Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 35
Filter
1.
Elife ; 122023 06 01.
Article in English | MEDLINE | ID: mdl-37261974

ABSTRACT

Mutations in genes encoding components of chromatin modifying and remodeling complexes are among the most frequently observed somatic events in human cancers. For example, missense and nonsense mutations targeting the mixed lineage leukemia family member 3 (MLL3, encoded by KMT2C) histone methyltransferase occur in a range of solid tumors, and heterozygous deletions encompassing KMT2C occur in a subset of aggressive leukemias. Although MLL3 loss can promote tumorigenesis in mice, the molecular targets and biological processes by which MLL3 suppresses tumorigenesis remain poorly characterized. Here, we combined genetic, epigenomic, and animal modeling approaches to demonstrate that one of the mechanisms by which MLL3 links chromatin remodeling to tumor suppression is by co-activating the Cdkn2a tumor suppressor locus. Disruption of Kmt2c cooperates with Myc overexpression in the development of murine hepatocellular carcinoma (HCC), in which MLL3 binding to the Cdkn2a locus is blunted, resulting in reduced H3K4 methylation and low expression levels of the locus-encoded tumor suppressors p16/Ink4a and p19/Arf. Conversely, elevated KMT2C expression increases its binding to the CDKN2A locus and co-activates gene transcription. Endogenous Kmt2c restoration reverses these chromatin and transcriptional effects and triggers Ink4a/Arf-dependent apoptosis. Underscoring the human relevance of this epistasis, we found that genomic alterations in KMT2C and CDKN2A were associated with similar transcriptional profiles in human HCC samples. These results collectively point to a new mechanism for disrupting CDKN2A activity during cancer development and, in doing so, link MLL3 to an established tumor suppressor network.


Subject(s)
Carcinoma, Hepatocellular , Liver Neoplasms , Humans , Animals , Mice , Liver Neoplasms/genetics , Liver Neoplasms/pathology , Tumor Suppressor Protein p14ARF/genetics , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/pathology , Cyclin-Dependent Kinase Inhibitor p16/genetics , Cyclin-Dependent Kinase Inhibitor p16/metabolism , Chromatin , Carcinogenesis
2.
Proc Natl Acad Sci U S A ; 119(17): e2110557119, 2022 04 26.
Article in English | MEDLINE | ID: mdl-35442775

ABSTRACT

Anticancer drug development campaigns often fail due to an incomplete understanding of the therapeutic index differentiating the efficacy of the agent against the cancer and its on-target toxicities to the host. To address this issue, we established a versatile preclinical platform in which genetically defined cancers are produced using somatic tissue engineering in transgenic mice harboring a doxycycline-inducible short hairpin RNA against the target of interest. In this system, target inhibition is achieved by the addition of doxycycline, enabling simultaneous assessment of efficacy and toxicity in the same animal. As proof of concept, we focused on CDK9­a cancer target whose clinical development has been hampered by compounds with poorly understood target specificity and unacceptable toxicities. We systematically compared phenotypes produced by genetic Cdk9 inhibition to those achieved using a recently developed highly specific small molecule CDK9 inhibitor and found that both perturbations led to robust antitumor responses. Remarkably, nontoxic levels of CDK9 inhibition could achieve significant treatment efficacy, and dose-dependent toxicities produced by prolonged CDK9 suppression were largely reversible upon Cdk9 restoration or drug withdrawal. Overall, these results establish a versatile in vivo target validation platform that can be employed for rapid triaging of therapeutic targets and lend support to efforts aimed at advancing CDK9 inhibitors for cancer therapy.


Subject(s)
Antineoplastic Agents , Neoplasms , Animals , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Cell Line, Tumor , Cyclin-Dependent Kinase 9/metabolism , Mice , Neoplasms/drug therapy , Neoplasms/genetics , RNA Interference
3.
Gut ; 71(8): 1613-1628, 2022 08.
Article in English | MEDLINE | ID: mdl-34509979

