Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 62
Filtrer
Plus de filtres











Base de données
Gamme d'année
1.
Nat Chem Biol ; 20(9): 1164-1175, 2024 Sep.
Article de Anglais | MEDLINE | ID: mdl-38773330

RÉSUMÉ

The C-terminal to LisH (CTLH) complex is a ubiquitin ligase complex that recognizes substrates with Pro/N-degrons via its substrate receptor Glucose-Induced Degradation 4 (GID4), but its function and substrates in humans remain unclear. Here, we report PFI-7, a potent, selective and cell-active chemical probe that antagonizes Pro/N-degron binding to human GID4. Use of PFI-7 in proximity-dependent biotinylation and quantitative proteomics enabled the identification of GID4 interactors and GID4-regulated proteins. GID4 interactors are enriched for nucleolar proteins, including the Pro/N-degron-containing RNA helicases DDX21 and DDX50. We also identified a distinct subset of proteins whose cellular levels are regulated by GID4 including HMGCS1, a Pro/N-degron-containing metabolic enzyme. These data reveal human GID4 Pro/N-degron targets regulated through a combination of degradative and nondegradative functions. Going forward, PFI-7 will be a valuable research tool for investigating CTLH complex biology and facilitating development of targeted protein degradation strategies that highjack CTLH E3 ligase activity.


Sujet(s)
Liaison aux protéines , Humains , Protéolyse , Cellules HEK293 , Sondes moléculaires/composition chimique , Sondes moléculaires/métabolisme , DEAD-box RNA helicases/métabolisme , Ubiquitin-protein ligases/métabolisme , , Récepteurs à l'interleukine-17
2.
Nat Methods ; 21(4): 635-647, 2024 Apr.
Article de Anglais | MEDLINE | ID: mdl-38532014

RÉSUMÉ

Most proteins are organized in macromolecular assemblies, which represent key functional units regulating and catalyzing most cellular processes. Affinity purification of the protein of interest combined with liquid chromatography coupled to tandem mass spectrometry (AP-MS) represents the method of choice to identify interacting proteins. The composition of complex isoforms concurrently present in the AP sample can, however, not be resolved from a single AP-MS experiment but requires computational inference from multiple time- and resource-intensive reciprocal AP-MS experiments. Here we introduce deep interactome profiling by mass spectrometry (DIP-MS), which combines AP with blue-native-PAGE separation, data-independent acquisition with mass spectrometry and deep-learning-based signal processing to resolve complex isoforms sharing the same bait protein in a single experiment. We applied DIP-MS to probe the organization of the human prefoldin family of complexes, resolving distinct prefoldin holo- and subcomplex variants, complex-complex interactions and complex isoforms with new subunits that were experimentally validated. Our results demonstrate that DIP-MS can reveal proteome modularity at unprecedented depth and resolution.


Sujet(s)
Protéome , Protéomique , Humains , Protéomique/méthodes , Chromatographie d'affinité , Protéome/analyse , Spectrométrie de masse en tandem , Isoformes de protéines
3.
Nat Immunol ; 24(12): 2135-2149, 2023 Dec.
Article de Anglais | MEDLINE | ID: mdl-37932456

RÉSUMÉ

Current US Food and Drug Administration-approved chimeric antigen receptor (CAR) T cells harbor the T cell receptor (TCR)-derived ζ chain as an intracellular activation domain in addition to costimulatory domains. The functionality in a CAR format of the other chains of the TCR complex, namely CD3δ, CD3ε and CD3γ, instead of ζ, remains unknown. In the present study, we have systematically engineered new CD3 CARs, each containing only one of the CD3 intracellular domains. We found that CARs containing CD3δ, CD3ε or CD3γ cytoplasmic tails outperformed the conventional ζ CAR T cells in vivo. Transcriptomic and proteomic analysis revealed differences in activation potential, metabolism and stimulation-induced T cell dysfunctionality that mechanistically explain the enhanced anti-tumor performance. Furthermore, dimerization of the CARs improved their overall functionality. Using these CARs as minimalistic and synthetic surrogate TCRs, we have identified the phosphatase SHP-1 as a new interaction partner of CD3δ that binds the CD3δ-ITAM on phosphorylation of its C-terminal tyrosine. SHP-1 attenuates and restrains activation signals and might thus prevent exhaustion and dysfunction. These new insights into T cell activation could promote the rational redesign of synthetic antigen receptors to improve cancer immunotherapy.


Sujet(s)
Protéomique , Récepteurs aux antigènes des cellules T , Antigènes CD3 , Récepteurs aux antigènes des cellules T/génétique , Récepteurs aux antigènes des cellules T/métabolisme , Membrane cellulaire/métabolisme , Activation des lymphocytes , Lymphocytes T
4.
Methods Mol Biol ; 2706: 177-190, 2023.
Article de Anglais | MEDLINE | ID: mdl-37558949

RÉSUMÉ

Limited proteolysis coupled to mass spectrometry (LiP-MS) is a recent proteomics technique that allows structure-based target engagement profiling on a proteome-wide level. To achieve this, native lysates are first incubated with a compound, followed by a short incubation with a nonspecific protease. Binding of a compound can change accessibility at the binding site or induce other structural changes in the target. This leads to treatment-specific proteolytic fingerprints upon limited proteolysis, which can be analyzed by standard bottom-up MS-based proteomics. Here, we describe a basic LiP-MS protocol using the natural product rapamycin as an example compound. Along with the provided LiP-MS reference data available via ProteomeXchange with identifier PXD035183, this enables the straightforward implementation of the method by scientists with a basic biochemistry and mass spectrometry background. We describe how the procedure can easily be adapted to other protein samples and small molecules.


Sujet(s)
Peptide hydrolases , Protéome , Protéolyse , Spectrométrie de masse/méthodes , Protéome/composition chimique , Sites de fixation
5.
Mol Syst Biol ; 19(4): e11024, 2023 04 12.
Article de Anglais | MEDLINE | ID: mdl-36896621

RÉSUMÉ

While several computational methods have been developed to predict the functional relevance of phosphorylation sites, experimental analysis of the interdependency between protein phosphorylation and Protein-Protein Interactions (PPIs) remains challenging. Here, we describe an experimental strategy to establish interdependencies between protein phosphorylation and complex formation. This strategy is based on three main steps: (i) systematically charting the phosphorylation landscape of a target protein; (ii) assigning distinct proteoforms of the target protein to different protein complexes by native complex separation (AP-BNPAGE) and protein correlation profiling; and (iii) analyzing proteoforms and complexes in cells lacking regulators of the target protein. We applied this strategy to YAP1, a transcriptional co-activator for the control of organ size and tissue homeostasis that is highly phosphorylated and among the most connected proteins in human cells. We identified multiple YAP1 phosphosites associated with distinct complexes and inferred how both are controlled by Hippo pathway members. We detected a PTPN14/LATS1/YAP1 complex and suggest a model how PTPN14 inhibits YAP1 via augmenting WW domain-dependent complex formation and phosphorylation by LATS1/2.


Sujet(s)
Protéines adaptatrices de la transduction du signal , Transduction du signal , Humains , Phosphorylation , Protéines adaptatrices de la transduction du signal/métabolisme , Protéines de signalisation YAP , Protein-Serine-Threonine Kinases/métabolisme , Facteurs de transcription/métabolisme , Protein Tyrosine Phosphatases, Non-Receptor/métabolisme
6.
Proc Natl Acad Sci U S A ; 119(40): e2117175119, 2022 10 04.
Article de Anglais | MEDLINE | ID: mdl-36179048

RÉSUMÉ

Protein-protein interactions (PPIs) represent the main mode of the proteome organization in the cell. In the last decade, several large-scale representations of PPI networks have captured generic aspects of the functional organization of network components but mostly lack the context of cellular states. However, the generation of context-dependent PPI networks is essential for structural and systems-level modeling of biological processes-a goal that remains an unsolved challenge. Here we describe an experimental/computational strategy to achieve a modeling of PPIs that considers contextual information. This strategy defines the composition, stoichiometry, temporal organization, and cellular requirements for the formation of target assemblies. We used this approach to generate an integrated model of the formation principles and architecture of a large signalosome, the TNF-receptor signaling complex (TNF-RSC). Overall, we show that the integration of systems- and structure-level information provides a generic, largely unexplored link between the modular proteome and cellular function.


Sujet(s)
Phénomènes biologiques , Protéomique , Cartographie d'interactions entre protéines , Cartes d'interactions protéiques/physiologie , Protéome/métabolisme
7.
J Am Chem Soc ; 144(14): 6326-6342, 2022 04 13.
Article de Anglais | MEDLINE | ID: mdl-35353516

RÉSUMÉ

Covalent protein kinase inhibitors exploit currently noncatalytic cysteines in the adenosine 5'-triphosphate (ATP)-binding site via electrophiles directly appended to a reversible-inhibitor scaffold. Here, we delineate a path to target solvent-exposed cysteines at a distance >10 Å from an ATP-site-directed core module and produce potent covalent phosphoinositide 3-kinase α (PI3Kα) inhibitors. First, reactive warheads are used to reach out to Cys862 on PI3Kα, and second, enones are replaced with druglike warheads while linkers are optimized. The systematic investigation of intrinsic warhead reactivity (kchem), rate of covalent bond formation and proximity (kinact and reaction space volume Vr), and integration of structure data, kinetic and structural modeling, led to the guided identification of high-quality, covalent chemical probes. A novel stochastic approach provided direct access to the calculation of overall reaction rates as a function of kchem, kinact, Ki, and Vr, which was validated with compounds with varied linker lengths. X-ray crystallography, protein mass spectrometry (MS), and NanoBRET assays confirmed covalent bond formation of the acrylamide warhead and Cys862. In rat liver microsomes, compounds 19 and 22 outperformed the rapidly metabolized CNX-1351, the only known PI3Kα irreversible inhibitor. Washout experiments in cancer cell lines with mutated, constitutively activated PI3Kα showed a long-lasting inhibition of PI3Kα. In SKOV3 cells, compounds 19 and 22 revealed PI3Kß-dependent signaling, which was sensitive to TGX221. Compounds 19 and 22 thus qualify as specific chemical probes to explore PI3Kα-selective signaling branches. The proposed approach is generally suited to develop covalent tools targeting distal, unexplored Cys residues in biologically active enzymes.


Sujet(s)
Cystéine , Phosphatidylinositol 3-kinase , Adénosine triphosphate , Animaux , Cystéine/composition chimique , Phosphatidylinositol 3-kinases/métabolisme , Inhibiteurs des phosphoinositide-3 kinases , Inhibiteurs de protéines kinases/composition chimique , Rats
8.
Nat Methods ; 18(5): 520-527, 2021 05.
Article de Anglais | MEDLINE | ID: mdl-33859439

RÉSUMÉ

Despite the availability of methods for analyzing protein complexes, systematic analysis of complexes under multiple conditions remains challenging. Approaches based on biochemical fractionation of intact, native complexes and correlation of protein profiles have shown promise. However, most approaches for interpreting cofractionation datasets to yield complex composition and rearrangements between samples depend considerably on protein-protein interaction inference. We introduce PCprophet, a toolkit built on size exclusion chromatography-sequential window acquisition of all theoretical mass spectrometry (SEC-SWATH-MS) data to predict protein complexes and characterize their changes across experimental conditions. We demonstrate improved performance of PCprophet over state-of-the-art approaches and introduce a Bayesian approach to analyze altered protein-protein interactions across conditions. We provide both command-line and graphical interfaces to support the application of PCprophet to any cofractionation MS dataset, independent of separation or quantitative liquid chromatography-MS workflow, for the detection and quantitative tracking of protein complexes and their physiological dynamics.


Sujet(s)
Apprentissage machine , Protéines/composition chimique , Protéomique , Logiciel , Théorème de Bayes , Chromatographie sur gel , Bases de données de protéines , Conformation des protéines
9.
Methods Mol Biol ; 2259: 269-294, 2021.
Article de Anglais | MEDLINE | ID: mdl-33687722

RÉSUMÉ

In living cells, most proteins are organized in stable or transient functional assemblies, protein complexes, which control a multitude of vital cellular processes such as cell cycle progression, metabolism, and signal transduction. Over several decades, specific protein complexes have been analyzed by structural biology methods, initially X-ray crystallography and more recently single particle cryoEM. In parallel, mass spectrometry (MS)-based methods including in vitro affinity-purification coupled to MS or in vivo protein proximity-dependent labeling methods have proven particularly effective to detect complexes, thus nominating new assemblies for structural analysis. Those approaches, however, are either of limited in throughput or require specifically engineered protein systems.In this chapter, we present protocols for a workflow that supports the parallel analysis of multiple complexes from the same biological sample with respect to abundance, subunit composition, and stoichiometry. It consists of the separation of native complexes by size-exclusion chromatography (SEC) and the subsequent mass spectrometric analysis of the proteins in consecutive SEC fractions. In particular, we describe (1) optimized conditions to achieve native protein complex separation by SEC, (2) the preparation of the SEC fractions for MS analysis, (3) the acquisition of the MS data at high throughput via SWATH/DIA (data-independent analysis) mass spectrometry and short chromatographic gradients, and (4) a set of bioinformatic tools for the targeted analysis of protein complexes. Altogether, the parallel measurement of a high number of complexes from a single biological sample results in unprecedented system-level insights into the remodeling of cellular protein complexes in response to perturbations of a broad range of cellular systems.


Sujet(s)
Chromatographie sur gel/méthodes , Spectrométrie de masse/méthodes , Protéines/analyse , Protéomique/méthodes , Chromatographie en phase liquide à haute performance/méthodes , Humains , Cellules Jurkat , Ultracentrifugation/méthodes , Flux de travaux
10.
Nat Commun ; 12(1): 1693, 2021 03 16.
Article de Anglais | MEDLINE | ID: mdl-33727531

RÉSUMÉ

Proteases are among the largest protein families and critical regulators of biochemical processes like apoptosis and blood coagulation. Knowledge of proteases has been expanded by the development of proteomic approaches, however, technology for multiplexed screening of proteases within native environments is currently lacking behind. Here we introduce a simple method to profile protease activity based on isolation of protease products from native lysates using a 96FASP filter, their analysis in a mass spectrometer and a custom data analysis pipeline. The method is significantly faster, cheaper, technically less demanding, easy to multiplex and produces accurate protease fingerprints. Using the blood cascade proteases as a case study, we obtain protease substrate profiles that can be used to map specificity, cleavage entropy and allosteric effects and to design protease probes. The data further show that protease substrate predictions enable the selection of potential physiological substrates for targeted validation in biochemical assays.


Sujet(s)
Entropie , Tests de criblage à haut débit , Peptide hydrolases/sang , Peptide hydrolases/métabolisme , Régulation allostérique , Séquence d'acides aminés , Coagulation sanguine , Fluorescence , Cellules HEK293 , Humains , Matrix metalloproteinases/métabolisme , Peptides/métabolisme , Spécificité du substrat , Thromboplastine/métabolisme
11.
Oncogene ; 40(9): 1659-1673, 2021 03.
Article de Anglais | MEDLINE | ID: mdl-33500549

RÉSUMÉ

The clinical benefit of MAPK pathway inhibition in melanoma patients carrying BRAF mutations is temporal. After the initial response to treatment, the majority of tumors will develop resistance and patients will relapse. Here we demonstrate that the endothelin-endothelin receptor B (ETBR) signaling pathway confers resistance to MAPK pathway inhibitors in BRAF mutated melanoma. MAPK blockade, in addition to being anti-proliferative, induces a phenotypic change which is characterized by increased expression of melanocyte-specific genes including ETBR. In the presence of MAPK inhibitors, activation of ETBR by endothelin enables the sustained proliferation of melanoma cells. In mouse models of melanoma, including patient-derived xenograft models, concurrent inhibition of the MAPK pathway and ETBR signaling resulted in a more effective anti-tumor response compared to MAPK pathway inhibition alone. The combination treatment significantly reduced tumor growth and prolonged survival compared to therapies with MAPK pathway inhibitors alone. The phosphoproteomic analysis revealed that ETBR signaling did not induce resistance towards MAPK pathway inhibitors by restoring MAPK activity, but instead via multiple alternative signaling pathways downstream of the small G proteins GNAq/11. Together these data indicate that a combination of MAPK pathway inhibitors with ETBR antagonists could have a synergistically beneficial effect in melanoma patients with hyperactivated MAPK signaling pathways.


Sujet(s)
Mélanome/traitement médicamenteux , Récidive tumorale locale/traitement médicamenteux , Protéines proto-oncogènes B-raf/génétique , Récepteur de l'endothéline de type B/génétique , Animaux , Apoptose/effets des médicaments et des substances chimiques , Prolifération cellulaire/effets des médicaments et des substances chimiques , Résistance aux médicaments antinéoplasiques/génétique , Antagonistes du récepteur de type B de l'endothéline/pharmacologie , Humains , Mélanome/génétique , Mélanome/anatomopathologie , Souris , Mitogen-Activated Protein Kinase Kinases/antagonistes et inhibiteurs , Mutation/génétique , Récidive tumorale locale/génétique , Récidive tumorale locale/anatomopathologie , Inhibiteurs de protéines kinases/pharmacologie , Transduction du signal/effets des médicaments et des substances chimiques , Tests d'activité antitumorale sur modèle de xénogreffe
12.
Elife ; 92020 08 03.
Article de Anglais | MEDLINE | ID: mdl-32744500

RÉSUMÉ

The NuRD complex subunit CHD4 is essential for fusion-positive rhabdomyosarcoma (FP-RMS) survival, but the mechanisms underlying this dependency are not understood. Here, a NuRD-specific CRISPR screen demonstrates that FP-RMS is particularly sensitive to CHD4 amongst the NuRD members. Mechanistically, NuRD complex containing CHD4 localizes to super-enhancers where CHD4 generates a chromatin architecture permissive for the binding of the tumor driver and fusion protein PAX3-FOXO1, allowing downstream transcription of its oncogenic program. Moreover, CHD4 depletion removes HDAC2 from the chromatin, leading to an increase and spread of histone acetylation, and prevents the positioning of RNA Polymerase 2 at promoters impeding transcription initiation. Strikingly, analysis of genome-wide cancer dependency databases identifies CHD4 as a general cancer vulnerability. Our findings describe CHD4, a classically defined repressor, as positive regulator of transcription and super-enhancer accessibility as well as establish this remodeler as an unexpected broad tumor susceptibility and promising drug target for cancer therapy.


Sujet(s)
Régulation de l'expression des gènes tumoraux , Complexe Mi-2/NuRD/génétique , Rhabdomyosarcome/génétique , Lignée cellulaire tumorale , Humains , Complexe Mi-2/NuRD/métabolisme
13.
Nat Protoc ; 15(8): 2341-2386, 2020 08.
Article de Anglais | MEDLINE | ID: mdl-32690956

RÉSUMÉ

Most catalytic, structural and regulatory functions of the cell are carried out by functional modules, typically complexes containing or consisting of proteins. The composition and abundance of these complexes and the quantitative distribution of specific proteins across different modules are therefore of major significance in basic and translational biology. However, detection and quantification of protein complexes on a proteome-wide scale is technically challenging. We have recently extended the targeted proteomics rationale to the level of native protein complex analysis (complex-centric proteome profiling). The complex-centric workflow described herein consists of size exclusion chromatography (SEC) to fractionate native protein complexes, data-independent acquisition mass spectrometry to precisely quantify the proteins in each SEC fraction based on a set of proteotypic peptides and targeted, complex-centric analysis where prior information from generic protein interaction maps is used to detect and quantify protein complexes with high selectivity and statistical error control via the computational framework CCprofiler (https://github.com/CCprofiler/CCprofiler). Complex-centric proteome profiling captures most proteins in complex-assembled state and reveals their organization into hundreds of complexes and complex variants observable in a given cellular state. The protocol is applicable to cultured cells and can potentially also be adapted to primary tissue and does not require any genetic engineering of the respective sample sources. At present, it requires ~8 d of wet-laboratory work, 15 d of mass spectrometry measurement time and 7 d of computational analysis.


Sujet(s)
Chromatographie sur gel , Spectrométrie de masse , Protéines/isolement et purification , Protéines/métabolisme , Protéomique/méthodes , Cellules HEK293 , Humains
14.
Nat Commun ; 11(1): 3563, 2020 07 16.
Article de Anglais | MEDLINE | ID: mdl-32678104

RÉSUMÉ

Rapidly increasing availability of genomic data and ensuing identification of disease associated mutations allows for an unbiased insight into genetic drivers of disease development. However, determination of molecular mechanisms by which individual genomic changes affect biochemical processes remains a major challenge. Here, we develop a multilayered proteomic workflow to explore how genetic lesions modulate the proteome and are translated into molecular phenotypes. Using this workflow we determine how expression of a panel of disease-associated mutations in the Dyrk2 protein kinase alter the composition, topology and activity of this kinase complex as well as the phosphoproteomic state of the cell. The data show that altered protein-protein interactions caused by the mutations are associated with topological changes and affected phosphorylation of known cancer driver proteins, thus linking Dyrk2 mutations with cancer-related biochemical processes. Overall, we discover multiple mutation-specific functionally relevant changes, thus highlighting the extensive plasticity of molecular responses to genetic lesions.


Sujet(s)
Tumeurs/génétique , Tumeurs/anatomopathologie , Protein kinases/génétique , Protéomique/méthodes , Lignée cellulaire , Humains , Spectrométrie de masse , Complexes multiprotéiques , Mutation , Protéines tumorales/métabolisme , Tumeurs/métabolisme , Phénotype , Phosphoprotéines/métabolisme , Phosphorylation , Conformation des protéines , Cartes d'interactions protéiques , Protein kinases/composition chimique , Protein kinases/métabolisme , Protein-Serine-Threonine Kinases/composition chimique , Protein-Serine-Threonine Kinases/génétique , Protein-Serine-Threonine Kinases/métabolisme , Protein-tyrosine kinases/composition chimique , Protein-tyrosine kinases/génétique , Protein-tyrosine kinases/métabolisme , Protéome/métabolisme ,
15.
Mol Cell ; 79(3): 504-520.e9, 2020 08 06.
Article de Anglais | MEDLINE | ID: mdl-32707033

RÉSUMÉ

Protein kinases are essential for signal transduction and control of most cellular processes, including metabolism, membrane transport, motility, and cell cycle. Despite the critical role of kinases in cells and their strong association with diseases, good coverage of their interactions is available for only a fraction of the 535 human kinases. Here, we present a comprehensive mass-spectrometry-based analysis of a human kinase interaction network covering more than 300 kinases. The interaction dataset is a high-quality resource with more than 5,000 previously unreported interactions. We extensively characterized the obtained network and were able to identify previously described, as well as predict new, kinase functional associations, including those of the less well-studied kinases PIM3 and protein O-mannose kinase (POMK). Importantly, the presented interaction map is a valuable resource for assisting biomedical studies. We uncover dozens of kinase-disease associations spanning from genetic disorders to complex diseases, including cancer.


Sujet(s)
Réseaux de régulation génique , Maladies génétiques congénitales/génétique , Tumeurs/génétique , Protein kinases/génétique , Protein-Serine-Threonine Kinases/génétique , Protéines proto-oncogènes/génétique , Biologie informatique/méthodes , Jeux de données comme sujet , Régulation de l'expression des gènes , Gene Ontology , Maladies génétiques congénitales/enzymologie , Maladies génétiques congénitales/anatomopathologie , Humains , Voies et réseaux métaboliques/génétique , Annotation de séquence moléculaire , Dystrophies musculaires/enzymologie , Dystrophies musculaires/génétique , Dystrophies musculaires/anatomopathologie , Tumeurs/enzymologie , Tumeurs/anatomopathologie , Maladies neurodégénératives/enzymologie , Maladies neurodégénératives/génétique , Maladies neurodégénératives/anatomopathologie , Cartographie d'interactions entre protéines/méthodes , Protein kinases/composition chimique , Protein kinases/classification , Protein kinases/métabolisme , Protein-Serine-Threonine Kinases/composition chimique , Protein-Serine-Threonine Kinases/métabolisme , Protéines proto-oncogènes/composition chimique , Protéines proto-oncogènes/métabolisme , Transduction du signal
16.
Cell Syst ; 10(2): 133-155.e6, 2020 02 26.
Article de Anglais | MEDLINE | ID: mdl-32027860

RÉSUMÉ

Living systems integrate biochemical reactions that determine the functional state of each cell. Reactions are primarily mediated by proteins. In proteomic studies, these have been treated as independent entities, disregarding their higher-level organization into complexes that affects their activity and/or function and is thus of great interest for biological research. Here, we describe the implementation of an integrated technique to quantify cell-state-specific changes in the physical arrangement of protein complexes concurrently for thousands of proteins and hundreds of complexes. Applying this technique to a comparison of human cells in interphase and mitosis, we provide a systematic overview of mitotic proteome reorganization. The results recall key hallmarks of mitotic complex remodeling and suggest a model of nuclear pore complex disassembly, which we validate by orthogonal methods. To support the interpretation of quantitative SEC-SWATH-MS datasets, we extend the software CCprofiler and provide an interactive exploration tool, SECexplorer-cc.


Sujet(s)
Mitose/génétique , Protéomique/méthodes , Spectrométrie de masse en tandem/méthodes , Humains
17.
Cancer Cell ; 36(6): 630-644.e9, 2019 12 09.
Article de Anglais | MEDLINE | ID: mdl-31735627

RÉSUMÉ

The chimeric transcription factor TCF3-HLF defines an incurable acute lymphoblastic leukemia subtype. Here we decipher the regulome of endogenous TCF3-HLF and dissect its essential transcriptional components and targets by functional genomics. We demonstrate that TCF3-HLF recruits HLF binding sites at hematopoietic stem cell/myeloid lineage associated (super-) enhancers to drive lineage identity and self-renewal. Among direct targets, hijacking an HLF binding site in a MYC enhancer cluster by TCF3-HLF activates a conserved MYC-driven transformation program crucial for leukemia propagation in vivo. TCF3-HLF pioneers the cooperation with ERG and recruits histone acetyltransferase p300 (EP300), conferring susceptibility to EP300 inhibition. Our study provides a framework for targeting driving transcriptional dependencies in this fatal leukemia.


Sujet(s)
Protéine p300-E1A/génétique , Protéines de fusion oncogènes/génétique , Leucémie-lymphome lymphoblastique à précurseurs B et T/génétique , Facteurs de transcription à motif basique et à glissière à leucines/génétique , Protéines de liaison à l'ADN/génétique , Humains , Translocation génétique
18.
Mol Syst Biol ; 15(8): e8828, 2019 08.
Article de Anglais | MEDLINE | ID: mdl-31464372

RÉSUMÉ

Endothelins (EDN) are peptide hormones that activate a GPCR signalling system and contribute to several diseases, including hypertension and cancer. Current knowledge about EDN signalling is fragmentary, and no systems level understanding is available. We investigated phosphoproteomic changes caused by endothelin B receptor (ENDRB) activation in the melanoma cell lines UACC257 and A2058 and built an integrated model of EDNRB signalling from the phosphoproteomics data. More than 5,000 unique phosphopeptides were quantified. EDN induced quantitative changes in more than 800 phosphopeptides, which were all strictly dependent on EDNRB. Activated kinases were identified based on high confidence EDN target sites and validated by Western blot. The data were combined with prior knowledge to construct the first comprehensive logic model of EDN signalling. Among the kinases predicted by the signalling model, AKT, JNK, PKC and AMP could be functionally linked to EDN-induced cell migration. The model contributes to the system-level understanding of the mechanisms underlying the pleiotropic effects of EDN signalling and supports the rational selection of kinase inhibitors for combination treatments with EDN receptor antagonists.


Sujet(s)
Endothélines/pharmacologie , Régulation de l'expression des gènes tumoraux , Mélanocytes/métabolisme , Phosphoprotéines/génétique , Maturation post-traductionnelle des protéines , Transduction du signal , AMP-Activated Protein Kinases/génétique , AMP-Activated Protein Kinases/métabolisme , Lignée cellulaire tumorale , Mouvement cellulaire/effets des médicaments et des substances chimiques , Endothélines/génétique , Endothélines/métabolisme , Réseaux de régulation génique , Humains , MAP Kinase Kinase 4/génétique , MAP Kinase Kinase 4/métabolisme , Mélanocytes/effets des médicaments et des substances chimiques , Mélanocytes/anatomopathologie , Phosphoprotéines/métabolisme , Phosphorylation , Protéine kinase C/génétique , Protéine kinase C/métabolisme , Protéomique/méthodes , Protéines proto-oncogènes c-akt/génétique , Protéines proto-oncogènes c-akt/métabolisme , Récepteur de l'endothéline de type B/génétique , Récepteur de l'endothéline de type B/métabolisme
19.
Cell Rep ; 27(11): 3315-3330.e7, 2019 06 11.
Article de Anglais | MEDLINE | ID: mdl-31189114

RÉSUMÉ

Deciphering how TCR signals are modulated by coinhibitory receptors is of fundamental and clinical interest. Using quantitative interactomics, we define the composition and dynamics of the PD-1 and BTLA coinhibitory signalosomes in primary effector T cells and at the T cell-antigen-presenting cell interface. We also solve the existing controversy regarding the role of the SHP-1 and SHP-2 protein-tyrosine phosphatases in mediating PD-1 coinhibition. PD-1 predominantly recruits SHP-2, but when absent, it recruits SHP-1 and remains functional. In contrast, BTLA predominantly recruits SHP-1 and to a lesser extent SHP-2. By separately analyzing the PD-1-SHP-1 and PD-1-SHP-2 complexes, we show that both dampen the TCR and CD28 signaling pathways equally. Therefore, our study illustrates how comparison of coinhibitory receptor signaling via quantitative interactomics in primary T cells unveils their extent of redundancy and provides a rationale for designing combinations of blocking antibodies in cancer immunotherapy on the basis of undisputed modes of action.


Sujet(s)
Récepteur-1 de mort cellulaire programmée/métabolisme , Récepteurs immunologiques/métabolisme , Transduction du signal , Lymphocytes T/métabolisme , Animaux , Femelle , Humains , Immunothérapie , Cellules Jurkat , Mâle , Souris , Souris de lignée C57BL , Récepteur-1 de mort cellulaire programmée/génétique , Liaison aux protéines , Protein Tyrosine Phosphatase, Non-Receptor Type 11/métabolisme , Protein Tyrosine Phosphatase, Non-Receptor Type 6/métabolisme , Récepteurs aux antigènes des cellules T/métabolisme
20.
Sci Rep ; 9(1): 7779, 2019 05 23.
Article de Anglais | MEDLINE | ID: mdl-31123282

RÉSUMÉ

Despite the great success of small molecule inhibitors in the treatment of patients with BRAFV600E mutated melanoma, the response to these drugs remains transient and patients eventually relapse within a few months, highlighting the need to develop novel combination therapies based on the understanding of the molecular changes induced by BRAFV600E inhibitors. The acute inhibition of oncogenic signaling can rewire entire cellular signaling pathways and thereby create novel cancer cell vulnerabilities. Here, we demonstrate that inhibition of BRAFV600E oncogenic signaling in melanoma cell lines leads to destabilization of the large subunit of RNA polymerase II POLR2A (polymerase RNA II DNA-directed polypeptide A), thereby preventing its binding to the unconventional prefoldin RPB5 interactor (URI1) chaperone complex and the successful assembly of RNA polymerase II holoenzymes. Furthermore, in melanoma cell lines treated with mitogen-activated protein kinase (MAPK) inhibitors, α-amanitin, a specific and irreversible inhibitor of RNA polymerase II, induced massive apoptosis. Pre-treatment of melanoma cell lines with MAPK inhibitors significantly reduced IC50 values to α-amanitin, creating a state of collateral vulnerability similar to POLR2A hemizygous deletions. Thus, the development of melanoma specific α-amanitin antibody-drug conjugates could represent an interesting therapeutic approach for combination therapies with BRAFV600E inhibitors.


Sujet(s)
alpha-Amanitine/pharmacologie , Mort cellulaire/effets des médicaments et des substances chimiques , Mélanome/métabolisme , Inhibiteurs de la synthèse d'acide nucléique/pharmacologie , Protéines proto-oncogènes B-raf/antagonistes et inhibiteurs , RNA polymerase II/métabolisme , Apoptose/effets des médicaments et des substances chimiques , Lignée cellulaire tumorale , Humains , Transduction du signal/effets des médicaments et des substances chimiques
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE