Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
1.
Nat Cell Biol ; 8(11): 1246-54, 2006 Nov.
Article in English | MEDLINE | ID: mdl-17013377

ABSTRACT

Many proteins contain ubiquitin-binding domains or motifs (UBDs), such as the UIM (ubiquitin-interacting motif) and are referred to as ubiquitin receptors. Ubiquitin receptors themselves are frequently monoubiquitinated by a process that requires the presence of a UBD and is referred to as coupled monoubiquitination. Using a UIM-containing protein, eps15, as a model, we show here that coupled monoubiquitination strictly depends on the ability of the UIM to bind to monoubiquitin (mUb). We found that the underlying molecular mechanism is based on interaction between the UIM and a ubiquitin ligase (E3), which has itself been modified by ubiquitination. Furthermore, we demonstrate that the in vivo ubiquitination of members of the Nedd4 family of E3 ligases correlates with their ability to monoubiquitinate eps15. Thus, our results clarify the mechanism of coupled monoubiquitination and identify the ubiquitination of E3 ligases as a critical determinant in this process.


Subject(s)
Calcium-Binding Proteins/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Phosphoproteins/metabolism , Ubiquitin-Protein Ligases/metabolism , Ubiquitin/metabolism , Adaptor Proteins, Signal Transducing , Binding Sites/genetics , Calcium-Binding Proteins/genetics , Catalysis , Endosomal Sorting Complexes Required for Transport , HeLa Cells , Humans , Immunoblotting , Intracellular Signaling Peptides and Proteins/genetics , Models, Biological , Mutation/genetics , Nedd4 Ubiquitin Protein Ligases , Phosphoproteins/genetics , Protein Binding , Transfection , Ubiquitin-Protein Ligases/genetics
2.
Proc Natl Acad Sci U S A ; 107(14): 6459-64, 2010 Apr 06.
Article in English | MEDLINE | ID: mdl-20308550

ABSTRACT

The phosphatase and tensin homolog (PTEN) is a tumor suppressor that is inactivated in many human cancers. PTEN loss has been associated with resistance to inhibitors of the epidermal growth factor receptor (EGFR), but the molecular basis of this resistance is unclear. It is believed that unopposed phosphatidylinositol-3-kinase (PI3K) activation through multiple receptor tyrosine kinases (RTKs) can relieve PTEN-deficient cancers from their "dependence" on EGFR or any other single RTK for survival. Here we report a distinct resistance mechanism whereby PTEN inactivation specifically raises EGFR activity by impairing the ligand-induced ubiquitylation and degradation of the activated receptor through destabilization of newly formed ubiquitin ligase Cbl complexes. PTEN-associated resistance to EGFR kinase inhibitors is phenocopied by expression of dominant negative Cbl and can be overcome by more complete EGFR kinase inhibition. PTEN inactivation does not confer resistance to inhibitors of the MET or PDGFRA kinase. Our study identifies a critical role for PTEN in EGFR signal termination and suggests that more potent EGFR inhibition should overcome resistance caused by PI3K pathway activation.


Subject(s)
ErbB Receptors/antagonists & inhibitors , ErbB Receptors/metabolism , PTEN Phosphohydrolase/metabolism , Protein Kinase Inhibitors/pharmacology , Animals , Apoptosis , Cell Line , Enzyme Activation , Humans , Mice , Mice, Knockout , PTEN Phosphohydrolase/deficiency , PTEN Phosphohydrolase/genetics , Protein Binding , Proto-Oncogene Proteins c-cbl/metabolism , RNA Interference , Signal Transduction/drug effects , Ubiquitination
3.
Mol Syst Biol ; 7: 462, 2011 Jan 18.
Article in English | MEDLINE | ID: mdl-21245847

ABSTRACT

The activity, localization and fate of many cellular proteins are regulated through ubiquitination, a process whereby one or more ubiquitin (Ub) monomers or chains are covalently attached to target proteins. While Ub-conjugated and Ub-associated proteomes have been described, we lack a high-resolution picture of the dynamics of ubiquitination in response to signaling. In this study, we describe the epidermal growth factor (EGF)-regulated Ubiproteome, as obtained by two complementary purification strategies coupled to quantitative proteomics. Our results unveil the complex impact of growth factor signaling on Ub-based intracellular networks to levels that extend well beyond what might have been expected. In addition to endocytic proteins, the EGF-regulated Ubiproteome includes a large number of signaling proteins, ubiquitinating and deubiquitinating enzymes, transporters and proteins involved in translation and transcription. The Ub-based signaling network appears to intersect both housekeeping and regulatory circuitries of cellular physiology. Finally, as proof of principle of the biological relevance of the EGF-Ubiproteome, we demonstrated that EphA2 is a novel, downstream ubiquitinated target of epidermal growth factor receptor (EGFR), critically involved in EGFR biological responses.


Subject(s)
Epidermal Growth Factor/metabolism , Proteome/metabolism , Proteomics/methods , Systems Biology/methods , Ubiquitin/metabolism , Animals , Blotting, Western , Cell Line , Cluster Analysis , Epidermal Growth Factor/chemistry , HeLa Cells , Humans , Mass Spectrometry , Mice , Microscopy, Fluorescence , Proteome/chemistry , Receptor, EphA2/metabolism , Signal Transduction , Ubiquitin/chemistry
4.
Elife ; 102021 08 17.
Article in English | MEDLINE | ID: mdl-34399888

ABSTRACT

The molecular basis underlying glioblastoma (GBM) heterogeneity and plasticity is not fully understood. Using transcriptomic data of human patient-derived brain tumor stem cell lines (BTSCs), classified based on GBM-intrinsic signatures, we identify the AP-1 transcription factor FOSL1 as a key regulator of the mesenchymal (MES) subtype. We provide a mechanistic basis to the role of the neurofibromatosis type 1 gene (NF1), a negative regulator of the RAS/MAPK pathway, in GBM mesenchymal transformation through the modulation of FOSL1 expression. Depletion of FOSL1 in NF1-mutant human BTSCs and Kras-mutant mouse neural stem cells results in loss of the mesenchymal gene signature and reduction in stem cell properties and in vivo tumorigenic potential. Our data demonstrate that FOSL1 controls GBM plasticity and aggressiveness in response to NF1 alterations.


Subject(s)
Brain Neoplasms/genetics , Gene Expression Regulation, Neoplastic , Glioblastoma/genetics , Neoplastic Stem Cells/pathology , Neurofibromin 1/genetics , Proto-Oncogene Proteins c-fos/genetics , Cell Line, Tumor , Humans , Neurofibromin 1/metabolism , Proto-Oncogene Proteins c-fos/metabolism
5.
Nat Commun ; 11(1): 2977, 2020 06 12.
Article in English | MEDLINE | ID: mdl-32532995

ABSTRACT

Independent scientific achievements have led to the discovery of aberrant splicing patterns in oncogenesis, while more recent advances have uncovered novel gene fusions involving neurotrophic tyrosine receptor kinases (NTRKs) in gliomas. The exploration of NTRK splice variants in normal and neoplastic brain provides an intersection of these two rapidly evolving fields. Tropomyosin receptor kinase B (TrkB), encoded NTRK2, is known for critical roles in neuronal survival, differentiation, molecular properties associated with memory, and exhibits intricate splicing patterns and post-translational modifications. Here, we show a role for a truncated NTRK2 splice variant, TrkB.T1, in human glioma. TrkB.T1 enhances PDGF-driven gliomas in vivo, augments PDGF-induced Akt and STAT3 signaling in vitro, while next generation sequencing broadly implicates TrkB.T1 in the PI3K signaling cascades in a ligand-independent fashion. These TrkB.T1 findings highlight the importance of expanding upon whole gene and gene fusion analyses to include splice variants in basic and translational neuro-oncology research.