ABSTRACT

OBJECTIVE: Large-scale genome sequencing efforts of human tumours identified epigenetic modifiers as one of the most frequently mutated gene class in human cancer. However, how these mutations drive tumour development and tumour progression are largely unknown. Here, we investigated the function of the histone demethylase KDM6A in gastrointestinal cancers, such as liver cancer and pancreatic cancer. DESIGN: Genetic alterations as well as expression analyses of KDM6A were performed in patients with liver cancer. Genetic mouse models of liver and pancreatic cancer coupled with Kdm6a-deficiency were investigated, transcriptomic and epigenetic profiling was performed, and in vivo and in vitro drug treatments were conducted. RESULTS: KDM6A expression was lost in 30% of patients with liver cancer. Kdm6a deletion significantly accelerated tumour development in murine liver and pancreatic cancer models. Kdm6a-deficient tumours showed hyperactivation of mTORC1 signalling, whereas endogenous Kdm6a re-expression by inducible RNA-interference in established Kdm6a-deficient tumours diminished mTORC1 activity resulting in attenuated tumour progression. Genome-wide transcriptional and epigenetic profiling revealed direct binding of Kdm6a to crucial negative regulators of mTORC1, such as Deptor, and subsequent transcriptional activation by epigenetic remodelling. Moreover, in vitro and in vivo genetic epistasis experiments illustrated a crucial function of Deptor and mTORC1 in Kdm6a-dependent tumour suppression. Importantly, KDM6A expression in human tumours correlates with mTORC1 activity and KDM6A-deficient tumours exhibit increased sensitivity to mTORC1 inhibition. CONCLUSION: KDM6A is an important tumour suppressor in gastrointestinal cancers and acts as an epigenetic toggle for mTORC1 signalling. Patients with KDM6A-deficient tumours could benefit of targeted therapy focusing on mTORC1 inhibition.


Subject(s)
Histone Demethylases/metabolism , Liver Neoplasms , Pancreatic Neoplasms , Animals , Epigenesis, Genetic , Histone Demethylases/genetics , Histones/genetics , Liver Neoplasms/genetics , Mechanistic Target of Rapamycin Complex 1/metabolism , Mice , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms
4.
Cell Rep ; 30(10): 3280-3295.e6, 2020 03 10.
Article in English | MEDLINE | ID: mdl-32160537

ABSTRACT

Genome editing technologies have transformed our ability to engineer desired genomic changes within living systems. However, detecting precise genomic modifications often requires sophisticated, expensive, and time-consuming experimental approaches. Here, we describe DTECT (Dinucleotide signaTurE CapTure), a rapid and versatile detection method that relies on the capture of targeted dinucleotide signatures resulting from the digestion of genomic DNA amplicons by the type IIS restriction enzyme AcuI. DTECT enables the accurate quantification of marker-free precision genome editing events introduced by CRISPR-dependent homology-directed repair, base editing, or prime editing in various biological systems, such as mammalian cell lines, organoids, and tissues. Furthermore, DTECT allows the identification of oncogenic mutations in cancer mouse models, patient-derived xenografts, and human cancer patient samples. The ease, speed, and cost efficiency by which DTECT identifies genomic signatures should facilitate the generation of marker-free cellular and animal models of human disease and expedite the detection of human pathogenic variants.


Subject(s)
Gene Editing , Genetic Variation , Genomics , Animals , BRCA1 Protein/genetics , BRCA2 Protein/genetics , Base Sequence , DNA/genetics , Disease Models, Animal , Genetic Loci , Genetic Markers , Genotype , HEK293 Cells , Humans , Mice , Mutation/genetics , NIH 3T3 Cells , Neoplasms/genetics , Nucleotides/genetics , Oncogenes , Recombinational DNA Repair/genetics , Restriction Mapping
5.
Cell Stem Cell ; 25(2): 241-257.e8, 2019 08 01.
Article in English | MEDLINE | ID: mdl-31303549

ABSTRACT

Brain tumor stem cells (BTSCs) are a chemoresistant population that can drive tumor growth and relapse, but the lack of BTSC-specific markers prevents selective targeting that spares resident stem cells. Through a ribosome-profiling analysis of mouse neural stem cells (NSCs) and BTSCs, we find glycerol-3-phosphate dehydrogenase 1 (GPD1) expression specifically in BTSCs and not in NSCs. GPD1 expression is present in the dormant BTSC population, which is enriched at tumor borders and drives tumor relapse after chemotherapy. GPD1 inhibition prolongs survival in mouse models of glioblastoma in part through altering cellular metabolism and protein translation, compromising BTSC maintenance. Metabolomic and lipidomic analyses confirm that GPD1+ BTSCs have a profile distinct from that of NSCs, which is dependent on GPD1 expression. Similar GPD1 expression patterns and prognostic associations are observed in human gliomas. This study provides an attractive therapeutic target for treating brain tumors and new insights into mechanisms regulating BTSC dormancy.


Subject(s)
Brain Neoplasms/metabolism , Glioblastoma/metabolism , Glioma/metabolism , Glycerolphosphate Dehydrogenase/metabolism , Neoplastic Stem Cells/physiology , Neural Stem Cells/physiology , Neurons/physiology , Animals , Biomarkers, Tumor/metabolism , Brain/pathology , Brain Neoplasms/pathology , Disease Models, Animal , Drug Resistance, Neoplasm , Gene Expression Regulation, Neoplastic , Glioblastoma/pathology , Glioma/pathology , Glycerolphosphate Dehydrogenase/genetics , Humans , Metabolome , Mice , Recurrence , Tumor Cells, Cultured
6.
Nat Methods ; 16(1): 51-54, 2019 01.
Article in English | MEDLINE | ID: mdl-30559432

ABSTRACT

CRISPR-Cas9-based combinatorial perturbation approaches for orthogonal knockout and gene activation have been impeded by complex vector designs and co-delivery of multiple constructs. Here, we demonstrate that catalytically active CRISPR-Cas12a fused to a transcriptional-activator domain enables flexible switching between genome editing and transcriptional activation by altering guide length. By leveraging Cas12a-mediated CRISPR-RNA array processing, we illustrate that Cas12a-VPR enables simplified multiplexed knockout and transcriptional activation in vitro and in vivo.


Subject(s)
CRISPR-Cas Systems , Gene Editing , Transcriptional Activation , Animals , Cell Line, Tumor , HEK293 Cells , Humans , Mice
7.
Cell ; 176(3): 564-580.e19, 2019 01 24.
Article in English | MEDLINE | ID: mdl-30580964

ABSTRACT

There are still gaps in our understanding of the complex processes by which p53 suppresses tumorigenesis. Here we describe a novel role for p53 in suppressing the mevalonate pathway, which is responsible for biosynthesis of cholesterol and nonsterol isoprenoids. p53 blocks activation of SREBP-2, the master transcriptional regulator of this pathway, by transcriptionally inducing the ABCA1 cholesterol transporter gene. A mouse model of liver cancer reveals that downregulation of mevalonate pathway gene expression by p53 occurs in premalignant hepatocytes, when p53 is needed to actively suppress tumorigenesis. Furthermore, pharmacological or RNAi inhibition of the mevalonate pathway restricts the development of murine hepatocellular carcinomas driven by p53 loss. Like p53 loss, ablation of ABCA1 promotes murine liver tumorigenesis and is associated with increased SREBP-2 maturation. Our findings demonstrate that repression of the mevalonate pathway is a crucial component of p53-mediated liver tumor suppression and outline the mechanism by which this occurs.


Subject(s)
Mevalonic Acid/metabolism , Tumor Suppressor Protein p53/metabolism , ATP Binding Cassette Transporter 1/metabolism , Animals , Cell Line , Cholesterol/metabolism , Female , Genes, Tumor Suppressor , HCT116 Cells , Hepatocytes/metabolism , Humans , Liver/metabolism , Male , Mice , Mice, Inbred C57BL , Neoplasms/genetics , Promoter Regions, Genetic , Sterol Regulatory Element Binding Protein 2/metabolism , Terpenes/metabolism
9.
Nat Biotechnol ; 36(9): 888-893, 2018 10.
Article in English | MEDLINE | ID: mdl-29969439

ABSTRACT

CRISPR base editing enables the creation of targeted single-base conversions without generating double-stranded breaks. However, the efficiency of current base editors is very low in many cell types. We reengineered the sequences of BE3, BE4Gam, and xBE3 by codon optimization and incorporation of additional nuclear-localization sequences. Our collection of optimized constitutive and inducible base-editing vector systems dramatically improves the efficiency by which single-nucleotide variants can be created. The reengineered base editors enable target modification in a wide range of mouse and human cell lines, and intestinal organoids. We also show that the optimized base editors mediate efficient in vivo somatic editing in the liver in adult mice.


Subject(s)
CRISPR-Cas Systems , Gene Editing , Animals , Cell Line , Genetic Variation , Humans , Mice
10.
Cancer Cell ; 33(3): 527-541.e8, 2018 03 12.
Article in English | MEDLINE | ID: mdl-29502955

ABSTRACT

Synovial sarcoma is an aggressive cancer invariably associated with a chromosomal translocation involving genes encoding the SWI-SNF complex component SS18 and an SSX (SSX1 or SSX2) transcriptional repressor. Using functional genomics, we identify KDM2B, a histone demethylase and component of a non-canonical polycomb repressive complex 1 (PRC1.1), as selectively required for sustaining synovial sarcoma cell transformation. SS18-SSX1 physically interacts with PRC1.1 and co-associates with SWI/SNF and KDM2B complexes on unmethylated CpG islands. Via KDM2B, SS18-SSX1 binds and aberrantly activates expression of developmentally regulated genes otherwise targets of polycomb-mediated repression, which is restored upon KDM2B depletion, leading to irreversible mesenchymal differentiation. Thus, SS18-SSX1 deregulates developmental programs to drive transformation by hijacking a transcriptional repressive complex to aberrantly activate gene expression.


Subject(s)
F-Box Proteins/metabolism , Jumonji Domain-Containing Histone Demethylases/metabolism , Neoplasm Proteins/metabolism , Polycomb Repressive Complex 1/metabolism , Proto-Oncogene Proteins/metabolism , Repressor Proteins/metabolism , Sarcoma, Synovial/metabolism , Cell Nucleus/metabolism , Cell Transformation, Neoplastic/genetics , Gene Expression/physiology , Humans , Neoplasm Proteins/genetics , Oncogene Proteins, Fusion/genetics , Repressor Proteins/genetics , Sarcoma, Synovial/genetics
11.
Nat Mater ; 17(4): 361-368, 2018 04.
Article in English | MEDLINE | ID: mdl-29403054

ABSTRACT

Development of targeted nanoparticle drug carriers often requires complex synthetic schemes involving both supramolecular self-assembly and chemical modification. These processes are generally difficult to predict, execute, and control. We describe herein a targeted drug delivery system that is accurately and quantitatively predicted to self-assemble into nanoparticles based on the molecular structures of precursor molecules, which are the drugs themselves. The drugs assemble with the aid of sulfated indocyanines into particles with ultrahigh drug loadings of up to 90%. We devised quantitative structure-nanoparticle assembly prediction (QSNAP) models to identify and validate electrotopological molecular descriptors as highly predictive indicators of nano-assembly and nanoparticle size. The resulting nanoparticles selectively targeted kinase inhibitors to caveolin-1-expressing human colon cancer and autochthonous liver cancer models to yield striking therapeutic effects while avoiding pERK inhibition in healthy skin. This finding enables the computational design of nanomedicines based on quantitative models for drug payload selection.


Subject(s)
Drug Carriers/chemistry , Nanomedicine/methods , Animals , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Drug Carriers/metabolism , Drug Carriers/pharmacokinetics , Endocytosis , Indoles/chemistry , Mice , Nanoparticles/chemistry , Particle Size , Tissue Distribution
12.
Mol Cancer Res ; 16(3): 390-402, 2018 03.
Article in English | MEDLINE | ID: mdl-29330288

ABSTRACT

Most lung cancer deaths are related to metastases, which indicates the necessity of detecting and inhibiting tumor cell dissemination. Here, we aimed to identify miRNAs involved in metastasis of lung adenocarcinoma as prognostic biomarkers and therapeutic targets. To that end, lymph node metastasis-associated miRNAs were identified in The Cancer Genome Atlas lung adenocarcinoma patient cohort (sequencing data; n = 449) and subsequently validated by qRT-PCR in an independent clinical cohort (n = 108). Overexpression of miRNAs located on chromosome 14q32 was associated with metastasis in lung adenocarcinoma patients. Importantly, Kaplan-Meier analysis and log-rank test revealed that higher expression levels of individual 14q32 miRNAs (mir-539, mir-323b, and mir-487a) associated with worse disease-free survival of never-smoker patients. Epigenetic analysis including DNA methylation microarray data and bisulfite sequencing validation demonstrated that the induction of 14q32 cluster correlated with genomic hypomethylation of the 14q32 locus. CRISPR activation technology, applied for the first time to functionally study the increase of clustered miRNA levels in a coordinated manner, showed that simultaneous overexpression of 14q32 miRNAs promoted tumor cell migratory and invasive properties. Analysis of individual miRNAs by mimic transfection further illustrated that miR-323b-3p, miR-487a-3p, and miR-539-5p significantly contributed to the invasive phenotype through the indirect regulation of different target genes. In conclusion, overexpression of 14q32 miRNAs, associated with the respective genomic hypomethylation, promotes metastasis and correlates with poor patient prognosis in lung adenocarcinoma.Implications: This study points to chromosome 14q32 miRNAs as promising targets to inhibit tumor cell dissemination and to predict patient prognosis in lung adenocarcinoma. Mol Cancer Res; 16(3); 390-402. ©2018 AACR.


Subject(s)
Adenocarcinoma of Lung/genetics , Chromosomes, Human, Pair 14 , MicroRNAs/genetics , Adenocarcinoma of Lung/metabolism , Adenocarcinoma of Lung/pathology , Cohort Studies , Epigenesis, Genetic , Female , Humans , Male , Neoplasm Metastasis , Prognosis
13.
Proc Natl Acad Sci U S A ; 114(50): 13076-13084, 2017 12 12.
Article in English | MEDLINE | ID: mdl-29162699

ABSTRACT

A segmental deletion resulting in DNAJB1-PRKACA gene fusion is now recognized as the signature genetic event of fibrolamellar hepatocellular carcinoma (FL-HCC), a rare but lethal liver cancer that primarily affects adolescents and young adults. Here we implement CRISPR-Cas9 genome editing and transposon-mediated somatic gene transfer to demonstrate that expression of either the endogenous fusion protein or a chimeric cDNA leads to the formation of indolent liver tumors in mice that closely resemble human FL-HCC. Notably, overexpression of the wild-type PRKACA was unable to fully recapitulate the oncogenic activity of DNAJB1-PRKACA, implying that FL-HCC does not simply result from enhanced PRKACA expression. Tumorigenesis was significantly enhanced by genetic activation of ß-catenin, an observation supported by evidence of recurrent Wnt pathway mutations in human FL-HCC, as well as treatment with the hepatotoxin 3,5-diethoxycarbonyl-1,4-dihydrocollidine, which causes tissue injury, inflammation, and fibrosis. Our study validates the DNAJB1-PRKACA fusion kinase as an oncogenic driver and candidate drug target for FL-HCC, and establishes a practical model for preclinical studies to identify strategies to treat this disease.


Subject(s)
Carcinoma, Hepatocellular/genetics , Cyclic AMP-Dependent Protein Kinase Catalytic Subunits/genetics , HSP40 Heat-Shock Proteins/genetics , Liver Neoplasms, Experimental/genetics , Liver Neoplasms/genetics , Liver Regeneration/genetics , Liver/physiology , Oncogene Proteins, Fusion/genetics , beta Catenin/genetics , Adult , Animals , Base Sequence , Carcinogenesis/chemically induced , Carcinogenesis/genetics , Carcinoma, Hepatocellular/pathology , Chromosomes, Human, Pair 19/genetics , Clustered Regularly Interspaced Short Palindromic Repeats/genetics , Cohort Studies , Female , Gene Expression Regulation, Neoplastic , Humans , Liver/drug effects , Liver/pathology , Liver Neoplasms/pathology , Liver Neoplasms, Experimental/chemically induced , Mice , Mice, Inbred C57BL , Pyridines/toxicity , Sequence Deletion/genetics , Young Adult
14.
Cancer Cell ; 31(6): 771-789.e6, 2017 06 12.
Article in English | MEDLINE | ID: mdl-28609656

ABSTRACT

Intrahepatic cholangiocarcinoma (ICC) is a highly malignant, heterogeneous cancer with poor treatment options. We found that mitochondrial dysfunction and oxidative stress trigger a niche favoring cholangiocellular overgrowth and tumorigenesis. Liver damage, reactive oxygen species (ROS) and paracrine tumor necrosis factor (Tnf) from Kupffer cells caused JNK-mediated cholangiocellular proliferation and oncogenic transformation. Anti-oxidant treatment, Kupffer cell depletion, Tnfr1 deletion, or JNK inhibition reduced cholangiocellular pre-neoplastic lesions. Liver-specific JNK1/2 deletion led to tumor reduction and enhanced survival in Akt/Notch- or p53/Kras-induced ICC models. In human ICC, high Tnf expression near ICC lesions, cholangiocellular JNK-phosphorylation, and ROS accumulation in surrounding hepatocytes are present. Thus, Kupffer cell-derived Tnf favors cholangiocellular proliferation/differentiation and carcinogenesis. Targeting the ROS/Tnf/JNK axis may provide opportunities for ICC therapy.


Subject(s)
Bile Duct Neoplasms/metabolism , Cholangiocarcinoma/metabolism , Kupffer Cells/metabolism , MAP Kinase Signaling System , Tumor Necrosis Factor-alpha/metabolism , Animals , Bile Duct Neoplasms/pathology , Butylated Hydroxyanisole/therapeutic use , Carcinogenesis , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Transformation, Neoplastic/drug effects , Cholangiocarcinoma/pathology , Humans , Kupffer Cells/drug effects , Liver/drug effects , Liver/pathology , Mice , Mitochondria/drug effects , Mitochondria/metabolism , Mitochondria/physiology , Oxidative Stress/drug effects , Reactive Oxygen Species/metabolism , Receptors, Tumor Necrosis Factor/genetics , Receptors, Tumor Necrosis Factor/metabolism , Signal Transduction , Tumor Microenvironment
15.
Cancer Res ; 77(16): 4355-4364, 2017 08 15.
Article in English | MEDLINE | ID: mdl-28630053

ABSTRACT

Different pyruvate kinase isoforms are expressed in a tissue-specific manner, with pyruvate kinase M2 (PKM2) suggested to be the predominant isoform in proliferating cells and cancer cells. Because of differential regulation of enzymatic activities, PKM2, but not PKM1, has been thought to favor cell proliferation. However, the role of PKM2 in tumorigenesis has been recently challenged. Here we report that increased glucose catabolism through glycolysis and increased pyruvate kinase activity in c-MYC-driven liver tumors are associated with increased expression of both PKM1 and PKM2 isoforms and decreased expression of the liver-specific isoform of pyruvate kinase, PKL. Depletion of PKM2 at the time of c-MYC overexpression in murine livers did not affect c-MYC-induced tumorigenesis and resulted in liver tumor formation with decreased pyruvate kinase activity and decreased catabolism of glucose into alanine and the Krebs cycle. An increased PKM1/PKM2 ratio by ectopic PKM1 expression further decreased glucose flux into serine biosynthesis and increased flux into lactate and the Krebs cycle, resulting in reduced total levels of serine. However, these changes also did not affect c-MYC-induced liver tumor development. These results suggest that increased expression of PKM2 is not required to support c-MYC-induced tumorigenesis in the liver and that various PKM1/PKM2 ratios and pyruvate kinase activities can sustain glucose catabolism required for this process. Cancer Res; 77(16); 4355-64. ©2017 AACR.


Subject(s)
Glucose/metabolism , Liver Neoplasms, Experimental/metabolism , Proto-Oncogene Proteins c-myc/metabolism , Pyruvate Kinase/metabolism , Animals , Genes, myc , Isoenzymes , Liver Neoplasms, Experimental/genetics , Liver Neoplasms, Experimental/pathology , Mice , Phosphorylation , Proto-Oncogene Proteins c-myc/genetics , Pyruvate Kinase/genetics
16.
Nat Biotechnol ; 35(4): 350-353, 2017 04.
Article in English | MEDLINE | ID: mdl-28263295

ABSTRACT

We present SplashRNA, a sequential classifier to predict potent microRNA-based short hairpin RNAs (shRNAs). Trained on published and novel data sets, SplashRNA outperforms previous algorithms and reliably predicts the most efficient shRNAs for a given gene. Combined with an optimized miR-E backbone, >90% of high-scoring SplashRNA predictions trigger >85% protein knockdown when expressed from a single genomic integration. SplashRNA can significantly improve the accuracy of loss-of-function genetics studies and facilitates the generation of compact shRNA libraries.


Subject(s)
Algorithms , Clustered Regularly Interspaced Short Palindromic Repeats/genetics , Gene Silencing , Machine Learning , RNA, Small Interfering/genetics , Software , CRISPR-Cas Systems/genetics , Chromosome Mapping/methods , Sequence Analysis, RNA/methods
18.
Cancer Cell ; 29(5): 617-619, 2016 05 09.
Article in English | MEDLINE | ID: mdl-27165739

ABSTRACT

In this issue of Cancer Cell, Cai et al. use genome editing to study 8p deletions in a mammary epithelial cell model and show that 8p loss of heterozygosity (LOH) attenuates the action of several genes that collectively promote cell invasion and enhance cellular sensitivity to autophagy inhibitors.


Subject(s)
Chromosome Deletion , Loss of Heterozygosity , Chromosome Mapping , Genes, Tumor Suppressor , Humans , Neoplasms/genetics
19.
FEBS J ; 283(17): 3194-203, 2016 09.
Article in English | MEDLINE | ID: mdl-27149548

ABSTRACT

The recent discovery of the CRISPR/Cas system and repurposing of this technology to edit a variety of different genomes have revolutionized an array of scientific fields, from genetics and translational research, to agriculture and bioproduction. In particular, the prospect of rapid and precise genome editing in laboratory animals by CRISPR/Cas has generated an immense interest in the scientific community. Here we review current in vivo applications of CRISPR/Cas and how this technology can improve our knowledge of gene function and our understanding of biological processes in animal models.


Subject(s)
CRISPR-Cas Systems , Animals , Chromosome Aberrations , DNA End-Joining Repair , Disease Models, Animal , Embryonic Stem Cells/cytology , Embryonic Stem Cells/metabolism , Epigenetic Repression , Gene Editing/methods , Germ-Line Mutation , Humans , Mice , Models, Genetic , Transcriptional Activation
20.
Cancer Discov ; 6(6): 612-29, 2016 06.
Article in English | MEDLINE | ID: mdl-27099234

ABSTRACT

UNLABELLED: Oncogene-induced senescence is a potent barrier to tumorigenesis that limits cellular expansion following certain oncogenic events. Senescent cells display a repressive chromatin configuration thought to stably silence proliferation-promoting genes while simultaneously activating an unusual form of immune surveillance involving a secretory program referred to as the senescence-associated secretory phenotype (SASP). Here, we demonstrate that senescence also involves a global remodeling of the enhancer landscape with recruitment of the chromatin reader BRD4 to newly activated super-enhancers adjacent to key SASP genes. Transcriptional profiling and functional studies indicate that BRD4 is required for the SASP and downstream paracrine signaling. Consequently, BRD4 inhibition disrupts immune cell-mediated targeting and elimination of premalignant senescent cells in vitro and in vivo Our results identify a critical role for BRD4-bound super-enhancers in senescence immune surveillance and in the proper execution of a tumor-suppressive program. SIGNIFICANCE: This study reveals how cells undergoing oncogene-induced senescence acquire a distinctive enhancer landscape that includes formation of super-enhancers adjacent to immune-modulatory genes required for paracrine immune activation. This process links BRD4 and super-enhancers to a tumor-suppressive immune surveillance program that can be disrupted by small molecule inhibitors of the bromo and extra terminal domain family of proteins. Cancer Discov; 6(6); 612-29. ©2016 AACR.See related commentary by Vizioli and Adams, p. 576This article is highlighted in the In This Issue feature, p. 561.


Subject(s)
Cellular Senescence/genetics , Chromatin Assembly and Disassembly , Enhancer Elements, Genetic , Immunologic Surveillance/genetics , Nuclear Proteins/metabolism , Transcription Factors/metabolism , Animals , Binding Sites , Cell Cycle/genetics , Cell Cycle Proteins , Cell Line, Tumor , Cell Proliferation , Chromatin Immunoprecipitation , Cluster Analysis , Computational Biology/methods , Fibroblasts , GTP Phosphohydrolases/genetics , GTP Phosphohydrolases/metabolism , Gene Expression Profiling , Gene Expression Regulation , Hepatocytes/metabolism , High-Throughput Nucleotide Sequencing , Histones/metabolism , Humans , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mice , Nucleotide Motifs , Oncogenes , Paracrine Communication , Position-Specific Scoring Matrices , Protein Binding
SELECTION OF CITATIONS
SEARCH DETAIL
...