Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 14 de 14
Filtrer
Plus de filtres










Base de données
Gamme d'année
1.
Cell Rep ; 43(5): 114229, 2024 May 28.
Article de Anglais | MEDLINE | ID: mdl-38758649

RÉSUMÉ

GPR133 (ADGRD1) is an adhesion G-protein-coupled receptor that signals through Gαs/cyclic AMP (cAMP) and is required for the growth of glioblastoma (GBM), an aggressive brain malignancy. The regulation of GPR133 signaling is incompletely understood. Here, we use proximity biotinylation proteomics to identify ESYT1, a Ca2+-dependent mediator of endoplasmic reticulum-plasma membrane bridge formation, as an intracellular interactor of GPR133. ESYT1 knockdown or knockout increases GPR133 signaling, while its overexpression has the opposite effect, without altering GPR133 levels in the plasma membrane. The GPR133-ESYT1 interaction requires the Ca2+-sensing C2C domain of ESYT1. Thapsigargin-mediated increases in cytosolic Ca2+ relieve signaling-suppressive effects of ESYT1 by promoting ESYT1-GPR133 dissociation. ESYT1 knockdown or knockout in GBM slows tumor growth, suggesting tumorigenic functions of ESYT1. Our findings demonstrate a mechanism for the modulation of GPR133 signaling by increased cytosolic Ca2+, which reduces the signaling-suppressive interaction between GPR133 and ESYT1 to raise cAMP levels.


Sujet(s)
Calcium , Glioblastome , Récepteurs couplés aux protéines G , Transduction du signal , Récepteurs couplés aux protéines G/métabolisme , Récepteurs couplés aux protéines G/génétique , Humains , Animaux , Calcium/métabolisme , Glioblastome/métabolisme , Glioblastome/anatomopathologie , Glioblastome/génétique , Souris , AMP cyclique/métabolisme , Lignée cellulaire tumorale , Cellules HEK293 , Liaison aux protéines , Souris nude , Protéines oncogènes
2.
Cell Rep ; 42(11): 113374, 2023 11 28.
Article de Anglais | MEDLINE | ID: mdl-37938973

RÉSUMÉ

Glioblastoma (GBM) is the most common and aggressive primary brain malignancy. Adhesion G protein-coupled receptors (aGPCRs) have attracted interest for their potential as treatment targets. Here, we show that CD97 (ADGRE5) is the most promising aGPCR target in GBM, by virtue of its de novo expression compared to healthy brain tissue. CD97 knockdown or knockout significantly reduces the tumor initiation capacity of patient-derived GBM cultures (PDGCs) in vitro and in vivo. We find that CD97 promotes glycolytic metabolism via the mitogen-activated protein kinase (MAPK) pathway, which depends on phosphorylation of its C terminus and recruitment of ß-arrestin. We also demonstrate that THY1/CD90 is a likely CD97 ligand in GBM. Lastly, we show that an anti-CD97 antibody-drug conjugate selectively kills tumor cells in vitro. Our studies identify CD97 as a regulator of tumor metabolism, elucidate mechanisms of receptor activation and signaling, and provide strong scientific rationale for developing biologics to target it therapeutically in GBM.


Sujet(s)
Glioblastome , Humains , Glioblastome/anatomopathologie , Phosphorylation , Récepteurs couplés aux protéines G/métabolisme , Transduction du signal
3.
Cell Rep ; 42(7): 112679, 2023 07 25.
Article de Anglais | MEDLINE | ID: mdl-37354459

RÉSUMÉ

The adhesion G-protein-coupled receptor GPR133 (ADGRD1) supports growth of the brain malignancy glioblastoma. How the extracellular interactome of GPR133 in glioblastoma modulates signaling remains unknown. Here, we use affinity proteomics to identify the transmembrane protein PTK7 as an extracellular binding partner of GPR133 in glioblastoma. PTK7 binds the autoproteolytically generated N-terminal fragment of GPR133 and its expression in trans increases GPR133 signaling. This effect requires the intramolecular cleavage of GPR133 and PTK7's anchoring in the plasma membrane. PTK7's allosteric action on GPR133 signaling is additive with but topographically distinct from orthosteric activation by soluble peptide mimicking the endogenous tethered Stachel agonist. GPR133 and PTK7 are expressed in adjacent cells in glioblastoma, where their knockdown phenocopies each other. We propose that this ligand-receptor interaction is relevant to the pathogenesis of glioblastoma and possibly other physiological processes in healthy tissues.


Sujet(s)
Glioblastome , Humains , Transduction du signal , Récepteurs couplés aux protéines G/métabolisme , Membrane cellulaire/métabolisme , Régulation allostérique , Ligands , Site allostérique , Molécules d'adhérence cellulaire/métabolisme , Récepteurs à activité tyrosine kinase/métabolisme
4.
bioRxiv ; 2023 Feb 09.
Article de Anglais | MEDLINE | ID: mdl-36798364

RÉSUMÉ

GPR133 (ADGRD1) is an adhesion G protein-coupled receptor that signals through Gαs and is required for growth of glioblastoma (GBM), an aggressive brain malignancy. The regulation of GPR133 signaling is incompletely understood. Here, we use proximity biotinylation proteomics to identify ESYT1, a Ca2+-dependent mediator of endoplasmic reticulum-plasma membrane bridge formation, as an intracellular interactor of GPR133. ESYT1 knockdown or knockout increases GPR133 signaling, while its overexpression has the opposite effect, without altering GPR133 levels in the plasma membrane. The GPR133-ESYT1 interaction requires the Ca2+-sensing C2C domain of ESYT1. Thapsigargin-mediated increases in cytosolic Ca2+ relieve signaling-suppressive effects of ESYT1 by promoting ESYT1-GPR133 dissociation. ESYT1 knockdown or knockout in GBM impairs tumor growth in vitro, suggesting functions of ESYT1 beyond the interaction with GPR133. Our findings suggest a novel mechanism for modulation of GPR133 signaling by increased cytosolic Ca2+, which reduces the signaling-suppressive interaction between GPR133 and ESYT1 to raise cAMP levels.

5.
Front Cell Dev Biol ; 10: 873278, 2022.
Article de Anglais | MEDLINE | ID: mdl-35813217

RÉSUMÉ

The adhesion G protein-coupled receptor (aGPCR) GPR126/ADGRG6 plays an important role in several physiological functions, such as myelination or peripheral nerve repair. This renders the receptor an attractive pharmacological target. GPR126 is a mechano-sensor that translates the binding of extracellular matrix (ECM) molecules to its N terminus into a metabotropic intracellular signal. To date, the structural requirements and the character of the forces needed for this ECM-mediated receptor activation are largely unknown. In this study, we provide this information by combining classic second-messenger detection with single-cell atomic force microscopy. We established a monoclonal antibody targeting the N terminus to stimulate GPR126 and compared it to the activation through its known ECM ligands, collagen IV and laminin 211. As each ligand uses a distinct mode of action, the N terminus can be regarded as an allosteric module that can fine-tune receptor activation in a context-specific manner.

6.
J Biol Chem ; 298(6): 101949, 2022 06.
Article de Anglais | MEDLINE | ID: mdl-35447113

RÉSUMÉ

We recently demonstrated that GPR133 (ADGRD1), an adhesion G protein-coupled receptor involved in raising cytosolic cAMP levels, is necessary for growth of glioblastoma (GBM) and is de novo expressed in GBM relative to normal brain tissue. Our previous work suggested that dissociation of autoproteolytically generated N-terminal and C-terminal fragments of GPR133 at the plasma membrane correlates with receptor activation and signaling. To promote the goal of developing biologics that modulate GPR133 function, we investigated the effects of antibodies against the N-terminus of GPR133 on receptor signaling. Here, we show that treatment of HEK293T cells overexpressing GPR133 with these antibodies increased cAMP levels in a concentration-dependent manner. Analysis of culture medium following antibody treatment further indicated the presence of complexes of these antibodies with the autoproteolytically cleaved N-terminal fragments of GPR133. In addition, cells expressing a cleavage-deficient mutant of GPR133 (H543R) did not respond to antibody stimulation, suggesting that the effect is cleavage dependent. Finally, we demonstrate the antibody-mediated stimulation of WT GPR133, but not the cleavage-deficient H543R mutant, was reproducible in patient-derived GBM cells. These findings provide a paradigm for modulation of GPR133 function with biologics and support the hypothesis that the intramolecular cleavage in the N-terminus modulates receptor activation and signaling.


Sujet(s)
Anticorps , Glioblastome , Récepteurs couplés aux protéines G , Anticorps/métabolisme , Anticorps/pharmacologie , Glioblastome/métabolisme , Cellules HEK293 , Humains , Récepteurs couplés aux protéines G/génétique , Récepteurs couplés aux protéines G/métabolisme , Transduction du signal/effets des médicaments et des substances chimiques
7.
Neurooncol Adv ; 3(1): vdab046, 2021.
Article de Anglais | MEDLINE | ID: mdl-33959717

RÉSUMÉ

BACKGROUND: Members of the adhesion family of G protein-coupled receptors (GPCRs) have received attention for their roles in health and disease, including cancer. Over the past decade, several members of the family have been implicated in the pathogenesis of glioblastoma. METHODS: Here, we discuss the basic biology of adhesion GPCRs and review in detail specific members of the receptor family with known functions in glioblastoma. Finally, we discuss the potential use of adhesion GPCRs as novel treatment targets in neuro-oncology.

8.
J Biol Chem ; 296: 100798, 2021.
Article de Anglais | MEDLINE | ID: mdl-34022221

RÉSUMÉ

GPR133 (ADGRD1), an adhesion G protein-coupled receptor (GPCR) whose canonical signaling activates GαS-mediated generation of cytosolic cAMP, has been shown to be necessary for the growth of glioblastoma (GBM), a brain malignancy. The extracellular N terminus of GPR133 is thought to be autoproteolytically cleaved into N-terminal and C- terminal fragments (NTF and CTF, respectively). However, the role of this cleavage in receptor activation remains unclear. Here, we used subcellular fractionation and immunoprecipitation approaches to show that the WT GPR133 receptor is cleaved shortly after protein synthesis and generates significantly more canonical signaling than an uncleavable point mutant GPR133 (H543R) in patient-derived GBM cultures and HEK293T cells. After cleavage, the resulting NTF and CTF remain noncovalently bound to each other until the receptor is trafficked to the plasma membrane, where we demonstrated NTF-CTF dissociation occurs. Using a fusion of the CTF of GPR133 and the N terminus of thrombin-activated human protease-activated receptor 1 as a controllable proxy system to test the effect of intramolecular cleavage and dissociation, we also showed that thrombin-induced cleavage and shedding of the human protease-activated receptor 1 NTF increased intracellular cAMP levels. These results support a model wherein dissociation of the NTF from the CTF at the plasma membrane promotes GPR133 activation and downstream signaling. These findings add depth to our understanding of the molecular life cycle and mechanism of action of GPR133 and provide critical insights that will inform therapeutic targeting of GPR133 in GBM.


Sujet(s)
Récepteurs couplés aux protéines G/métabolisme , Transduction du signal , AMP cyclique/métabolisme , Glioblastome/métabolisme , Humains , Protéolyse , Récepteurs couplés aux protéines G/composition chimique , Cellules cancéreuses en culture
9.
Neurooncol Adv ; 2(1): vdaa053, 2020.
Article de Anglais | MEDLINE | ID: mdl-32642706

RÉSUMÉ

BACKGROUND: Glioma is a family of primary brain malignancies with limited treatment options and in need of novel therapies. We previously demonstrated that the adhesion G protein-coupled receptor GPR133 (ADGRD1) is necessary for tumor growth in adult glioblastoma, the most advanced malignancy within the glioma family. However, the expression pattern of GPR133 in other types of adult glioma is unknown. METHODS: We used immunohistochemistry in tumor specimens and non-neoplastic cadaveric brain tissue to profile GPR133 expression in adult gliomas. RESULTS: We show that GPR133 expression increases as a function of WHO grade and peaks in glioblastoma, where all tumors ubiquitously express it. Importantly, GPR133 is expressed within the tumor bulk, as well as in the brain-infiltrating tumor margin. Furthermore, GPR133 is expressed in both isocitrate dehydrogenase (IDH) wild-type and mutant gliomas, albeit at higher levels in IDH wild-type tumors. CONCLUSION: The fact that GPR133 is absent from non-neoplastic brain tissue but de novo expressed in glioma suggests that it may be exploited therapeutically.

10.
Nat Commun ; 9(1): 3351, 2018 08 17.
Article de Anglais | MEDLINE | ID: mdl-30120221

RÉSUMÉ

The originally published version of this article contained an error in the name of the author Flóra Gölöncsér, which was incorrectly given as Flóra Göröncsér. This has now been corrected in both the PDF and HTML versions of the article.

11.
Nat Commun ; 9(1): 1354, 2018 04 10.
Article de Anglais | MEDLINE | ID: mdl-29636447

RÉSUMÉ

Two subclasses of acid-sensing ion channels (ASIC3) and of ATP-sensitive P2X receptors (P2X3Rs) show a partially overlapping expression in sensory neurons. Here we report that both recombinant and native receptors interact with each other in multiple ways. Current measurements with the patch-clamp technique prove that ASIC3 stimulation strongly inhibits the P2X3R current partly by a Ca2+-dependent mechanism. The proton-binding site is critical for this effect and the two receptor channels appear to switch their ionic permeabilities during activation. Co-immunoprecipation proves the close association of the two protein structures. BN-PAGE and SDS-PAGE analysis is also best reconciled with the view that ASIC3 and P2X3Rs form a multiprotein structure. Finally, in vivo measurements in rats reveal the summation of pH and purinergically induced pain. In conclusion, the receptor subunits do not appear to form a heteromeric channel, but tightly associate with each other to form a protein complex, mediating unidirectional inhibition.


Sujet(s)
Canaux ioniques sensibles à l'acidité/génétique , Calcium/métabolisme , Ganglions sensitifs des nerfs spinaux/métabolisme , Hyperalgésie/génétique , Douleur/génétique , Protons , Récepteurs purinergiques P2X3/génétique , Canaux ioniques sensibles à l'acidité/métabolisme , Animaux , Animaux nouveau-nés , Cellules CHO , Cricetulus , Ganglions sensitifs des nerfs spinaux/cytologie , Concentration en ions d'hydrogène , Hyperalgésie/métabolisme , Hyperalgésie/anatomopathologie , Ouverture et fermeture des portes des canaux ioniques , Mâle , Ovocytes/cytologie , Ovocytes/métabolisme , Douleur/métabolisme , Douleur/anatomopathologie , Techniques de patch-clamp , Liaison aux protéines , Sous-unités de protéines/génétique , Sous-unités de protéines/métabolisme , Rats , Rat Wistar , Récepteurs purinergiques P2X3/métabolisme , Cellules réceptrices sensorielles/métabolisme , Cellules réceptrices sensorielles/anatomopathologie , Xenopus laevis
12.
Purinergic Signal ; 12(2): 221-33, 2016 06.
Article de Anglais | MEDLINE | ID: mdl-26825305

RÉSUMÉ

P2X3 receptors (P2X3R) are trimeric ATP-gated cation channels involved in sensory neurotransmission and inflammatory pain. We used homology modeling and molecular dynamic simulations of the hP2X3R to identify inter-subunit interactions of residues that are instrumental to elucidate conformational changes associated with gating of the hPX3R. We identified an ionic interaction between E112 and R198 of the head domain and dorsal fin domain, respectively, and E57 and T263 of the lower body domains of adjacent subunits and detected a marked rearrangement of these domains during gating of the hP3X3R. Double-mutant cycle analysis of the inter-subunit residue pairs E112/R198 and E57/T263 revealed significant interaction-free energies. Disulfide locking of the hP2X3R E112C/R198C or the E57C/T263C double cysteine mutants markedly reduced the ATP-induced current responses. The decreased current amplitude following inter-subunit disulfide cross-linking indicates that disulfide locking of the head and dorsal fin domains or at the level of the lower body domains of the hP2X3R prevents the gating-induced conformational rearrangement of the subunits with respect to each other. The distinct reorganization of the subunit interfaces during gating of the hP2X3R is generally consistent with the gating mechanism of other P2XRs. Charge-reversal mutagenesis and methanethiosulfonate (MTS)-modification of substituted cysteines demonstrated that E112 and R198 interact electrostatically. Both disulfide locking and salt bridge breaking of the E112/R198 interaction reduced the hP2X3R function. We conclude that the inter-subunit salt bridge between E112 and R198 of the head and dorsal fin domains, respectively, serves to control the mobility of these domains during agonist-activation of the hP2X3R.


Sujet(s)
Ouverture et fermeture des portes des canaux ioniques/physiologie , Récepteurs purinergiques P2X3/composition chimique , Techniques de knock-down de gènes , Humains , Modèles moléculaires , Conformation moléculaire , Mutagenèse dirigée , Techniques de patch-clamp
13.
Neuropharmacology ; 99: 115-30, 2015 Dec.
Article de Anglais | MEDLINE | ID: mdl-26184350

RÉSUMÉ

The aim of the present work was to clarify whether heterotrimeric P2X2/3 receptors have a fixed subunit stoichiometry consisting of one P2X2 and two P2X3 subunits as previously suggested, or a flexible stoichiometry containing also the inverse subunit composition. For this purpose we transfected HEK293 cells with P2X2 and P2X3 encoding cDNA at the ratios of 1:2 and 4:1, and analysed the biophysical and pharmacological properties of the generated receptors by means of the whole-cell patch-clamp technique. The concentration-response curves for the selective agonist α,ß-meATP did not differ from each other under the two transfection ratios. However, co-expression of an inactive P2X2 mutant and the wild type P2X3 subunit and vice versa resulted in characteristic distortions of the α,ß-meATP concentration-response relationships, depending on which subunit was expressed in excess, suggesting that HEK293 cells express mixtures of (P2X2)1/(P2X3)2 and (P2X2)2/(P2X3)1 receptors. Whereas the allosteric modulators H+ and Zn2+ failed to discriminate between the two possible heterotrimeric receptor variants, the α,ß-meATP-induced responses were blocked more potently by the competitive antagonist A317491, when the P2X2 subunit was expressed in deficit of the P2X3 subunit. Furthermore, blue-native PAGE analysis of P2X2 and P2X3 subunits co-expressed in Xenopus laevis oocytes and HEK293 cells revealed that plasma membrane-bound P2X2/3 receptors appeared in two clearly distinct heterotrimeric complexes: a (P2X2-GFP)2/(P2X3)1 complex and a (P2X2-GFP)1/(P2X3)2 complex. These data strongly indicate that the stoichiometry of the heteromeric P2X2/3 receptor is not fixed, but determined in a permutational manner by the relative availability of P2X2 and P2X3 subunits.


Sujet(s)
Récepteurs purinergiques P2X2/métabolisme , Récepteurs purinergiques P2X3/métabolisme , Adénosine triphosphate/analogues et dérivés , Adénosine triphosphate/pharmacologie , Régulation allostérique , Animaux , Calcium/métabolisme , Cations divalents/métabolisme , Membrane cellulaire/effets des médicaments et des substances chimiques , Membrane cellulaire/physiologie , Relation dose-effet des médicaments , Cellules HEK293 , Humains , Potentiels de membrane/effets des médicaments et des substances chimiques , Potentiels de membrane/physiologie , Mutation , Ovocytes , Techniques de patch-clamp , Phénols/pharmacologie , Composés polycycliques/pharmacologie , Agonistes des récepteurs purinergiques P2X/pharmacologie , Antagonistes des récepteurs purinergiques P2X/pharmacologie , Récepteurs purinergiques P2X2/génétique , Récepteurs purinergiques P2X3/génétique , Transfection , Xenopus laevis
14.
Prax Kinderpsychol Kinderpsychiatr ; 54(1): 59-69, 2005 Jan.
Article de Allemand | MEDLINE | ID: mdl-15730151

RÉSUMÉ

The paper explores the meaning of childrens' pictures as a symbol for growing autonomy. The psychotherapy of a 7-years old turkish boy, who came to treatment with a destroyed identity, will be described. The parents failed to support the boy's development by adequate mothering. The aggressive symptomatic as it emerged at the beginning of the psychotherapy is shown in a shipwreck. During therapy he succeeds to develop age-appropriate autonomy that is shown in a series of drawings.


Sujet(s)
Thérapie par l'art , Fantasme , Individuation , Relations mère-enfant , Psychothérapie analytique , Trouble réactionnel de l'attachement/thérapie , Concept du soi , Symbolisme , Agressivité/psychologie , Enfant , Émigration et immigration , Humains , Événements de vie , Mâle , Ludothérapie , Trouble réactionnel de l'attachement/psychologie , Relations dans la fratrie , Turquie/ethnologie , États-Unis
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE
...