Subject(s)
Brain Neoplasms/genetics , Glioma/genetics , Membrane Glycoproteins/genetics , Oncogenes/genetics , RNA Isoforms/genetics , RNA Splicing , Receptor, trkB/genetics , Animals , Brain/metabolism , Brain/pathology , Brain Neoplasms/metabolism , Brain Neoplasms/pathology , Carcinogenesis/genetics , Cells, Cultured , Gene Expression Profiling , Gene Ontology , Glioma/metabolism , Glioma/pathology , High-Throughput Nucleotide Sequencing , Humans , Membrane Glycoproteins/metabolism , Mice , NIH 3T3 Cells , Neural Stem Cells/metabolism , Oncogene Proteins, Fusion/genetics , Oncogene Proteins, Fusion/metabolism , Phosphatidylinositol 3-Kinases/metabolism , RNA Isoforms/metabolism , Receptor, trkB/metabolism , Signal Transduction/genetics
6.
Nat Commun ; 11(1): 3883, 2020 08 04.
Article in English | MEDLINE | ID: mdl-32753598

ABSTRACT

Temozolomide (TMZ) is an oral alkylating agent used for the treatment of glioblastoma and is now becoming a chemotherapeutic option in patients diagnosed with high-risk low-grade gliomas. The O-6-methylguanine-DNA methyltransferase (MGMT) is responsible for the direct repair of the main TMZ-induced toxic DNA adduct, the O6-Methylguanine lesion. MGMT promoter hypermethylation is currently the only known biomarker for TMZ response in glioblastoma patients. Here we show that a subset of recurrent gliomas carries MGMT genomic rearrangements that lead to MGMT overexpression, independently from changes in its promoter methylation. By leveraging the CRISPR/Cas9 technology we generated some of these MGMT rearrangements in glioma cells and demonstrated that the MGMT genomic rearrangements contribute to TMZ resistance both in vitro and in vivo. Lastly, we showed that such fusions can be detected in tumor-derived exosomes and could potentially represent an early detection marker of tumor recurrence in a subset of patients treated with TMZ.


Subject(s)
Brain Neoplasms/drug therapy , DNA Modification Methylases/genetics , DNA Repair Enzymes/genetics , Drug Resistance, Neoplasm/genetics , Gene Rearrangement , Glioma/drug therapy , Neoplasm Recurrence, Local/genetics , Temozolomide/pharmacology , Tumor Suppressor Proteins/genetics , Adolescent , Adult , Aged , Animals , Brain Neoplasms/genetics , Cell Line, Tumor , DNA Adducts/drug effects , DNA Adducts/metabolism , DNA Methylation , DNA Modification Methylases/metabolism , DNA Repair Enzymes/metabolism , Female , Gene Expression Regulation, Neoplastic , Glioma/genetics , Humans , Male , Mice , Middle Aged , Neoplasm Recurrence, Local/prevention & control , Promoter Regions, Genetic/genetics , RNA-Seq , Temozolomide/therapeutic use , Tumor Suppressor Proteins/metabolism , Up-Regulation , Whole Genome Sequencing , Xenograft Model Antitumor Assays , Young Adult
7.
Nat Commun ; 9(1): 1466, 2018 04 13.
Article in English | MEDLINE | ID: mdl-29654229

ABSTRACT

To accurately recapitulate the heterogeneity of human diseases, animal models require to recreate multiple complex genetic alterations. Here, we combine the RCAS-TVA system with the CRISPR-Cas9 genome editing tools for precise modeling of human tumors. We show that somatic deletion in neural stem cells of a variety of known tumor suppressor genes (Trp53, Cdkn2a, and Pten) leads to high-grade glioma formation. Moreover, by simultaneous delivery of pairs of guide RNAs we generate different gene fusions with oncogenic potential, either by chromosomal deletion (Bcan-Ntrk1) or by chromosomal translocation (Myb-Qk). Lastly, using homology-directed-repair, we also produce tumors carrying the homologous mutation to human BRAF V600E, frequently identified in a variety of tumors, including different types of gliomas. In summary, we have developed an extremely versatile mouse model for in vivo somatic genome editing, that will elicit the generation of more accurate cancer models particularly appropriate for pre-clinical testing.


Subject(s)
Brain Neoplasms/genetics , CRISPR-Cas Systems , Gene Editing , RNA, Guide, Kinetoplastida/genetics , Animals , Antigens, Neoplasm/genetics , Benzamides/pharmacology , Brain Neoplasms/drug therapy , Brevican/genetics , DNA Repair , False Positive Reactions , Gene Frequency , Gene Transfer Techniques , Glioma/metabolism , Humans , In Situ Hybridization, Fluorescence , Indazoles/pharmacology , Mice , Mice, SCID , Mice, Transgenic , Mutation , NIH 3T3 Cells , Receptor, trkA/genetics
8.
Nat Commun ; 8(1): 2035, 2017 12 11.
Article in English | MEDLINE | ID: mdl-29229958

ABSTRACT

Transport of macromolecules through the nuclear pore by importins and exportins plays a critical role in the spatial regulation of protein activity. How cancer cells co-opt this process to promote tumorigenesis remains unclear. The epidermal growth factor receptor (EGFR) plays a critical role in normal development and in human cancer. Here we describe a mechanism of EGFR regulation through the importin ß family member RAN-binding protein 6 (RanBP6), a protein of hitherto unknown functions. We show that RanBP6 silencing impairs nuclear translocation of signal transducer and activator of transcription 3 (STAT3), reduces STAT3 binding to the EGFR promoter, results in transcriptional derepression of EGFR, and increased EGFR pathway output. Focal deletions of the RanBP6 locus on chromosome 9p were found in a subset of glioblastoma (GBM) and silencing of RanBP6 promoted glioma growth in vivo. Our results provide an example of EGFR deregulation in cancer through silencing of components of the nuclear import pathway.


Subject(s)
ErbB Receptors/genetics , Gene Expression Regulation, Neoplastic , Glioma/genetics , beta Karyopherins/genetics , ran GTP-Binding Protein/genetics , Active Transport, Cell Nucleus/genetics , Animals , Antibiotics, Antineoplastic/pharmacology , Cell Line, Tumor , Cells, Cultured , Doxorubicin/pharmacology , ErbB Receptors/metabolism , Feedback, Physiological , Female , Gene Knockdown Techniques , Glioma/drug therapy , Glioma/metabolism , HEK293 Cells , Humans , Mice, Knockout , Mice, SCID , STAT3 Transcription Factor/genetics , STAT3 Transcription Factor/metabolism , Xenograft Model Antitumor Assays , beta Karyopherins/metabolism , ran GTP-Binding Protein/metabolism
9.
Cancer Cell ; 28(6): 681-682, 2015 Dec 14.
Article in English | MEDLINE | ID: mdl-26678333

ABSTRACT

In this issue of Cancer Cell, Giachino and colleagues, employing various approaches, describe a tumor suppressor function for Notch signaling in forebrain tumors and suggest that decreased Notch activity could be a key molecular event in supratentorial primitive neuroectodermal tumors (sPNET).


Subject(s)
Brain Neoplasms/metabolism , Glioma/metabolism , Neoplastic Stem Cells/metabolism , Neural Stem Cells/metabolism , Prosencephalon/metabolism , Receptors, Notch/metabolism , Signal Transduction , Tumor Suppressor Proteins/metabolism , Animals , Humans
10.
Elife ; 32014 Dec 31.
Article in English | MEDLINE | ID: mdl-25551293

ABSTRACT

The serine-threonine kinase AKT regulates proliferation and survival by phosphorylating a network of protein substrates. In this study, we describe a kinase-independent function of AKT. In cancer cells harboring gain-of-function alterations in MET, HER2, or Phosphatidyl-Inositol-3-Kinase (PI3K), catalytically inactive AKT (K179M) protected from drug induced cell death in a PH-domain dependent manner. An AKT kinase domain mutant found in human melanoma (G161V) lacked enzymatic activity in vitro and in AKT1/AKT2 double knockout cells, but promoted growth factor independent survival of primary human melanocytes. ATP-competitive AKT inhibitors failed to block the kinase-independent function of AKT, a liability that limits their effectiveness compared to allosteric AKT inhibitors. Our results broaden the current view of AKT function and have important implications for the development of AKT inhibitors for cancer.


Subject(s)
Cell Survival , Melanoma/pathology , Proto-Oncogene Proteins c-akt/metabolism , Cell Line, Tumor , Humans , Melanoma/enzymology
11.
Science ; 340(6132): 626-30, 2013 May 03.
Article in English | MEDLINE | ID: mdl-23558169

ABSTRACT

The recent discovery of mutations in metabolic enzymes has rekindled interest in harnessing the altered metabolism of cancer cells for cancer therapy. One potential drug target is isocitrate dehydrogenase 1 (IDH1), which is mutated in multiple human cancers. Here, we examine the role of mutant IDH1 in fully transformed cells with endogenous IDH1 mutations. A selective R132H-IDH1 inhibitor (AGI-5198) identified through a high-throughput screen blocked, in a dose-dependent manner, the ability of the mutant enzyme (mIDH1) to produce R-2-hydroxyglutarate (R-2HG). Under conditions of near-complete R-2HG inhibition, the mIDH1 inhibitor induced demethylation of histone H3K9me3 and expression of genes associated with gliogenic differentiation. Blockade of mIDH1 impaired the growth of IDH1-mutant--but not IDH1-wild-type--glioma cells without appreciable changes in genome-wide DNA methylation. These data suggest that mIDH1 may promote glioma growth through mechanisms beyond its well-characterized epigenetic effects.


Subject(s)
Benzeneacetamides/pharmacology , Cell Differentiation , Enzyme Inhibitors/pharmacology , Glioma/enzymology , Glioma/pathology , Imidazoles/pharmacology , Isocitrate Dehydrogenase/antagonists & inhibitors , Isocitrate Dehydrogenase/genetics , Animals , Benzeneacetamides/administration & dosage , Benzeneacetamides/toxicity , Cell Differentiation/drug effects , Cell Transformation, Neoplastic , Enzyme Inhibitors/toxicity , Gene Expression Profiling , Gene Expression Regulation, Neoplastic/drug effects , Glioma/drug therapy , Glioma/genetics , Glutarates/metabolism , Histones/metabolism , Imidazoles/administration & dosage , Imidazoles/toxicity , Isocitrate Dehydrogenase/chemistry , Isocitrate Dehydrogenase/metabolism , Methylation , Mice , Mice, SCID , Mutant Proteins/antagonists & inhibitors , Mutant Proteins/chemistry , Mutant Proteins/metabolism , Protein Multimerization , RNA Interference , Xenograft Model Antitumor Assays
12.
Cancer Discov ; 2(5): 458-71, 2012 May.
Article in English | MEDLINE | ID: mdl-22588883

ABSTRACT

UNLABELLED: Activation of the epidermal growth factor receptor (EGFR) in glioblastoma (GBM) occurs through mutations or deletions in the extracellular (EC) domain. Unlike lung cancers with EGFR kinase domain (KD) mutations, GBMs respond poorly to the EGFR inhibitor erlotinib. Using RNAi, we show that GBM cells carrying EGFR EC mutations display EGFR addiction. In contrast to KD mutants found in lung cancer, glioma-specific EGFR EC mutants are poorly inhibited by EGFR inhibitors that target the active kinase conformation (e.g., erlotinib). Inhibitors that bind to the inactive EGFR conformation, however, potently inhibit EGFR EC mutants and induce cell death in EGFR-mutant GBM cells. Our results provide first evidence for single kinase addiction in GBM and suggest that the disappointing clinical activity of first-generation EGFR inhibitors in GBM versus lung cancer may be attributed to the different conformational requirements of mutant EGFR in these 2 cancer types. SIGNIFICANCE: Approximately 40% of human glioblastomas harbor oncogenic EGFR alterations, but attempts to therapeutically target EGFR with first-generation EGFR kinase inhibitors have failed. Here, we demonstrate selective sensitivity of glioma-specific EGFR mutants to ATP-site competitive EGFR kinase inhibitors that target the inactive conformation of the catalytic domain.


Subject(s)
Brain Neoplasms/genetics , ErbB Receptors/antagonists & inhibitors , ErbB Receptors/genetics , Glioma/genetics , Lung Neoplasms/genetics , Protein Kinase Inhibitors/pharmacology , Adenosine Triphosphate/metabolism , Animals , Antineoplastic Agents/pharmacology , Brain Neoplasms/metabolism , Cell Line, Tumor , ErbB Receptors/metabolism , Erlotinib Hydrochloride , Glioma/metabolism , Humans , Lapatinib , Lung Neoplasms/metabolism , Mice , Mutation , Quinazolines/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL