Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
1.
Mol Cell ; 79(1): 167-179.e11, 2020 07 02.
Article in English | MEDLINE | ID: mdl-32497496

ABSTRACT

The identification of microRNA (miRNA) targets by Ago2 crosslinking-immunoprecipitation (CLIP) methods has provided major insights into the biology of this important class of non-coding RNAs. However, these methods are technically challenging and not easily applicable to an in vivo setting. To overcome these limitations and facilitate the investigation of miRNA functions in vivo, we have developed a method based on a genetically engineered mouse harboring a conditional Halo-Ago2 allele expressed from the endogenous Ago2 locus. By using a resin conjugated to the HaloTag ligand, Ago2-miRNA-mRNA complexes can be purified from cells and tissues expressing the endogenous Halo-Ago2 allele. We demonstrate the reproducibility and sensitivity of this method in mouse embryonic stem cells, developing embryos, adult tissues, and autochthonous mouse models of human brain and lung cancers. This method and the datasets we have generated will facilitate the characterization of miRNA-mRNA networks in vivo under physiological and pathological conditions.


Subject(s)
Argonaute Proteins/physiology , Embryonic Stem Cells/metabolism , Glioma/metabolism , MicroRNAs/metabolism , RNA, Messenger/metabolism , Recombinant Fusion Proteins/metabolism , Animals , Embryonic Stem Cells/cytology , Female , Gene Expression Regulation , Glioma/genetics , Glioma/pathology , High-Throughput Nucleotide Sequencing , Hydrolases/genetics , Mice , Mice, Knockout , MicroRNAs/genetics , Protein Binding , RNA, Messenger/genetics , Recombinant Fusion Proteins/genetics
2.
Nature ; 516(7531): 423-7, 2014 Dec 18.
Article in English | MEDLINE | ID: mdl-25337876

ABSTRACT

Chromosomal rearrangements have a central role in the pathogenesis of human cancers and often result in the expression of therapeutically actionable gene fusions. A recently discovered example is a fusion between the genes echinoderm microtubule-associated protein like 4 (EML4) and anaplastic lymphoma kinase (ALK), generated by an inversion on the short arm of chromosome 2: inv(2)(p21p23). The EML4-ALK oncogene is detected in a subset of human non-small cell lung cancers (NSCLC) and is clinically relevant because it confers sensitivity to ALK inhibitors. Despite their importance, modelling such genetic events in mice has proven challenging and requires complex manipulation of the germ line. Here we describe an efficient method to induce specific chromosomal rearrangements in vivo using viral-mediated delivery of the CRISPR/Cas9 system to somatic cells of adult animals. We apply it to generate a mouse model of Eml4-Alk-driven lung cancer. The resulting tumours invariably harbour the Eml4-Alk inversion, express the Eml4-Alk fusion gene, display histopathological and molecular features typical of ALK(+) human NSCLCs, and respond to treatment with ALK inhibitors. The general strategy described here substantially expands our ability to model human cancers in mice and potentially in other organisms.


Subject(s)
CRISPR-Associated Proteins/genetics , CRISPR-Cas Systems , Genetic Engineering/methods , Translocation, Genetic/genetics , Anaplastic Lymphoma Kinase , Animals , Antineoplastic Agents/therapeutic use , Cells, Cultured , Chromosome Inversion/genetics , Clustered Regularly Interspaced Short Palindromic Repeats , Crizotinib , Disease Models, Animal , Lung Neoplasms/drug therapy , Lung Neoplasms/enzymology , Lung Neoplasms/pathology , Mice , NIH 3T3 Cells , Protein Kinase Inhibitors/therapeutic use , Pyrazoles/therapeutic use , Pyridines/therapeutic use , Receptor Protein-Tyrosine Kinases/metabolism
4.
PLoS Genet ; 8(7): e1002797, 2012.
Article in English | MEDLINE | ID: mdl-22844244

ABSTRACT

MicroRNAs belonging to the miR-34 family have been proposed as critical modulators of the p53 pathway and potential tumor suppressors in human cancers. To formally test these hypotheses, we have generated mice carrying targeted deletion of all three members of this microRNA family. We show that complete inactivation of miR-34 function is compatible with normal development in mice. Surprisingly, p53 function appears to be intact in miR-34-deficient cells and tissues. Although loss of miR-34 expression leads to a slight increase in cellular proliferation in vitro, it does not impair p53-induced cell cycle arrest or apoptosis. Furthermore, in contrast to p53-deficient mice, miR-34-deficient animals do not display increased susceptibility to spontaneous, irradiation-induced, or c-Myc-initiated tumorigenesis. We also show that expression of members of the miR-34 family is particularly high in the testes, lungs, and brains of mice and that it is largely p53-independent in these tissues. These findings indicate that miR-34 plays a redundant function in the p53 pathway and suggest additional p53-independent functions for this family of miRNAs.


Subject(s)
Cell Transformation, Neoplastic/genetics , Gene Expression Regulation, Neoplastic , MicroRNAs/genetics , Tumor Suppressor Protein p53 , Animals , Apoptosis/genetics , Cell Cycle Checkpoints/genetics , Cell Proliferation , Cells, Cultured , Fibroblasts/cytology , Fibroblasts/metabolism , Gene Expression Regulation, Neoplastic/genetics , Humans , Mice , Mice, Knockout , MicroRNAs/metabolism , Tumor Suppressor Protein p53/deficiency , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism
5.
Proc Natl Acad Sci U S A ; 108(52): 21099-104, 2011 Dec 27.
Article in English | MEDLINE | ID: mdl-22160706

ABSTRACT

The p53 family member TAp73 is a transcription factor that plays a key role in many biological processes, including neuronal development. In particular, we have shown that p73 drives the expression of miR-34a, but not miR-34b and c, in mouse cortical neurons. miR-34a in turn modulates the expression of synaptic targets including synaptotagmin-1 and syntaxin-1A. Here we show that this axis is retained in mouse ES cells committed to differentiate toward a neurological phenotype. Moreover, overexpression of miR-34a alters hippocampal spinal morphology, and results in electrophysiological changes consistent with a reduction in spinal function. Therefore, the TAp73/miR-34a axis has functional relevance in primary neurons. These data reinforce a role for miR-34a in neuronal development.


Subject(s)
Cell Differentiation/physiology , Embryonic Stem Cells/cytology , MicroRNAs/metabolism , Neurites/physiology , Nuclear Proteins/metabolism , Spine/cytology , Animals , Blotting, Western , Cell Differentiation/genetics , Electrophysiology , Embryonic Stem Cells/metabolism , Mice , Mice, Knockout , Nuclear Proteins/genetics , Real-Time Polymerase Chain Reaction , Spine/physiology , Synaptotagmin I/metabolism , Syntaxin 1/metabolism
6.
Cancer Discov ; 12(2): 562-585, 2022 02.
Article in English | MEDLINE | ID: mdl-34561242

ABSTRACT

SMARCA4/BRG1 encodes for one of two mutually exclusive ATPases present in mammalian SWI/SNF chromatin remodeling complexes and is frequently mutated in human lung adenocarcinoma. However, the functional consequences of SMARCA4 mutation on tumor initiation, progression, and chromatin regulation in lung cancer remain poorly understood. Here, we demonstrate that loss of Smarca4 sensitizes club cell secretory protein-positive cells within the lung in a cell type-dependent fashion to malignant transformation and tumor progression, resulting in highly advanced dedifferentiated tumors and increased metastatic incidence. Consistent with these phenotypes, Smarca4-deficient primary tumors lack lung lineage transcription factor activities and resemble a metastatic cell state. Mechanistically, we show that Smarca4 loss impairs the function of all three classes of SWI/SNF complexes, resulting in decreased chromatin accessibility at lung lineage motifs and ultimately accelerating tumor progression. Thus, we propose that the SWI/SNF complex via Smarca4 acts as a gatekeeper for lineage-specific cellular transformation and metastasis during lung cancer evolution. SIGNIFICANCE: We demonstrate cell-type specificity in the tumor-suppressive functions of SMARCA4 in the lung, pointing toward a critical role of the cell-of-origin in driving SWI/SNF-mutant lung adenocarcinoma. We further show the direct effects of SMARCA4 loss on SWI/SNF function and chromatin regulation that cause aggressive malignancy during lung cancer evolution.This article is highlighted in the In This Issue feature, p. 275.


Subject(s)
Adenocarcinoma of Lung/genetics , Cell Transformation, Neoplastic , DNA Helicases/genetics , Lung Neoplasms/genetics , Neoplasm Metastasis , Nuclear Proteins/genetics , Transcription Factors/genetics , Adenocarcinoma of Lung/secondary , Animals , Disease Models, Animal , Humans , Lung Neoplasms/pathology , Mice
7.
Clin Cancer Res ; 26(21): 5701-5708, 2020 11 01.
Article in English | MEDLINE | ID: mdl-32709715

ABSTRACT

PURPOSE: SMARCA4 mutations are among the most common recurrent alterations in non-small cell lung cancer (NSCLC), but the relationship to other genomic abnormalities and clinical impact has not been established. EXPERIMENTAL DESIGN: To characterize SMARCA4 alterations in NSCLC, we analyzed the genomic, protein expression, and clinical outcome data of patients with SMARCA4 alterations treated at Memorial Sloan Kettering. RESULTS: In 4,813 tumors from patients with NSCLC, we identified 8% (n = 407) of patients with SMARCA4-mutant lung cancer. We describe two categories of SMARCA4 mutations: class 1 mutations (truncating mutations, fusions, and homozygous deletion) and class 2 mutations (missense mutations). Protein expression loss was associated with class 1 mutation (81% vs. 0%, P < 0.001). Both classes of mutation co-occurred more frequently with KRAS, STK11, and KEAP1 mutations compared with SMARCA4 wild-type tumors (P < 0.001). In patients with metastatic NSCLC, SMARCA4 alterations were associated with shorter overall survival, with class 1 alterations associated with shortest survival times (P < 0.001). Conversely, we found that treatment with immune checkpoint inhibitors (ICI) was associated with improved outcomes in patients with SMARCA4-mutant tumors (P = 0.01), with class 1 mutations having the best response to ICIs (P = 0.027). CONCLUSIONS: SMARCA4 alterations can be divided into two clinically relevant genomic classes associated with differential protein expression as well as distinct prognostic and treatment implications. Both classes co-occur with KEAP1, STK11, and KRAS mutations, but individually represent independent predictors of poor prognosis. Despite association with poor outcomes, SMARCA4-mutant lung cancers may be more sensitive to immunotherapy.


Subject(s)
Carcinoma, Non-Small-Cell Lung/genetics , DNA Helicases/genetics , Kelch-Like ECH-Associated Protein 1/genetics , Nuclear Proteins/genetics , Protein Serine-Threonine Kinases/genetics , Proto-Oncogene Proteins p21(ras)/genetics , Transcription Factors/genetics , AMP-Activated Protein Kinase Kinases , Aged , Carcinoma, Non-Small-Cell Lung/classification , Carcinoma, Non-Small-Cell Lung/epidemiology , Carcinoma, Non-Small-Cell Lung/pathology , Disease-Free Survival , Female , Gene Expression Regulation, Neoplastic/genetics , Genome, Human/genetics , Genomics , Humans , Immunotherapy , Male , Middle Aged , Mutation/genetics , Prognosis , Treatment Outcome
8.
Cancer Res ; 80(18): 3841-3854, 2020 09 15.
Article in English | MEDLINE | ID: mdl-32690724

ABSTRACT

Inactivation of SMARCA4/BRG1, the core ATPase subunit of mammalian SWI/SNF complexes, occurs at very high frequencies in non-small cell lung cancers (NSCLC). There are no targeted therapies for this subset of lung cancers, nor is it known how mutations in BRG1 contribute to lung cancer progression. Using a combination of gain- and loss-of-function approaches, we demonstrate that deletion of BRG1 in lung cancer leads to activation of replication stress responses. Single-molecule assessment of replication fork dynamics in BRG1-deficient cells revealed increased origin firing mediated by the prelicensing protein, CDC6. Quantitative mass spectrometry and coimmunoprecipitation assays showed that BRG1-containing SWI/SNF complexes interact with RPA complexes. Finally, BRG1-deficient lung cancers were sensitive to pharmacologic inhibition of ATR. These findings provide novel mechanistic insight into BRG1-mutant lung cancers and suggest that their dependency on ATR can be leveraged therapeutically and potentially expanded to BRG1-mutant cancers in other tissues. SIGNIFICANCE: These findings indicate that inhibition of ATR is a promising therapy for the 10% of non-small cell lung cancer patients harboring mutations in SMARCA4/BRG1. GRAPHICAL ABSTRACT: http://cancerres.aacrjournals.org/content/canres/80/18/3841/F1.large.jpg.


Subject(s)
Ataxia Telangiectasia Mutated Proteins/antagonists & inhibitors , Carcinoma, Non-Small-Cell Lung/genetics , DNA Helicases/genetics , Gene Deletion , Lung Neoplasms/genetics , Nuclear Proteins/genetics , Transcription Factors/genetics , Animals , Cell Cycle Proteins/metabolism , Chromosomal Proteins, Non-Histone , DNA Helicases/deficiency , Disease Progression , Female , Forkhead Transcription Factors , Gene Editing , Humans , Immunoprecipitation , Mass Spectrometry , Mice , Mice, Inbred C57BL , Mice, Nude , Nuclear Proteins/deficiency , Nuclear Proteins/metabolism , Sequence Analysis, RNA , Transcription Factors/deficiency
9.
Nat Genet ; 47(7): 766-75, 2015 Jul.
Article in English | MEDLINE | ID: mdl-26029871

ABSTRACT

Polycistronic microRNA (miRNA) clusters are a common feature of vertebrate genomes. The coordinated expression of miRNAs belonging to different seed families from a single transcriptional unit suggests functional cooperation, but this hypothesis has not been experimentally tested. Here we report the characterization of an allelic series of genetically engineered mice harboring selective targeted deletions of individual components of the miR-17 ∼ 92 cluster. Our results demonstrate the coexistence of functional cooperation and specialization among members of this cluster, identify a previously undescribed function for the miR-17 seed family in controlling axial patterning in vertebrates and show that loss of miR-19 selectively impairs Myc-driven tumorigenesis in two models of human cancer. By integrating phenotypic analysis and gene expression profiling, we provide a genome-wide view of how the components of a polycistronic miRNA cluster affect gene expression in vivo. The reagents and data sets reported here will accelerate exploration of the complex biological functions of this important miRNA cluster.


Subject(s)
MicroRNAs/genetics , Animals , Apoptosis , B-Lymphocytes/physiology , Carcinogenesis/genetics , Cells, Cultured , Eyelids/abnormalities , Gene Frequency , Genes, Lethal , Genome-Wide Association Study , Intellectual Disability/genetics , Limb Deformities, Congenital/genetics , Male , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Transgenic , Microcephaly/genetics , Multigene Family , Mutation , Tracheoesophageal Fistula/genetics
10.
Cancer J ; 18(3): 262-7, 2012.
Article in English | MEDLINE | ID: mdl-22647363

ABSTRACT

Overwhelming experimental evidence accumulated over the past decade indicates that microRNAs (miRNAs) are key regulators of gene expression in animals and plants and play important roles in development, homeostasis, and disease. The miR-17-92 family of miRNA clusters is composed of 3 related, highly conserved, polycistronic miRNA genes that collectively encode for a total of 15 miRNAs. We discuss recent studies demonstrating that these miRNAs are essential for vertebrate development and homeostasis. We also show how their mutation or deregulation contributes to the pathogenesis of a variety of human diseases, including cancer and congenital developmental defects. Finally, we discuss the current evidence suggesting how the different miRNAs encoded by these 3 clusters can functionally cooperate to fine-tune signaling and developmental pathways.


Subject(s)
MicroRNAs/genetics , Neoplasms/genetics , Vertebrates/embryology , Vertebrates/genetics , Animals , Cell Transformation, Neoplastic/genetics , Gene Expression Regulation , Humans , Multigene Family , Oncogenes , RNA Processing, Post-Transcriptional , RNA, Long Noncoding , Signal Transduction/genetics , Transcription, Genetic
SELECTION OF CITATIONS
SEARCH DETAIL