Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
1.
Nature ; 535(7611): 246-51, 2016 07 14.
Article in English | MEDLINE | ID: mdl-27383785

ABSTRACT

Poor prognosis in neuroblastoma is associated with genetic amplification of MYCN. MYCN is itself a target of let-7, a tumour suppressor family of microRNAs implicated in numerous cancers. LIN28B, an inhibitor of let-7 biogenesis, is overexpressed in neuroblastoma and has been reported to regulate MYCN. Here we show, however, that LIN28B is dispensable in MYCN-amplified neuroblastoma cell lines, despite de-repression of let-7. We further demonstrate that MYCN messenger RNA levels in amplified disease are exceptionally high and sufficient to sponge let-7, which reconciles the dispensability of LIN28B. We found that genetic loss of let-7 is common in neuroblastoma, inversely associated with MYCN amplification, and independently associated with poor outcomes, providing a rationale for chromosomal loss patterns in neuroblastoma. We propose that let-7 disruption by LIN28B, MYCN sponging, or genetic loss is a unifying mechanism of neuroblastoma development with broad implications for cancer pathogenesis.


Subject(s)
Gene Amplification/genetics , MicroRNAs/genetics , Neuroblastoma/genetics , Nuclear Proteins/genetics , Oncogene Proteins/genetics , RNA-Binding Proteins/genetics , 3' Untranslated Regions/genetics , Animals , Chromosome Deletion , Female , Gene Deletion , Genes, Neoplasm/genetics , Humans , Mice , MicroRNAs/metabolism , Models, Genetic , N-Myc Proto-Oncogene Protein , Neuroblastoma/pathology , Xenograft Model Antitumor Assays
2.
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
3.
Nature ; 511(7510): 483-7, 2014 Jul 24.
Article in English | MEDLINE | ID: mdl-25043018

ABSTRACT

In mammalian cells, the MYC oncoprotein binds to thousands of promoters. During mitogenic stimulation of primary lymphocytes, MYC promotes an increase in the expression of virtually all genes. In contrast, MYC-driven tumour cells differ from normal cells in the expression of specific sets of up- and downregulated genes that have considerable prognostic value. To understand this discrepancy, we studied the consequences of inducible expression and depletion of MYC in human cells and murine tumour models. Changes in MYC levels activate and repress specific sets of direct target genes that are characteristic of MYC-transformed tumour cells. Three factors account for this specificity. First, the magnitude of response parallels the change in occupancy by MYC at each promoter. Functionally distinct classes of target genes differ in the E-box sequence bound by MYC, suggesting that different cellular responses to physiological and oncogenic MYC levels are controlled by promoter affinity. Second, MYC both positively and negatively affects transcription initiation independent of its effect on transcriptional elongation. Third, complex formation with MIZ1 (also known as ZBTB17) mediates repression of multiple target genes by MYC and the ratio of MYC and MIZ1 bound to each promoter correlates with the direction of response.


Subject(s)
Down-Regulation/genetics , Gene Expression Regulation, Neoplastic/genetics , Genes, myc/genetics , Neoplasms/genetics , Transcriptome , Up-Regulation/genetics , Animals , Binding Sites , Cell Line, Tumor , E-Box Elements/genetics , Humans , Kruppel-Like Transcription Factors/metabolism , Mice , Nuclear Proteins/metabolism , Promoter Regions, Genetic/genetics , Protein Inhibitors of Activated STAT/metabolism , Proto-Oncogene Proteins c-myc/genetics , Proto-Oncogene Proteins c-myc/metabolism , RNA Polymerase II/metabolism , Ubiquitin-Protein Ligases
4.
EMBO J ; 31(20): 3961-75, 2012 Oct 17.
Article in English | MEDLINE | ID: mdl-22909821

ABSTRACT

Following genotoxic stress, cells activate a complex signalling network to arrest the cell cycle and initiate DNA repair or apoptosis. The tumour suppressor p53 lies at the heart of this DNA damage response. However, it remains incompletely understood, which signalling molecules dictate the choice between these different cellular outcomes. Here, we identify the transcriptional regulator apoptosis-antagonizing transcription factor (AATF)/Che-1 as a critical regulator of the cellular outcome of the p53 response. Upon genotoxic stress, AATF is phosphorylated by the checkpoint kinase MK2. Phosphorylation results in the release of AATF from cytoplasmic MRLC3 and subsequent nuclear translocation where AATF binds to the PUMA, BAX and BAK promoter regions to repress p53-driven expression of these pro-apoptotic genes. In xenograft experiments, mice exhibit a dramatically enhanced response of AATF-depleted tumours following genotoxic chemotherapy with adriamycin. The exogenous expression of a phospho-mimicking AATF point mutant results in marked adriamycin resistance in vivo. Nuclear AATF enrichment appears to be selected for in p53-proficient endometrial cancers. Furthermore, focal copy number gains at the AATF locus in neuroblastoma, which is known to be almost exclusively p53-proficient, correlate with an adverse prognosis and reduced overall survival. These data identify the p38/MK2/AATF signalling module as a critical repressor of p53-driven apoptosis and commend this pathway as a target for DNA damage-sensitizing therapeutic regimens.


Subject(s)
Apoptosis Regulatory Proteins/physiology , Apoptosis/physiology , DNA Damage/physiology , Repressor Proteins/physiology , Tumor Suppressor Protein p53/physiology , Active Transport, Cell Nucleus , Amino Acid Sequence , Animals , Apoptosis Regulatory Proteins/genetics , Cell Cycle Checkpoints , DNA Damage/genetics , Doxorubicin/pharmacology , Drug Resistance, Neoplasm/genetics , Endometrial Neoplasms/genetics , Female , Gene Amplification , Gene Dosage , HEK293 Cells , Humans , Mice , Molecular Sequence Data , Multiprotein Complexes , Myosin Light Chains/metabolism , Neuroblastoma/genetics , Neuroblastoma/mortality , Osmotic Pressure , Phosphorylation , Prognosis , Protein Processing, Post-Translational , Repressor Proteins/genetics
5.
Blood ; 120(13): 2620-30, 2012 Sep 27.
Article in English | MEDLINE | ID: mdl-22517906

ABSTRACT

Multiple myeloma (MM) is a clonal plasma cell disorder frequently accompanied by hematopoietic impairment. We show that hematopoietic stem and progenitor cells (HSPCs), in particular megakaryocyte-erythrocyte progenitors, are diminished in the BM of MM patients. Genomic profiling of HSPC subsets revealed deregulations of signaling cascades, most notably TGFß signaling, and pathways involved in cytoskeletal organization, migration, adhesion, and cell-cycle regulation in the patients. Functionally, proliferation, colony formation, and long-term self-renewal were impaired as a consequence of activated TGFß signaling. In accordance, TGFß levels in the BM extracellular fluid were elevated and mesenchymal stromal cells (MSCs) had a reduced capacity to support long-term hematopoiesis of HSPCs that completely recovered on blockade of TGFß signaling. Furthermore, we found defective actin assembly and down-regulation of the adhesion receptor CD44 in MM HSPCs functionally reflected by impaired migration and adhesion. Still, transplantation into myeloma-free NOG mice revealed even enhanced engraftment and normal differentiation capacities of MM HSPCs, which underlines that functional impairment of HSPCs depends on MM-related microenvironmental cues and is reversible. Taken together, these data implicate that hematopoietic suppression in MM emerges from the HSPCs as a result of MM-related microenvironmental alterations.


Subject(s)
Antigens, CD34/metabolism , Biomarkers/metabolism , Bone Marrow/pathology , Hematopoietic Stem Cells/pathology , Megakaryocyte-Erythroid Progenitor Cells/pathology , Multiple Myeloma/metabolism , Multiple Myeloma/pathology , Animals , Blotting, Western , Bone Marrow/metabolism , Case-Control Studies , Cell Adhesion , Cell Cycle , Cell Differentiation , Cell Movement , Cell Proliferation , Cells, Cultured , Female , Flow Cytometry , Fluorescent Antibody Technique , Gene Expression Profiling , Hematopoietic Stem Cells/metabolism , Humans , Immunoenzyme Techniques , Male , Megakaryocyte-Erythroid Progenitor Cells/metabolism , Mesenchymal Stem Cells/metabolism , Mesenchymal Stem Cells/pathology , Mice , Mice, Inbred NOD , Multiple Myeloma/genetics , Oligonucleotide Array Sequence Analysis , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction
6.
BMC Cancer ; 14: 840, 2014 Nov 18.
Article in English | MEDLINE | ID: mdl-25406647

ABSTRACT

BACKGROUND: Segmental genomic copy number alterations, such as loss of 11q or 3p and gain of 17q, are well established markers of poor outcome in neuroblastoma, and have been suggested to comprise tumor suppressor genes or oncogenes, respectively. The gene forkhead box P1 (FOXP1) maps to chromosome 3p14.1, a tumor suppressor locus deleted in many human cancers including neuroblastoma. FoxP1 belongs to a family of winged-helix transcription factors that are involved in processes of cellular proliferation, differentiation and neoplastic transformation. METHODS: Microarray expression profiles of 476 neuroblastoma specimens were generated and genes differentially expressed between favorable and unfavorable neuroblastoma were identified. FOXP1 expression was correlated to clinical markers and patient outcome. To determine whether hypermethylation is involved in silencing of FOXP1, methylation analysis of the 5' region of FOXP1 in 47 neuroblastomas was performed. Furthermore, FOXP1 was re-expressed in three neuroblastoma cell lines to study the effect of FOXP1 on growth characteristics of neuroblastoma cells. RESULTS: Low expression of FOXP1 is associated with markers of unfavorable prognosis like stage 4, age >18 months and MYCN amplification and unfavorable gene expression-based classification (P < 0.001 each). Moreover, FOXP1 expression predicts patient outcome accurately and independently from well-established prognostic markers. Array-based CGH analysis of 159 neuroblastomas revealed that heterozygous loss of the FOXP1 locus was a rare event (n = 4), but if present, was associated with low FOXP1 expression. By contrast, DNA methylation analysis in 47 neuroblastomas indicated that hypermethylation is not regularly involved in FOXP1 gene silencing. Re-expression of FoxP1 significantly impaired cell proliferation, viability and colony formation in soft agar. Furthermore, induction of FOXP1 expression led to cell cycle arrest and apoptotic cell death of neuroblastoma cells. CONCLUSIONS: Our results suggest that down-regulation of FOXP1 expression is a common event in high-risk neuroblastoma pathogenesis and may contribute to tumor progression and unfavorable patient outcome.


Subject(s)
Cell Transformation, Neoplastic/genetics , Forkhead Transcription Factors/genetics , Neuroblastoma/genetics , Repressor Proteins/genetics , Apoptosis/genetics , Biomarkers, Tumor , Cell Line, Tumor , Cell Movement/genetics , Cell Proliferation , Child, Preschool , Cluster Analysis , Comparative Genomic Hybridization , DNA Methylation , Epigenesis, Genetic , Gene Expression Profiling , Humans , Infant , Neoplasm Staging , Neuroblastoma/mortality , Neuroblastoma/pathology , Phenotype , Prognosis , Promoter Regions, Genetic , Transcription, Genetic
7.
Mol Oncol ; 10(2): 344-59, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26598443

ABSTRACT

Neuroblastoma is an embryonal pediatric tumor that originates from the developing sympathetic nervous system and shows a broad range of clinical behavior, ranging from fatal progression to differentiation into benign ganglioneuroma. In experimental neuroblastoma systems, retinoic acid (RA) effectively induces neuronal differentiation, and RA treatment has been therefore integrated in current therapies. However, the molecular mechanisms underlying differentiation are still poorly understood. We here investigated the role of transcription factor activating protein 2 beta (TFAP2B), a key factor in sympathetic nervous system development, in neuroblastoma pathogenesis and differentiation. Microarray analyses of primary neuroblastomas (n = 649) demonstrated that low TFAP2B expression was significantly associated with unfavorable prognostic markers as well as adverse patient outcome. We also found that low TFAP2B expression was strongly associated with CpG methylation of the TFAP2B locus in primary neuroblastomas (n = 105) and demethylation with 5-aza-2'-deoxycytidine resulted in induction of TFAP2B expression in vitro, suggesting that TFAP2B is silenced by genomic methylation. Tetracycline inducible re-expression of TFAP2B in IMR-32 and SH-EP neuroblastoma cells significantly impaired proliferation and cell cycle progression. In IMR-32 cells, TFAP2B induced neuronal differentiation, which was accompanied by up-regulation of the catecholamine biosynthesizing enzyme genes DBH and TH, and down-regulation of MYCN and REST, a master repressor of neuronal genes. By contrast, knockdown of TFAP2B by lentiviral transduction of shRNAs abrogated RA-induced neuronal differentiation of SH-SY5Y and SK-N-BE(2)c neuroblastoma cells almost completely. Taken together, our results suggest that TFAP2B is playing a vital role in retaining RA responsiveness and mediating noradrenergic neuronal differentiation in neuroblastoma.


Subject(s)
Adrenergic Neurons/pathology , Neuroblastoma/pathology , Transcription Factor AP-2/metabolism , Adolescent , Adrenergic Neurons/metabolism , Adult , Azacitidine/analogs & derivatives , Azacitidine/pharmacology , Cell Cycle , Cell Differentiation/drug effects , Cell Line, Tumor , Child , Child, Preschool , CpG Islands/genetics , DNA Methylation/drug effects , Decitabine , Dopamine beta-Hydroxylase/metabolism , Down-Regulation , Gene Knockdown Techniques , Humans , Infant , Infant, Newborn , N-Myc Proto-Oncogene Protein , Neuroblastoma/genetics , Neuroblastoma/metabolism , Nuclear Proteins/metabolism , Oncogene Proteins/metabolism , Prognosis , Promoter Regions, Genetic , RNA, Small Interfering/metabolism , Repressor Proteins/metabolism , Transcription Factor AP-2/genetics , Tretinoin/pharmacology , Tyrosine 3-Monooxygenase/metabolism , Up-Regulation , Young Adult
8.
Oncotarget ; 7(39): 62799-62813, 2016 09 27.
Article in English | MEDLINE | ID: mdl-27588497

ABSTRACT

As high-risk neuroblastoma (NB) has a poor prognosis, new therapeutic modalities are needed. We therefore investigated the susceptibility of NB cells to γ-secretase inhibitor I (GSI-I). NOTCH signaling activity, the cellular effects of GSI-I and its mechanisms of cytotoxicity were evaluated in NB cells in vitro and in vivo. The results show that NOTCH signaling is relevant for human NB cells. Of the GSIs screened in vitro GSI-I was the most effective inhibitor of NB cells. Both MYCN-amplified and non-amplified NB cells were susceptible to GSI-I. Among the targets of GSI-I in NB cells were NOTCH and the proteasome. GSI-I caused G2/M arrest that was enhanced by acute activation of MYCN and led to mitotic dysfunction. GSI-I also induced proapoptotic NOXA. Survival of mice bearing an MYCN non-amplified orthotopic patient-derived NB xenograft was significantly prolonged by systemic GSI-I, associated with mitotic catastrophe and reduced angiogenesis, and without evidence of intestinal toxicity. In conclusion, the activity of GSI-I on multiple targets in NB cells and the lack of gastrointestinal toxicity in mice are advantageous and merit further investigations of GSI-I in NB.


Subject(s)
Brain Neoplasms/metabolism , Neuroblastoma/metabolism , Proteasome Endopeptidase Complex/metabolism , Receptor, Notch1/metabolism , Amyloid Precursor Protein Secretases/antagonists & inhibitors , Amyloid Precursor Protein Secretases/metabolism , Animals , Apoptosis , Brain Neoplasms/drug therapy , Carbamates/pharmacology , Cell Cycle , Cell Line, Tumor , Dipeptides/pharmacology , Enzyme Inhibitors/pharmacology , Female , Humans , In Situ Hybridization, Fluorescence , Mice , Mitosis , N-Myc Proto-Oncogene Protein/metabolism , Neoplasm Transplantation , Neovascularization, Pathologic , Neuroblastoma/drug therapy , Oligopeptides/pharmacology , Receptors, Notch/metabolism , Signal Transduction
9.
Med Sci Sports Exerc ; 48(5): 829-38, 2016 May.
Article in English | MEDLINE | ID: mdl-26694850

ABSTRACT

PURPOSE: Maternal obesity is known to predispose the offspring to impaired glucose metabolism and obesity associated with low-grade inflammation and hypothalamic dysfunction. Because preventive approaches in this context are missing to date, we aimed to identify molecular mechanisms in the offspring that are affected by maternal exercise during pregnancy. METHODS: Diet-induced obese mouse dams were divided into a sedentary obese (high-fat diet [HFD]) group and an obese intervention (HFD-running intervention [RUN]) group, which performed voluntary wheel running throughout gestation. Male offspring were compared with the offspring of a sedentary lean control group at postnatal day 21. RESULTS: HFD and HFD-RUN offspring showed increased body weight and white adipose tissue mass. Glucose tolerance testing showed mild impairment only in HFD offspring. Serum interleukin-6 (IL-6) levels, hypothalamic and white adipose tissue IL-6 gene expressions, and phosphorylation of signal transducer and activator of transcription 3 in HFD offspring were significantly increased, whereas HFD-RUN was protected against these changes. The altered hypothalamic global gene expression in HFD offspring showed partial normalization in HFD-RUN offspring, especially with respect to IL-6 action. CONCLUSION: Maternal exercise in obese pregnancies effectively reduces IL-6 trans-signaling and might be the underlying mechanism for the amelioration of glucose metabolism at postnatal day 21 independent of body composition.


Subject(s)
Interleukin-6/metabolism , Obesity/physiopathology , Physical Conditioning, Animal , Signal Transduction , Adipose Tissue, White/metabolism , Adiposity , Animals , Body Weight , Diet, High-Fat , Female , Glucose/metabolism , Glucose Tolerance Test , Hypothalamus/metabolism , Insulin/blood , Interleukin-6/blood , Leptin/blood , Male , Mice , Motor Activity , Phenotype , Pregnancy , STAT3 Transcription Factor/metabolism , Transcriptome
10.
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
11.
Genome Biol ; 16: 133, 2015 Jun 25.
Article in English | MEDLINE | ID: mdl-26109056

ABSTRACT

BACKGROUND: Gene expression profiling is being widely applied in cancer research to identify biomarkers for clinical endpoint prediction. Since RNA-seq provides a powerful tool for transcriptome-based applications beyond the limitations of microarrays, we sought to systematically evaluate the performance of RNA-seq-based and microarray-based classifiers in this MAQC-III/SEQC study for clinical endpoint prediction using neuroblastoma as a model. RESULTS: We generate gene expression profiles from 498 primary neuroblastomas using both RNA-seq and 44 k microarrays. Characterization of the neuroblastoma transcriptome by RNA-seq reveals that more than 48,000 genes and 200,000 transcripts are being expressed in this malignancy. We also find that RNA-seq provides much more detailed information on specific transcript expression patterns in clinico-genetic neuroblastoma subgroups than microarrays. To systematically compare the power of RNA-seq and microarray-based models in predicting clinical endpoints, we divide the cohort randomly into training and validation sets and develop 360 predictive models on six clinical endpoints of varying predictability. Evaluation of factors potentially affecting model performances reveals that prediction accuracies are most strongly influenced by the nature of the clinical endpoint, whereas technological platforms (RNA-seq vs. microarrays), RNA-seq data analysis pipelines, and feature levels (gene vs. transcript vs. exon-junction level) do not significantly affect performances of the models. CONCLUSIONS: We demonstrate that RNA-seq outperforms microarrays in determining the transcriptomic characteristics of cancer, while RNA-seq and microarray-based models perform similarly in clinical endpoint prediction. Our findings may be valuable to guide future studies on the development of gene expression-based predictive models and their implementation in clinical practice.


Subject(s)
Gene Expression Profiling , Neuroblastoma/genetics , Oligonucleotide Array Sequence Analysis , Sequence Analysis, RNA , Adolescent , Adult , Child , Child, Preschool , Endpoint Determination , Female , Humans , Infant , Infant, Newborn , Male , Models, Genetic , Neuroblastoma/classification , Neuroblastoma/diagnosis , Tumor Cells, Cultured , Young Adult
12.
Cancer Res ; 74(9): 2604-16, 2014 May 01.
Article in English | MEDLINE | ID: mdl-24419085

ABSTRACT

Neuroblastoma is an embryonic solid tumor of neural crest origin and accounts for 11% of all cancer-related deaths in children. Novel therapeutic strategies are therefore urgently required. MYCN oncogene amplification, which occurs in 20% of neuroblastomas, is a hallmark of high risk. Here, we aimed to exploit molecular mechanisms that can be pharmacologically addressed with epigenetically modifying drugs, such as histone deacetylase (HDAC) inhibitors. Grainyhead-like 1 (GRHL1), a gene critical for Drosophila neural development, belonged to the genes most strongly responding to HDAC inhibitor treatment of neuroblastoma cells in a genome-wide screen. An increase in the histone H4 pan-acetylation associated with its promoter preceded transcriptional activation. Physically adjacent, HDAC3 and MYCN colocalized to the GRHL1 promoter and repressed its transcription. High-level GRHL1 expression in primary neuroblastomas correlated on transcriptional and translational levels with favorable patient survival and established clinical and molecular markers for favorable tumor biology, including lack of MYCN amplification. Enforced GRHL1 expression in MYCN-amplified neuroblastoma cells with low endogenous GRHL1 levels abrogated anchorage-independent colony formation, inhibited proliferation, and retarded xenograft growth in mice. GRHL1 knockdown in MYCN single-copy cells with high endogenous GRHL1 levels promoted colony formation. GRHL1 regulated 170 genes genome-wide, and most were involved in pathways regulated during neuroblastomagenesis, including nervous system development, proliferation, cell-cell adhesion, cell spreading, and cellular differentiation. In summary, the data presented here indicate a significant role of HDAC3 in the MYCN-mediated repression of GRHL1 and suggest drugs that block HDAC3 activity and suppress MYCN expression as promising candidates for novel treatment strategies of high-risk neuroblastoma.


Subject(s)
Histone Deacetylases/physiology , Neuroblastoma/genetics , Nuclear Proteins/physiology , Oncogene Proteins/physiology , Repressor Proteins/genetics , Animals , Antineoplastic Agents/pharmacology , Cell Line, Tumor , Cell Proliferation , Disease-Free Survival , Female , Gene Expression Regulation, Neoplastic , Gene Silencing , Genes, Tumor Suppressor , Histone Deacetylase Inhibitors/pharmacology , Humans , Hydroxamic Acids/pharmacology , Indoles/pharmacology , Infant , Kaplan-Meier Estimate , Mice , Mice, SCID , N-Myc Proto-Oncogene Protein , Neoplasm Transplantation , Neuroblastoma/metabolism , Neuroblastoma/mortality , Neuroblastoma/pathology , Panobinostat , Repressor Proteins/metabolism , Transcription, Genetic , Tumor Burden
13.
J Clin Oncol ; 31(7): 944-51, 2013 Mar 01.
Article in English | MEDLINE | ID: mdl-23341514

ABSTRACT

PURPOSE: Radioiodinated metaiodobenzylguanidine ((123)I-mIBG) scintigraphy is an established imaging method in neuroblastoma. Semiquantitative scoring systems have been developed to assess the extent of disease and response to chemotherapy. We present the results of the comparison between the SIOPEN [International Society of Pediatric Oncology Europe Neuroblastoma Group] score and the modified Curie score. PATIENTS AND METHODS: We retrospectively analyzed 147 mIBG scans of 58 patients older than 1 year of age with stage 4 neuroblastoma from German Neuroblastoma Trial NB97 that were assessed according to the SIOPEN and the Curie scoring method. mIBG examinations were performed at diagnosis and after four and six cycles of chemotherapy. RESULTS: Scoring results were highly correlated between both methods, and interobserver reliability was excellent. A Curie score ≤ 2 and a SIOPEN score ≤ 4 (best cutoff) at diagnosis were correlated to significantly better event-free and overall survival compared with higher scores. After four cycles of chemotherapy, overall survival was significantly better for mIBG-negative patients compared with those with any residual mIBG-positive metastases. After six cycles of chemotherapy, there was no difference in survival between mIBG-negative patients and patients with residual mIBG-positive metastases. Patients without mIBG-positive metastases after four and six cycles of chemotherapy had a better overall survival, but late clearance of mIBG-positive metastases did not improve outcome. CONCLUSION: Higher mIBG scores at diagnosis and occurrence of any residual mIBG-positive metastases after four cycles of chemotherapy predicted unfavorable outcome for patients with stage 4 neuroblastoma. Later clearance of metastases did not improve prognosis. The Curie and the SIOPEN score were equally reliable and predictive.


Subject(s)
3-Iodobenzylguanidine , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Neuroblastoma/diagnostic imaging , Neuroblastoma/pathology , Radiopharmaceuticals , Child, Preschool , Disease-Free Survival , Female , Germany , Humans , Infant , Kaplan-Meier Estimate , Male , Neoplasm Staging , Neuroblastoma/drug therapy , Observer Variation , Predictive Value of Tests , Prognosis , Radionuclide Imaging , Retrospective Studies , Risk Factors , Treatment Outcome
14.
Cancer Cell ; 24(1): 75-89, 2013 Jul 08.
Article in English | MEDLINE | ID: mdl-23792191

ABSTRACT

Amplification of MYCN is a driver mutation in a subset of human neuroendocrine tumors, including neuroblastoma. No small molecules that target N-Myc, the protein encoded by MYCN, are clinically available. N-Myc forms a complex with the Aurora-A kinase, which protects N-Myc from proteasomal degradation. Although stabilization of N-Myc does not require the catalytic activity of Aurora-A, we show here that two Aurora-A inhibitors, MLN8054 and MLN8237, disrupt the Aurora-A/N-Myc complex and promote degradation of N-Myc mediated by the Fbxw7 ubiquitin ligase. Disruption of the Aurora-A/N-Myc complex inhibits N-Myc-dependent transcription, correlating with tumor regression and prolonged survival in a mouse model of MYCN-driven neuroblastoma. We conclude that Aurora-A is an accessible target that makes destabilization of N-Myc a viable therapeutic strategy.


Subject(s)
Antineoplastic Agents/pharmacology , Azepines/pharmacology , Benzazepines/pharmacology , Neuroblastoma/drug therapy , Proteasome Endopeptidase Complex/physiology , Protein Serine-Threonine Kinases/antagonists & inhibitors , Proto-Oncogene Proteins c-myc/metabolism , Pyrimidines/pharmacology , Animals , Aurora Kinase A , Aurora Kinases , Cell Cycle Proteins/physiology , Cell Line, Tumor , Cyclohexanecarboxylic Acids/pharmacology , F-Box Proteins/physiology , F-Box-WD Repeat-Containing Protein 7 , Humans , Mice , Neuroblastoma/genetics , Neuroblastoma/pathology , Proto-Oncogene Proteins c-myc/genetics , Thiazoles/pharmacology , Ubiquitin-Protein Ligases/physiology
SELECTION OF CITATIONS
SEARCH DETAIL