Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
1.
Nature ; 526(7575): 700-4, 2015 Oct 29.
Article in English | MEDLINE | ID: mdl-26466568

ABSTRACT

Neuroblastoma is a malignant paediatric tumour of the sympathetic nervous system. Roughly half of these tumours regress spontaneously or are cured by limited therapy. By contrast, high-risk neuroblastomas have an unfavourable clinical course despite intensive multimodal treatment, and their molecular basis has remained largely elusive. Here we have performed whole-genome sequencing of 56 neuroblastomas (high-risk, n = 39; low-risk, n = 17) and discovered recurrent genomic rearrangements affecting a chromosomal region at 5p15.33 proximal of the telomerase reverse transcriptase gene (TERT). These rearrangements occurred only in high-risk neuroblastomas (12/39, 31%) in a mutually exclusive fashion with MYCN amplifications and ATRX mutations, which are known genetic events in this tumour type. In an extended case series (n = 217), TERT rearrangements defined a subgroup of high-risk tumours with particularly poor outcome. Despite a large structural diversity of these rearrangements, they all induced massive transcriptional upregulation of TERT. In the remaining high-risk tumours, TERT expression was also elevated in MYCN-amplified tumours, whereas alternative lengthening of telomeres was present in neuroblastomas without TERT or MYCN alterations, suggesting that telomere lengthening represents a central mechanism defining this subtype. The 5p15.33 rearrangements juxtapose the TERT coding sequence to strong enhancer elements, resulting in massive chromatin remodelling and DNA methylation of the affected region. Supporting a functional role of TERT, neuroblastoma cell lines bearing rearrangements or amplified MYCN exhibited both upregulated TERT expression and enzymatic telomerase activity. In summary, our findings show that remodelling of the genomic context abrogates transcriptional silencing of TERT in high-risk neuroblastoma and places telomerase activation in the centre of transformation in a large fraction of these tumours.


Subject(s)
Gene Expression Regulation, Neoplastic/genetics , Genome, Human/genetics , Neuroblastoma/genetics , Neuroblastoma/pathology , Recombination, Genetic/genetics , Telomerase/genetics , Telomerase/metabolism , Cell Line, Tumor , Cell Transformation, Neoplastic/genetics , Chromatin/genetics , Chromatin/metabolism , Chromosomes, Human, Pair 5/genetics , DNA Helicases/genetics , DNA Methylation , Enhancer Elements, Genetic/genetics , Enzyme Activation/genetics , Gene Amplification/genetics , Gene Silencing , Humans , Infant , N-Myc Proto-Oncogene Protein , Neuroblastoma/classification , Neuroblastoma/enzymology , Nuclear Proteins/genetics , Oncogene Proteins/genetics , Prognosis , RNA, Messenger/analysis , RNA, Messenger/genetics , Risk , Translocation, Genetic/genetics , Up-Regulation/genetics , X-linked Nuclear Protein
2.
Nat Cancer ; 3(4): 471-485, 2022 04.
Article in English | MEDLINE | ID: mdl-35484422

ABSTRACT

Aberrant expression of MYC transcription factor family members predicts poor clinical outcome in many human cancers. Oncogenic MYC profoundly alters metabolism and mediates an antioxidant response to maintain redox balance. Here we show that MYCN induces massive lipid peroxidation on depletion of cysteine, the rate-limiting amino acid for glutathione (GSH) biosynthesis, and sensitizes cells to ferroptosis, an oxidative, non-apoptotic and iron-dependent type of cell death. The high cysteine demand of MYCN-amplified childhood neuroblastoma is met by uptake and transsulfuration. When uptake is limited, cysteine usage for protein synthesis is maintained at the expense of GSH triggering ferroptosis and potentially contributing to spontaneous tumor regression in low-risk neuroblastomas. Pharmacological inhibition of both cystine uptake and transsulfuration combined with GPX4 inactivation resulted in tumor remission in an orthotopic MYCN-amplified neuroblastoma model. These findings provide a proof of concept of combining multiple ferroptosis targets as a promising therapeutic strategy for aggressive MYCN-amplified tumors.


Subject(s)
Ferroptosis , Neuroblastoma , Cell Death , Child , Cysteine/therapeutic use , Ferroptosis/genetics , Glutathione/therapeutic use , Humans , N-Myc Proto-Oncogene Protein/genetics , Neuroblastoma/genetics
3.
Nat Genet ; 53(5): 683-693, 2021 05.
Article in English | MEDLINE | ID: mdl-33767450

ABSTRACT

Neuroblastoma is a pediatric tumor of the developing sympathetic nervous system. However, the cellular origin of neuroblastoma has yet to be defined. Here we studied the single-cell transcriptomes of neuroblastomas and normal human developing adrenal glands at various stages of embryonic and fetal development. We defined normal differentiation trajectories from Schwann cell precursors over intermediate states to neuroblasts or chromaffin cells and showed that neuroblastomas transcriptionally resemble normal fetal adrenal neuroblasts. Importantly, neuroblastomas with varying clinical phenotypes matched different temporal states along normal neuroblast differentiation trajectories, with the degree of differentiation corresponding to clinical prognosis. Our work highlights the roles of oncogenic MYCN and loss of TFAP2B in blocking differentiation and may provide the basis for designing therapeutic interventions to overcome differentiation blocks.


Subject(s)
Gene Expression Profiling , Neuroblastoma/genetics , Neuroblastoma/pathology , Single-Cell Analysis , Adrenal Glands/embryology , Adrenal Glands/pathology , Cell Differentiation , Cell Line, Tumor , Cohort Studies , Gene Expression Regulation, Neoplastic , Humans , Transcriptome/genetics , Treatment Outcome
4.
Life Sci Alliance ; 4(5)2021 05.
Article in English | MEDLINE | ID: mdl-33658318

ABSTRACT

The migrational propensity of neuroblastoma is affected by cell identity, but the mechanisms behind the divergence remain unknown. Using RNAi and time-lapse imaging, we show that ADRN-type NB cells exhibit RAC1- and kalirin-dependent nucleokinetic (NUC) migration that relies on several integral components of neuronal migration. Inhibition of NUC migration by RAC1 and kalirin-GEF1 inhibitors occurs without hampering cell proliferation and ADRN identity. Using three clinically relevant expression dichotomies, we reveal that most of up-regulated mRNAs in RAC1- and kalirin-GEF1-suppressed ADRN-type NB cells are associated with low-risk characteristics. The computational analysis shows that, in a context of overall gene set poverty, the upregulomes in RAC1- and kalirin-GEF1-suppressed ADRN-type cells are a batch of AU-rich element-containing mRNAs, which suggests a link between NUC migration and mRNA stability. Gene set enrichment analysis-based search for vulnerabilities reveals prospective weak points in RAC1- and kalirin-GEF1-suppressed ADRN-type NB cells, including activities of H3K27- and DNA methyltransferases. Altogether, these data support the introduction of NUC inhibitors into cancer treatment research.


Subject(s)
Guanine Nucleotide Exchange Factors/metabolism , Neuroblastoma/metabolism , Protein Serine-Threonine Kinases/metabolism , rac1 GTP-Binding Protein/metabolism , Adrenergic Neurons/metabolism , Cell Line, Tumor , Cell Movement/genetics , Cells, Cultured , Child, Preschool , Databases, Genetic , Female , Guanine Nucleotide Exchange Factors/physiology , Humans , Male , Neuroblastoma/pathology , Prospective Studies , Protein Serine-Threonine Kinases/physiology , rac1 GTP-Binding Protein/physiology
5.
Nat Cancer ; 2(1): 114-128, 2021 01.
Article in English | MEDLINE | ID: mdl-35121888

ABSTRACT

Half of the children diagnosed with neuroblastoma (NB) have high-risk disease, disproportionately contributing to overall childhood cancer-related deaths. In addition to recurrent gene mutations, there is increasing evidence supporting the role of epigenetic deregulation in disease pathogenesis. Yet, comprehensive cis-regulatory network descriptions from NB are lacking. Here, using genome-wide H3K27ac profiles across 60 NBs, covering the different clinical and molecular subtypes, we identified four major super-enhancer-driven epigenetic subtypes and their underlying master regulatory networks. Three of these subtypes recapitulated known clinical groups; namely, MYCN-amplified, MYCN non-amplified high-risk and MYCN non-amplified low-risk NBs. The fourth subtype, exhibiting mesenchymal characteristics, shared cellular identity with multipotent Schwann cell precursors, was induced by RAS activation and was enriched in relapsed disease. Notably, CCND1, an essential gene in NB, was regulated by both mesenchymal and adrenergic regulatory networks converging on distinct super-enhancer modules. Overall, this study reveals subtype-specific super-enhancer regulation in NBs.


Subject(s)
Neuroblastoma , Child , Humans , Mutation , N-Myc Proto-Oncogene Protein/genetics , Neuroblastoma/genetics , Regulatory Sequences, Nucleic Acid
6.
Nat Commun ; 12(1): 1269, 2021 02 24.
Article in English | MEDLINE | ID: mdl-33627664

ABSTRACT

Telomere maintenance by telomerase activation or alternative lengthening of telomeres (ALT) is a major determinant of poor outcome in neuroblastoma. Here, we screen for ALT in primary and relapsed neuroblastomas (n = 760) and characterize its features using multi-omics profiling. ALT-positive tumors are molecularly distinct from other neuroblastoma subtypes and enriched in a population-based clinical sequencing study cohort for relapsed cases. They display reduced ATRX/DAXX complex abundance, due to either ATRX mutations (55%) or low protein expression. The heterochromatic histone mark H3K9me3 recognized by ATRX is enriched at the telomeres of ALT-positive tumors. Notably, we find a high frequency of telomeric repeat loci with a neuroblastoma ALT-specific hotspot on chr1q42.2 and loss of the adjacent chromosomal segment forming a neo-telomere. ALT-positive neuroblastomas proliferate slowly, which is reflected by a protracted clinical course of disease. Nevertheless, children with an ALT-positive neuroblastoma have dismal outcome.


Subject(s)
Whole Genome Sequencing/methods , Blotting, Western , Exons/genetics , Flow Cytometry , Humans , Proteome/metabolism , Retrospective Studies , Sequence Analysis, RNA/methods , Telomere/genetics , Telomere/metabolism , Telomere Homeostasis/genetics , X-linked Nuclear Protein/genetics
7.
Nat Commun ; 10(1): 4128, 2019 09 11.
Article in English | MEDLINE | ID: mdl-31511524

ABSTRACT

Pediatric malignancies including Ewing sarcoma (EwS) feature a paucity of somatic alterations except for pathognomonic driver-mutations that cannot explain overt variations in clinical outcome. Here, we demonstrate in EwS how cooperation of dominant oncogenes and regulatory germline variants determine tumor growth, patient survival and drug response. Binding of the oncogenic EWSR1-FLI1 fusion transcription factor to a polymorphic enhancer-like DNA element controls expression of the transcription factor MYBL2 mediating these phenotypes. Whole-genome and RNA sequencing reveals that variability at this locus is inherited via the germline and is associated with variable inter-tumoral MYBL2 expression. High MYBL2 levels sensitize EwS cells for inhibition of its upstream activating kinase CDK2 in vitro and in vivo, suggesting MYBL2 as a putative biomarker for anti-CDK2-therapy. Collectively, we establish cooperation of somatic mutations and regulatory germline variants as a major determinant of tumor progression and highlight the importance of integrating the regulatory genome in precision medicine.


Subject(s)
Germ-Line Mutation/genetics , Neoplasms/genetics , Neoplasms/therapy , Animals , Cell Cycle Proteins , Cell Line, Tumor , Cell Proliferation , Cell Survival , Cyclin-Dependent Kinase 2/antagonists & inhibitors , Cyclin-Dependent Kinase 2/metabolism , Gene Expression Regulation, Neoplastic , HEK293 Cells , Humans , Mice , Microsatellite Repeats/genetics , Neoplasm Proteins/metabolism , Oncogene Proteins, Fusion/metabolism , Phenotype , Polymorphism, Genetic , Trans-Activators , Treatment Outcome , Up-Regulation/genetics
8.
Nat Commun ; 9(1): 4866, 2018 11 19.
Article in English | MEDLINE | ID: mdl-30451831

ABSTRACT

Chromosome 17q gains are almost invariably present in high-risk neuroblastoma cases. Here, we perform an integrative epigenomics search for dosage-sensitive transcription factors on 17q marked by H3K27ac defined super-enhancers and identify TBX2 as top candidate gene. We show that TBX2 is a constituent of the recently established core regulatory circuitry in neuroblastoma with features of a cell identity transcription factor, driving proliferation through activation of p21-DREAM repressed FOXM1 target genes. Combined MYCN/TBX2 knockdown enforces cell growth arrest suggesting that TBX2 enhances MYCN sustained activation of FOXM1 targets. Targeting transcriptional addiction by combined CDK7 and BET bromodomain inhibition shows synergistic effects on cell viability with strong repressive effects on CRC gene expression and p53 pathway response as well as several genes implicated in transcriptional regulation. In conclusion, we provide insight into the role of the TBX2 CRC gene in transcriptional dependency of neuroblastoma cells warranting clinical trials using BET and CDK7 inhibitors.


Subject(s)
Brain Neoplasms/genetics , Forkhead Box Protein M1/genetics , Gene Expression Regulation, Neoplastic , Kv Channel-Interacting Proteins/genetics , N-Myc Proto-Oncogene Protein/genetics , Neuroblastoma/genetics , Repressor Proteins/genetics , T-Box Domain Proteins/genetics , Antineoplastic Agents/pharmacology , Azepines/pharmacology , Brain Neoplasms/drug therapy , Brain Neoplasms/metabolism , Brain Neoplasms/pathology , Cell Line, Tumor , Cell Survival/drug effects , Cyclin-Dependent Kinases/genetics , Cyclin-Dependent Kinases/metabolism , DNA Copy Number Variations , Epigenesis, Genetic , Forkhead Box Protein M1/metabolism , HEK293 Cells , Histones/genetics , Histones/metabolism , Humans , Kv Channel-Interacting Proteins/metabolism , N-Myc Proto-Oncogene Protein/metabolism , Neuroblastoma/drug therapy , Neuroblastoma/metabolism , Neuroblastoma/pathology , Organoids/drug effects , Organoids/metabolism , Organoids/pathology , Panobinostat/pharmacology , Phenylenediamines/pharmacology , Pyrimidines/pharmacology , Repressor Proteins/metabolism , Signal Transduction , T-Box Domain Proteins/metabolism , Triazoles/pharmacology , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism , Cyclin-Dependent Kinase-Activating Kinase
9.
Science ; 362(6419): 1165-1170, 2018 12 07.
Article in English | MEDLINE | ID: mdl-30523111

ABSTRACT

Neuroblastoma is a pediatric tumor of the sympathetic nervous system. Its clinical course ranges from spontaneous tumor regression to fatal progression. To investigate the molecular features of the divergent tumor subtypes, we performed genome sequencing on 416 pretreatment neuroblastomas and assessed telomere maintenance mechanisms in 208 of these tumors. We found that patients whose tumors lacked telomere maintenance mechanisms had an excellent prognosis, whereas the prognosis of patients whose tumors harbored telomere maintenance mechanisms was substantially worse. Survival rates were lowest for neuroblastoma patients whose tumors harbored telomere maintenance mechanisms in combination with RAS and/or p53 pathway mutations. Spontaneous tumor regression occurred both in the presence and absence of these mutations in patients with telomere maintenance-negative tumors. On the basis of these data, we propose a mechanistic classification of neuroblastoma that may benefit the clinical management of patients.


Subject(s)
Neuroblastoma/classification , Neuroblastoma/mortality , Telomere Homeostasis/genetics , Child , Child, Preschool , Disease-Free Survival , Exome/genetics , Genome, Human , Humans , Metabolic Networks and Pathways/genetics , Mutation , Neuroblastoma/drug therapy , Neuroblastoma/genetics , Prognosis , Sequence Analysis, DNA , Tumor Suppressor Protein p53/genetics , ras Proteins/genetics
10.
Oncoimmunology ; 5(7): e1116674, 2016 Jul.
Article in English | MEDLINE | ID: mdl-27622013

ABSTRACT

Natural Killer (NK) cells are innate effector cells that are able to recognize and eliminate tumor cells through engagement of their surface receptors. NKp30 is a potent activating NK cell receptor that elicits efficient NK cell-mediated target cell killing. Recently, B7-H6 was identified as tumor cell surface expressed ligand for NKp30. Enhanced B7-H6 mRNA levels are frequently detected in tumor compared to healthy tissues. To gain insight in the regulation of expression of B7-H6 in tumors, we investigated transcriptional mechanisms driving B7-H6 expression by promoter analyses. Using luciferase reporter assays and chromatin immunoprecipitation we mapped a functional binding site for Myc, a proto-oncogene overexpressed in certain tumors, in the B7-H6 promoter. Pharmacological inhibition or siRNA/shRNA-mediated knock-down of c-Myc or N-Myc significantly decreased B7-H6 expression on a variety of tumor cells including melanoma, pancreatic carcinoma and neuroblastoma cell lines. In tumor cell lines from different origin and primary tumor tissues of hepatocellular carcinoma (HCC), lymphoma and neuroblastoma, mRNA levels of c-Myc positively correlated with B7-H6 expression. Most importantly, upon inhibition or knock-down of c-Myc in tumor cells impaired NKp30-mediated degranulation of NK cells was observed. Thus, our data imply that Myc driven tumors could be targets for cancer immunotherapy exploiting the NKp30/B7-H6 axis.

11.
Cancer Res ; 76(18): 5523-37, 2016 09 15.
Article in English | MEDLINE | ID: mdl-27635046

ABSTRACT

The broad clinical spectrum of neuroblastoma ranges from spontaneous regression to rapid progression despite intensive multimodal therapy. This diversity is not fully explained by known genetic aberrations, suggesting the possibility of epigenetic involvement in pathogenesis. In pursuit of this hypothesis, we took an integrative approach to analyze the methylomes, transcriptomes, and copy number variations in 105 cases of neuroblastoma, complemented by primary tumor- and cell line-derived global histone modification analyses and epigenetic drug treatment in vitro We found that DNA methylation patterns identify divergent patient subgroups with respect to survival and clinicobiologic variables, including amplified MYCN Transcriptome integration and histone modification-based definition of enhancer elements revealed intragenic enhancer methylation as a mechanism for high-risk-associated transcriptional deregulation. Furthermore, in high-risk neuroblastomas, we obtained evidence for cooperation between PRC2 activity and DNA methylation in blocking tumor-suppressive differentiation programs. Notably, these programs could be re-activated by combination treatments, which targeted both PRC2 and DNA methylation. Overall, our results illuminate how epigenetic deregulation contributes to neuroblastoma pathogenesis, with novel implications for its diagnosis and therapy. Cancer Res; 76(18); 5523-37. ©2016 AACR.


Subject(s)
DNA Methylation/genetics , Epigenesis, Genetic/genetics , Neuroblastoma/genetics , Adolescent , Cell Line, Tumor , Child , Child, Preschool , Chromatin Immunoprecipitation , Cluster Analysis , Female , Genome-Wide Association Study , High-Throughput Nucleotide Sequencing , Humans , Infant , Infant, Newborn , Kaplan-Meier Estimate , Male , N-Myc Proto-Oncogene Protein/genetics , Neuroblastoma/mortality , Neuroblastoma/pathology , Oligonucleotide Array Sequence Analysis , Transcription, Genetic , Transcriptome , Young Adult
12.
Cancer Lett ; 366(1): 123-32, 2015 Sep 28.
Article in English | MEDLINE | ID: mdl-26123663

ABSTRACT

LIN28B has been identified as an oncogene in various tumor entities, including neuroblastoma, a childhood cancer that originates from neural crest-derived cells, and is characterized by amplification of the MYCN oncogene. Recently, elevated LIN28B expression levels were shown to contribute to neuroblastoma tumorigenesis via let-7 dependent de-repression of MYCN. However, additional insight in the regulation of LIN28B in neuroblastoma is lacking. Therefore, we have performed a comprehensive analysis of the regulation of LIN28B in neuroblastoma, with a specific focus on the contribution of miRNAs. We show that MYCN regulates LIN28B expression in neuroblastoma tumors via two distinct parallel mechanisms. First, through an unbiased LIN28B-3'UTR reporter screen, we found that miR-26a-5p and miR-26b-5p regulate LIN28B expression. Next, we demonstrated that MYCN indirectly affects the expression of miR-26a-5p, and hence regulates LIN28B, therefore establishing an MYCN-miR-26a-5p-LIN28B regulatory axis. Second, we provide evidence that MYCN regulates LIN28B expression via interaction with the LIN28B promoter, establishing a direct MYCN-LIN28B regulatory axis. We believe that these findings mark LIN28B as an important effector of the MYCN oncogenic phenotype and underline the importance of MYCN-regulated miRNAs in establishing the MYCN-driven oncogenic process.


Subject(s)
Neuroblastoma/pathology , Nuclear Proteins/physiology , Oncogene Proteins/physiology , RNA-Binding Proteins/genetics , Cell Line, Tumor , Humans , MicroRNAs/genetics , N-Myc Proto-Oncogene Protein , Transcription, Genetic
13.
Nat Genet ; 47(8): 872-7, 2015 Aug.
Article in English | MEDLINE | ID: mdl-26121086

ABSTRACT

Neuroblastoma is a malignancy of the developing sympathetic nervous system that is often lethal when relapse occurs. We here used whole-exome sequencing, mRNA expression profiling, array CGH and DNA methylation analysis to characterize 16 paired samples at diagnosis and relapse from individuals with neuroblastoma. The mutational burden significantly increased in relapsing tumors, accompanied by altered mutational signatures and reduced subclonal heterogeneity. Global allele frequencies at relapse indicated clonal mutation selection during disease progression. Promoter methylation patterns were consistent over disease course and were patient specific. Recurrent alterations at relapse included mutations in the putative CHD5 neuroblastoma tumor suppressor, chromosome 9p losses, DOCK8 mutations, inactivating mutations in PTPN14 and a relapse-specific activity pattern for the PTPN14 target YAP. Recurrent new mutations in HRAS, KRAS and genes mediating cell-cell interaction in 13 of 16 relapse tumors indicate disturbances in signaling pathways mediating mesenchymal transition. Our data shed light on genetic alteration frequency, identity and evolution in neuroblastoma.


Subject(s)
Gene Expression Regulation, Neoplastic , Mutation , Neoplasm Recurrence, Local/genetics , Neuroblastoma/genetics , Adaptor Proteins, Signal Transducing/genetics , Cell Line, Tumor , Comparative Genomic Hybridization , DNA Copy Number Variations , DNA Helicases/genetics , Exome/genetics , Gene Expression Profiling/methods , Gene Frequency , Guanine Nucleotide Exchange Factors/genetics , Hippo Signaling Pathway , Humans , In Situ Hybridization, Fluorescence , Nerve Tissue Proteins/genetics , Neuroblastoma/pathology , Oligonucleotide Array Sequence Analysis , Phosphoproteins/genetics , Protein Serine-Threonine Kinases/genetics , Protein Tyrosine Phosphatases, Non-Receptor/genetics , Sequence Analysis, DNA/methods , Signal Transduction/genetics , Transcription Factors , YAP-Signaling Proteins
SELECTION OF CITATIONS
SEARCH DETAIL