor receptor (FGFR), driving PI3K/AKT/mTOR pathway hyperactivation, and overall metabolic upregulation. As a result, ERBB3 inhibition by specific antibodies is lethal for GBM stem-like cells and xenotransplants. These findings highlight a subset of patients eligible for ERBB3-targeted therapy.


Asunto(s)
Glioblastoma/genética , MicroARNs/metabolismo , Receptor ErbB-3/metabolismo , Anticuerpos/metabolismo , Apoptosis , Línea Celular Tumoral , Factor 2 de Crecimiento de Fibroblastos , Regulación Neoplásica de la Expresión Génica , Genes Supresores de Tumor , Humanos , MicroARNs/genética , Oligodendroglía/metabolismo , Fosfatidilinositol 3-Quinasas/metabolismo , Pronóstico , Proteínas Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Receptor ErbB-3/antagonistas & inhibidores , Receptores de Factores de Crecimiento de Fibroblastos/metabolismo , Transducción de Señal , Esferoides Celulares/patología , Serina-Treonina Quinasas TOR/metabolismo
9.
Crit Rev Oncol Hematol ; 146: 102879, 2020 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-32005411

RESUMEN

Cell-free circulating tumor DNA (ct-DNA) reflecting the whole tumor spatial and temporal heterogeneity currently represents the most promising candidate for liquid biopsy strategy in glioma. Unlike other solid tumors, it is now widely accepted that the best source of ct-DNA for glioma patients is the cerebrospinal fluid, since blood levels are usually low and detectable only in few cases. A cerebrospinal fluid ct-DNA liquid biopsy approach may virtually support all the stages of glioma management, from facilitating molecular diagnosis when surgery is not feasible, to monitoring tumor response, identifying early recurrence, tracking longitudinal genomic evolution, providing a new molecular characterization at recurrence and allowing patient selection for targeted therapies. This review traces the history of ct-DNA liquid biopsy in the field of diffuse malignant gliomas, describes its current status and analyzes what are the future perspectives and pitfalls of this potentially revolutionary molecular tool.


Asunto(s)
Biomarcadores de Tumor/metabolismo , Neoplasias Encefálicas/líquido cefalorraquídeo , ADN Tumoral Circulante/líquido cefalorraquídeo , ADN de Neoplasias/metabolismo , Glioma/líquido cefalorraquídeo , Biopsia Líquida/métodos , Células Neoplásicas Circulantes/metabolismo , Biomarcadores de Tumor/genética , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/patología , ADN de Neoplasias/genética , Genes Relacionados con las Neoplasias/genética , Glioblastoma/líquido cefalorraquídeo , Glioblastoma/genética , Glioblastoma/patología , Glioma/genética , Glioma/patología , Humanos , Mutación , Clasificación del Tumor , Recurrencia Local de Neoplasia/diagnóstico , Recurrencia Local de Neoplasia/genética , Recurrencia Local de Neoplasia/metabolismo , Células Neoplásicas Circulantes/patología
10.
Neurogenetics ; 9(2): 95-100, 2008 May.
Artículo en Inglés | MEDLINE | ID: mdl-18196300

RESUMEN

Neurofibromatosis type 1 (NF1) is a common genetic disease caused by haploinsufficiency of the NF1 tumor-suppressor gene. Different pathogenetic mechanisms have been identified, with the majority (95%) causing intragenic lesions. Single or multiexon NF1 copy number changes occur in about 2% of patients, but little is known about the molecular mechanisms behind these intragenic deletions. We report here on the molecular characterization of a novel NF1 multiexonic deletion. The application of a multidisciplinary approach including multiplex ligation-dependent probe amplification, allelic segregation analysis, and fluorescent in situ hybridization allowed us to map the breakpoints in IVS27b and IVS48. Furthermore, the breakpoint junction was characterized by sequencing. Using bioinformatic analysis, we identified some recombinogenic motifs in close proximity to the centromeric and telomeric breakpoints and predicted the presence of a mutated messenger ribonucleic acid, which was deleted between exons 28 and 48 and encodes a neurofibromin that lacks some domains essential for its function. Through reverse transcriptase-polymerase chain reaction, the expression of the mutated allele was verified, showing the junction between exons 27b and 49 and, as expected, was not subjected to nonsense-mediated decay. Multiexonic deletions represent 2% of NF1 mutations, and until now, the breakpoint has been identified in only a few cases. The fine characterization of multiexonic deletions broadens the mutational repertoire of the NF1 gene, allowing for the identification of different pathogenetic mechanisms causing NF1.


Asunto(s)
Genes de Neurofibromatosis 1 , Neurofibromatosis 1/genética , Eliminación de Secuencia , Adolescente , Alelos , Secuencia de Aminoácidos , Secuencia de Bases , Cromosomas Humanos Par 17/genética , Roturas del ADN , Exones , Expresión Génica , Humanos , Hibridación Fluorescente in Situ , Masculino , Datos de Secuencia Molecular
11.
Neuro Oncol ; 10(1): 52-60, 2008 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-18094369

RESUMEN

Chordomas are rare embryogenetic tumors, arising from remnants of the notochord, characterized by local invasiveness and variable tendency for recurrence. No molecular markers are currently used in a clinical setting to distinguish chordomas with an indolent or an aggressive pattern. Among the genetic lesions observed in this tumor, one of the most commonly detected is 1p loss. In a previous study we observed 1p36 loss of heterozygosity (LOH) in 85% of the analyzed chordomas. We studied a group of 16 homogeneously treated skull base chordomas (SBCs), reporting 1p36 LOH in 75% of them and determining the expression pattern of eight apoptotic genes mapped at 1p36. No tumors shared a common expression profile with nucleus pulposus, which is considered the only adult normal tissue deriving from notochord. In particular, tumor necrosis factor receptor superfamily genes TNFRSF8, TNFRSF9, and TNFRSF14 were differently expressed compared with control in a higher percentage of tumors (40%-53%) than were the remaining analyzed genes, suggesting that the deregulation of these three genes might have a role in chordoma tumorigenesis. The presence/absence of LOH and the expression/nonexpression of each apoptotic gene were studied in a survival analysis. Our results suggest that the lack of 1p36 LOH or the presence of TNFRSF8 expression might be associated with a better prognosis in patients with SBCs.


Asunto(s)
Biomarcadores de Tumor/genética , Cordoma/genética , Cromosomas Humanos Par 1/genética , Neoplasias de la Base del Cráneo/genética , Adulto , Anciano , Cordoma/mortalidad , Cordoma/patología , Femenino , Expresión Génica , Perfilación de la Expresión Génica , Humanos , Estimación de Kaplan-Meier , Antígeno Ki-1/genética , Antígeno Ki-1/metabolismo , Pérdida de Heterocigocidad , Masculino , Persona de Mediana Edad , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Neoplasias de la Base del Cráneo/mortalidad , Neoplasias de la Base del Cráneo/patología , Análisis de Supervivencia , Tasa de Supervivencia , Miembro 14 de la Superfamilia de Ligandos de Factores de Necrosis Tumoral/genética , Miembro 14 de la Superfamilia de Ligandos de Factores de Necrosis Tumoral/metabolismo , Miembro 9 de la Superfamilia de Receptores de Factores de Necrosis Tumoral/genética , Miembro 9 de la Superfamilia de Receptores de Factores de Necrosis Tumoral/metabolismo
12.
Clin Cancer Res ; 24(4): 807-820, 2018 02 15.
Artículo en Inglés | MEDLINE | ID: mdl-28974546

RESUMEN

Purpose: Patient-derived xenografts ("xenopatients") of colorectal cancer metastases have been essential to identify genetic determinants of resistance to the anti-EGFR antibody cetuximab and to explore new therapeutic strategies. From xenopatients, a genetically annotated collection of stem-like cultures ("xenospheres") was generated and characterized for response to targeted therapies.Experimental Design: Xenospheres underwent exome-sequencing analysis, gene expression profile, and in vitro targeted treatments to assess genetic, biological, and pharmacologic correspondence with xenopatients, and to investigate nongenetic biomarkers of therapeutic resistance. The outcome of EGFR family inhibition was tested in an NRG1-expressing in vivo model.Results: Xenospheres faithfully retained the genetic make-up of their matched xenopatients over in vitro and in vivo passages. Frequent and rare genetic lesions triggering primary resistance to cetuximab through constitutive activation of the RAS signaling pathway were conserved, as well as the vulnerability to their respective targeted treatments. Xenospheres lacking such alterations (RASwt) were highly sensitive to cetuximab, but were protected by ligands activating the EGFR family, mostly NRG1. Upon reconstitution of NRG1 expression, xenospheres displayed increased tumorigenic potential in vivo and generated tumors completely resistant to cetuximab, and sensitive only to comprehensive EGFR family inhibition.Conclusions: Xenospheres are a reliable model to identify both genetic and nongenetic mechanisms of response and resistance to targeted therapies in colorectal cancer. In the absence of RAS pathway mutations, NRG1 and other EGFR ligands can play a major role in conferring primary cetuximab resistance, indicating that comprehensive inhibition of the EGFR family is required to achieve a significant therapeutic response. Clin Cancer Res; 24(4); 807-20. ©2017 AACRSee related commentary by Napolitano and Ciardiello, p. 727.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/farmacología , Neoplasias del Colon/tratamiento farmacológico , Resistencia a Antineoplásicos/genética , Receptores ErbB/antagonistas & inhibidores , Células Madre Neoplásicas/efectos de los fármacos , Ensayos Antitumor por Modelo de Xenoinjerto/métodos , Animales , Cetuximab/administración & dosificación , Neoplasias del Colon/genética , Neoplasias del Colon/patología , Receptores ErbB/genética , Receptores ErbB/metabolismo , Perfilación de la Expresión Génica/métodos , Humanos , Ratones Endogámicos NOD , Ratones SCID , Terapia Molecular Dirigida/métodos , Células Madre Neoplásicas/metabolismo , Carga Tumoral/efectos de los fármacos , Carga Tumoral/genética , Células Tumorales Cultivadas , Secuenciación del Exoma/métodos
13.
BMC Evol Biol ; 7: 39, 2007 Mar 14.
Artículo en Inglés | MEDLINE | ID: mdl-17359533

RESUMEN

BACKGROUND: In order to obtain insights into the functionality of the human 4q35.2 domain harbouring the facioscapulohumeral muscular dystrophy (FSHD) locus, we investigated in African apes genomic and chromatin organisations, and the nuclear topology of orthologous regions. RESULTS: A basic block consisting of short D4Z4 arrays (10-15 repeats), 4q35.2 specific sequences, and approximately 35 kb of interspersed repeats from different LINE subfamilies was repeated at least twice in the gorilla 4qter. This genomic organisation has undergone evolutionary remodelling, leading to the single representation of both the D4Z4 array and LINE block in chimpanzee, and the loss of the LINE block in humans. The genomic remodelling has had an impact on 4qter chromatin organisation, but not its interphase nuclear topology. In comparison with humans, African apes show very low or undetectable levels of FRG1 and FRG2 histone 4 acetylation and gene transcription, although histone deacetylase inhibition restores gene transcription to levels comparable with those of human cells, thus indicating that the 4qter region is capable of acquiring a more open chromatin structure. Conversely, as in humans, the 4qter region in African apes has a very peripheral nuclear localisation. CONCLUSION: The 4q subtelomere has undergone substantial genomic changes during evolution that have had an impact on chromatin condensation and the region's transcriptional regulation. Consequently, the 4qter genes in African apes and humans seem to be subjected to a different strategy of regulation in which LINE and D4Z4 sequences may play a pivotal role. However, the effect of peripheral nuclear anchoring of 4qter on these regulation mechanisms is still unclear. The observed differences in the regulation of 4qter gene expression between African apes and humans suggest that the human 4q35.2 locus has acquired a novel functional relevance.


Asunto(s)
Cromosomas Humanos Par 4 , Cromosomas de los Mamíferos , Gorilla gorilla/genética , Distrofia Muscular Facioescapulohumeral/genética , Pan troglodytes/genética , Animales , Southern Blotting , Cromatina/genética , Clonación Molecular , Evolución Molecular , Marcadores Genéticos , Biblioteca Genómica , Humanos , Hibridación Fluorescente in Situ , Mapeo Físico de Cromosoma , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
14.
Oncol Rep ; 18(1): 249-52, 2007 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-17549375

RESUMEN

Chordomas are rare neoplasms arising along the axial skeleton. Up to now, the most suitable therapeutic approach is based on a combination of surgical excision and radiotherapy. Chemotherapy in not applied due to its reported low efficacy. Recently, evidence on the efficacy of Imatinib mesylate in two patients has been reported. We analyzed 14 chordoma samples for the expression of the Imatinib mesylate targets by means of RT-PCR and immunohistochemistry and found that PDGFR alpha and PDGFR beta are in some cases expressed in neoplastic cells, while the stromal counterpart of the same tumor shows the above receptors. Findings on the PDGFA/PDGFB expression suggest a receptor-activated status. Our study provides new insights into the specific localization of Imatinib mesylate targets in skull base chordomas that could be taken into account for the setting up of a pharmacological treatment for this tumor.


Asunto(s)
Cordoma/metabolismo , Piperazinas/uso terapéutico , Proteínas Proto-Oncogénicas c-kit/metabolismo , Pirimidinas/uso terapéutico , Receptor alfa de Factor de Crecimiento Derivado de Plaquetas/metabolismo , Receptor beta de Factor de Crecimiento Derivado de Plaquetas/metabolismo , Neoplasias de la Base del Cráneo/metabolismo , Adulto , Anciano , Antineoplásicos/uso terapéutico , Benzamidas , Cordoma/genética , Cordoma/patología , Femenino , Humanos , Mesilato de Imatinib , Técnicas para Inmunoenzimas , Masculino , Persona de Mediana Edad , Proteínas Proto-Oncogénicas c-kit/genética , ARN Mensajero/genética , ARN Mensajero/metabolismo , ARN Neoplásico/genética , ARN Neoplásico/metabolismo , Proteínas Tirosina Quinasas Receptoras/antagonistas & inhibidores , Receptor alfa de Factor de Crecimiento Derivado de Plaquetas/genética , Receptor beta de Factor de Crecimiento Derivado de Plaquetas/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Neoplasias de la Base del Cráneo/genética , Neoplasias de la Base del Cráneo/patología , Células del Estroma/metabolismo , Células del Estroma/patología
15.
Nat Commun ; 8: 15107, 2017 05 31.
Artículo en Inglés | MEDLINE | ID: mdl-28561063

RESUMEN

Stromal content heavily impacts the transcriptional classification of colorectal cancer (CRC), with clinical and biological implications. Lineage-dependent stromal transcriptional components could therefore dominate over more subtle expression traits inherent to cancer cells. Since in patient-derived xenografts (PDXs) stromal cells of the human tumour are substituted by murine counterparts, here we deploy human-specific expression profiling of CRC PDXs to assess cancer-cell intrinsic transcriptional features. Through this approach, we identify five CRC intrinsic subtypes (CRIS) endowed with distinctive molecular, functional and phenotypic peculiarities: (i) CRIS-A: mucinous, glycolytic, enriched for microsatellite instability or KRAS mutations; (ii) CRIS-B: TGF-ß pathway activity, epithelial-mesenchymal transition, poor prognosis; (iii) CRIS-C: elevated EGFR signalling, sensitivity to EGFR inhibitors; (iv) CRIS-D: WNT activation, IGF2 gene overexpression and amplification; and (v) CRIS-E: Paneth cell-like phenotype, TP53 mutations. CRIS subtypes successfully categorize independent sets of primary and metastatic CRCs, with limited overlap on existing transcriptional classes and unprecedented predictive and prognostic performances.


Asunto(s)
Neoplasias Colorrectales/clasificación , Neoplasias Colorrectales/genética , Células del Estroma/metabolismo , Transcriptoma , Animales , Antineoplásicos Inmunológicos/farmacología , Linaje de la Célula , Cetuximab/farmacología , Neoplasias Colorrectales/patología , Transición Epitelial-Mesenquimal , Receptores ErbB/antagonistas & inhibidores , Receptores ErbB/inmunología , Receptores ErbB/metabolismo , Femenino , Perfilación de la Expresión Génica , Genes p53 , Glucólisis , Xenoinjertos , Humanos , Factor II del Crecimiento Similar a la Insulina/genética , Masculino , Ratones , Inestabilidad de Microsatélites , Mutación , Pronóstico , Transducción de Señal , Células del Estroma/patología , Factor de Crecimiento Transformador beta/metabolismo
16.
EMBO Mol Med ; 8(5): 550-68, 2016 05.
Artículo en Inglés | MEDLINE | ID: mdl-27138567

RESUMEN

Glioblastoma (GBM) contains stem-like cells (GSCs) known to be resistant to ionizing radiation and thus responsible for therapeutic failure and rapidly lethal tumor recurrence. It is known that GSC radioresistance relies on efficient activation of the DNA damage response, but the mechanisms linking this response with the stem status are still unclear. Here, we show that the MET receptor kinase, a functional marker of GSCs, is specifically expressed in a subset of radioresistant GSCs and overexpressed in human GBM recurring after radiotherapy. We elucidate that MET promotes GSC radioresistance through a novel mechanism, relying on AKT activity and leading to (i) sustained activation of Aurora kinase A, ATM kinase, and the downstream effectors of DNA repair, and (ii) phosphorylation and cytoplasmic retention of p21, which is associated with anti-apoptotic functions. We show that MET pharmacological inhibition causes DNA damage accumulation in irradiated GSCs and their depletion in vitro and in GBMs generated by GSC xenotransplantation. Preclinical evidence is thus provided that MET inhibitors can radiosensitize tumors and convert GSC-positive selection, induced by radiotherapy, into GSC eradication.


Asunto(s)
Glioblastoma/radioterapia , Proteínas Proto-Oncogénicas c-met/antagonistas & inhibidores , Células Madre/fisiología , Células Madre/efectos de la radiación , Animales , Proteínas de la Ataxia Telangiectasia Mutada/metabolismo , Aurora Quinasa A/metabolismo , Supervivencia Celular , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/metabolismo , Reparación del ADN , Xenoinjertos , Humanos , Ratones , Proteína Oncogénica v-akt/metabolismo
17.
Cancer Res ; 74(6): 1857-69, 2014 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-24448239

RESUMEN

Metastatic colorectal cancer remains largely incurable, although in a subset of patients, survival is prolonged by new targeting agents such as anti-EGF receptor (anti-EGFR) antibodies. This disease is believed to be supported by a subpopulation of stem-like cells termed colon cancer-initiating cell (CCIC), which may also confer therapeutic resistance. However, how CCICs respond to EGFR inhibition has not been fully characterized. To explore this question, we systematically generated CCICs through spheroid cultures of patient-derived xenografts of metastatic colorectal cancer. These cultures, termed "xenospheres," were capable of long-term self-propagation in vitro and phenocopied the original patient tumors in vivo, thus operationally defining CCICs. Xenosphere CCICs retained the genetic determinants for EGFR therapeutic response in vitro and in xenografts; like the original tumors, xenospheres harboring a mutated KRAS gene were resistant to EGFR therapy, whereas those harboring wild-type RAS pathway genes (RAS(wt)) were sensitive. Notably, the effects of EGFR inhibition in sensitive CCICs could be counteracted by cytokines secreted by cancer-associated fibroblasts. In particular, we found that the MET receptor ligand hepatocyte growth factor (HGF) was especially active in supporting in vitro CCIC proliferation and resistance to EGFR inhibition. Ectopic production of human HGF in CCIC xenografts rendered the xenografts susceptible to MET inhibition, which sensitized the response to EGFR therapy. By showing that RAS(wt) CCICs rely on both EGFR and MET signaling, our results offer a strong preclinical proof-of-concept for concurrent targeting of these two pathways in the clinical setting.


Asunto(s)
Anticuerpos Monoclonales Humanizados/farmacología , Antineoplásicos/farmacología , Neoplasias del Colon/enzimología , Receptores ErbB/antagonistas & inhibidores , Células Madre Neoplásicas/enzimología , Proteínas Proto-Oncogénicas c-met/metabolismo , Animales , Proliferación Celular , Supervivencia Celular , Cetuximab , Neoplasias del Colon/tratamiento farmacológico , Neoplasias del Colon/patología , Resistencia a Antineoplásicos , Sinergismo Farmacológico , Receptores ErbB/metabolismo , Femenino , Factor de Crecimiento de Hepatocito/fisiología , Humanos , Ratones , Ratones Endogámicos NOD , Ratones SCID , Ratones Transgénicos , Células Madre Neoplásicas/efectos de los fármacos , Proteínas Proto-Oncogénicas c-met/antagonistas & inhibidores , Transducción de Señal , Esferoides Celulares/enzimología , Carga Tumoral/efectos de los fármacos , Células Tumorales Cultivadas , Ensayos Antitumor por Modelo de Xenoinjerto
18.
Cancer Res ; 72(17): 4537-50, 2012 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-22738909

RESUMEN

The existence of treatment-resistant cancer stem cells contributes to the aggressive phenotype of glioblastoma. However, the molecular alterations that drive stem cell proliferation in these tumors remain unknown. In this study, we found that expression of the MET oncogene was associated with neurospheres expressing the gene signature of mesenchymal and proneural subtypes of glioblastoma. Met expression was almost absent from neurospheres expressing the signature of the classical subtype and was mutually exclusive with amplification and expression of the EGF receptor (EGFR) gene. Met-positive and Met-negative neurospheres displayed distinct growth factor requirements, differentiated along divergent pathways, and generated tumors with distinctive features. The Met(high) subpopulation within Met-pos neurospheres displayed clonogenic potential and long-term self-renewal ability in vitro and enhanced growth kinetics in vivo. In Met(high) cells, the Met ligand HGF further sustained proliferation, clonogenicity, expression of self-renewal markers, migration, and invasion in vitro. Together, our findings suggest that Met is a functional marker of glioblastoma stem cells and a candidate target for identification and therapy of a subset of glioblastomas.


Asunto(s)
Glioblastoma/genética , Glioblastoma/metabolismo , Células Madre Neoplásicas/metabolismo , Proteínas Proto-Oncogénicas c-met/genética , Adolescente , Adulto , Anciano , Animales , Biomarcadores de Tumor/genética , Biomarcadores de Tumor/metabolismo , Proliferación Celular , Transformación Celular Neoplásica/genética , Receptores ErbB/genética , Receptores ErbB/metabolismo , Femenino , Perfilación de la Expresión Génica , Glioblastoma/mortalidad , Glioblastoma/patología , Humanos , Ligandos , Masculino , Ratones , Ratones Endogámicos NOD , Ratones SCID , Persona de Mediana Edad , Células Madre Neoplásicas/patología , Proteínas Proto-Oncogénicas c-met/metabolismo , Transcripción Genética , Adulto Joven
19.
Neuro Oncol ; 12(4): 377-88, 2010 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-20308315

RESUMEN

Pilot data showed that adding intratumoral (IT) injection of dendritic cells (DCs) prolongs survival of patients affected by glioblastoma multiforme (GBM) treated by subcutaneous (SC) delivery of DCs. Using a murine model resembling GBM, we investigated the immunological mechanisms underlying this effect. C57BL6/N mice received brain injections of GL261 glioma cells. Seven days later, mice were treated by 3 SC injections of DCs with or without 1 IT injection of DCs. DC maturation, induced by pulsing with GL261 lysates, was necessary to develop effective immune responses. IT injection of pulsed (pDC), but not unpulsed DCs (uDC), increased significantly the survival, either per se or in combination with SC-pDC (P < .001 vs controls). Mice treated by IT-pDC plus SC-pDC survived longer than mice treated by SC-pDC only (P = .03). Injected pDC were detectable in tumor parenchyma, but not in cervical lymph nodes. In gliomas injected with IT-pDC, CD8+ cells were significantly more abundant and Foxp3+ cells were significantly less abundant than in other groups. Using real-time polymerase chain reaction, we also found enhanced expression of IFN-gamma and TNF-alpha and decreased expression of transforming growth factor-beta (TGF-beta) and Foxp3 in mice treated with SC-pDC and IT-pDC. In vitro, pDC produced more TNF-alpha than uDC: addition of TNF-alpha to the medium decreased the proliferation of glioma cells. Overall, the results suggest that IT-pDC potentiates the anti-tumor immune response elicited by SC-pDC by pro-immune modulation of cytokines in the tumor microenvironment, decrease of Treg cells, and direct inhibition of tumor proliferation by TNF-alpha.


Asunto(s)
Neoplasias Encefálicas/prevención & control , Células Dendríticas/trasplante , Glioma/terapia , Inmunoterapia , Vacunación , Animales , Western Blotting , Neoplasias Encefálicas/inmunología , Neoplasias Encefálicas/metabolismo , Pruebas Inmunológicas de Citotoxicidad , Células Dendríticas/inmunología , Ensayo de Inmunoadsorción Enzimática , Citometría de Flujo , Glioma/inmunología , Técnicas para Inmunoenzimas , Interferón gamma/metabolismo , Ganglios Linfáticos , Linfocitos Infiltrantes de Tumor/inmunología , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , ARN Mensajero/genética , ARN Mensajero/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Tasa de Supervivencia , Linfocitos T/inmunología , Factor de Necrosis Tumoral alfa/genética , Factor de Necrosis Tumoral alfa/inmunología , Factor de Necrosis Tumoral alfa/metabolismo
20.
Genomics ; 85(2): 273-9, 2005 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-15676286

RESUMEN

Neurofibromatosis type 1 (NF1) microdeletion syndrome is caused by haploinsufficiency of the NF1 gene and of gene(s) located in adjacent flanking regions. Most of the NF1 deletions originate by nonallelic homologous recombination between repeated sequences (REP-P and -M) mapped to 17q11.2, while a few uncommon deletions show unusual breakpoints. We characterized an uncommon 1.5-Mb deletion of an NF1 patient displaying a mild phenotype. We applied high-resolution FISH analysis allowing us to obtain the sequence of the first junction fragment of an uncommon deletion showing the telomeric breakpoint inside the IVS23a of the NF1 gene. Sequence analysis of the centromeric and telomeric boundaries revealed that the breakpoints were present in the AluJb and AluSx regions, respectively, showing 85% homology. The centromeric breakpoint is localized inside a chi-like element; a few copies of this sequence are also located very close to both breakpoints. The in silico analysis of the breakpoint intervals, aimed at identifying consensus sequences of several motifs usually involved in deletions and translocations, suggests that Alu sequences, probably associated with the chi-like element, might be the only recombinogenic motif directly mediating this large deletion.


Asunto(s)
Elementos Alu , Genes de Neurofibromatosis 1 , Neurofibromatosis 1/genética , Eliminación de Secuencia , Secuencia de Bases , Rotura Cromosómica , Cromosomas Humanos Par 17 , Humanos , Hibridación Fluorescente in Situ , Datos de Secuencia Molecular
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA