Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
1.
Nature ; 483(7391): 589-93, 2012 Feb 22.
Artigo em Inglês | MEDLINE | ID: mdl-22367537

RESUMO

Neuroblastoma is a childhood tumour of the peripheral sympathetic nervous system. The pathogenesis has for a long time been quite enigmatic, as only very few gene defects were identified in this often lethal tumour. Frequently detected gene alterations are limited to MYCN amplification (20%) and ALK activations (7%). Here we present a whole-genome sequence analysis of 87 neuroblastoma of all stages. Few recurrent amino-acid-changing mutations were found. In contrast, analysis of structural defects identified a local shredding of chromosomes, known as chromothripsis, in 18% of high-stage neuroblastoma. These tumours are associated with a poor outcome. Structural alterations recurrently affected ODZ3, PTPRD and CSMD1, which are involved in neuronal growth cone stabilization. In addition, ATRX, TIAM1 and a series of regulators of the Rac/Rho pathway were mutated, further implicating defects in neuritogenesis in neuroblastoma. Most tumours with defects in these genes were aggressive high-stage neuroblastomas, but did not carry MYCN amplifications. The genomic landscape of neuroblastoma therefore reveals two novel molecular defects, chromothripsis and neuritogenesis gene alterations, which frequently occur in high-risk tumours.


Assuntos
Cromossomos Humanos/genética , Neuritos/metabolismo , Neuroblastoma/genética , Neuroblastoma/patologia , Envelhecimento/genética , Análise por Conglomerados , DNA Helicases/genética , Análise Mutacional de DNA , Regulação Neoplásica da Expressão Gênica , Genoma Humano/genética , Cones de Crescimento/metabolismo , Cones de Crescimento/patologia , Fatores de Troca do Nucleotídeo Guanina/genética , Humanos , Mutação , Estadiamento de Neoplasias , Neuroblastoma/diagnóstico , Neuroblastoma/metabolismo , Proteínas Nucleares/genética , Prognóstico , Proteína 1 Indutora de Invasão e Metástase de Linfoma de Células T , Proteína Nuclear Ligada ao X , Proteínas rac de Ligação ao GTP/metabolismo , Proteínas rho de Ligação ao GTP/metabolismo
2.
Proc Natl Acad Sci U S A ; 109(47): 19190-5, 2012 Nov 20.
Artigo em Inglês | MEDLINE | ID: mdl-23091029

RESUMO

Neuroblastoma is a pediatric tumor of the sympathetic nervous system. MYCN (V-myc myelocytomatosis viral-related oncogene, neuroblastoma derived [avian]) is amplified in 20% of neuroblastomas, and these tumors carry a poor prognosis. However, tumors without MYCN amplification also may have a poor outcome. Here, we identified downstream targets of MYCN by shRNA-mediated silencing MYCN in neuroblastoma cells. From these targets, 157 genes showed an expression profile correlating with MYCN mRNA levels in NB88, a series of 88 neuroblastoma tumors, and therefore represent in vivo relevant MYCN pathway genes. This 157-gene signature identified very poor prognosis tumors in NB88 and independent neuroblastoma cohorts and was more powerful than MYCN amplification or MYCN expression alone. Remarkably, this signature also identified poor outcome of a group of tumors without MYCN amplification. Most of these tumors have low MYCN mRNA levels but high nuclear MYCN protein levels, suggesting stabilization of MYCN at the protein level. One tumor has an MYC amplification and high MYC expression. Chip-on-chip analyses showed that most genes in this signature are directly regulated by MYCN. MYCN induces genes functioning in cell cycle and DNA repair while repressing neuronal differentiation genes. The functional MYCN-157 signature recognizes classical neuroblastoma with MYCN amplification, as well as a newly identified group marked by MYCN protein stabilization.


Assuntos
Amplificação de Genes/genética , Perfilação da Expressão Gênica , Neuroblastoma/genética , Proteínas Nucleares/genética , Proteínas Oncogênicas/genética , Diferenciação Celular/genética , Análise por Conglomerados , Reparo do DNA/genética , Regulação Neoplásica da Expressão Gênica , Genes Neoplásicos/genética , Humanos , Proteína Proto-Oncogênica N-Myc , Neuroblastoma/patologia , Neurônios/patologia , Proteínas Nucleares/metabolismo , Proteínas Oncogênicas/metabolismo , Prognóstico , Regiões Promotoras Genéticas/genética , Ligação Proteica/genética , Resultado do Tratamento , Regulação para Cima/genética
3.
Cancer Res ; 82(3): 484-496, 2022 02 01.
Artigo em Inglês | MEDLINE | ID: mdl-34853072

RESUMO

Cancer therapy frequently fails due to the emergence of resistance. Many tumors include phenotypically immature tumor cells, which have been implicated in therapy resistance. Neuroblastoma cells can adopt a lineage-committed adrenergic (ADRN) or an immature mesenchymal (MES) state. They differ in epigenetic landscape and transcription factors, and MES cells are more resistant to chemotherapy. Here we analyzed the response of MES cells to targeted drugs. Activating anaplastic lymphoma kinase (ALK) mutations are frequently found in neuroblastoma and ALK inhibitors (ALKi) are in clinical trials. ALKi treatment of ADRN neuroblastoma cells with a tumor-driving ALK mutation induced cell death. Conversely, MES cells did not express either mutant or wild-type ALK and were resistant to ALKi, and MES cells formed tumors that progressed under ALKi therapy. In assessing the role of MES cells in relapse development, TRAIL was identified to specifically induce apoptosis in MES cells and to suppress MES tumor growth. Addition of TRAIL to ALKi treatment of neuroblastoma xenografts delayed relapses in a subset of the animals, suggesting a role for MES cells in relapse formation. While ADRN cells resembled normal embryonal neuroblasts, MES cells resembled immature precursor cells, which also lacked ALK expression. Resistance to targeted drugs can therefore be an intrinsic property of immature cancer cells based on their resemblance to developmental precursors. SIGNIFICANCE: In neuroblastoma, mesenchymal tumor cells lack expression of the tumor-driving ALK oncogene and are resistant to ALKi, but dual treatment with ALKi and mesenchymal cell-targeting TRAIL delays tumor relapse.


Assuntos
Quinase do Linfoma Anaplásico/antagonistas & inibidores , Neuroblastoma/genética , Linhagem Celular Tumoral , Humanos , Neuroblastoma/patologia
4.
Nat Commun ; 10(1): 1530, 2019 04 04.
Artigo em Inglês | MEDLINE | ID: mdl-30948783

RESUMO

Transition between differentiation states in development occurs swift but the mechanisms leading to epigenetic and transcriptional reprogramming are poorly understood. The pediatric cancer neuroblastoma includes adrenergic (ADRN) and mesenchymal (MES) tumor cell types, which differ in phenotype, super-enhancers (SEs) and core regulatory circuitries. These cell types can spontaneously interconvert, but the mechanism remains largely unknown. Here, we unravel how a NOTCH3 intracellular domain reprogrammed the ADRN transcriptional landscape towards a MES state. A transcriptional feed-forward circuitry of NOTCH-family transcription factors amplifies the NOTCH signaling levels, explaining the swift transition between two semi-stable cellular states. This transition induces genome-wide remodeling of the H3K27ac landscape and a switch from ADRN SEs to MES SEs. Once established, the NOTCH feed-forward loop maintains the induced MES state. In vivo reprogramming of ADRN cells shows that MES and ADRN cells are equally oncogenic. Our results elucidate a swift transdifferentiation between two semi-stable epigenetic cellular states.


Assuntos
Neurônios Adrenérgicos/patologia , Reprogramação Celular/genética , Células-Tronco Mesenquimais/patologia , Neuroblastoma/patologia , Receptor Notch3/fisiologia , Neurônios Adrenérgicos/metabolismo , Linhagem Celular Tumoral , Epigênese Genética , Retroalimentação Fisiológica , Perfilação da Expressão Gênica , Regulação Neoplásica da Expressão Gênica , Humanos , Células-Tronco Mesenquimais/metabolismo , Neuroblastoma/metabolismo , Receptor Notch3/genética , Receptor Notch3/metabolismo
5.
Cancer Lett ; 256(2): 218-28, 2007 Oct 28.
Artigo em Inglês | MEDLINE | ID: mdl-17643814

RESUMO

Neuroblastomas are tumors of the developing peripheral sympathetic nervous system, which originates from the neural crest. Twenty percent of neuroblastomas show amplification of the MYCN oncogene, which correlates with poor prognosis. The MYCN transcription factor can activate and repress gene expression. To broaden our insight in the spectrum of genes down-regulated by MYCN, we generated gene expression profiles of the neuroblastoma cell lines SHEP-21N and SKNAS-NmycER, in which MYCN activity can be regulated. In this study, we show that MYCN suppresses the expression of Dickkopf-1 (DKK1) in both cell lines. DKK1 is a potent inhibitor of the wnt/beta-catenin signalling cascade, which is known to function in neural crest cell migration. We generated a DKK1 inducible cell line, IMR32-DKK1, which showed impaired proliferation upon DKK1 expression. Surprisingly, DKK1 expression did not inhibit the canonical wnt/beta-catenin signalling, suggesting a role of DKK1 in an alternative route of the wnt pathway. Gene expression profiling of two IMR32-DKK1 clones showed that only a few genes, amongst which SYNPO2, were up-regulated by DKK1. SYNPO2 encodes an actin-binding protein and was previously found to inhibit proliferation and invasiveness of prostate cancer cells. These results suggest that MYCN might stimulate cell proliferation by inhibiting the expression of DKK1. DKK1 might exert part of its growth suppressive effect by induction of SYNPO2 expression.


Assuntos
Proliferação de Células , Regulação Neoplásica da Expressão Gênica , Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Neuroblastoma/metabolismo , Proteínas Nucleares/metabolismo , Proteínas Oncogênicas/metabolismo , Transdução de Sinais , Regulação para Baixo , Perfilação da Expressão Gênica/métodos , Humanos , Peptídeos e Proteínas de Sinalização Intercelular/genética , Proteínas dos Microfilamentos/metabolismo , Proteína Proto-Oncogênica N-Myc , Neuroblastoma/genética , Neuroblastoma/patologia , Proteínas Nucleares/genética , Análise de Sequência com Séries de Oligonucleotídeos , Proteínas Oncogênicas/genética , Regiões Promotoras Genéticas , RNA Mensageiro/metabolismo , Transdução de Sinais/genética , Fatores de Tempo , Transfecção , Proteínas Wnt/metabolismo , beta Catenina/metabolismo
6.
Cancer Res ; 65(8): 3136-45, 2005 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-15833843

RESUMO

The best studied oncogenic mechanisms are inactivating defects in both alleles of tumor suppressor genes and activating mutations in oncogenes. Chromosomal gains and losses are frequent in human tumors, but for many regions, like 1p36 and 17q in neuroblastoma, no mutated tumor suppressor genes or oncogenes were identified. Amplification of N-myc in neuroblastoma is strongly correlated with loss of 1p36 and gain of 17q. Here we report that N-myc down-regulates the mRNA expression of many genes with a role in cell architecture. One of them is the 1p36 gene Cdc42. Restoring the Cdc42 expression in neuroblastoma cells strongly induced differentiation. N-myc also inhibited Cdc42 functioning at the protein level. This was mediated by nm23-H1 and nm23-H2, which are located in the amplified 17q region. Nm23-H1 and nm23-H2 are strongly up-regulated downstream targets of N-myc. Nm23-H1 was shown to bind Cdc42 and prevented the induction of differentiation. Overexpression of Nm23 due to gain of 17q and induction by N-myc combined with weak expression of Cdc42 due to loss of 1p36 and down-regulation by N-myc can thus block differentiation. Although this marks Cdc42 as a candidate tumor suppressor gene, no mutations were found. Further silencing of Cdc42 by small interfering RNA induced massive apoptosis, indicating that tumor cell survival requires a minimal Cdc42 activity. Three regions of chromosomal gain and loss thus affect genes functioning in one pathway in neuroblastoma. They converge to bring the pathway out of balance and prevent Cdc42 mediated differentiation.


Assuntos
Diferenciação Celular/genética , Genes myc/genética , Neuroblastoma/genética , Núcleosídeo-Difosfato Quinase/genética , Proteína cdc42 de Ligação ao GTP/genética , Divisão Celular/genética , Linhagem Celular Tumoral , Regulação para Baixo , Dosagem de Genes , Regulação Neoplásica da Expressão Gênica , Genes Supressores de Tumor , Guanosina Difosfato/metabolismo , Guanosina Trifosfato/metabolismo , Humanos , Nucleosídeo NM23 Difosfato Quinases , Neuroblastoma/enzimologia , Neuroblastoma/patologia , Neurônios/citologia , Neurônios/fisiologia , Núcleosídeo-Difosfato Quinase/biossíntese , Interferência de RNA , RNA Mensageiro/biossíntese , RNA Mensageiro/genética , Transfecção , Proteína cdc42 de Ligação ao GTP/antagonistas & inibidores , Proteína cdc42 de Ligação ao GTP/biossíntese , Proteína cdc42 de Ligação ao GTP/metabolismo
8.
Nat Genet ; 49(8): 1261-1266, 2017 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-28650485

RESUMO

Neuroblastoma and other pediatric tumors show a paucity of gene mutations, which has sparked an interest in their epigenetic regulation. Several tumor types include phenotypically divergent cells, resembling cells from different lineage development stages. It has been proposed that super-enhancer-associated transcription factor (TF) networks underlie lineage identity, but the role of these enhancers in intratumoral heterogeneity is unknown. Here we show that most neuroblastomas include two types of tumor cells with divergent gene expression profiles. Undifferentiated mesenchymal cells and committed adrenergic cells can interconvert and resemble cells from different lineage differentiation stages. ChIP-seq analysis of isogenic pairs of mesenchymal and adrenergic cells identified a distinct super-enhancer landscape and super-enhancer-associated TF network for each cell type. Expression of the mesenchymal TF PRRX1 could reprogram the super-enhancer and mRNA landscapes of adrenergic cells toward a mesenchymal state. Mesenchymal cells were more chemoresistant in vitro and were enriched in post-therapy and relapse tumors. Two super-enhancer-associated TF networks, which probably mediate lineage control in normal development, thus dominate epigenetic control of neuroblastoma and shape intratumoral heterogeneity.


Assuntos
Diferenciação Celular/genética , Epigênese Genética , Neuroblastoma/genética , Neuroblastoma/patologia , Antígeno AC133/genética , Neurônios Adrenérgicos/citologia , Linhagem Celular Tumoral , Linhagem da Célula , Proteínas de Homeodomínio/genética , Humanos , Mesoderma/citologia , Fatores de Transcrição/metabolismo , Transcriptoma
9.
Nat Genet ; 44(11): 1199-206, 2012 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-23042116

RESUMO

LIN28B regulates developmental processes by modulating microRNAs (miRNAs) of the let-7 family. A role for LIN28B in cancer has been proposed but has not been established in vivo. Here, we report that LIN28B showed genomic aberrations and extensive overexpression in high-risk neuroblastoma compared to several other tumor entities and normal tissues. High LIN28B expression was an independent risk factor for adverse outcome in neuroblastoma. LIN28B signaled through repression of the let-7 miRNAs and consequently resulted in elevated MYCN protein expression in neuroblastoma cells. LIN28B-let-7-MYCN signaling blocked differentiation of normal neuroblasts and neuroblastoma cells. These findings were fully recapitulated in a mouse model in which LIN28B expression in the sympathetic adrenergic lineage induced development of neuroblastomas marked by low let-7 miRNA levels and high MYCN protein expression. Interference with this pathway might offer therapeutic perspectives.


Assuntos
Proteínas de Ligação a DNA/genética , MicroRNAs , Neuroblastoma , Proteínas Nucleares , Proteínas Oncogênicas , Animais , Diferenciação Celular , Proteínas de Ligação a DNA/metabolismo , Regulação Neoplásica da Expressão Gênica , Inativação Gênica , Humanos , Camundongos , Camundongos Transgênicos , MicroRNAs/genética , MicroRNAs/metabolismo , Proteína Proto-Oncogênica N-Myc , Neuroblastoma/genética , Neuroblastoma/metabolismo , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Proteínas Oncogênicas/genética , Proteínas Oncogênicas/metabolismo , Proteínas de Ligação a RNA , Transdução de Sinais
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa