Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 23
Filter
1.
Nat Commun ; 14(1): 1364, 2023 03 13.
Article in English | MEDLINE | ID: mdl-36914634

ABSTRACT

Robust, generalizable approaches to identify compounds efficiently with undesirable mechanisms of action in complex cellular assays remain elusive. Such a process would be useful for hit triage during high-throughput screening and, ultimately, predictive toxicology during drug development. Here we generate cell painting and cellular health profiles for 218 prototypical cytotoxic and nuisance compounds in U-2 OS cells in a concentration-response format. A diversity of compounds that cause cellular damage produces bioactive cell painting morphologies, including cytoskeletal poisons, genotoxins, nonspecific electrophiles, and redox-active compounds. Further, we show that lower quality lysine acetyltransferase inhibitors and nonspecific electrophiles can be distinguished from more selective counterparts. We propose that the purposeful inclusion of cytotoxic and nuisance reference compounds such as those profiled in this resource will help with assay optimization and compound prioritization in complex cellular assays like cell painting.


Subject(s)
High-Throughput Screening Assays , Oxidation-Reduction
2.
J Mol Biol ; 433(23): 167294, 2021 11 19.
Article in English | MEDLINE | ID: mdl-34662547

ABSTRACT

Activating mutations in the epidermal growth factor receptor (EGFR) are common driver mutations in non-small cell lung cancer (NSCLC). First, second and third generation EGFR tyrosine kinase inhibitors (TKIs) are effective at inhibiting mutant EGFR NSCLC, however, acquired resistance is a major issue, leading to disease relapse. Here, we characterize a small molecule, EMI66, an analog of a small molecule which we previously identified to inhibit mutant EGFR signalling via a novel mechanism of action. We show that EMI66 attenuates receptor tyrosine kinase (RTK) expression and signalling and alters the electrophoretic mobility of Coatomer Protein Complex Beta 2 (COPB2) protein in mutant EGFR NSCLC cells. Moreover, we demonstrate that EMI66 can alter the subcellular localization of EGFR and COPB2 within the early secretory pathway. Furthermore, we find that COPB2 knockdown reduces the growth of mutant EGFR lung cancer cells, alters the post-translational processing of RTKs, and alters the endoplasmic reticulum (ER) stress response pathway. Lastly, we show that EMI66 treatment also alters the ER stress response pathway and inhibits the growth of mutant EGFR lung cancer cells and organoids. Our results demonstrate that targeting of COPB2 with EMI66 presents a viable approach to attenuate mutant EGFR signalling and growth in NSCLC.


Subject(s)
Coatomer Protein/genetics , Coatomer Protein/metabolism , Drug Discovery , Endoplasmic Reticulum Stress/drug effects , Endoplasmic Reticulum Stress/genetics , Gene Expression Regulation, Neoplastic/drug effects , Receptor Protein-Tyrosine Kinases/genetics , Drug Discovery/methods , Drug Screening Assays, Antitumor , ErbB Receptors/antagonists & inhibitors , ErbB Receptors/genetics , ErbB Receptors/metabolism , Humans , Mutation , Protein Kinase Inhibitors/pharmacology , Protein Processing, Post-Translational , Receptor Protein-Tyrosine Kinases/metabolism , Signal Transduction/drug effects
3.
Oncogene ; 40(12): 2243-2257, 2021 03.
Article in English | MEDLINE | ID: mdl-33649538

ABSTRACT

Mdm2 antagonizes the tumor suppressor p53. Targeting the Mdm2-p53 interaction represents an attractive approach for the treatment of cancers with functional p53. Investigating mechanisms underlying Mdm2-p53 regulation is therefore important. The scaffold protein ß-arrestin2 (ß-arr2) regulates tumor suppressor p53 by counteracting Mdm2. ß-arr2 nucleocytoplasmic shuttling displaces Mdm2 from the nucleus to the cytoplasm resulting in enhanced p53 signaling. ß-arr2 is constitutively exported from the nucleus, via a nuclear export signal, but mechanisms regulating its nuclear entry are not completely elucidated. ß-arr2 can be SUMOylated, but no information is available on how SUMO may regulate ß-arr2 nucleocytoplasmic shuttling. While we found ß-arr2 SUMOylation to be dispensable for nuclear import, we identified a non-covalent interaction between SUMO and ß-arr2, via a SUMO interaction motif (SIM), that is required for ß-arr2 cytonuclear trafficking. This SIM promotes association of ß-arr2 with the multimolecular RanBP2/RanGAP1-SUMO nucleocytoplasmic transport hub that resides on the cytoplasmic filaments of the nuclear pore complex. Depletion of RanBP2/RanGAP1-SUMO levels result in defective ß-arr2 nuclear entry. Mutation of the SIM inhibits ß-arr2 nuclear import, its ability to delocalize Mdm2 from the nucleus to the cytoplasm and enhanced p53 signaling in lung and breast tumor cell lines. Thus, a ß-arr2 SIM nuclear entry checkpoint, coupled with active ß-arr2 nuclear export, regulates its cytonuclear trafficking function to control the Mdm2-p53 signaling axis.


Subject(s)
GTPase-Activating Proteins/genetics , Neoplasms/genetics , Proto-Oncogene Proteins c-mdm2/genetics , SUMO-1 Protein/genetics , Tumor Suppressor Protein p53/genetics , beta-Arrestin 2/genetics , Cell Line, Tumor , Cell Nucleus/genetics , Cell Nucleus/metabolism , Cytoskeleton/genetics , Cytoskeleton/metabolism , Humans , Mutation/genetics , Neoplasms/drug therapy , Neoplasms/pathology , Nuclear Export Signals/genetics , Signal Transduction/genetics , Sumoylation/genetics
4.
Cell ; 184(1): 226-242.e21, 2021 01 07.
Article in English | MEDLINE | ID: mdl-33417860

ABSTRACT

Cancer cells enter a reversible drug-tolerant persister (DTP) state to evade death from chemotherapy and targeted agents. It is increasingly appreciated that DTPs are important drivers of therapy failure and tumor relapse. We combined cellular barcoding and mathematical modeling in patient-derived colorectal cancer models to identify and characterize DTPs in response to chemotherapy. Barcode analysis revealed no loss of clonal complexity of tumors that entered the DTP state and recurred following treatment cessation. Our data fit a mathematical model where all cancer cells, and not a small subpopulation, possess an equipotent capacity to become DTPs. Mechanistically, we determined that DTPs display remarkable transcriptional and functional similarities to diapause, a reversible state of suspended embryonic development triggered by unfavorable environmental conditions. Our study provides insight into how cancer cells use a developmentally conserved mechanism to drive the DTP state, pointing to novel therapeutic opportunities to target DTPs.


Subject(s)
Antineoplastic Agents/therapeutic use , Colorectal Neoplasms/drug therapy , Diapause , Drug Resistance, Neoplasm , Animals , Antineoplastic Agents/pharmacology , Autophagy/drug effects , Autophagy/genetics , Cell Line, Tumor , Clone Cells , Colorectal Neoplasms/genetics , Colorectal Neoplasms/pathology , Drug Resistance, Neoplasm/drug effects , Embryo, Mammalian/drug effects , Embryo, Mammalian/metabolism , Gene Expression Profiling , Gene Expression Regulation, Neoplastic/drug effects , Genetic Heterogeneity/drug effects , Humans , Irinotecan/pharmacology , Irinotecan/therapeutic use , Mice, Inbred NOD , Mice, SCID , Models, Biological , Signal Transduction/drug effects , Up-Regulation/drug effects , Up-Regulation/genetics , Xenograft Model Antitumor Assays
5.
Nat Commun ; 11(1): 4205, 2020 08 21.
Article in English | MEDLINE | ID: mdl-32826891

ABSTRACT

Triple negative breast cancer (TNBC) is a deadly form of breast cancer due to the development of resistance to chemotherapy affecting over 30% of patients. New therapeutics and companion biomarkers are urgently needed. Recognizing the elevated expression of glucose transporter 1 (GLUT1, encoded by SLC2A1) and associated metabolic dependencies in TNBC, we investigated the vulnerability of TNBC cell lines and patient-derived samples to GLUT1 inhibition. We report that genetic or pharmacological inhibition of GLUT1 with BAY-876 impairs the growth of a subset of TNBC cells displaying high glycolytic and lower oxidative phosphorylation (OXPHOS) rates. Pathway enrichment analysis of gene expression data suggests that the functionality of the E2F pathway may reflect to some extent OXPHOS activity. Furthermore, the protein levels of retinoblastoma tumor suppressor (RB1) strongly correlate with the degree of sensitivity to GLUT1 inhibition in TNBC, where RB1-negative cells are insensitive to GLUT1 inhibition. Collectively, our results highlight a strong and targetable RB1-GLUT1 metabolic axis in TNBC and warrant clinical evaluation of GLUT1 inhibition in TNBC patients stratified according to RB1 protein expression levels.


