Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 24
Filter
1.
Nature ; 569(7757): 503-508, 2019 05.
Article in English | MEDLINE | ID: mdl-31068700

ABSTRACT

Large panels of comprehensively characterized human cancer models, including the Cancer Cell Line Encyclopedia (CCLE), have provided a rigorous framework with which to study genetic variants, candidate targets, and small-molecule and biological therapeutics and to identify new marker-driven cancer dependencies. To improve our understanding of the molecular features that contribute to cancer phenotypes, including drug responses, here we have expanded the characterizations of cancer cell lines to include genetic, RNA splicing, DNA methylation, histone H3 modification, microRNA expression and reverse-phase protein array data for 1,072 cell lines from individuals of various lineages and ethnicities. Integration of these data with functional characterizations such as drug-sensitivity, short hairpin RNA knockdown and CRISPR-Cas9 knockout data reveals potential targets for cancer drugs and associated biomarkers. Together, this dataset and an accompanying public data portal provide a resource for the acceleration of cancer research using model cancer cell lines.


Subject(s)
Cell Line, Tumor , Neoplasms/genetics , Neoplasms/pathology , Antineoplastic Agents/pharmacology , Biomarkers, Tumor , DNA Methylation , Drug Resistance, Neoplasm , Ethnicity/genetics , Gene Editing , Histones/metabolism , Humans , MicroRNAs/genetics , Molecular Targeted Therapy , Neoplasms/metabolism , Protein Array Analysis , RNA Splicing
2.
Stem Cells ; 34(5): 1321-31, 2016 05.
Article in English | MEDLINE | ID: mdl-26756672

ABSTRACT

Inhibitor of DNA binding proteins (Id1-Id4) function to inhibit differentiation and promote proliferation of many different cell types. Among the Id family members, Id2 has been most extensively studied in the central nervous system (CNS). Id2 contributes to cultured neural precursor cell (NPC) proliferation as well as to the proliferation of CNS tumors such as glioblastoma that are likely to arise from NPC-like cells. We identified three phosphorylation sites near the N-terminus of Id2 in NPCs. To interrogate the importance of Id2 phosphorylation, Id2(-/-) NPCs were modified to express wild type (WT) Id2 or an Id2 mutant protein that could not be phosphorylated at the identified sites. We observed that NPCs expressing this mutant lacking phosphorylation near the N-terminus had higher steady-state levels of Id2 when compared to NPCs expressing WT Id2. This elevated level was the result of a longer half-life and reduced proteasome-mediated degradation. Moreover, NPCs expressing constitutively de-phosphorylated Id2 proliferated more rapidly than NPCs expressing WT Id2, a finding consistent with the well-characterized function of Id2 in driving proliferation. Observing that phosphorylation of Id2 modulates the degradation of this important cell-cycle regulator, we sought to identify a phosphatase that would stabilize Id2 enhancing its activity in NPCs and extended our analysis to include human glioblastoma-derived stem cells (GSCs). We found that expression of the phosphatase PP2A altered Id2 levels. Our findings suggest that inhibition of PP2A may be a novel strategy to regulate the proliferation of normal NPCs and malignant GSCs by decreasing Id2 levels. Stem Cells 2016;34:1321-1331.


Subject(s)
Inhibitor of Differentiation Protein 2/metabolism , Neural Stem Cells/cytology , Neural Stem Cells/metabolism , Proteolysis , Amino Acid Sequence , Animals , Cell Cycle , Cell Proliferation , Glioblastoma/pathology , Inhibitor of Differentiation Protein 2/chemistry , Mice , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology , Phosphorylation , Proteasome Endopeptidase Complex/metabolism , Protein Phosphatase 2/metabolism
3.
Genome Biol ; 24(1): 192, 2023 08 23.
Article in English | MEDLINE | ID: mdl-37612728

ABSTRACT

BACKGROUND: Hundreds of functional genomic screens have been performed across a diverse set of cancer contexts, as part of efforts such as the Cancer Dependency Map, to identify gene dependencies-genes whose loss of function reduces cell viability or fitness. Recently, large-scale screening efforts have shifted from RNAi to CRISPR-Cas9, due to superior efficacy and specificity. However, many effective oncology drugs only partially inhibit their protein targets, leading us to question whether partial suppression of genes using RNAi could reveal cancer vulnerabilities that are missed by complete knockout using CRISPR-Cas9. Here, we compare CRISPR-Cas9 and RNAi dependency profiles of genes across approximately 400 matched cancer cell lines. RESULTS: We find that CRISPR screens accurately identify more gene dependencies per cell line, but the majority of each cell line's dependencies are part of a set of 1867 genes that are shared dependencies across the entire collection (pan-lethals). While RNAi knockdown of about 30% of these genes is also pan-lethal, approximately 50% have selective dependency patterns across cell lines, suggesting they could still be cancer vulnerabilities. The accuracy of the unique RNAi selectivity is supported by associations to multi-omics profiles, drug sensitivity, and other expected co-dependencies. CONCLUSIONS: Incorporating RNAi data for genes that are pan-lethal knockouts facilitates the discovery of a wider range of gene targets than could be detected using the CRISPR dataset alone. This can aid in the interpretation of contrasting results obtained from CRISPR and RNAi screens and reinforce the importance of partial gene suppression methods in building a cancer dependency map.


Subject(s)
CRISPR-Cas Systems , Neoplasms , Humans , Neoplasms/genetics , Genetic Techniques , RNA Interference , Cell Line
4.
Stem Cells ; 29(7): 1090-101, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21608079

ABSTRACT

Neural progenitor cells (NPCs) have the capacity to proliferate and give rise to all major central nervous system cell types and represent a possible cell of origin in gliomagenesis. Deletion of the tumor suppressor gene Tp53 (p53) results in increased proliferation and self-renewal of NPCs and is a common genetic mutation found in glioma. We have identified inhibitor of DNA binding 2 (Id2) as a novel target gene directly repressed by p53 to maintain normal NPC proliferation. p53((-/-)) NPCs express elevated levels of Id2 and suppression of Id2 expression is sufficient to inhibit the increased proliferation and self-renewal which results from p53 loss. Elevated expression of Id2 in wild-type NPCs phenocopies the behavior of p53((-/-)) NPCs by enhancing NPC proliferation and self-renewal. Interestingly, p53 directly binds to a conserved site within the Id2 promoter to mediate these effects. Finally, we have identified elevated Id2 expression in glioma cell lines with mutated p53 and demonstrated that constitutive expression of Id2 plays a key role in the proliferation of glioma stem-like cells. These findings indicate that Id2 functions as a proproliferative gene that antagonizes p53-mediated cell cycle regulation in NPCs and may contribute to the malignant proliferation of glioma-derived tumor stem cells.


Subject(s)
Genes, p53 , Inhibitor of Differentiation Protein 2/genetics , Neural Stem Cells/cytology , Stem Cells/cytology , Animals , Cell Growth Processes/physiology , Cell Line, Tumor , Gene Expression Regulation, Neoplastic , Gene Knockdown Techniques , Glioma/genetics , Glioma/metabolism , Glioma/pathology , Humans , Inhibitor of Differentiation Protein 2/biosynthesis , Mice , Mice, Inbred C57BL , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology , Promoter Regions, Genetic
5.
Nat Commun ; 13(1): 5495, 2022 09 20.
Article in English | MEDLINE | ID: mdl-36127368

ABSTRACT

Conditional degron tags (CDTs) are a powerful tool for target validation that combines the kinetics and reversible action of pharmacological agents with the generalizability of genetic manipulation. However, successful design of a CDT fusion protein often requires a prolonged, ad hoc cycle of construct design, failure, and re-design. To address this limitation, we report here a system to rapidly compare the activity of five unique CDTs: AID/AID2, IKZF3d, dTAG, HaloTag, and SMASh. We demonstrate the utility of this system against 16 unique protein targets. We find that expression and degradation are highly dependent on the specific CDT, the construct design, and the target. None of the CDTs leads to efficient expression and/or degradation across all targets; however, our systematic approach enables the identification of at least one optimal CDT fusion for each target. To enable the adoption of CDT strategies more broadly, we have made these reagents, and a detailed protocol, available as a community resource.


Subject(s)
Proteolysis , Kinetics
6.
Nat Cancer ; 3(6): 681-695, 2022 06.
Article in English | MEDLINE | ID: mdl-35437317

ABSTRACT

Despite advances in precision medicine, the clinical prospects for patients with ovarian and uterine cancers have not substantially improved. Here, we analyzed genome-scale CRISPR-Cas9 loss-of-function screens across 851 human cancer cell lines and found that frequent overexpression of SLC34A2-encoding a phosphate importer-is correlated with sensitivity to loss of the phosphate exporter XPR1, both in vitro and in vivo. In patient-derived tumor samples, we observed frequent PAX8-dependent overexpression of SLC34A2, XPR1 copy number amplifications and XPR1 messenger RNA overexpression. Mechanistically, in SLC34A2-high cancer cell lines, genetic or pharmacologic inhibition of XPR1-dependent phosphate efflux leads to the toxic accumulation of intracellular phosphate. Finally, we show that XPR1 requires the novel partner protein KIDINS220 for proper cellular localization and activity, and that disruption of this protein complex results in acidic "vacuolar" structures preceding cell death. These data point to the XPR1-KIDINS220 complex and phosphate dysregulation as a therapeutic vulnerability in ovarian cancer.


Subject(s)
Membrane Proteins , Nerve Tissue Proteins , Ovarian Neoplasms , Female , Humans , Membrane Proteins/genetics , Membrane Proteins/metabolism , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Ovarian Neoplasms/drug therapy , Ovarian Neoplasms/genetics , Ovarian Neoplasms/metabolism , Phosphates/pharmacology , Receptors, G-Protein-Coupled/genetics , Receptors, Virus/genetics , Xenotropic and Polytropic Retrovirus Receptor/genetics , Xenotropic and Polytropic Retrovirus Receptor/metabolism
7.
Nat Genet ; 53(12): 1664-1672, 2021 12.
Article in English | MEDLINE | ID: mdl-34857952

ABSTRACT

Although single-gene perturbation screens have revealed a number of new targets, vulnerabilities specific to frequently altered drivers have not been uncovered. An important question is whether the compensatory relationship between functionally redundant genes masks potential therapeutic targets in single-gene perturbation studies. To identify digenic dependencies, we developed a CRISPR paralog targeting library to investigate the viability effects of disrupting 3,284 genes, 5,065 paralog pairs and 815 paralog families. We identified that dual inactivation of DUSP4 and DUSP6 selectively impairs growth in NRAS and BRAF mutant cells through the hyperactivation of MAPK signaling. Furthermore, cells resistant to MAPK pathway therapeutics become cross-sensitized to DUSP4 and DUSP6 perturbations such that the mechanisms of resistance to the inhibitors reinforce this mechanism of vulnerability. Together, multigene perturbation technologies unveil previously unrecognized digenic vulnerabilities that may be leveraged as new therapeutic targets in cancer.


Subject(s)
Dual Specificity Phosphatase 6/genetics , Dual-Specificity Phosphatases/genetics , MAP Kinase Signaling System , Mitogen-Activated Protein Kinase Phosphatases/genetics , Neoplasms/genetics , Cell Line, Tumor , Clustered Regularly Interspaced Short Palindromic Repeats , Enzyme Activation , GTP Phosphohydrolases/genetics , Gene Knockout Techniques , Humans , Melanoma, Experimental/genetics , Melanoma, Experimental/therapy , Membrane Proteins/genetics , Neoplasms/enzymology , Neoplasms/metabolism , Neoplasms/therapy , Proto-Oncogene Proteins B-raf/genetics
8.
Cancer Discov ; 11(9): 2282-2299, 2021 09.
Article in English | MEDLINE | ID: mdl-33883167

ABSTRACT

Cancer dependency maps, which use CRISPR/Cas9 depletion screens to profile the landscape of genetic dependencies in hundreds of cancer cell lines, have identified context-specific dependencies that could be therapeutically exploited. An ideal therapy is both lethal and precise, but these depletion screens cannot readily distinguish between gene effects that are cytostatic or cytotoxic. Here, we use a diverse panel of functional genomic screening assays to identify NXT1 as a selective and rapidly lethal in vivo relevant genetic dependency in MYCN-amplified neuroblastoma. NXT1 heterodimerizes with NXF1, and together they form the principal mRNA nuclear export machinery. We describe a previously unrecognized mechanism of synthetic lethality between NXT1 and its paralog NXT2: their common essential binding partner NXF1 is lost only in the absence of both. We propose a potential therapeutic strategy for tumor-selective elimination of a protein that, if targeted directly, is expected to cause widespread toxicity. SIGNIFICANCE: We provide a framework for identifying new therapeutic targets from functional genomic screens. We nominate NXT1 as a selective lethal target in neuroblastoma and propose a therapeutic approach where the essential protein NXF1 can be selectively eliminated in tumor cells by exploiting the NXT1-NXT2 paralog relationship.See related commentary by Wang and Abdel-Wahab, p. 2129.This article is highlighted in the In This Issue feature, p. 2113.


Subject(s)
Neoplasms/drug therapy , Nucleocytoplasmic Transport Proteins/genetics , Cell Line, Tumor , Humans , Neoplasms/genetics
9.
Nat Genet ; 53(4): 529-538, 2021 04.
Article in English | MEDLINE | ID: mdl-33753930

ABSTRACT

Exciting therapeutic targets are emerging from CRISPR-based screens of high mutational-burden adult cancers. A key question, however, is whether functional genomic approaches will yield new targets in pediatric cancers, known for remarkably few mutations, which often encode proteins considered challenging drug targets. To address this, we created a first-generation pediatric cancer dependency map representing 13 pediatric solid and brain tumor types. Eighty-two pediatric cancer cell lines were subjected to genome-scale CRISPR-Cas9 loss-of-function screening to identify genes required for cell survival. In contrast to the finding that pediatric cancers harbor fewer somatic mutations, we found a similar complexity of genetic dependencies in pediatric cancer cell lines compared to that in adult models. Findings from the pediatric cancer dependency map provide preclinical support for ongoing precision medicine clinical trials. The vulnerabilities observed in pediatric cancers were often distinct from those in adult cancer, indicating that repurposing adult oncology drugs will be insufficient to address childhood cancers.


Subject(s)
Chromosome Mapping/methods , Gene Expression Regulation, Neoplastic , Genome, Human , Mutation , Neoplasm Proteins/genetics , Neoplasms/genetics , Adult , CRISPR-Associated Protein 9/genetics , CRISPR-Associated Protein 9/metabolism , CRISPR-Cas Systems , Cell Line, Tumor , Child , Clustered Regularly Interspaced Short Palindromic Repeats , Gene Editing , Gene Expression Profiling , Genetic Predisposition to Disease , Humans , Neoplasm Proteins/classification , Neoplasm Proteins/metabolism , Neoplasms/metabolism , Neoplasms/pathology , RNA, Guide, Kinetoplastida/genetics , RNA, Guide, Kinetoplastida/metabolism
10.
ACS Chem Biol ; 15(6): 1358-1369, 2020 06 19.
Article in English | MEDLINE | ID: mdl-32348107

ABSTRACT

Antisense oligonucleotide therapies are important cancer treatments, which can suppress genes in cancer cells that are critical for cell survival. SF3B1 has recently emerged as a promising gene target that encodes a key splicing factor in the SF3B protein complex. Over 10% of cancers have lost one or more copies of the SF3B1 gene, rendering these cancers vulnerable after further suppression. SF3B1 is just one example of a CYCLOPS (Copy-number alterations Yielding Cancer Liabilities Owing to Partial losS) gene, but over 120 additional candidate CYCLOPS genes are known. Antisense oligonucleotide therapies for cancer offer the promise of effective suppression for CYCLOPS genes, but developing these treatments is difficult due to their limited permeability into cells and poor cytosolic stability. Here, we develop an effective approach to suppress CYCLOPS genes by delivering antisense peptide nucleic acids (PNAs) into the cytosol of cancer cells. We achieve efficient cytosolic PNA delivery with the two main nontoxic components of the anthrax toxin: protective antigen (PA) and the 263-residue N-terminal domain of lethal factor (LFN). Sortase-mediated ligation readily enables the conjugation of PNAs to the C terminus of the LFN protein. LFN and PA work together in concert to translocate PNAs into the cytosol of mammalian cells. Antisense SF3B1 PNAs delivered with the LFN/PA system suppress the SF3B1 gene and decrease cell viability, particularly of cancer cells with partial copy-number loss of SF3B1. Moreover, antisense SF3B1 PNAs delivered with a HER2-binding PA variant selectively target cancer cells that overexpress the HER2 cell receptor, demonstrating receptor-specific targeting of cancer cells. Taken together, our efforts illustrate how PA-mediated delivery of PNAs provides an effective and general approach for delivering antisense PNA therapeutics and for targeting gene dependencies in cancer.


Subject(s)
Antigens, Bacterial/chemistry , Bacterial Toxins/chemistry , Drug Carriers/chemistry , Oligonucleotides, Antisense/administration & dosage , Peptide Nucleic Acids/administration & dosage , Cell Line, Tumor , Cell Survival/drug effects , Drug Delivery Systems , Genetic Therapy , Humans , Neoplasms/genetics , Neoplasms/therapy , Oligonucleotides, Antisense/pharmacology , Peptide Nucleic Acids/pharmacology , Phosphoproteins/genetics , RNA Splicing Factors/genetics
11.
Nat Commun ; 11(1): 2517, 2020 05 20.
Article in English | MEDLINE | ID: mdl-32433464

ABSTRACT

Alterations in non-driver genes represent an emerging class of potential therapeutic targets in cancer. Hundreds to thousands of non-driver genes undergo loss of heterozygosity (LOH) events per tumor, generating discrete differences between tumor and normal cells. Here we interrogate LOH of polymorphisms in essential genes as a novel class of therapeutic targets. We hypothesized that monoallelic inactivation of the allele retained in tumors can selectively kill cancer cells but not somatic cells, which retain both alleles. We identified 5664 variants in 1278 essential genes that undergo LOH in cancer and evaluated the potential for each to be targeted using allele-specific gene-editing, RNAi, or small-molecule approaches. We further show that allele-specific inactivation of either of two essential genes (PRIM1 and EXOSC8) reduces growth of cells harboring that allele, while cells harboring the non-targeted allele remain intact. We conclude that LOH of essential genes represents a rich class of non-driver cancer vulnerabilities.


Subject(s)
Genes, Essential , Loss of Heterozygosity , Neoplasms/genetics , Alleles , Cell Proliferation , DNA Primase/genetics , Exosome Multienzyme Ribonuclease Complex/genetics , Humans , Models, Genetic , Neoplasms/physiopathology , RNA-Binding Proteins/genetics
12.
Nat Commun ; 11(1): 4296, 2020 08 27.
Article in English | MEDLINE | ID: mdl-32855387

ABSTRACT

Assays to study cancer cell responses to pharmacologic or genetic perturbations are typically restricted to using simple phenotypic readouts such as proliferation rate. Information-rich assays, such as gene-expression profiling, have generally not permitted efficient profiling of a given perturbation across multiple cellular contexts. Here, we develop MIX-Seq, a method for multiplexed transcriptional profiling of post-perturbation responses across a mixture of samples with single-cell resolution, using SNP-based computational demultiplexing of single-cell RNA-sequencing data. We show that MIX-Seq can be used to profile responses to chemical or genetic perturbations across pools of 100 or more cancer cell lines. We combine it with Cell Hashing to further multiplex additional experimental conditions, such as post-treatment time points or drug doses. Analyzing the high-content readout of scRNA-seq reveals both shared and context-specific transcriptional response components that can identify drug mechanism of action and enable prediction of long-term cell viability from short-term transcriptional responses to treatment.


Subject(s)
Gene Expression Profiling/methods , Neoplasms/genetics , Single-Cell Analysis/methods , Antineoplastic Agents/pharmacology , Base Sequence , Cell Line, Tumor , Cell Survival/genetics , Gene Expression Regulation, Neoplastic/drug effects , Humans , Models, Statistical , Neoplasms/drug therapy , Neoplasms/pathology , Polymorphism, Single Nucleotide , Pyridones/pharmacology , Pyrimidinones/pharmacology
13.
Cell Rep ; 33(11): 108493, 2020 12 15.
Article in English | MEDLINE | ID: mdl-33326793

ABSTRACT

Few therapies target the loss of tumor suppressor genes in cancer. We examine CRISPR-SpCas9 and RNA-interference loss-of-function screens to identify new therapeutic targets associated with genomic loss of tumor suppressor genes. The endosomal sorting complexes required for transport (ESCRT) ATPases VPS4A and VPS4B score as strong synthetic lethal dependencies. VPS4A is essential in cancers harboring loss of VPS4B adjacent to SMAD4 on chromosome 18q and VPS4B is required in tumors with co-deletion of VPS4A and CDH1 (E-cadherin) on chromosome 16q. We demonstrate that more than 30% of cancers selectively require VPS4A or VPS4B. VPS4A suppression in VPS4B-deficient cells selectively leads to ESCRT-III filament accumulation, cytokinesis defects, nuclear deformation, G2/M arrest, apoptosis, and potent tumor regression. CRISPR-SpCas9 screening and integrative genomic analysis reveal other ESCRT members, regulators of abscission, and interferon signaling as modifiers of VPS4A dependency. We describe a compendium of synthetic lethal vulnerabilities and nominate VPS4A and VPS4B as high-priority therapeutic targets for cancers with 18q or 16q loss.


Subject(s)
ATPases Associated with Diverse Cellular Activities/metabolism , Endosomal Sorting Complexes Required for Transport/metabolism , Neoplasms/genetics , Vacuolar Proton-Translocating ATPases/metabolism , Cell Line, Tumor , Humans
14.
J Neurosci ; 28(52): 14074-86, 2008 Dec 24.
Article in English | MEDLINE | ID: mdl-19109490

ABSTRACT

Understanding the biology of adult neural stem cells has important implications for nervous system development and may contribute to our understanding of neurodegenerative disorders and their treatment. We have characterized the process of olfactory neurogenesis in adult mice lacking inhibitor of DNA binding 2(-/-) (Id2(-/-)). We found a diminished olfactory bulb containing reduced numbers of granular and periglomerular neurons with a distinct paucity of dopaminergic periglomerular neurons. While no deficiency of the stem cell compartment was detectable, migrating neuroblasts in Id2(-/-) mutant mice prematurely undergo astroglial differentiation within a disorganized rostral migratory stream. Further, when evaluated in vitro loss of Id2 results in decreased proliferation of neural progenitors and decreased expression of the Hes1 and Ascl1 (Mash1) transcription factors, known mediators of neuronal differentiation. These data support a novel role for sustained Id2 expression in migrating neural progenitors mediating olfactory dopaminergic neuronal differentiation in adult animals.


Subject(s)
Dopamine/metabolism , Inhibitor of Differentiation Protein 2/physiology , Neurogenesis/genetics , Neurons/physiology , Olfactory Bulb/cytology , Adult Stem Cells/physiology , Animals , Animals, Newborn , Astrocytes/physiology , Basic Helix-Loop-Helix Transcription Factors/genetics , Basic Helix-Loop-Helix Transcription Factors/metabolism , Bromodeoxyuridine/metabolism , Cell Count/methods , Cell Differentiation/genetics , Cells, Cultured , Discrimination, Psychological/physiology , Gene Expression Regulation, Developmental/genetics , Glial Fibrillary Acidic Protein/metabolism , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Inhibitor of Differentiation Protein 2/deficiency , Mice , Mice, Knockout , Neurogenesis/physiology , Olfactory Bulb/growth & development , Olfactory Pathways/cytology , Olfactory Pathways/physiology , Smell/genetics , Statistics, Nonparametric , Transcription Factor HES-1 , Tyrosine 3-Monooxygenase/metabolism
15.
Nat Commun ; 10(1): 3731, 2019 08 19.
Article in English | MEDLINE | ID: mdl-31427603

ABSTRACT

Pilocytic astrocytoma (PA), the most common childhood brain tumor, is a low-grade glioma with a single driver BRAF rearrangement. Here, we perform scRNAseq in six PAs using methods that enabled detection of the rearrangement. When compared to higher-grade gliomas, a strikingly higher proportion of the PA cancer cells exhibit a differentiated, astrocyte-like phenotype. A smaller proportion of cells exhibit a progenitor-like phenotype with evidence of proliferation. These express a mitogen-activated protein kinase (MAPK) programme that was absent from higher-grade gliomas. Immune cells, especially microglia, comprise 40% of all cells in the PAs and account for differences in bulk expression profiles between tumor locations and subtypes. These data indicate that MAPK signaling is restricted to relatively undifferentiated cancer cells in PA, with implications for investigational therapies directed at this pathway.


Subject(s)
Astrocytoma/genetics , Astrocytoma/pathology , Brain Neoplasms/pathology , Neural Stem Cells/cytology , Proto-Oncogene Proteins B-raf/genetics , Animals , Brain Neoplasms/genetics , Humans , MAP Kinase Signaling System/genetics , Mice , Microglia/pathology , Mitogen-Activated Protein Kinases/genetics , Mitogen-Activated Protein Kinases/metabolism , Oligodendroglia/cytology , Oncogene Proteins, Fusion/metabolism , Tumor Cells, Cultured
16.
Nat Commun ; 10(1): 2400, 2019 06 03.
Article in English | MEDLINE | ID: mdl-31160565

ABSTRACT

BET-bromodomain inhibition (BETi) has shown pre-clinical promise for MYC-amplified medulloblastoma. However, the mechanisms for its action, and ultimately for resistance, have not been fully defined. Here, using a combination of expression profiling, genome-scale CRISPR/Cas9-mediated loss of function and ORF/cDNA driven rescue screens, and cell-based models of spontaneous resistance, we identify bHLH/homeobox transcription factors and cell-cycle regulators as key genes mediating BETi's response and resistance. Cells that acquire drug tolerance exhibit a more neuronally differentiated cell-state and expression of lineage-specific bHLH/homeobox transcription factors. However, they do not terminally differentiate, maintain expression of CCND2, and continue to cycle through S-phase. Moreover, CDK4/CDK6 inhibition delays acquisition of resistance. Therefore, our data provide insights about the mechanisms underlying BETi effects and the appearance of resistance and support the therapeutic use of combined cell-cycle inhibitors with BETi in MYC-amplified medulloblastoma.


Subject(s)
Azepines/pharmacology , Cell Cycle/drug effects , Cerebellar Neoplasms/drug therapy , Medulloblastoma/drug therapy , Neurogenesis/drug effects , Proteins/antagonists & inhibitors , Triazoles/pharmacology , Animals , Basic Helix-Loop-Helix Transcription Factors/drug effects , Basic Helix-Loop-Helix Transcription Factors/metabolism , CRISPR-Cas Systems , Cell Cycle Proteins/drug effects , Cell Cycle Proteins/metabolism , Cell Line, Tumor , Cell Lineage , Cerebellar Neoplasms/genetics , Cyclin D2/drug effects , Cyclin D2/metabolism , Cyclin-Dependent Kinase 4/antagonists & inhibitors , Cyclin-Dependent Kinase 6/antagonists & inhibitors , Drug Resistance, Neoplasm , Gene Expression Profiling , Humans , Medulloblastoma/genetics , Mice , Neural Stem Cells/drug effects , Neural Stem Cells/metabolism , Proto-Oncogene Proteins c-myc/genetics , S Phase/drug effects
17.
Elife ; 62017 02 08.
Article in English | MEDLINE | ID: mdl-28177281

ABSTRACT

Genomic instability is a hallmark of human cancer, and results in widespread somatic copy number alterations. We used a genome-scale shRNA viability screen in human cancer cell lines to systematically identify genes that are essential in the context of particular copy-number alterations (copy-number associated gene dependencies). The most enriched class of copy-number associated gene dependencies was CYCLOPS (Copy-number alterations Yielding Cancer Liabilities Owing to Partial losS) genes, and spliceosome components were the most prevalent. One of these, the pre-mRNA splicing factor SF3B1, is also frequently mutated in cancer. We validated SF3B1 as a CYCLOPS gene and found that human cancer cells harboring partial SF3B1 copy-loss lack a reservoir of SF3b complex that protects cells with normal SF3B1 copy number from cell death upon partial SF3B1 suppression. These data provide a catalog of copy-number associated gene dependencies and identify partial copy-loss of wild-type SF3B1 as a novel, non-driver cancer gene dependency.


Subject(s)
Gene Dosage , Neoplasms/genetics , Neoplasms/pathology , Phosphoproteins/genetics , RNA Splicing Factors/genetics , Cell Line, Tumor , Humans
18.
Cancer Cell ; 29(6): 778-780, 2016 06 13.
Article in English | MEDLINE | ID: mdl-27300432

ABSTRACT

An emerging body of data highlights trophic functions of neurotransmitters on proliferation and differentiation of normal neural progenitors. In this issue of Cancer Cell, Dolma et al. document pro-survival functions of a neurotransmitter receptor in glioma progenitor cells, with practical overtones for therapy.


Subject(s)
Glioma/therapy , Stem Cells , Brain Neoplasms , Cell Differentiation , Humans
20.
Nat Genet ; 48(3): 273-82, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26829751

ABSTRACT

Angiocentric gliomas are pediatric low-grade gliomas (PLGGs) without known recurrent genetic drivers. We performed genomic analysis of new and published data from 249 PLGGs, including 19 angiocentric gliomas. We identified MYB-QKI fusions as a specific and single candidate driver event in angiocentric gliomas. In vitro and in vivo functional studies show that MYB-QKI rearrangements promote tumorigenesis through three mechanisms: MYB activation by truncation, enhancer translocation driving aberrant MYB-QKI expression and hemizygous loss of the tumor suppressor QKI. To our knowledge, this represents the first example of a single driver rearrangement simultaneously transforming cells via three genetic and epigenetic mechanisms in a tumor.


Subject(s)
Glioma/genetics , Oncogene Proteins v-myb/genetics , Oncogene Proteins, Fusion/genetics , RNA-Binding Proteins/genetics , Carcinogenesis/genetics , Cell Line, Tumor , Child , Comparative Genomic Hybridization , Exome/genetics , Gene Expression Regulation, Neoplastic , Gene Rearrangement , Glioma/pathology , High-Throughput Nucleotide Sequencing , Humans , Mutation , Oncogene Proteins v-myb/biosynthesis , Oncogene Proteins, Fusion/biosynthesis , RNA-Binding Proteins/biosynthesis
SELECTION OF CITATIONS
SEARCH DETAIL