Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
Add more filters











Database
Language
Publication year range
1.
J Proteome Res ; 19(4): 1533-1547, 2020 04 03.
Article in English | MEDLINE | ID: mdl-32159963

ABSTRACT

Acquisition of drug resistance remains a chief impediment to successful cancer therapy, and we previously described a transient drug-tolerant cancer cell population (DTPs) whose survival is in part dependent on the activities of the histone methyltransferases G9a/EHMT2 and EZH2, the latter being the catalytic component of the polycomb repressive complex 2 (PRC2). Here, we apply multiple proteomic techniques to better understand the role of these histone methyltransferases (HMTs) in the establishment of the DTP state. Proteome-wide comparisons of lysine methylation patterns reveal that DTPs display an increase in methylation on K116 of PRC member Jarid2, an event that helps stabilize and recruit PRC2 to chromatin. We also find that EZH2, in addition to methylating histone H3K27, also can methylate G9a at K185, and that methylated G9a better recruits repressive complexes to chromatin. These complexes are similar to complexes recruited by histone H3 methylated at K9. Finally, a detailed histone post-translational modification (PTM) analysis shows that EZH2, either directly or through its ability to methylate G9a, alters H3K9 methylation in the context of H3 serine 10 phosphorylation, primarily in a cancer cell subpopulation that serves as DTP precursors. We also show that combinations of histone PTMs recruit a different set of complexes to chromatin, shedding light on the temporal mechanisms that contribute to drug tolerance.


Subject(s)
Neoplasms , Proteomics , Drug Tolerance , Enhancer of Zeste Homolog 2 Protein/genetics , Enhancer of Zeste Homolog 2 Protein/metabolism , Histones/metabolism , Methylation , Polycomb Repressive Complex 2/genetics , Polycomb Repressive Complex 2/metabolism
2.
Cancer Cell ; 32(2): 221-237.e13, 2017 08 14.
Article in English | MEDLINE | ID: mdl-28781121

ABSTRACT

Maintenance of phenotypic heterogeneity within cell populations is an evolutionarily conserved mechanism that underlies population survival upon stressful exposures. We show that the genomes of a cancer cell subpopulation that survives treatment with otherwise lethal drugs, the drug-tolerant persisters (DTPs), exhibit a repressed chromatin state characterized by increased methylation of histone H3 lysines 9 and 27 (H3K9 and H3K27). We also show that survival of DTPs is, in part, maintained by regulators of H3K9me3-mediated heterochromatin formation and that the observed increase in H3K9me3 in DTPs is most prominent over long interspersed repeat element 1 (LINE-1). Disruption of the repressive chromatin over LINE-1 elements in DTPs results in DTP ablation, which is partially rescued by reducing LINE-1 expression or function.


Subject(s)
Chromatin/genetics , Drug Resistance, Neoplasm/genetics , Epigenetic Repression/drug effects , Long Interspersed Nucleotide Elements/genetics , Neoplasms/pathology , Animals , Antineoplastic Agents/pharmacology , Gene Expression Regulation, Neoplastic , Genomic Instability/drug effects , Histone-Lysine N-Methyltransferase/metabolism , Histones/metabolism , Humans , Methylation , Mice , Mice, Nude , Mice, SCID , Neoplasms/drug therapy , Neoplasms/genetics , Stress, Physiological , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
3.
Nat Chem Biol ; 12(7): 531-8, 2016 07.
Article in English | MEDLINE | ID: mdl-27214401

ABSTRACT

The KDM5 family of histone demethylases catalyzes the demethylation of histone H3 on lysine 4 (H3K4) and is required for the survival of drug-tolerant persister cancer cells (DTPs). Here we report the discovery and characterization of the specific KDM5 inhibitor CPI-455. The crystal structure of KDM5A revealed the mechanism of inhibition of CPI-455 as well as the topological arrangements of protein domains that influence substrate binding. CPI-455 mediated KDM5 inhibition, elevated global levels of H3K4 trimethylation (H3K4me3) and decreased the number of DTPs in multiple cancer cell line models treated with standard chemotherapy or targeted agents. These findings show that pretreatment of cancer cells with a KDM5-specific inhibitor results in the ablation of a subpopulation of cancer cells that can serve as the founders for therapeutic relapse.


Subject(s)
Antineoplastic Agents/pharmacology , Drug Resistance, Neoplasm/drug effects , Enzyme Inhibitors/pharmacology , Neoplasms/drug therapy , Neoplasms/pathology , Retinoblastoma-Binding Protein 2/antagonists & inhibitors , Antineoplastic Agents/chemistry , Cell Survival/drug effects , Dose-Response Relationship, Drug , Drug Screening Assays, Antitumor , Enzyme Inhibitors/chemistry , Humans , Models, Molecular , Molecular Structure , Retinoblastoma-Binding Protein 2/metabolism , Structure-Activity Relationship
4.
Mol Cell ; 48(6): 888-99, 2012 Dec 28.
Article in English | MEDLINE | ID: mdl-23142077

ABSTRACT

Apoptotic caspase activation mechanisms are well defined, yet inactivation modes remain unclear. The death receptors (DRs), DR4, DR5, and Fas, transduce cell-extrinsic apoptotic signals by recruiting caspase-8 into a death-inducing signaling complex (DISC). At the DISC, Cullin3-dependent polyubiquitination on the small catalytic subunit of caspase-8 augments stimulation. Here we report that tumor necrosis factor receptor-associated factor 2 (TRAF2) interacts with caspase-8 at the DISC, downstream of Cullin3. TRAF2 directly mediates RING-dependent, K48-linked polyubiquitination on the large catalytic domain of caspase-8. This modification destines activated caspase-8 molecules to rapid proteasomal degradation upon autoprocessing and cytoplasmic translocation. TRAF2 depletion lowers the signal threshold for DR-mediated apoptosis, altering cell life versus death decisions in vitro and in vivo. Thus, TRAF2 sets a critical barrier for cell-extrinsic apoptosis commitment by tagging activated caspase-8 with a K48-ubiquitin shutoff timer. These results may have important implications for caspase regulation mechanisms.


Subject(s)
Apoptosis , Caspase 8/metabolism , Protein Processing, Post-Translational , Proteolysis , TNF Receptor-Associated Factor 2/physiology , Amino Acid Sequence , Animals , Catalytic Domain , Cell Survival , Cullin Proteins/metabolism , Death Domain Receptor Signaling Adaptor Proteins/metabolism , Enzyme Activation , HCT116 Cells , Humans , Leupeptins/pharmacology , Mice , Mice, Inbred C57BL , Mice, Knockout , Molecular Sequence Data , Peptide Mapping , Proteasome Endopeptidase Complex/metabolism , Proteasome Inhibitors/pharmacology , TNF Receptor-Associated Factor 2/genetics , TNF Receptor-Associated Factor 2/metabolism , Ubiquitination
5.
Cancer Cell ; 22(1): 80-90, 2012 Jul 10.
Article in English | MEDLINE | ID: mdl-22789540

ABSTRACT

The proapoptotic death receptor DR5 has been studied extensively in cancer cells, but its action in the tumor microenvironment is not well defined. Here, we uncover a role for DR5 signaling in tumor endothelial cells (ECs). We detected DR5 expression in ECs within tumors but not normal tissues. Treatment of tumor-bearing mice with an oligomeric form of the DR5 ligand Apo2L/TRAIL induced apoptosis in tumor ECs, collapsing blood vessels and reducing tumor growth: Vascular disruption and antitumor activity required DR5 expression on tumor ECs but not malignant cells. These results establish a therapeutic paradigm for proapoptotic receptor agonists as selective tumor vascular disruption agents, providing an alternative, perhaps complementary, strategy to their use as activators of apoptosis in malignant cells.


Subject(s)
Apoptosis , Cell Division , Endothelium, Vascular/metabolism , Neoplasms/blood supply , Receptors, TNF-Related Apoptosis-Inducing Ligand/metabolism , Animals , Humans , Mice , Neoplasms/pathology
6.
J Proteome Res ; 11(5): 2947-54, 2012 May 04.
Article in English | MEDLINE | ID: mdl-22432722

ABSTRACT

Proteolysis is a key regulatory event that controls intracellular and extracellular signaling through irreversible changes in a protein's structure that greatly alters its function. Here we describe a platform for profiling caspase substrates which encompasses two highly complementary proteomic techniques--the first is a differential gel based approach termed Global Analyzer of SILAC-derived Substrates of Proteolysis (GASSP) and the second involves affinity enrichment of peptides containing a C-terminal aspartic acid residue. In combination, these techniques have enabled the profiling of a large cellular pool of apoptotic-mediated proteolytic events across a wide dynamic range. By applying this integrated proteomic work flow to analyze proteolytic events resulting from the induction of intrinsic apoptosis in Jurkat cells via etoposide treatment, 3346 proteins were quantified, of which 360 proteins were identified as etoposide-induced proteolytic substrates, including 160 previously assigned caspase substrates. In addition to global profiling, a targeted approach using BAX HCT116 isogenic cell lines was utilized to dissect pre- and post-mitochondrial extrinsic apoptotic cleavage events. By employing apoptotic activation with a pro-apoptotic receptor agonist (PARA), a limited set of apoptotic substrates including known caspase substrates such as BH3 interacting-domain death agonist (BID) and Poly (ADP-ribose) polymerase (PARP)-1, and novel substrates such as Basic Transcription Factor 3, TRK-fused gene protein (TFG), and p62/Sequestosome were also identified.


Subject(s)
Apoptosis/drug effects , Proteolysis , Proteomics/methods , Adaptor Proteins, Signal Transducing/chemistry , Aspartic Acid/chemistry , BH3 Interacting Domain Death Agonist Protein/chemistry , Caspases/chemistry , Computational Biology , Etoposide/pharmacology , HCT116 Cells , Humans , Jurkat Cells , Nuclear Proteins/chemistry , Peptides/chemistry , Poly (ADP-Ribose) Polymerase-1 , Poly(ADP-ribose) Polymerases/chemistry , Proteins/chemistry , RNA-Binding Proteins/chemistry , Sequestosome-1 Protein , Substrate Specificity , Transcription Factors/chemistry
7.
Cancer Cell ; 19(1): 101-13, 2011 Jan 18.
Article in English | MEDLINE | ID: mdl-21251615

ABSTRACT

Antibodies to cell-surface antigens trigger activatory Fcγ receptor (FcγR)-mediated retrograde signals in leukocytes to control immune effector functions. Here, we uncover an FcγR mechanism that drives antibody-dependent forward signaling in target cells. Agonistic antibodies to death receptor 5 (DR5) induce cancer-cell apoptosis and are in clinical trials; however, their mechanism of action in vivo is not fully defined. Interaction of the DR5-agonistic antibody drozitumab with leukocyte FcγRs promoted DR5-mediated tumor-cell apoptosis. Whereas the anti-CD20 antibody rituximab required activatory FcγRs for tumoricidal function, drozitumab was effective in the context of either activatory or inhibitory FcγRs. A CD40-agonistic antibody required similar FcγR interactions to stimulate nuclear factor-κB activity in B cells. Thus, FcγRs can drive antibody-mediated receptor signaling in target cells.


Subject(s)
Antibodies, Monoclonal/metabolism , Neoplasms/metabolism , Receptors, IgG/metabolism , Receptors, TNF-Related Apoptosis-Inducing Ligand/metabolism , Signal Transduction/immunology , Animals , Antibodies, Monoclonal/genetics , Antibodies, Monoclonal/immunology , Antibodies, Monoclonal/pharmacology , Antibodies, Monoclonal/therapeutic use , Antibodies, Monoclonal, Humanized , Apoptosis/immunology , B-Lymphocytes/drug effects , B-Lymphocytes/metabolism , CD40 Antigens/agonists , CD40 Antigens/immunology , Cell Line, Tumor , Female , HCT116 Cells , Humans , Immunoglobulin Fc Fragments/genetics , Immunoglobulin G/genetics , Immunoglobulin G/immunology , Immunoglobulin G/pharmacology , Killer Cells, Natural/immunology , Leukocytes/immunology , Leukocytes/metabolism , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Mutation/genetics , Mutation/immunology , Myeloid Cells/immunology , NF-kappa B/metabolism , Neoplasms/drug therapy , Neoplasms/pathology , Polymorphism, Single Nucleotide/genetics , Polymorphism, Single Nucleotide/immunology , Protein Binding/genetics , Protein Binding/immunology , Receptor Aggregation/immunology , Receptors, IgG/genetics , Receptors, TNF-Related Apoptosis-Inducing Ligand/agonists , Receptors, TNF-Related Apoptosis-Inducing Ligand/immunology , Xenograft Model Antitumor Assays
8.
Cell ; 137(4): 721-35, 2009 May 15.
Article in English | MEDLINE | ID: mdl-19427028

ABSTRACT

Cell-surface death receptors such as DR4 and DR5 trigger apoptosis through a death-inducing signaling complex (DISC) that recruits the apical protease caspase-8. Apoptosis commitment requires efficient activation and autocatalytic release of caspase-8 into the cytoplasm to engage executioner caspases. While DISC recruitment initiates caspase-8 stimulation, full activation of the protease depends on further molecular aggregation events that are not fully understood. Here, we show that death receptor ligation induces polyubiquitination of caspase-8, through a previously unknown interaction of the DISC with a cullin3 (CUL3)-based E3 ligase. CUL3-mediated caspase-8 polyubiquitination required the RING box protein RBX1, whereas the deubiquitinase A20 reversed this modification. The ubiquitin-binding protein p62/sequestosome-1 promoted aggregation of CUL3-modified caspase-8 within p62-dependent foci, leading to full activation and processing of the enzyme and driving commitment to cell death. These results identify a mechanism that positively controls apoptosis signaling by polyubiquitination and aggregation of a key initiator caspase.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Caspase 8/metabolism , Cullin Proteins/metabolism , Apoptosis , Carrier Proteins/metabolism , Cell Line, Tumor , Death Domain Receptor Signaling Adaptor Proteins/metabolism , Gene Knockdown Techniques , Humans , Protein Transport , Sequestosome-1 Protein , Ubiquitin/metabolism , Ubiquitination
9.
Nat Med ; 13(9): 1070-7, 2007 Sep.
Article in English | MEDLINE | ID: mdl-17767167

ABSTRACT

Apo2L/TRAIL stimulates cancer cell death through the proapoptotic receptors DR4 and DR5, but the determinants of tumor susceptibility to this ligand are not fully defined. mRNA expression of the peptidyl O-glycosyltransferase GALNT14 correlated with Apo2L/TRAIL sensitivity in pancreatic carcinoma, non-small-cell lung carcinoma and melanoma cell lines, and up to 30% of samples from various human malignancies showed GALNT14 overexpression. RNA interference of GALNT14 reduced cellular Apo2L/TRAIL sensitivity, whereas overexpression increased responsiveness. Biochemical analysis of DR5 identified several ectodomain O-(N-acetyl galactosamine-galactose-sialic acid) structures. Sequence comparison predicted conserved extracellular DR4 and DR5 O-glycosylation sites; progressive mutation of the DR5 sites attenuated apoptotic signaling. O-glycosylation promoted ligand-stimulated clustering of DR4 and DR5, which mediated recruitment and activation of the apoptosis-initiating protease caspase-8. These results uncover a new link between death-receptor O-glycosylation and apoptotic signaling, providing potential predictive biomarkers for Apo2L/TRAIL-based cancer therapy.


Subject(s)
Receptors, Death Domain/physiology , TNF-Related Apoptosis-Inducing Ligand/physiology , Amino Acid Sequence , Animals , Apoptosis , Carcinoma, Non-Small-Cell Lung , Cell Line, Tumor , Cell Survival , Genetic Predisposition to Disease , Glycosylation , Humans , Lung Neoplasms , Melanoma , Mice , Molecular Sequence Data , Oligonucleotide Array Sequence Analysis , Pancreatic Neoplasms , RNA, Messenger/genetics , Receptors, TNF-Related Apoptosis-Inducing Ligand/genetics , Receptors, TNF-Related Apoptosis-Inducing Ligand/metabolism , Transplantation, Heterologous
SELECTION OF CITATIONS
SEARCH DETAIL