Subject(s)
Glucose Transporter Type 1/antagonists & inhibitors , Glucose Transporter Type 1/metabolism , Retinoblastoma Binding Proteins/metabolism , Triple Negative Breast Neoplasms/metabolism , Ubiquitin-Protein Ligases/metabolism , Animals , Apoptosis/drug effects , Biomarkers, Tumor , Breast Neoplasms/metabolism , Cell Cycle , Cell Line, Tumor , Cell Proliferation , Female , Gene Expression Regulation, Neoplastic/drug effects , Glucose Transporter Type 1/genetics , Humans , Mice , Oxidative Phosphorylation , Proteomics , Pyrazoles/pharmacology , Pyridines/pharmacology , Quinolines , RNA, Messenger/metabolism , Triple Negative Breast Neoplasms/genetics , Ubiquitin-Protein Ligases/genetics
6.
Cancer Discov ; 10(9): 1312-1329, 2020 09.
Article in English | MEDLINE | ID: mdl-32546577

ABSTRACT

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


Subject(s)
Antineoplastic Agents/pharmacology , Epigenesis, Genetic/immunology , Molecular Mimicry/immunology , Triple Negative Breast Neoplasms/immunology , Tumor Escape/genetics , Animals , Antineoplastic Agents/therapeutic use , Cell Line, Tumor , Chromatin Immunoprecipitation Sequencing , DNA Methylation/drug effects , DNA Methylation/immunology , DNA Transposable Elements/genetics , Disease Progression , Drug Resistance, Neoplasm/genetics , Drug Resistance, Neoplasm/immunology , Enhancer of Zeste Homolog 2 Protein/antagonists & inhibitors , Enhancer of Zeste Homolog 2 Protein/metabolism , Epigenesis, Genetic/drug effects , Female , Humans , Mice , Molecular Mimicry/genetics , Paclitaxel/pharmacology , Paclitaxel/therapeutic use , RNA, Double-Stranded/immunology , RNA, Double-Stranded/metabolism , Triple Negative Breast Neoplasms/drug therapy , Triple Negative Breast Neoplasms/genetics , Triple Negative Breast Neoplasms/pathology , Xenograft Model Antitumor Assays
7.
Nat Commun ; 10(1): 5759, 2019 12 17.
Article in English | MEDLINE | ID: mdl-31848333

ABSTRACT

PRDM9 is a PR domain containing protein which trimethylates histone 3 on lysine 4 and 36. Its normal expression is restricted to germ cells and attenuation of its activity results in altered meiotic gene transcription, impairment of double-stranded breaks and pairing between homologous chromosomes. There is growing evidence for a role of aberrant expression of PRDM9 in oncogenesis and genome instability. Here we report the discovery of MRK-740, a potent (IC50: 80 ± 16 nM), selective and cell-active PRDM9 inhibitor (Chemical Probe). MRK-740 binds in the substrate-binding pocket, with unusually extensive interactions with the cofactor S-adenosylmethionine (SAM), conferring SAM-dependent substrate-competitive inhibition. In cells, MRK-740 specifically and directly inhibits H3K4 methylation at endogenous PRDM9 target loci, whereas the closely related inactive control compound, MRK-740-NC, does not. The discovery of MRK-740 as a chemical probe for the PRDM subfamily of methyltransferases highlights the potential for exploiting SAM in targeting SAM-dependent methyltransferases.


Subject(s)
Drug Discovery/methods , Enzyme Inhibitors/pharmacology , Histone-Lysine N-Methyltransferase/antagonists & inhibitors , Molecular Probes/pharmacology , Crystallography, X-Ray , DNA Methylation/drug effects , Enzyme Inhibitors/chemistry , HEK293 Cells , Histone-Lysine N-Methyltransferase/metabolism , Histone-Lysine N-Methyltransferase/ultrastructure , Histones/metabolism , Humans , Inhibitory Concentration 50 , Molecular Dynamics Simulation , Molecular Probes/chemistry , Protein Domains , S-Adenosylmethionine/metabolism
8.
Nat Commun ; 10(1): 1915, 2019 04 23.
Article in English | MEDLINE | ID: mdl-31015424

ABSTRACT

Bromodomains (BRDs) are conserved protein interaction modules which recognize (read) acetyl-lysine modifications, however their role(s) in regulating cellular states and their potential as targets for the development of targeted treatment strategies is poorly understood. Here we present a set of 25 chemical probes, selective small molecule inhibitors, covering 29 human bromodomain targets. We comprehensively evaluate the selectivity of this probe-set using BROMOscan and demonstrate the utility of the set identifying roles of BRDs in cellular processes and potential translational applications. For instance, we discovered crosstalk between histone acetylation and the glycolytic pathway resulting in a vulnerability of breast cancer cell lines under conditions of glucose deprivation or GLUT1 inhibition to inhibition of BRPF2/3 BRDs. This chemical probe-set will serve as a resource for future applications in the discovery of new physiological roles of bromodomain proteins in normal and disease states, and as a toolset for bromodomain target validation.


Subject(s)
Antineoplastic Agents/pharmacology , Epithelial Cells/drug effects , Gene Expression Regulation, Neoplastic , Protein Processing, Post-Translational/drug effects , Small Molecule Libraries/pharmacology , Acetylation , Amino Acid Sequence , Antineoplastic Agents/chemistry , Cell Line, Tumor , Epigenesis, Genetic , Epithelial Cells/metabolism , Epithelial Cells/pathology , Glucose/deficiency , Glucose Transporter Type 1/antagonists & inhibitors , Glucose Transporter Type 1/genetics , Glucose Transporter Type 1/metabolism , Glycolysis/drug effects , Glycolysis/genetics , High-Throughput Screening Assays , Histone Acetyltransferases , Histone Chaperones , Histones/genetics , Histones/metabolism , Humans , Mammary Glands, Human/metabolism , Mammary Glands, Human/pathology , Nuclear Proteins/antagonists & inhibitors , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Signal Transduction , Small Molecule Libraries/chemistry , Structure-Activity Relationship
9.
Nat Commun ; 10(1): 1436, 2019 03 29.
Article in English | MEDLINE | ID: mdl-30926792

ABSTRACT

In embryonic stem cells, promoters of key lineage-specific differentiation genes are found in a bivalent state, having both activating H3K4me3 and repressive H3K27me3 histone marks, making them poised for transcription upon loss of H3K27me3. Whether cancer-initiating cells (C-ICs) have similar epigenetic mechanisms that prevent lineage commitment is unknown. Here we show that colorectal C-ICs (CC-ICs) are maintained in a stem-like state through a bivalent epigenetic mechanism. Disruption of the bivalent state through inhibition of the H3K27 methyltransferase EZH2, resulted in decreased self-renewal of patient-derived C-ICs. Epigenomic analyses revealed that the promoter of Indian Hedgehog (IHH), a canonical driver of normal colonocyte differentiation, exists in a bivalent chromatin state. Inhibition of EZH2 resulted in de-repression of IHH, decreased self-renewal, and increased sensitivity to chemotherapy in vivo. Our results reveal an epigenetic block to differentiation in CC-ICs and demonstrate the potential for epigenetic differentiation therapy of a solid tumour through EZH2 inhibition.


Subject(s)
Cell Self Renewal , Colorectal Neoplasms/metabolism , Colorectal Neoplasms/pathology , Hedgehog Proteins/metabolism , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology , Animals , Cell Differentiation/drug effects , Cell Differentiation/genetics , Cell Proliferation/drug effects , Cell Self Renewal/drug effects , Enhancer of Zeste Homolog 2 Protein/metabolism , Female , Fluorouracil/pharmacology , Humans , Male , Mice, Inbred NOD , Mice, SCID , Neoplastic Stem Cells/drug effects , Pyridones/pharmacology
10.
Nucleic Acids Res ; 47(3): 1225-1238, 2019 02 20.
Article in English | MEDLINE | ID: mdl-30462309

ABSTRACT

Aberrant isoform expression of chromatin-associated proteins can induce epigenetic programs related to disease. The MDS1 and EVI1 complex locus (MECOM) encodes PRDM3, a protein with an N-terminal PR-SET domain, as well as a shorter isoform, EVI1, lacking the N-terminus containing the PR-SET domain (ΔPR). Imbalanced expression of MECOM isoforms is observed in multiple malignancies, implicating EVI1 as an oncogene, while PRDM3 has been suggested to function as a tumor suppressor through an unknown mechanism. To elucidate functional characteristics of these N-terminal residues, we compared the protein interactomes of the full-length and ΔPR isoforms of PRDM3 and its closely related paralog, PRDM16. Unlike the ΔPR isoforms, both full-length isoforms exhibited a significantly enriched association with components of the NuRD chromatin remodeling complex, especially RBBP4. Typically, RBBP4 facilitates chromatin association of the NuRD complex by binding to histone H3 tails. We show that RBBP4 binds to the N-terminal amino acid residues of PRDM3 and PRDM16, with a dissociation constant of 3.0 µM, as measured by isothermal titration calorimetry. Furthermore, high-resolution X-ray crystal structures of PRDM3 and PRDM16 N-terminal peptides in complex with RBBP4 revealed binding to RBBP4 within the conserved histone H3-binding groove. These data support a mechanism of isoform-specific interaction of PRDM3 and PRDM16 with the NuRD chromatin remodeling complex.


Subject(s)
DNA-Binding Proteins/chemistry , DNA-Binding Proteins/metabolism , MDS1 and EVI1 Complex Locus Protein/chemistry , MDS1 and EVI1 Complex Locus Protein/metabolism , Mi-2 Nucleosome Remodeling and Deacetylase Complex/metabolism , Transcription Factors/chemistry , Transcription Factors/metabolism , Animals , Cell Line , Crystallography, X-Ray , Humans , MDS1 and EVI1 Complex Locus Protein/genetics , Mice , Models, Molecular , Neoplasms/genetics , Neoplasms/metabolism , Protein Interaction Domains and Motifs , Retinoblastoma-Binding Protein 4/chemistry , Retinoblastoma-Binding Protein 4/metabolism , Tumor Suppressor Proteins/chemistry , Tumor Suppressor Proteins/genetics , Tumor Suppressor Proteins/metabolism
11.
Clin Cancer Res ; 24(9): 2116-2127, 2018 05 01.
Article in English | MEDLINE | ID: mdl-29476017

ABSTRACT

Purpose: Cancer-initiating cells (C-IC) have been described in multiple cancer types, including colorectal cancer. C-ICs are defined by their capacity to self-renew, thereby driving tumor growth. C-ICs were initially thought to be static entities; however, recent studies have determined these cells to be dynamic and influenced by microenvironmental cues such as hypoxia. If hypoxia drives the formation of C-ICs, then therapeutic targeting of hypoxia could represent a novel means to target C-ICs.Experimental Design: Patient-derived colorectal cancer xenografts were treated with evofosfamide, a hypoxia-activated prodrug (HAP), in combination with 5-fluorouracil (5-FU) or chemoradiotherapy (5-FU and radiation; CRT). Treatment groups included both concurrent and sequential dosing regimens. Effects on the colorectal cancer-initiating cell (CC-IC) fraction were assessed by serial passage in vivo limiting dilution assays. FAZA-PET imaging was utilized as a noninvasive method to assess intratumoral hypoxia.Results: Hypoxia was sufficient to drive the formation of CC-ICs and colorectal cancer cells surviving conventional therapy were more hypoxic and C-IC-like. Using a novel approach to combination therapy, we show that sequential treatment with 5-FU or CRT followed by evofosfamide not only inhibits tumor growth of xenografts compared with 5-FU or CRT alone, but also significantly decreases the CC-IC fraction. Furthermore, noninvasive FAZA-PET hypoxia imaging was predictive of a tumor's response to evofosfamide.Conclusions: Our data demonstrate a novel means to target the CC-IC fraction by adding a HAP sequentially after conventional adjuvant therapy, as well as the use of FAZA-PET as a biomarker for hypoxia to identify tumors that will benefit most from this approach. Clin Cancer Res; 24(9); 2116-27. ©2018 AACR.


Subject(s)
Colorectal Neoplasms/metabolism , Hypoxia/metabolism , Neoplastic Stem Cells/drug effects , Neoplastic Stem Cells/metabolism , Nitroimidazoles/administration & dosage , Phosphoramide Mustards/administration & dosage , Prodrugs/administration & dosage , Animals , Biomarkers , Caspases/metabolism , Cell Hypoxia/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Chemoradiotherapy , Colorectal Neoplasms/drug therapy , Colorectal Neoplasms/pathology , Colorectal Neoplasms/radiotherapy , Disease Models, Animal , Drug Evaluation, Preclinical , Drug Synergism , Female , Humans , Male , Mice , Phenotype , Positron-Emission Tomography , Standard of Care , Wnt Signaling Pathway , Xenograft Model Antitumor Assays
13.
Nat Chem Biol ; 13(4): 389-395, 2017 04.
Article in English | MEDLINE | ID: mdl-28135237

ABSTRACT

Polycomb repressive complex 2 (PRC2) is a regulator of epigenetic states required for development and homeostasis. PRC2 trimethylates histone H3 at lysine 27 (H3K27me3), which leads to gene silencing, and is dysregulated in many cancers. The embryonic ectoderm development (EED) protein is an essential subunit of PRC2 that has both a scaffolding function and an H3K27me3-binding function. Here we report the identification of A-395, a potent antagonist of the H3K27me3 binding functions of EED. Structural studies demonstrate that A-395 binds to EED in the H3K27me3-binding pocket, thereby preventing allosteric activation of the catalytic activity of PRC2. Phenotypic effects observed in vitro and in vivo are similar to those of known PRC2 enzymatic inhibitors; however, A-395 retains potent activity against cell lines resistant to the catalytic inhibitors. A-395 represents a first-in-class antagonist of PRC2 protein-protein interactions (PPI) for use as a chemical probe to investigate the roles of EED-containing protein complexes.


Subject(s)
Antineoplastic Agents/pharmacology , Indans/pharmacology , Polycomb Repressive Complex 2/antagonists & inhibitors , Sulfonamides/pharmacology , Antineoplastic Agents/chemistry , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Dose-Response Relationship, Drug , Drug Screening Assays, Antitumor , Humans , Indans/chemistry , Models, Molecular , Molecular Structure , Polycomb Repressive Complex 2/chemistry , Polycomb Repressive Complex 2/metabolism , Protein Binding/drug effects , Structure-Activity Relationship , Sulfonamides/chemistry , Tumor Cells, Cultured
14.
J Biol Chem ; 291(26): 13509-19, 2016 Jun 24.
Article in English | MEDLINE | ID: mdl-27129774

ABSTRACT

PR domain-containing protein 7 (PRDM7) is a primate-specific histone methyltransferase that is the result of a recent gene duplication of PRDM9. The two proteins are highly homologous, especially in the catalytic PR/SET domain, where they differ by only three amino acid residues. Here we report that PRDM7 is an efficient methyltransferase that selectively catalyzes the trimethylation of H3 lysine 4 (H3K4) both in vitro and in cells. Through selective mutagenesis we have dissected the functional roles of each of the three divergent residues between the PR domains of PRDM7 and PRDM9. These studies indicate that after a single serine to tyrosine mutation at residue 357 (S357Y), PRDM7 regains the substrate specificities and catalytic activities similar to its evolutionary predecessor, including the ability to efficiently methylate H3K36.


Subject(s)
Histone-Lysine N-Methyltransferase/chemistry , Histone-Lysine N-Methyltransferase/metabolism , Amino Acid Substitution , Gene Duplication , HEK293 Cells , Histone-Lysine N-Methyltransferase/genetics , Histones/chemistry , Histones/genetics , Histones/metabolism , Humans , Methylation , Mutagenesis , Mutation, Missense , Substrate Specificity
15.
Methods Mol Biol ; 1388: 95-110, 2016.
Article in English | MEDLINE | ID: mdl-27033073

ABSTRACT

Tumor suppressor PTEN phosphatase acts to inhibit the PI3K/AKT pathway and thus regulates cell proliferation, survival, and migration. Dysregulation of PTEN function is observed in a wide range of cancers. In addition to alterations of the PTEN gene, repression of PTEN function can also occur at the protein level through changes in PTEN conformation, localization, activity, and stability. The ability to follow switches in PTEN conformation in live cells provides a rapid approach to study changes in PTEN function and may provide a basis to screen pharmacological agents aimed at enhancing or reestablishing PTEN-dependent signaling pathways that have gone awry in cancer. Here, we describe methods to use an intramolecular bioluminescent resonance energy transfer (BRET)-based biosensor that reports dynamic signal-dependent changes in PTEN conformational rearrangement and function.


Subject(s)
Bioluminescence Resonance Energy Transfer Techniques/methods , PTEN Phosphohydrolase/chemistry , Cell Line, Tumor , HEK293 Cells , HeLa Cells , Humans
16.
Biochim Biophys Acta ; 1850(9): 1842-8, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26002201

ABSTRACT

BACKGROUND: Dysregulation of methylation of lysine 36 on histone H3 (H3K36) have been implicated in a variety of diseases including cancers. ASH1L and SETD2 are two enzymes among others that catalyze H3K36 methylation. H3K4 methylation has also been reported for ASH1L. METHODS: Radioactivity-based enzyme assays, Western and immunoblotting using specific antibodies and molecular modeling were used to characterize substrate specificity of ASH1L and SETD2. RESULTS: Here we report on the assay development and kinetic characterization of ASH1L and SETD2 and their substrate specificities in vitro. Both enzymes were active with recombinant nucleosome as substrate. However, SETD2 but not ASH1L methylated histone peptides as well indicating that the interaction of the basic post-SET extension with substrate may not be critical for SETD2 activity. Both enzymes were not active with nucleosome containing a H3K36A mutation indicating their specificity for H3K36. Analyzing the methylation state of the products of ASH1L and SETD2 reactions also confirmed that both enzymes mono- and dimethylate H3K36 and are inactive with H3K4 as substrate, and that only SETD2 is able to trimethylate H3K36 in vitro. CONCLUSIONS: We determined the kinetic parameters for ASH1L and SETD2 activity enabling screening for inhibitors that can be used to further investigate the roles of these two proteins in health and disease. Both ASH1L and SETD2 are H3K36 specific methyltransferases but only SETD2 can trimethylate this mark. The basic post-SET extension is critical for ASH1L but not SETD2 activity. GENERAL SIGNIFICANCE: We provide full kinetic characterization of ASH1L and SETD2 activity.


Subject(s)
DNA-Binding Proteins/metabolism , Histone-Lysine N-Methyltransferase/metabolism , Transcription Factors/metabolism , Amino Acid Sequence , DNA-Binding Proteins/chemistry , Histone-Lysine N-Methyltransferase/chemistry , Humans , Kinetics , Methylation , Models, Molecular , Molecular Sequence Data , Substrate Specificity , Transcription Factors/chemistry
17.
Nat Commun ; 5: 4431, 2014 Jul 16.
Article in English | MEDLINE | ID: mdl-25028204

ABSTRACT

Tumour suppressor PTEN is a phosphatase that negatively regulates the PI3K/AKT pathway. The ability to directly monitor PTEN conformation and function in a rapid, sensitive manner is a key step towards developing anti-cancer drugs aimed at enhancing or restoring PTEN-dependent pathways. Here we developed an intramolecular bioluminescence resonance energy transfer (BRET)-based biosensor, capable of detecting signal-dependent PTEN conformational changes in live cells. The biosensor retains intrinsic properties of PTEN, enabling structure-function and kinetic analyses. BRET shifts, indicating conformational change, were detected following mutations that disrupt intramolecular PTEN interactions, promoting plasma membrane targeting and also following physiological PTEN activation. Using the biosensor as a reporter, we uncovered PTEN activation by several G protein-coupled receptors, previously unknown as PTEN regulators. Trastuzumab, used to treat ERBB2-overexpressing breast cancers also elicited activation-associated PTEN conformational rearrangement. We propose the biosensor can be used to identify pathways regulating PTEN or molecules that enhance its anti-tumour activity.


Subject(s)
Biosensing Techniques/methods , PTEN Phosphohydrolase/metabolism , Cell Line , Cell Line, Tumor , Flow Cytometry , Humans , Immunoblotting , Immunoprecipitation , PTEN Phosphohydrolase/genetics , Receptor, ErbB-2/genetics , Receptor, ErbB-2/metabolism
18.
J Biol Chem ; 289(27): 19042-52, 2014 Jul 04.
Article in English | MEDLINE | ID: mdl-24855645

ABSTRACT

CCR5 binds the chemokines CCL3, CCL4, and CCL5 and is the major coreceptor for HIV-1 entry into target cells. Chemokines are supposed to form a natural barrier against human immunodeficiency virus, type 1 (HIV-1) infection. However, we showed that their antiviral activity is limited by CCR5 adopting low-chemokine affinity conformations at the cell surface. Here, we investigated whether a pool of CCR5 that is not stabilized by chemokines could represent a target for inhibiting HIV infection. We exploited the characteristics of the chemokine analog PSC-RANTES (N-α-(n-nonanoyl)-des-Ser(1)-[l-thioprolyl(2), l-cyclohexylglycyl(3)]-RANTES(4-68)), which displays potent anti-HIV-1 activity. We show that native chemokines fail to prevent high-affinity binding of PSC-RANTES, analog-mediated calcium release (in desensitization assays), and analog-mediated CCR5 internalization. These results indicate that a pool of spare CCR5 may bind PSC-RANTES but not native chemokines. Improved recognition of CCR5 by PSC-RANTES may explain why the analog promotes higher amounts of ß-arrestin 2·CCR5 complexes, thereby increasing CCR5 down-regulation and HIV-1 inhibition. Together, these results highlight that spare CCR5, which might permit HIV-1 to escape from chemokines, should be targeted for efficient viral blockade.


Subject(s)
Anti-HIV Agents/pharmacology , Chemokine CCL5/pharmacology , HIV-1/drug effects , HIV-1/physiology , Receptors, CCR5/metabolism , Virus Internalization/drug effects , Arrestins/metabolism , HEK293 Cells , Humans , Intracellular Space/drug effects , Intracellular Space/metabolism , Signal Transduction/drug effects , beta-Arrestin 2 , beta-Arrestins
19.
J Biol Chem ; 289(17): 12177-12188, 2014 Apr 25.
Article in English | MEDLINE | ID: mdl-24634223

ABSTRACT

PRDM9 (PR domain-containing protein 9) is a meiosis-specific protein that trimethylates H3K4 and controls the activation of recombination hot spots. It is an essential enzyme in the progression of early meiotic prophase. Disruption of the PRDM9 gene results in sterility in mice. In human, several PRDM9 SNPs have been implicated in sterility as well. Here we report on kinetic studies of H3K4 methylation by PRDM9 in vitro indicating that PRDM9 is a highly active histone methyltransferase catalyzing mono-, di-, and trimethylation of the H3K4 mark. Screening for other potential histone marks, we identified H3K36 as a second histone residue that could also be mono-, di-, and trimethylated by PRDM9 as efficiently as H3K4. Overexpression of PRDM9 in HEK293 cells also resulted in a significant increase in trimethylated H3K36 and H3K4 further confirming our in vitro observations. Our findings indicate that PRDM9 may play critical roles through H3K36 trimethylation in cells.


Subject(s)
DNA Methylation , Histone-Lysine N-Methyltransferase/metabolism , Histones/metabolism , Lysine/metabolism , Calorimetry , Histones/chemistry , Humans , Kinetics , Mass Spectrometry , Substrate Specificity
20.
Nat Med ; 20(1): 29-36, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24292392

ABSTRACT

Tumor recurrence following treatment remains a major clinical challenge. Evidence from xenograft models and human trials indicates selective enrichment of cancer-initiating cells (CICs) in tumors that survive therapy. Together with recent reports showing that CIC gene signatures influence patient survival, these studies predict that targeting self-renewal, the key 'stemness' property unique to CICs, may represent a new paradigm in cancer therapy. Here we demonstrate that tumor formation and, more specifically, human colorectal CIC function are dependent on the canonical self-renewal regulator BMI-1. Downregulation of BMI-1 inhibits the ability of colorectal CICs to self-renew, resulting in the abrogation of their tumorigenic potential. Treatment of primary colorectal cancer xenografts with a small-molecule BMI-1 inhibitor resulted in colorectal CIC loss with long-term and irreversible impairment of tumor growth. Targeting the BMI-1-related self-renewal machinery provides the basis for a new therapeutic approach in the treatment of colorectal cancer.


Subject(s)
Colorectal Neoplasms/drug therapy , Heterocyclic Compounds, 2-Ring/pharmacology , Neoplasm Recurrence, Local/metabolism , Neoplastic Stem Cells/metabolism , Polycomb Repressive Complex 1/metabolism , Thiazoles/pharmacology , Animals , Blotting, Western , Bromodeoxyuridine , Cell Line, Tumor , Flow Cytometry , Genetic Vectors/genetics , Heterocyclic Compounds, 2-Ring/therapeutic use , Humans , Luciferases , Mice, Inbred NOD , Mice, SCID , Polycomb Repressive Complex 1/antagonists & inhibitors , RNA Interference , RNA, Small Interfering/genetics , Thiazoles/therapeutic use
SELECTION OF CITATIONS
SEARCH DETAIL
...