Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 25
Filter
1.
Mol Cancer Res ; 20(1): 102-113, 2022 01.
Article in English | MEDLINE | ID: mdl-34556523

ABSTRACT

Whole-genome sequencing of primary breast tumors enabled the identification of cancer driver genes and noncoding cancer driver plexuses from somatic mutations. However, differentiating driver from passenger events among noncoding genetic variants remains a challenge. Herein, we reveal cancer-driver cis-regulatory elements linked to transcription factors previously shown to be involved in development of luminal breast cancers by defining a tumor-enriched catalogue of approximately 100,000 unique cis-regulatory elements from 26 primary luminal estrogen receptor (ER)+ progesterone receptor (PR)+ breast tumors. Integrating this catalog with somatic mutations from 350 publicly available breast tumor whole genomes, we uncovered cancer driver cistromes, defined as the sum of binding sites for a transcription factor, for ten transcription factors in luminal breast cancer such as FOXA1 and ER, nine of which are essential for growth in breast cancer with four exclusive to the luminal subtype. Collectively, we present a strategy to find cancer driver cistromes relying on quantifying the enrichment of noncoding mutations over cis-regulatory elements concatenated into a functional unit. IMPLICATIONS: Mapping the accessible chromatin of luminal breast cancer led to discovery of an accumulation of mutations within cistromes of transcription factors essential to luminal breast cancer. This demonstrates coopting of regulatory networks to drive cancer and provides a framework to derive insight into the noncoding space of cancer.


Subject(s)
Breast Neoplasms/genetics , Chromatin/metabolism , Gene Expression Regulation, Neoplastic/genetics , Whole Genome Sequencing/methods , Breast Neoplasms/pathology , Female , Humans , Mutation
2.
Nat Commun ; 12(1): 979, 2021 02 12.
Article in English | MEDLINE | ID: mdl-33579912

ABSTRACT

Glioblastoma (GBM) is a deadly cancer in which cancer stem cells (CSCs) sustain tumor growth and contribute to therapeutic resistance. Protein arginine methyltransferase 5 (PRMT5) has recently emerged as a promising target in GBM. Using two orthogonal-acting inhibitors of PRMT5 (GSK591 or LLY-283), we show that pharmacological inhibition of PRMT5 suppresses the growth of a cohort of 46 patient-derived GBM stem cell cultures, with the proneural subtype showing greater sensitivity. We show that PRMT5 inhibition causes widespread disruption of splicing across the transcriptome, particularly affecting cell cycle gene products. We identify a GBM splicing signature that correlates with the degree of response to PRMT5 inhibition. Importantly, we demonstrate that LLY-283 is brain-penetrant and significantly prolongs the survival of mice with orthotopic patient-derived xenografts. Collectively, our findings provide a rationale for the clinical development of brain penetrant PRMT5 inhibitors as treatment for GBM.


Subject(s)
Antineoplastic Agents/pharmacology , Brain Neoplasms/metabolism , Glioblastoma/metabolism , Protein-Arginine N-Methyltransferases/metabolism , Animals , Apoptosis , Brain Neoplasms/drug therapy , Brain Neoplasms/genetics , Brain Neoplasms/pathology , Cell Cycle , Cell Line, Tumor , Cell Proliferation/drug effects , Drug Discovery , Epigenomics , Female , Gene Expression Regulation, Neoplastic , Glioblastoma/drug therapy , Glioblastoma/genetics , Glioblastoma/pathology , Humans , Mice , Neoplastic Stem Cells/metabolism , Protein-Arginine N-Methyltransferases/drug effects , Protein-Arginine N-Methyltransferases/genetics , RNA Splicing , Xenograft Model Antitumor Assays
3.
Elife ; 102021 01 11.
Article in English | MEDLINE | ID: mdl-33427645

ABSTRACT

Chromatin accessibility discriminates stem from mature cell populations, enabling the identification of primitive stem-like cells in primary tumors, such as glioblastoma (GBM) where self-renewing cells driving cancer progression and recurrence are prime targets for therapeutic intervention. We show, using single-cell chromatin accessibility, that primary human GBMs harbor a heterogeneous self-renewing population whose diversity is captured in patient-derived glioblastoma stem cells (GSCs). In-depth characterization of chromatin accessibility in GSCs identifies three GSC states: Reactive, Constructive, and Invasive, each governed by uniquely essential transcription factors and present within GBMs in varying proportions. Orthotopic xenografts reveal that GSC states associate with survival, and identify an invasive GSC signature predictive of low patient survival, in line with the higher invasive properties of Invasive state GSCs compared to Reactive and Constructive GSCs as shown by in vitro and in vivo assays. Our chromatin-driven characterization of GSC states improves prognostic precision and identifies dependencies to guide combination therapies.


Subject(s)
Cell Self Renewal , Chromatin/metabolism , Glioblastoma/secondary , Neoplastic Stem Cells/physiology , Cell Line, Tumor , Female , Humans , Male , Single-Cell Analysis
4.
Nat Cancer ; 2(2): 157-173, 2021 02.
Article in English | MEDLINE | ID: mdl-35122077

ABSTRACT

Glioblastomas harbor diverse cell populations, including rare glioblastoma stem cells (GSCs) that drive tumorigenesis. To characterize functional diversity within this population, we performed single-cell RNA sequencing on >69,000 GSCs cultured from the tumors of 26 patients. We observed a high degree of inter- and intra-GSC transcriptional heterogeneity that could not be fully explained by DNA somatic alterations. Instead, we found that GSCs mapped along a transcriptional gradient spanning two cellular states reminiscent of normal neural development and inflammatory wound response. Genome-wide CRISPR-Cas9 dropout screens independently recapitulated this observation, with each state characterized by unique essential genes. Further single-cell RNA sequencing of >56,000 malignant cells from primary tumors found that the majority organize along an orthogonal astrocyte maturation gradient yet retain expression of founder GSC transcriptional programs. We propose that glioblastomas grow out of a fundamental GSC-based neural wound response transcriptional program, which is a promising target for new therapy development.


Subject(s)
Brain Neoplasms , Glioblastoma , Brain Neoplasms/genetics , Carcinogenesis/genetics , Glioblastoma/genetics , Humans , Neoplastic Stem Cells/metabolism
5.
Cancer Discov ; 10(9): 1312-1329, 2020 09.
Article in English | MEDLINE | ID: mdl-32546577

ABSTRACT

Tumor progression upon treatment arises from preexisting resistant cancer cells and/or adaptation of persister cancer cells committing to an expansion phase. Here, we show that evasion from viral mimicry response allows the growth of taxane-resistant triple-negative breast cancer (TNBC). This is enabled by an epigenetic state adapted to taxane-induced metabolic stress, where DNA hypomethylation over loci enriched in transposable elements (TE) is compensated by large chromatin domains of H3K27me3 to warrant TE repression. This epigenetic state creates a vulnerability to epigenetic therapy against EZH2, the H3K27me3 methyltransferase, which alleviates TE repression in taxane-resistant TNBC, leading to double-stranded RNA production and growth inhibition through viral mimicry response. Collectively, our results illustrate how epigenetic states over TEs promote cancer progression under treatment and can inform about vulnerabilities to epigenetic therapy. SIGNIFICANCE: Drug-resistant cancer cells represent a major barrier to remission for patients with cancer. Here we show that drug-induced metabolic perturbation and epigenetic states enable evasion from the viral mimicry response induced by chemotherapy in TNBC. These epigenetic states define a vulnerability to epigenetic therapy using EZH2 inhibitors in taxane-resistant TNBC.See related commentary by Janin and Esteller, p. 1258.This article is highlighted in the In This Issue feature, p. 1241.


Subject(s)
Antineoplastic Agents/pharmacology , Epigenesis, Genetic/immunology , Molecular Mimicry/immunology , Triple Negative Breast Neoplasms/immunology , Tumor Escape/genetics , Animals , Antineoplastic Agents/therapeutic use , Cell Line, Tumor , Chromatin Immunoprecipitation Sequencing , DNA Methylation/drug effects , DNA Methylation/immunology , DNA Transposable Elements/genetics , Disease Progression , Drug Resistance, Neoplasm/genetics , Drug Resistance, Neoplasm/immunology , Enhancer of Zeste Homolog 2 Protein/antagonists & inhibitors , Enhancer of Zeste Homolog 2 Protein/metabolism , Epigenesis, Genetic/drug effects , Female , Humans , Mice , Molecular Mimicry/genetics , Paclitaxel/pharmacology , Paclitaxel/therapeutic use , RNA, Double-Stranded/immunology , RNA, Double-Stranded/metabolism , Triple Negative Breast Neoplasms/drug therapy , Triple Negative Breast Neoplasms/genetics , Triple Negative Breast Neoplasms/pathology , Xenograft Model Antitumor Assays
6.
Cell ; 181(6): 1329-1345.e24, 2020 06 11.
Article in English | MEDLINE | ID: mdl-32445698

ABSTRACT

Posterior fossa A (PFA) ependymomas are lethal malignancies of the hindbrain in infants and toddlers. Lacking highly recurrent somatic mutations, PFA ependymomas are proposed to be epigenetically driven tumors for which model systems are lacking. Here we demonstrate that PFA ependymomas are maintained under hypoxia, associated with restricted availability of specific metabolites to diminish histone methylation, and increase histone demethylation and acetylation at histone 3 lysine 27 (H3K27). PFA ependymomas initiate from a cell lineage in the first trimester of human development that resides in restricted oxygen. Unlike other ependymomas, transient exposure of PFA cells to ambient oxygen induces irreversible cellular toxicity. PFA tumors exhibit a low basal level of H3K27me3, and, paradoxically, inhibition of H3K27 methylation specifically disrupts PFA tumor growth. Targeting metabolism and/or the epigenome presents a unique opportunity for rational therapy for infants with PFA ependymoma.


Subject(s)
Ependymoma/genetics , Ependymoma/metabolism , Epigenome/genetics , Infratentorial Neoplasms/genetics , Infratentorial Neoplasms/metabolism , Animals , Brain Neoplasms/genetics , Brain Neoplasms/metabolism , Cell Line , Cell Proliferation/genetics , DNA Methylation/genetics , Epigenomics/methods , Histones/genetics , Histones/metabolism , Humans , Infant , Lysine/genetics , Lysine/metabolism , Male , Mice, Inbred C57BL , Mutation/genetics
7.
Cancer Cell ; 36(6): 674-689.e6, 2019 12 09.
Article in English | MEDLINE | ID: mdl-31735626

ABSTRACT

Thousands of noncoding somatic single-nucleotide variants (SNVs) of unknown function are reported in tumors. Partitioning the genome according to cistromes reveals the enrichment of somatic SNVs in prostate tumors as opposed to adjacent normal tissue cistromes of master transcription regulators, including AR, FOXA1, and HOXB13. This parallels enrichment of prostate cancer genetic predispositions over these transcription regulators' tumor cistromes, exemplified at the 8q24 locus harboring both risk variants and somatic SNVs in cis-regulatory elements upregulating MYC expression. However, Massively Parallel Reporter Assays reveal that few SNVs can alter the transactivation potential of individual cis-regulatory elements. Instead, similar to inherited risk variants, SNVs accumulate in cistromes of master transcription regulators required for prostate cancer development.


Subject(s)
Gene Expression Regulation, Neoplastic/genetics , Hepatocyte Nuclear Factor 3-alpha/metabolism , Homeodomain Proteins/metabolism , Prostatic Neoplasms/metabolism , Cell Line, Tumor , Homeodomain Proteins/genetics , Humans , Male , Mutation/genetics , Prostatic Neoplasms/pathology , Up-Regulation/genetics
9.
Cancer Cell ; 36(1): 51-67.e7, 2019 07 08.
Article in English | MEDLINE | ID: mdl-31287992

ABSTRACT

Embryonal tumors with multilayered rosettes (ETMRs) are highly lethal infant brain cancers with characteristic amplification of Chr19q13.41 miRNA cluster (C19MC) and enrichment of pluripotency factor LIN28A. Here we investigated C19MC oncogenic mechanisms and discovered a C19MC-LIN28A-MYCN circuit fueled by multiple complex regulatory loops including an MYCN core transcriptional network and super-enhancers resulting from long-range MYCN DNA interactions and C19MC gene fusions. Our data show that this powerful oncogenic circuit, which entraps an early neural lineage network, is potently abrogated by bromodomain inhibitor JQ1, leading to ETMR cell death.


Subject(s)
Brain Neoplasms/etiology , Chromosomes, Human, Pair 19 , MicroRNAs/genetics , Multigene Family , N-Myc Proto-Oncogene Protein/genetics , Neoplasms, Germ Cell and Embryonal/etiology , RNA-Binding Proteins/genetics , Biomarkers, Tumor , Brain Neoplasms/diagnosis , Brain Neoplasms/therapy , Cell Cycle/genetics , Cell Transformation, Neoplastic/drug effects , Cell Transformation, Neoplastic/genetics , Chromosomes, Human, Pair 2 , DNA Copy Number Variations , Enhancer Elements, Genetic , Epigenesis, Genetic , Gene Expression Regulation , Gene Regulatory Networks , Genetic Association Studies , Genetic Predisposition to Disease , Humans , Models, Biological , Neoplasms, Germ Cell and Embryonal/diagnosis , Neoplasms, Germ Cell and Embryonal/therapy , Oncogenes
10.
Genome Res ; 29(8): 1211-1222, 2019 08.
Article in English | MEDLINE | ID: mdl-31249064

ABSTRACT

We investigated the role of 3D genome architecture in instructing functional properties of glioblastoma stem cells (GSCs) by generating sub-5-kb resolution 3D genome maps by in situ Hi-C. Contact maps at sub-5-kb resolution allow identification of individual DNA loops, domain organization, and large-scale genome compartmentalization. We observed differences in looping architectures among GSCs from different patients, suggesting that 3D genome architecture is a further layer of inter-patient heterogeneity for glioblastoma. Integration of DNA contact maps with chromatin and transcriptional profiles identified specific mechanisms of gene regulation, including the convergence of multiple super enhancers to individual stemness genes within individual cells. We show that the number of loops contacting a gene correlates with elevated transcription. These results indicate that stemness genes are hubs of interaction between multiple regulatory regions, likely to ensure their sustained expression. Regions of open chromatin common among the GSCs tested were poised for expression of immune-related genes, including CD276 We demonstrate that this gene is co-expressed with stemness genes in GSCs and that CD276 can be targeted with an antibody-drug conjugate to eliminate self-renewing cells. Our results demonstrate that integrated structural genomics data sets can be employed to rationally identify therapeutic vulnerabilities in self-renewing cells.


Subject(s)
Brain Neoplasms/genetics , Chromatin/ultrastructure , Chromosome Mapping/methods , Gene Expression Regulation, Neoplastic , Glioblastoma/genetics , Neoplasm Proteins/genetics , B7 Antigens/antagonists & inhibitors , B7 Antigens/genetics , B7 Antigens/metabolism , Brain Neoplasms/metabolism , Brain Neoplasms/pathology , Cell Proliferation , Chromatin/chemistry , Enhancer Elements, Genetic , Gene Expression Profiling , Genetic Heterogeneity , Genome, Human , Genomics/methods , Glioblastoma/metabolism , Glioblastoma/pathology , Humans , Molecular Targeted Therapy , Neoplasm Proteins/classification , Neoplasm Proteins/metabolism , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology , Primary Cell Culture , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Transcription, Genetic
11.
Cancer Cell ; 35(5): 782-797.e8, 2019 05 13.
Article in English | MEDLINE | ID: mdl-31085178

ABSTRACT

High-grade gliomas defined by histone 3 K27M driver mutations exhibit global loss of H3K27 trimethylation and reciprocal gain of H3K27 acetylation, respectively shaping repressive and active chromatin landscapes. We generated tumor-derived isogenic models bearing this mutation and show that it leads to pervasive H3K27ac deposition across the genome. In turn, active enhancers and promoters are not created de novo and instead reflect the epigenomic landscape of the cell of origin. H3K27ac is enriched at repeat elements, resulting in their increased expression, which in turn can be further amplified by DNA demethylation and histone deacetylase inhibitors providing an exquisite therapeutic vulnerability. These agents may therefore modulate anti-tumor immune responses as a therapeutic modality for this untreatable disease.


Subject(s)
Brain Neoplasms/metabolism , Glioma/metabolism , Histones/genetics , Histones/metabolism , Acetylation , Brain Neoplasms/drug therapy , Brain Neoplasms/genetics , Cell Line, Tumor , Chromatin/metabolism , Enhancer Elements, Genetic/drug effects , Epigenomics/methods , Gene Expression Regulation, Neoplastic/drug effects , Glioma/drug therapy , Glioma/genetics , Histone Deacetylase Inhibitors/pharmacology , Histone Deacetylase Inhibitors/therapeutic use , Humans , Mutation
12.
Bioinformatics ; 35(5): 877-879, 2019 03 01.
Article in English | MEDLINE | ID: mdl-30816925

ABSTRACT

MOTIVATION: The 3D genome architecture influences the regulation of genes by facilitating chromatin interactions between distal cis-regulatory elements and gene promoters. We implement Cross Cell-type Correlation based on DNA accessibility (C3D), a customizable computational tool that predicts chromatin interactions using an unsupervised algorithm that utilizes correlations in chromatin measurements, such as DNaseI hypersensitivity signals. RESULTS: C3D accurately predicts 32.7%, 18.3% and 24.1% of interactions, validated by ChIA-PET assays, between promoters and distal regions that overlie DNaseI hypersensitive sites in K562, MCF-7 and GM12878 cells, respectively. AVAILABILITY AND IMPLEMENTATION: Source code is open-source and freely available on GitHub (https://github.com/LupienLabOrganization/C3D) under the GNU GPLv3 license. C3D is implemented in Bash and R; it runs on any platform with Bash (≥4.0), R (≥3.1.1) and BEDTools (≥2.19.0). It requires the following R packages: GenomicRanges, Sushi, data.table, preprocessCore and dynamicTreeCut. SUPPLEMENTARY INFORMATION: Supplementary data are available at Bioinformatics online.


Subject(s)
Genome , Genomics , Chromatin Immunoprecipitation , Regulatory Sequences, Nucleic Acid , Software
13.
BMC Bioinformatics ; 19(1): 454, 2018 Nov 26.
Article in English | MEDLINE | ID: mdl-30477433

ABSTRACT

BACKGROUND: Single Nucleotide Variants (SNVs), including somatic point mutations and Single Nucleotide Polymorphisms (SNPs), in noncoding cis-regulatory elements (CREs) can affect gene regulation and lead to disease development. Several approaches have been developed to identify highly mutated regions, but these do not take into account the specific genomic context, and thus likelihood of mutation, of CREs. RESULTS: Here, we present SMuRF (Significantly Mutated Region Finder), a user-friendly command-line tool to identify these significantly mutated regions from user-defined genomic intervals and SNVs. We demonstrate this using publicly available datasets in which SMuRF identifies 72 significantly mutated CREs in liver cancer, including known mutated gene promoters as well as previously unreported regions. CONCLUSIONS: SMuRF is a helpful tool to allow the simple identification of significantly mutated regulatory elements. It is open-source and freely available on GitHub ( https://github.com/LupienLab/SMURF ).


Subject(s)
Point Mutation , Regulatory Sequences, Nucleic Acid , Software , Genomics , Humans , Liver Neoplasms/genetics
14.
Cell Rep ; 21(10): 2772-2784, 2017 Dec 05.
Article in English | MEDLINE | ID: mdl-29212025

ABSTRACT

We describe molecular convergence between BMI1 and CHD7 in the initiation of medulloblastoma. Identified in a functional genomic screen in mouse models, a BMI1High;CHD7Low expression signature within medulloblastoma characterizes patients with poor overall survival. We show that BMI1-mediated repression of the ERK1/2 pathway leads to increased proliferation and tumor burden in primary human MB cells and in a xenograft model, respectively. We provide evidence that repression of the ERK inhibitor DUSP4 by BMI1 is dependent on a more accessible chromatin configuration in G4 MB cells with low CHD7 expression. These findings extend current knowledge of the role of BMI1 and CHD7 in medulloblastoma pathogenesis, and they raise the possibility that pharmacological targeting of BMI1 or ERK may be particularly indicated in a subgroup of MB with low expression levels of CHD7.


Subject(s)
DNA-Binding Proteins/metabolism , Extracellular Signal-Regulated MAP Kinases/metabolism , Medulloblastoma/metabolism , Polycomb Repressive Complex 1/metabolism , Proto-Oncogene Proteins/metabolism , Animals , Blotting, Western , Cell Proliferation/genetics , Cell Proliferation/physiology , Chromatin/genetics , Chromatin/metabolism , DNA-Binding Proteins/genetics , Extracellular Signal-Regulated MAP Kinases/genetics , Female , Male , Medulloblastoma/genetics , Mice , Polycomb Repressive Complex 1/genetics , Protein Tyrosine Phosphatases/genetics , Protein Tyrosine Phosphatases/metabolism , Proto-Oncogene Proteins/genetics , Signal Transduction/genetics , Signal Transduction/physiology
16.
Nature ; 549(7671): 227-232, 2017 09 14.
Article in English | MEDLINE | ID: mdl-28854171

ABSTRACT

Human glioblastomas harbour a subpopulation of glioblastoma stem cells that drive tumorigenesis. However, the origin of intratumoural functional heterogeneity between glioblastoma cells remains poorly understood. Here we study the clonal evolution of barcoded glioblastoma cells in an unbiased way following serial xenotransplantation to define their individual fate behaviours. Independent of an evolving mutational signature, we show that the growth of glioblastoma clones in vivo is consistent with a remarkably neutral process involving a conserved proliferative hierarchy rooted in glioblastoma stem cells. In this model, slow-cycling stem-like cells give rise to a more rapidly cycling progenitor population with extensive self-maintenance capacity, which in turn generates non-proliferative cells. We also identify rare 'outlier' clones that deviate from these dynamics, and further show that chemotherapy facilitates the expansion of pre-existing drug-resistant glioblastoma stem cells. Finally, we show that functionally distinct glioblastoma stem cells can be separately targeted using epigenetic compounds, suggesting new avenues for glioblastoma-targeted therapy.


Subject(s)
Cell Differentiation , Cell Lineage , Cell Tracking , Glioblastoma/pathology , Neoplastic Stem Cells/pathology , Animals , Cell Differentiation/drug effects , Cell Lineage/drug effects , Cell Proliferation , Clone Cells/drug effects , Clone Cells/pathology , Epigenesis, Genetic , Female , Glioblastoma/drug therapy , Heterografts , Humans , Mice , Neoplasm Invasiveness , Neoplasm Transplantation , Neoplastic Stem Cells/drug effects , Phenotype , Stochastic Processes
17.
Cell Stem Cell ; 21(2): 209-224.e7, 2017 08 03.
Article in English | MEDLINE | ID: mdl-28712938

ABSTRACT

Glioblastomas exhibit a hierarchical cellular organization, suggesting that they are driven by neoplastic stem cells that retain partial yet abnormal differentiation potential. Here, we show that a large subset of patient-derived glioblastoma stem cells (GSCs) express high levels of Achaete-scute homolog 1 (ASCL1), a proneural transcription factor involved in normal neurogenesis. ASCL1hi GSCs exhibit a latent capacity for terminal neuronal differentiation in response to inhibition of Notch signaling, whereas ASCL1lo GSCs do not. Increasing ASCL1 levels in ASCL1lo GSCs restores neuronal lineage potential, promotes terminal differentiation, and attenuates tumorigenicity. ASCL1 mediates these effects by functioning as a pioneer factor at closed chromatin, opening new sites to activate a neurogenic gene expression program. Directing GSCs toward terminal differentiation may provide therapeutic applications for a subset of GBM patients and strongly supports efforts to restore differentiation potential in GBM and other cancers.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/metabolism , Brain Neoplasms/pathology , Carcinogenesis/pathology , Cell Lineage , Chromatin/metabolism , Glioblastoma/pathology , Neurons/pathology , Base Sequence , Basic Helix-Loop-Helix Transcription Factors/genetics , Brain Neoplasms/genetics , Carcinogenesis/genetics , Cell Differentiation/genetics , Disease Progression , Enhancer Elements, Genetic/genetics , Gene Expression Regulation, Neoplastic , Glioblastoma/genetics , Humans , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology , Neurons/metabolism , Promoter Regions, Genetic/genetics , Protein Binding , Sequence Analysis, RNA , Up-Regulation/genetics
18.
Cancer Cell ; 30(6): 891-908, 2016 Dec 12.
Article in English | MEDLINE | ID: mdl-27960086

ABSTRACT

We recently reported that atypical teratoid rhabdoid tumors (ATRTs) comprise at least two transcriptional subtypes with different clinical outcomes; however, the mechanisms underlying therapeutic heterogeneity remained unclear. In this study, we analyzed 191 primary ATRTs and 10 ATRT cell lines to define the genomic and epigenomic landscape of ATRTs and identify subgroup-specific therapeutic targets. We found ATRTs segregated into three epigenetic subgroups with distinct genomic profiles, SMARCB1 genotypes, and chromatin landscape that correlated with differential cellular responses to a panel of signaling and epigenetic inhibitors. Significantly, we discovered that differential methylation of a PDGFRB-associated enhancer confers specific sensitivity of group 2 ATRT cells to dasatinib and nilotinib, and suggest that these are promising therapies for this highly lethal ATRT subtype.


Subject(s)
Central Nervous System Neoplasms/genetics , Chromatin/genetics , Epigenomics/methods , Receptor, Platelet-Derived Growth Factor beta/genetics , Rhabdoid Tumor/genetics , SMARCB1 Protein/genetics , Teratoma/genetics , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Central Nervous System Neoplasms/drug therapy , DNA Methylation , Dasatinib/pharmacology , Dasatinib/therapeutic use , Epigenesis, Genetic/drug effects , Humans , Mutation , Protein Kinase Inhibitors/pharmacology , Protein Kinase Inhibitors/therapeutic use , Pyrimidines/pharmacology , Pyrimidines/therapeutic use , Receptor, Platelet-Derived Growth Factor beta/antagonists & inhibitors , Rhabdoid Tumor/drug therapy , Teratoma/drug therapy
19.
Genome Med ; 7(1): 11, 2015.
Article in English | MEDLINE | ID: mdl-25763109

ABSTRACT

BACKGROUND: Wilms tumours (WTs) are characterised by several hallmarks that suggest epimutations such as aberrant DNA methylation are involved in tumour progression: loss of imprinting at 11p15, lack of recurrent mutations and formation of nephrogenic rests (NRs), which are lesions of retained undifferentiated embryonic tissue that can give rise to WTs. METHODS: To identify such epimutations, we performed a comprehensive methylome analysis on 20 matched trios of micro-dissected WTs, NRs and surrounding normal kidneys (NKs) using Illumina Infinium HumanMethylation450 Bead Chips and functionally validated findings using RNA sequencing. RESULTS: Comparison of NRs with NK revealed prominent tissue biomarkers: 629 differentially methylated regions, of which 55% were hypermethylated and enriched for domains that are bivalent in embryonic stem cells and for genes expressed during development (P = 2.49 × 10(-5)). Comparison of WTs with NRs revealed two WT subgroups; group-2 WTs and NRs were epigenetically indistinguishable whereas group-1 WTs showed an increase in methylation variability, hypomethylation of renal development genes, hypermethylation and relative loss of expression of cell adhesion genes and known and potential new WT tumour suppressor genes (CASP8, H19, MIR195, RB1 and TSPAN32) and was strongly associated with bilateral disease (P = 0.032). Comparison of WTs and NRs to embryonic kidney highlighted the significance of polycomb target methylation in Wilms tumourigenesis. CONCLUSIONS: Methylation levels vary during cancer evolution. We have described biomarkers related to WT evolution from its precursor NRs which may be useful to differentiate between these tissues for patients with bilateral disease.

20.
Methods ; 72: 9-15, 2015 Jan 15.
Article in English | MEDLINE | ID: mdl-25175075

ABSTRACT

DNA methylation analysis has become an integral part of biomedical research. For high-throughput applications such as epigenome-wide association studies, the Infinium HumanMethylation450 (450K) BeadChip is currently the platform of choice. However, BeadChip processing relies on traditional bisulfite (BS) based protocols which cannot discriminate between 5-methylcytosine (5mC) and 5-hydroxymethylcytosine (5hmC). Here, we report the adaptation of the recently developed oxidative bisulfite (oxBS) chemistry to specifically detect both 5mC and 5hmC in a single workflow using 450K BeadChips, termed oxBS-450K. Supported by validation using mass spectrometry and pyrosequencing, we demonstrate reproducible (R(2)>0.99) detection of 5hmC in human brain tissue using the optimised oxBS-450K protocol described here.


Subject(s)
DNA Methylation , Epigenomics/methods , Epigenomics/instrumentation , Humans , Oligonucleotide Array Sequence Analysis/instrumentation , Oligonucleotide Array Sequence Analysis/methods , Reproducibility of Results , Sequence Analysis, DNA/methods
SELECTION OF CITATIONS
SEARCH DETAIL
...