Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
1.
Mol Biol Evol ; 39(11)2022 11 03.
Article in English | MEDLINE | ID: mdl-36201625

ABSTRACT

Topologically associating domains (TADs) are thought to play an important role in preventing gene misexpression by spatially constraining enhancer-promoter contacts. The deleterious nature of gene misexpression implies that TADs should, therefore, be conserved among related species. Several early studies comparing chromosome conformation between species reported high levels of TAD conservation; however, more recent studies have questioned these results. Furthermore, recent work suggests that TAD reorganization is not associated with extensive changes in gene expression. Here, we investigate the evolutionary conservation of TADs among 11 species of Drosophila. We use Hi-C data to identify TADs in each species and employ a comparative phylogenetic approach to derive empirical estimates of the rate of TAD evolution. Surprisingly, we find that TADs evolve rapidly. However, we also find that the rate of evolution depends on the chromatin state of the TAD, with TADs enriched for developmentally regulated chromatin evolving significantly slower than TADs enriched for broadly expressed, active chromatin. We also find that, after controlling for differences in chromatin state, highly conserved TADs do not exhibit higher levels of gene expression constraint. These results suggest that, in general, most TADs evolve rapidly and their divergence is not associated with widespread changes in gene expression. However, higher levels of evolutionary conservation and gene expression constraints in TADs enriched for developmentally regulated chromatin suggest that these TAD subtypes may be more important for regulating gene expression, likely due to the larger number of long-distance enhancer-promoter contacts associated with developmental genes.


Subject(s)
Drosophila , Genome , Animals , Drosophila/genetics , Phylogeny , Chromatin/genetics , Evolution, Molecular
2.
Proc Natl Acad Sci U S A ; 115(25): E5746-E5755, 2018 06 19.
Article in English | MEDLINE | ID: mdl-29866822

ABSTRACT

Activating mutations in the KIT or PDGFRA receptor tyrosine kinases are hallmarks of gastrointestinal stromal tumor (GIST). The biological underpinnings of recurrence following resection or disease progression beyond kinase mutation are poorly understood. Utilizing chromatin immunoprecipitation with sequencing of tumor samples and cell lines, we describe the enhancer landscape of GIST, highlighting genes that reinforce and extend our understanding of these neoplasms. A group of core transcription factors can be distinguished from others unique to localized and metastatic disease. The transcription factor HAND1 emerges in metastatic disease, binds to established GIST-associated enhancers, and facilitates GIST cell proliferation and KIT gene expression. The pattern of transcription factor expression in primary tumors is predictive of metastasis-free survival in GIST patients. These results provide insight into the enhancer landscape and transcription factor network underlying GIST, and define a unique strategy for predicting clinical behavior of this disease.


Subject(s)
Gastrointestinal Stromal Tumors/genetics , Transcription Factors/genetics , Basic Helix-Loop-Helix Transcription Factors/genetics , Cell Line, Tumor , Cell Proliferation/genetics , Disease Progression , Disease-Free Survival , Gastrointestinal Stromal Tumors/pathology , HEK293 Cells , Humans , Mutation/genetics , Proto-Oncogene Proteins c-kit/genetics , Receptor Protein-Tyrosine Kinases/genetics , Receptor, Platelet-Derived Growth Factor alpha/genetics
3.
Nat Chem Biol ; 14(4): 405-412, 2018 04.
Article in English | MEDLINE | ID: mdl-29507391

ABSTRACT

The addressable pocket of a protein is often not functionally relevant in disease. This is true for the multidomain, bromodomain-containing transcriptional regulator TRIM24. TRIM24 has been posited as a dependency in numerous cancers, yet potent and selective ligands for the TRIM24 bromodomain do not exert effective anti-proliferative responses. We therefore repositioned these probes as targeting features for heterobifunctional protein degraders. Recruitment of the VHL E3 ubiquitin ligase by dTRIM24 elicits potent and selective degradation of TRIM24. Using dTRIM24 to probe TRIM24 function, we characterize the dynamic genome-wide consequences of TRIM24 loss on chromatin localization and gene control. Further, we identify TRIM24 as a novel dependency in acute leukemia. Pairwise study of TRIM24 degradation versus bromodomain inhibition reveals enhanced anti-proliferative response from degradation. We offer dTRIM24 as a chemical probe of an emerging cancer dependency, and establish a path forward for numerous selective yet ineffectual ligands for proteins of therapeutic interest.


Subject(s)
Carrier Proteins/chemistry , 3T3 Cells , Animals , Cell Line, Tumor , Cell Proliferation , Crystallography, X-Ray , Gene Expression Regulation, Neoplastic , HEK293 Cells , Humans , Leukemia, Myeloid, Acute/metabolism , Ligands , MCF-7 Cells , Mice , Mutagenesis , Nuclear Proteins/chemistry , Proteasome Endopeptidase Complex/chemistry , Protein Binding , Protein Domains , RNA, Small Interfering/metabolism , Transcription Factors/chemistry
4.
Nat Chem Biol ; 14(5): 431-441, 2018 05.
Article in English | MEDLINE | ID: mdl-29581585

ABSTRACT

Dissection of complex biological systems requires target-specific control of the function or abundance of proteins. Genetic perturbations are limited by off-target effects, multicomponent complexity, and irreversibility. Most limiting is the requisite delay between modulation to experimental measurement. To enable the immediate and selective control of single protein abundance, we created a chemical biology system that leverages the potency of cell-permeable heterobifunctional degraders. The dTAG system pairs a novel degrader of FKBP12F36V with expression of FKBP12F36V in-frame with a protein of interest. By transgene expression or CRISPR-mediated locus-specific knock-in, we exemplify a generalizable strategy to study the immediate consequence of protein loss. Using dTAG, we observe an unexpected superior antiproliferative effect of pan-BET bromodomain degradation over selective BRD4 degradation, characterize immediate effects of KRASG12V loss on proteomic signaling, and demonstrate rapid degradation in vivo. This technology platform will confer kinetic resolution to biological investigation and provide target validation in the context of drug discovery.


Subject(s)
CRISPR-Cas Systems , Nuclear Proteins/chemistry , Proto-Oncogene Proteins p21(ras)/genetics , Tacrolimus Binding Protein 1A/chemistry , Transcription Factors/genetics , Alleles , Animals , Cell Cycle Proteins , Cell Proliferation , Cytoplasm/metabolism , Dimerization , Gene Knock-In Techniques , HEK293 Cells , Homeostasis , Humans , Ligands , Mice , Mutation , NIH 3T3 Cells , Nuclear Proteins/genetics , Protein Binding , Protein Domains , Proteolysis , Proteomics , Signal Transduction , Transgenes
5.
Curr Opin Genet Dev ; 82: 102092, 2023 10.
Article in English | MEDLINE | ID: mdl-37517354

ABSTRACT

Transposable elements (TEs) are ubiquitous among eukaryotic species. Their evolutionary persistence is likely due to a combination of tolerogenic, evasive/antagonistic, and cooperative interactions with their host genomes. Here, we focus on metazoan species and review recent advances related to the harmful effects of TE insertions, including how epigenetic effects and TE-derived RNAs can damage host cells. We discuss new findings related to host pathways that silence TEs, such as the piRNA pathway and the APOBEC3 and Kruppel-associated box zinc finger gene families. Finally, we summarize novel strategies used by TEs to evade host silencing, including the Y chromosome as a permissive niche for TE mobilization and TE counterdefense strategies to block host silencing factors.


Subject(s)
DNA Transposable Elements , Gene Silencing , Animals , DNA Transposable Elements/genetics , RNA, Small Interfering/genetics , Evolution, Molecular , Biological Evolution
6.
Nat Commun ; 12(1): 6854, 2021 11 25.
Article in English | MEDLINE | ID: mdl-34824217

ABSTRACT

Transposable elements (TEs) must replicate in germline cells to pass novel insertions to offspring. In Drosophila melanogaster ovaries, TEs can exploit specific developmental windows of opportunity to evade host silencing and increase their copy numbers. However, TE activity and host silencing in the distinct cell types of Drosophila testis are not well understood. Here, we reanalyze publicly available single-cell RNA-seq datasets to quantify TE expression in the distinct cell types of the Drosophila testis. We develop a method for identification of TE and host gene expression modules and find that a distinct population of early spermatocytes expresses a large number of TEs at much higher levels than other germline and somatic components of the testes. This burst of TE expression coincides with the activation of Y chromosome fertility factors and spermatocyte-specific transcriptional regulators, as well as downregulation of many components of the piRNA pathway. The TEs expressed by this cell population are specifically enriched on the Y chromosome and depleted on the X chromosome, relative to other active TEs. These data suggest that some TEs may achieve high insertional activity in males by exploiting a window of opportunity for mobilization created by the activation of spermatocyte-specific and Y chromosome-specific transcriptional programs.


Subject(s)
DNA Transposable Elements/genetics , Drosophila melanogaster/genetics , Spermatogenesis/genetics , Y Chromosome/genetics , Animals , Drosophila melanogaster/cytology , Evolution, Molecular , Gene Expression , Gene Regulatory Networks , Genes, Y-Linked/genetics , Male , Mutagenesis, Insertional , RNA, Small Interfering/genetics , Spermatocytes/metabolism , Testis/cytology , Testis/metabolism , Y Chromosome/metabolism
7.
Blood Cancer Discov ; 2(1): 92-109, 2021 01.
Article in English | MEDLINE | ID: mdl-33458694

ABSTRACT

Long-range oncogenic enhancers play an important role in cancer. Yet, whether similar regulation of tumor suppressor genes is relevant remains unclear. Loss of expression of PTEN is associated with the pathogenesis of various cancers, including T-cell leukemia (T-ALL). Here, we identify a highly conserved distal enhancer (PE) that interacts with the PTEN promoter in multiple hematopoietic populations, including T-cells, and acts as a hub of relevant transcription factors in T-ALL. Consistently, loss of PE leads to reduced PTEN levels in T-ALL cells. Moreover, PE-null mice show reduced Pten levels in thymocytes and accelerated development of NOTCH1-induced T-ALL. Furthermore, secondary loss of PE in established leukemias leads to accelerated progression and a gene expression signature driven by Pten loss. Finally, we uncovered recurrent deletions encompassing PE in T-ALL, which are associated with decreased PTEN levels. Altogether, our results identify PE as the first long-range tumor suppressor enhancer directly implicated in cancer.


Subject(s)
Enhancer Elements, Genetic , PTEN Phosphohydrolase , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma , Receptor, Notch1 , Animals , Cell Differentiation , Genes, Tumor Suppressor , Mice , PTEN Phosphohydrolase/genetics , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/genetics , Receptor, Notch1/genetics , Signal Transduction
8.
Cancer Res ; 79(5): 994-1009, 2019 03 01.
Article in English | MEDLINE | ID: mdl-30630822

ABSTRACT

Gastrointestinal stromal tumor (GIST) is a mesenchymal neoplasm characterized by activating mutations in the related receptor tyrosine kinases KIT and PDGFRA. GIST relies on expression of these unamplified receptor tyrosine kinase (RTK) genes through a large enhancer domain, resulting in high expression levels of the oncogene required for tumor growth. Although kinase inhibition is an effective therapy for many patients with GIST, disease progression from kinase-resistant mutations is common and no other effective classes of systemic therapy exist. In this study, we identify regulatory regions of the KIT enhancer essential for KIT gene expression and GIST cell viability. Given the dependence of GIST upon enhancer-driven expression of RTKs, we hypothesized that the enhancer domains could be therapeutically targeted by a BET bromodomain inhibitor (BBI). Treatment of GIST cells with BBIs led to cell-cycle arrest, apoptosis, and cell death, with unique sensitivity in GIST cells arising from attenuation of the KIT enhancer domain and reduced KIT gene expression. BBI treatment in KIT-dependent GIST cells produced genome-wide changes in the H3K27ac enhancer landscape and gene expression program, which was also seen with direct KIT inhibition using a tyrosine kinase inhibitor (TKI). Combination treatment with BBI and TKI led to superior cytotoxic effects in vitro and in vivo, with BBI preventing tumor growth in TKI-resistant xenografts. Resistance to select BBI in GIST was attributable to drug efflux pumps. These results define a therapeutic vulnerability and clinical strategy for targeting oncogenic kinase dependency in GIST. SIGNIFICANCE: Expression and activity of mutant KIT is essential for driving the majority of GIST neoplasms, which can be therapeutically targeted using BET bromodomain inhibitors.


Subject(s)
Gastrointestinal Neoplasms/drug therapy , Gastrointestinal Neoplasms/metabolism , Gastrointestinal Stromal Tumors/drug therapy , Gastrointestinal Stromal Tumors/metabolism , Proteins/antagonists & inhibitors , Proto-Oncogene Proteins c-kit/biosynthesis , Animals , Apoptosis/drug effects , Azepines/pharmacology , Cell Cycle Checkpoints/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Female , Gastrointestinal Neoplasms/genetics , Gastrointestinal Neoplasms/pathology , Gastrointestinal Stromal Tumors/genetics , Gastrointestinal Stromal Tumors/pathology , Gene Expression , HEK293 Cells , Humans , Imatinib Mesylate/pharmacology , Mice , Mice, Nude , Protein Domains , Protein Kinase Inhibitors/pharmacology , Proteins/metabolism , Proto-Oncogene Proteins c-kit/antagonists & inhibitors , Proto-Oncogene Proteins c-kit/genetics , Triazoles/pharmacology , Xenograft Model Antitumor Assays
9.
Nat Med ; 25(2): 292-300, 2019 02.
Article in English | MEDLINE | ID: mdl-30664779

ABSTRACT

Chordoma is a primary bone cancer with no approved therapy1. The identification of therapeutic targets in this disease has been challenging due to the infrequent occurrence of clinically actionable somatic mutations in chordoma tumors2,3. Here we describe the discovery of therapeutically targetable chordoma dependencies via genome-scale CRISPR-Cas9 screening and focused small-molecule sensitivity profiling. These systematic approaches reveal that the developmental transcription factor T (brachyury; TBXT) is the top selectively essential gene in chordoma, and that transcriptional cyclin-dependent kinase (CDK) inhibitors targeting CDK7/12/13 and CDK9 potently suppress chordoma cell proliferation. In other cancer types, transcriptional CDK inhibitors have been observed to downregulate highly expressed, enhancer-associated oncogenic transcription factors4,5. In chordoma, we find that T is associated with a 1.5-Mb region containing 'super-enhancers' and is the most highly expressed super-enhancer-associated transcription factor. Notably, transcriptional CDK inhibition leads to preferential and concentration-dependent downregulation of cellular brachyury protein levels in all models tested. In vivo, CDK7/12/13-inhibitor treatment substantially reduces tumor growth. Together, these data demonstrate small-molecule targeting of brachyury transcription factor addiction in chordoma, identify a mechanism of T gene regulation that underlies this therapeutic strategy, and provide a blueprint for applying systematic genetic and chemical screening approaches to discover vulnerabilities in genomically quiet cancers.


Subject(s)
Chordoma/metabolism , Fetal Proteins/metabolism , T-Box Domain Proteins/metabolism , Transcription Factors/metabolism , Cell Proliferation/drug effects , Chordoma/genetics , Chordoma/pathology , Cyclin-Dependent Kinases/metabolism , Down-Regulation/drug effects , Genes, Essential , Humans , Protein Kinase Inhibitors/pharmacology , Small Molecule Libraries/pharmacology
10.
Cancer Cell ; 36(4): 369-384.e13, 2019 10 14.
Article in English | MEDLINE | ID: mdl-31543463

ABSTRACT

Mitochondrial apoptosis can be effectively targeted in lymphoid malignancies with the FDA-approved B cell lymphoma 2 (BCL-2) inhibitor venetoclax, but resistance to this agent is emerging. We show that venetoclax resistance in chronic lymphocytic leukemia is associated with complex clonal shifts. To identify determinants of resistance, we conducted parallel genome-scale screens of the BCL-2-driven OCI-Ly1 lymphoma cell line after venetoclax exposure along with integrated expression profiling and functional characterization of drug-resistant and engineered cell lines. We identified regulators of lymphoid transcription and cellular energy metabolism as drivers of venetoclax resistance in addition to the known involvement by BCL-2 family members, which were confirmed in patient samples. Our data support the implementation of combinatorial therapy with metabolic modulators to address venetoclax resistance.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacology , Bridged Bicyclo Compounds, Heterocyclic/pharmacology , Leukemia, Lymphocytic, Chronic, B-Cell/drug therapy , Mitochondria/pathology , Proto-Oncogene Proteins c-bcl-2/antagonists & inhibitors , Sulfonamides/pharmacology , Adult , Aged , Aged, 80 and over , Animals , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Apoptosis/drug effects , Apoptosis/genetics , Bridged Bicyclo Compounds, Heterocyclic/therapeutic use , Cell Line, Tumor , Clonal Evolution/drug effects , Disease Progression , Drug Resistance, Neoplasm/drug effects , Drug Resistance, Neoplasm/genetics , Energy Metabolism/drug effects , Energy Metabolism/genetics , Female , Gene Expression Regulation, Neoplastic , Humans , Leukemia, Lymphocytic, Chronic, B-Cell/pathology , Male , Mice , Middle Aged , Mitochondria/drug effects , Myeloid Cell Leukemia Sequence 1 Protein/metabolism , Oxidative Phosphorylation/drug effects , Proto-Oncogene Proteins c-bcl-2/metabolism , Sulfonamides/therapeutic use , Treatment Outcome , Xenograft Model Antitumor Assays
11.
Cell Rep ; 25(13): 3693-3705.e6, 2018 12 26.
Article in English | MEDLINE | ID: mdl-30590042

ABSTRACT

The relationship between promoter proximal transcription factor-associated gene expression and super-enhancer-driven transcriptional programs are not well defined. However, their distinct genomic occupancy suggests a mechanism for specific and separable gene control. We explored the transcriptional and functional interrelationship between E2F transcription factors and BET transcriptional co-activators in multiple myeloma. We found that the transcription factor E2F1 and its heterodimerization partner DP1 represent a dependency in multiple myeloma cells. Global chromatin analysis reveals distinct regulatory axes for E2F and BETs, with E2F predominantly localized to active gene promoters of growth and/or proliferation genes and BETs disproportionately at enhancer-regulated tissue-specific genes. These two separate gene regulatory axes can be simultaneously targeted to impair the myeloma proliferative program, providing an important molecular mechanism for combination therapy. This study therefore suggests a sequestered cellular functional control that may be perturbed in cancer with potential for development of a promising therapeutic strategy.


Subject(s)
Enhancer Elements, Genetic , Gene Expression Regulation, Neoplastic , Multiple Myeloma/genetics , Promoter Regions, Genetic , Transcriptome/genetics , Animals , Azepines/pharmacology , Carcinogenesis/genetics , Carcinogenesis/pathology , Cell Line, Tumor , Cell Proliferation/genetics , E2F1 Transcription Factor/metabolism , Humans , Mice, SCID , Multiple Myeloma/pathology , Protein Binding , Protein Domains , Protein Multimerization , Transcription Factor DP1/metabolism , Triazoles/pharmacology
12.
Cancer Res ; 78(10): 2705-2720, 2018 05 15.
Article in English | MEDLINE | ID: mdl-29490948

ABSTRACT

To date, no consistent oncogenic driver mutations have been identified in most adult soft tissue sarcomas; these tumors are thus generally insensitive to existing targeted therapies. Here we investigated alternate mechanisms underlying sarcomagenesis to identify potential therapeutic interventions. Undifferentiated pleomorphic sarcoma (UPS) is an aggressive tumor frequently found in skeletal muscle where deregulation of the Hippo pathway and aberrant stabilization of its transcriptional effector yes-associated protein 1 (YAP1) increases proliferation and tumorigenesis. However, the downstream mechanisms driving this deregulation are incompletely understood. Using autochthonous mouse models and whole genome analyses, we found that YAP1 was constitutively active in some sarcomas due to epigenetic silencing of its inhibitor angiomotin (AMOT). Epigenetic modulators vorinostat and JQ1 restored AMOT expression and wild-type Hippo pathway signaling, which induced a muscle differentiation program and inhibited sarcomagenesis. YAP1 promoted sarcomagenesis by inhibiting expression of ubiquitin-specific peptidase 31 (USP31), a newly identified upstream negative regulator of NFκB signaling. Combined treatment with epigenetic modulators effectively restored USP31 expression, resulting in decreased NFκB activity. Our findings highlight a key underlying molecular mechanism in UPS and demonstrate the potential impact of an epigenetic approach to sarcoma treatment.Significance: A new link between Hippo pathway signaling, NFκB, and epigenetic reprogramming is highlighted and has the potential for therapeutic intervention in soft tissue sarcomas. Cancer Res; 78(10); 2705-20. ©2018 AACR.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Cell Transformation, Neoplastic/pathology , NF-kappa B/metabolism , Phosphoproteins/metabolism , Sarcoma/pathology , Soft Tissue Neoplasms/pathology , Ubiquitin-Specific Proteases/antagonists & inhibitors , Ubiquitin-Specific Proteases/biosynthesis , Angiomotins , Animals , Antineoplastic Agents/pharmacology , Azepines/pharmacology , Cell Cycle Proteins , Cell Line, Tumor , Cell Transformation, Neoplastic/genetics , Gene Expression Regulation, Neoplastic , HEK293 Cells , Hippo Signaling Pathway , Humans , Intercellular Signaling Peptides and Proteins/genetics , Mice , Mice, Transgenic , Microfilament Proteins/genetics , Muscle, Skeletal/pathology , Protein Serine-Threonine Kinases/metabolism , RNA Interference , RNA, Small Interfering/genetics , Sarcoma/genetics , Signal Transduction/genetics , Soft Tissue Neoplasms/genetics , Transcription Factors , Triazoles/pharmacology , Vorinostat/pharmacology , YAP-Signaling Proteins
13.
Nat Genet ; 50(4): 515-523, 2018 04.
Article in English | MEDLINE | ID: mdl-29379199

ABSTRACT

Amplification of the locus encoding the oncogenic transcription factor MYCN is a defining feature of high-risk neuroblastoma. Here we present the first dynamic chromatin and transcriptional landscape of MYCN perturbation in neuroblastoma. At oncogenic levels, MYCN associates with E-box binding motifs in an affinity-dependent manner, binding to strong canonical E-boxes at promoters and invading abundant weaker non-canonical E-boxes clustered at enhancers. Loss of MYCN leads to a global reduction in transcription, which is most pronounced at MYCN target genes with the greatest enhancer occupancy. These highly occupied MYCN target genes show tissue-specific expression and are linked to poor patient survival. The activity of genes with MYCN-occupied enhancers is dependent on the tissue-specific transcription factor TWIST1, which co-occupies enhancers with MYCN and is required for MYCN-dependent proliferation. These data implicate tissue-specific enhancers in defining often highly tumor-specific 'MYC target gene signatures' and identify disruption of the MYCN enhancer regulatory axis as a promising therapeutic strategy in neuroblastoma.


Subject(s)
N-Myc Proto-Oncogene Protein/genetics , Neuroblastoma/genetics , Binding Sites/genetics , Cell Line, Tumor , Chromatin/genetics , Chromatin/metabolism , Enhancer Elements, Genetic , Gene Amplification , Genes, myc , Humans , Kinetics , N-Myc Proto-Oncogene Protein/metabolism , Neuroblastoma/metabolism , Nuclear Proteins/metabolism , Oncogenes , Promoter Regions, Genetic , Twist-Related Protein 1/metabolism
14.
Cancer Discov ; 6(9): 1006-21, 2016 09.
Article in English | MEDLINE | ID: mdl-27312177

ABSTRACT

UNLABELLED: As a master regulator of chromatin function, the lysine methyltransferase EZH2 orchestrates transcriptional silencing of developmental gene networks. Overexpression of EZH2 is commonly observed in human epithelial cancers, such as non-small cell lung carcinoma (NSCLC), yet definitive demonstration of malignant transformation by deregulated EZH2 remains elusive. Here, we demonstrate the causal role of EZH2 overexpression in NSCLC with new genetically engineered mouse models of lung adenocarcinoma. Deregulated EZH2 silences normal developmental pathways, leading to epigenetic transformation independent of canonical growth factor pathway activation. As such, tumors feature a transcriptional program distinct from KRAS- and EGFR-mutant mouse lung cancers, but shared with human lung adenocarcinomas exhibiting high EZH2 expression. To target EZH2-dependent cancers, we developed a potent open-source EZH2 inhibitor, JQEZ5, that promoted the regression of EZH2-driven tumors in vivo, confirming oncogenic addiction to EZH2 in established tumors and providing the rationale for epigenetic therapy in a subset of lung cancer. SIGNIFICANCE: EZH2 overexpression induces murine lung cancers that are similar to human NSCLC with high EZH2 expression and low levels of phosphorylated AKT and ERK, implicating biomarkers for EZH2 inhibitor sensitivity. Our EZH2 inhibitor, JQEZ5, promotes regression of these tumors, revealing a potential role for anti-EZH2 therapy in lung cancer. Cancer Discov; 6(9); 1006-21. ©2016 AACR.See related commentary by Frankel et al., p. 949This article is highlighted in the In This Issue feature, p. 932.


Subject(s)
Enhancer of Zeste Homolog 2 Protein/genetics , Gene Expression Regulation, Neoplastic , Lung Neoplasms/genetics , Animals , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Cell Line, Tumor , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/metabolism , Chromatin/genetics , Chromatin/metabolism , Disease Models, Animal , Drug Design , Enhancer Elements, Genetic , Enhancer of Zeste Homolog 2 Protein/antagonists & inhibitors , Enhancer of Zeste Homolog 2 Protein/metabolism , Gene Expression , Gene Expression Profiling , Gene Expression Regulation, Neoplastic/drug effects , Humans , Lung Neoplasms/diagnosis , Lung Neoplasms/drug therapy , Lung Neoplasms/metabolism , Magnetic Resonance Imaging , Mice , Models, Molecular , Molecular Conformation , Molecular Targeted Therapy , Promoter Regions, Genetic , Structure-Activity Relationship , Xenograft Model Antitumor Assays
SELECTION OF CITATIONS
SEARCH DETAIL