Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 17 de 17
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.
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
6.
Sci Adv ; 7(29)2021 Jul.
Article de Anglais | MEDLINE | ID: mdl-34261657

RÉSUMÉ

Histone H3K27M is a driving mutation in diffuse intrinsic pontine glioma (DIPG), a deadly pediatric brain tumor. H3K27M reshapes the epigenome through a global inhibition of PRC2 catalytic activity and displacement of H3K27me2/3, promoting oncogenesis of DIPG. As a consequence, a histone modification H3K36me2, antagonistic to H3K27me2/3, is aberrantly elevated. Here, we investigate the role of H3K36me2 in H3K27M-DIPG by tackling its upstream catalyzing enzymes (writers) and downstream binding factors (readers). We determine that NSD1 and NSD2 are the key writers for H3K36me2. Loss of NSD1/2 in H3K27M-DIPG impedes cellular proliferation and tumorigenesis by disrupting tumor-promoting transcriptional programs. Further, we demonstrate that LEDGF and HDGF2 are the main readers mediating the protumorigenic effects downstream of NSD1/2-H3K36me2. Treatment with a chemically modified peptide mimicking endogenous H3K36me2 dislodges LEDGF/HDGF2 from chromatin and specifically inhibits the proliferation of H3K27M-DIPG. Our results indicate a functional pathway of NSD1/2-H3K36me2-LEDGF/HDGF2 as an acquired dependency in H3K27M-DIPG.

7.
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
8.
Int J Hyperthermia ; 37(2): 20-26, 2020 07.
Article de Anglais | MEDLINE | ID: mdl-32672127

RÉSUMÉ

The concept of thermal therapy toward the treatment of brain tumors has gained traction in recent years. Traditionally, thermal therapy has been subdivided into hyperthermia, with mild elevation of temperature in treated tissue above the physiologic baseline; and thermal ablation, where even higher temperatures are achieved. The recent surge in interest has been driven by the use of novel thermal ablation technologies, including laser interstitial thermal therapy (LITT), that are implemented in brain tumor treatment. Here, we review previous scientific literature on the biologic effects of thermal therapy on brain tumors, with an emphasis on glioblastoma (GBM), an aggressive brain malignancy. In addition, we present in vitro evidence from our laboratory that even moderate elevations in temperature achieved in the penumbra around laser-ablated coagulum may also produce GBM cell death. While much remains to be elucidated in terms of the biology of thermal therapy, we propose that it is a welcome addition to the neuro-oncology armamentarium, in particular with regard to GBM, which is generally resistant to current chemoradiotherapeutic regimens.


Sujet(s)
Tumeurs du cerveau , Glioblastome , Hyperthermie provoquée , Thérapie laser , Tumeurs du cerveau/chirurgie , Mort cellulaire , Glioblastome/chirurgie , Humains , Lasers , Imagerie par résonance magnétique , Température , 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.
Clin Cancer Res ; 25(4): 1261-1271, 2019 02 15.
Article de Anglais | MEDLINE | ID: mdl-30397180

RÉSUMÉ

PURPOSE: Isocitrate dehydrogenase (IDH)-mutant glioma is a distinct glioma molecular subtype for which no effective molecularly directed therapy exists. Low-grade gliomas, which are 80%-90% IDH-mutant, have high RNA levels of the cell surface Notch ligand DLL3. We sought to determine DLL3 expression by IHC in glioma molecular subtypes and the potential efficacy of an anti-DLL3 antibody-drug conjugate (ADC), rovalpituzumab tesirine (Rova-T), in IDH-mutant glioma. EXPERIMENTAL DESIGN: We evaluated DLL3 expression by RNA using TCGA data and by IHC in a discovery set of 63 gliomas and 20 nontumor brain tissues and a validation set of 62 known IDH wild-type and mutant gliomas using a monoclonal anti-DLL3 antibody. Genotype was determined using a DNA methylation array classifier or by sequencing. The effect of Rova-T on patient-derived endogenous IDH-mutant glioma tumorspheres was determined by cell viability assay. RESULTS: Compared to IDH wild-type glioblastoma, IDH-mutant gliomas have significantly higher DLL3 RNA (P < 1 × 10-15) and protein by IHC (P = 0.0014 and P < 4.3 × 10-6 in the discovery and validation set, respectively). DLL3 immunostaining was intense and homogeneous in IDH-mutant gliomas, retained in all recurrent tumors, and detected in only 1 of 20 nontumor brains. Patient-derived IDH-mutant glioma tumorspheres overexpressed DLL3 and were potently sensitive to Rova-T in an antigen-dependent manner. CONCLUSIONS: DLL3 is selectively and homogeneously expressed in IDH-mutant gliomas and can be targeted with Rova-T in patient-derived IDH-mutant glioma tumorspheres. Our findings are potentially immediately translatable and have implications for therapeutic strategies that exploit cell surface tumor-associated antigens.


Sujet(s)
Gliome/traitement médicamenteux , Protéines et peptides de signalisation intracellulaire/génétique , Isocitrate dehydrogenases/génétique , Protéines membranaires/génétique , Thérapie moléculaire ciblée , Anticorps monoclonaux humanisés/génétique , Anticorps monoclonaux humanisés/usage thérapeutique , Benzodiazépinones/usage thérapeutique , Encéphale/anatomopathologie , Méthylation de l'ADN/génétique , Femelle , Régulation de l'expression des gènes tumoraux/effets des médicaments et des substances chimiques , Génotype , Gliome/génétique , Gliome/anatomopathologie , Humains , Immunoconjugués/génétique , Immunoconjugués/usage thérapeutique , Ligands , Mâle , Mutation , Récidive tumorale locale/traitement médicamenteux , Récidive tumorale locale/génétique , Récidive tumorale locale/anatomopathologie , ARN/génétique , Récepteurs Notch/génétique
11.
Methods Mol Biol ; 1741: 63-69, 2018.
Article de Anglais | MEDLINE | ID: mdl-29392690

RÉSUMÉ

In vitro propagation of patient-derived glioblastoma (GBM) cells can be achieved either by adherent monolayer culture, already described in Chapter 3 , or by tumorsphere culture in suspension. Here, we provide a detailed protocol for establishing patient-derived tumorsphere cultures. Such cultures are enriched for GBM stem cells (GSCs) and can be used to generate orthotopic tumor xenografts in the brain of immunocompromised mice. We also point out nuances in the protocol that can increase the yield of successful cultures from operative specimens.


Sujet(s)
Techniques de culture cellulaire , Glioblastome/anatomopathologie , Sphéroïdes de cellules , Cellules cancéreuses en culture , Lignée cellulaire tumorale , Humains
12.
Methods Mol Biol ; 1741: 71-79, 2018.
Article de Anglais | MEDLINE | ID: mdl-29392691

RÉSUMÉ

This chapter describes a straightforward method for isolating glioblastoma stem cells (GSCs) from in vitro tissue cultures via fluorescence-activated cell sorting (FACS) using CD133 as a surface marker. The use of a directly conjugated antibody to an APC fluorophore against the CD133 molecule provides sufficient and clear detection of positive cells from the rest of the population. This strategy avoids an unnecessary secondary antibody incubation step thereby minimizing loss and increasing yield. The same protocol can be applied to other GSC surface markers. The described method allows for quick and efficient purification of GSCs, which can then be used in several downstream applications.


Sujet(s)
Séparation cellulaire , Cytométrie en flux , Glioblastome/métabolisme , Glioblastome/anatomopathologie , Cellules souches tumorales/métabolisme , Antigène AC133/métabolisme , Marqueurs biologiques tumoraux , Techniques de culture cellulaire , Lignée cellulaire tumorale , Séparation cellulaire/méthodes , Humains , Cellules souches tumorales/anatomopathologie
13.
Methods Mol Biol ; 1741: 81-89, 2018.
Article de Anglais | MEDLINE | ID: mdl-29392692

RÉSUMÉ

This chapter provides detailed step-by-step instructions for the production of lentiviral particles and the transduction of primary human glioblastoma cultures. Lentiviruses stably transduce both dividing and non-dividing cells, such as quiescent cancer stem cell populations. The viral envelope is pseudotyped with the vesicular stomatitis virus envelope glycoprotein G (VSV-G), which renders the lentiviral particles pantropic, so that they can infect theoretically all cell types. The third generation packaging system used in this protocol produces lentiviruses with important safety features, including replication incompetence and self-inactivation (SIN). The protocol we describe here leads to transduction of primary human glioblastoma cultures with efficiencies of up to 90%.


Sujet(s)
Techniques de transfert de gènes , Vecteurs génétiques/génétique , Lentivirus/génétique , Transduction génétique , Techniques de culture cellulaire , Lignée cellulaire tumorale , Expression des gènes , Glioblastome/génétique , Glioblastome/métabolisme , Cellules HEK293 , Humains , Transgènes
14.
Methods Mol Biol ; 1741: 191-198, 2018.
Article de Anglais | MEDLINE | ID: mdl-29392701

RÉSUMÉ

Orthotopic rodent xenografts are an essential tool for studying glioblastoma in vivo. Xenograft growth as a function of time can only be monitored by noninvasive imaging. This chapter describes in detail how to assess xenograft size and growth using bioluminescent imaging with IVIS (in vivo imaging system). This form of imaging (a) can be performed without the help of a trained technician, (b) is a very quick procedure, allowing simultaneous imaging of up to five animals at a total experimental duration of 15 min, and (c) is cheaper than the alternatives (small animal MRI or CT). This technique relies on the stable expression of luciferase by the xenografted GBM cells. Luciferin, the substrate of luciferase, which is injected into host mice intraperitoneally, distributes throughout the mouse body and crosses the blood brain barrier. Luciferase expressed by the xenografted cells uses this substrate in a catalytic reaction, leading to the emission of visible light, which is detected by the CCD camera of the IVIS imaging system. The intensity of this emitted light correlates to the size of a given xenograft and allows comparisons of xenograft size across different animals, as well as within the same animal across different time points.


Sujet(s)
Tumeurs du cerveau/imagerie diagnostique , Tumeurs du cerveau/anatomopathologie , Glioblastome/imagerie diagnostique , Glioblastome/anatomopathologie , Mesures de luminescence , Imagerie optique , Animaux , Modèles animaux de maladie humaine , Expression des gènes , Gènes rapporteurs , Hétérogreffes , Humains , Traitement d'image par ordinateur , Mesures de luminescence/méthodes , Souris , Imagerie optique/méthodes
15.
Cell Rep ; 21(5): 1267-1280, 2017 Oct 31.
Article de Anglais | MEDLINE | ID: mdl-29091765

RÉSUMÉ

Low-grade astrocytomas (LGAs) carry neomorphic mutations in isocitrate dehydrogenase (IDH) concurrently with P53 and ATRX loss. To model LGA formation, we introduced R132H IDH1, P53 shRNA, and ATRX shRNA into human neural stem cells (NSCs). These oncogenic hits blocked NSC differentiation, increased invasiveness in vivo, and led to a DNA methylation and transcriptional profile resembling IDH1 mutant human LGAs. The differentiation block was caused by transcriptional silencing of the transcription factor SOX2 secondary to disassociation of its promoter from a putative enhancer. This occurred because of reduced binding of the chromatin organizer CTCF to its DNA motifs and disrupted chromatin looping. Our human model of IDH mutant LGA formation implicates impaired NSC differentiation because of repression of SOX2 as an early driver of gliomagenesis.


Sujet(s)
Isocitrate dehydrogenases/génétique , Facteurs de transcription SOX-B1/métabolisme , Protéine p53 suppresseur de tumeur/génétique , Protéine nucléaire liée à l'X/génétique , Animaux , Apoptose , Astrocytome/métabolisme , Astrocytome/anatomopathologie , Tumeurs du cerveau/métabolisme , Tumeurs du cerveau/anatomopathologie , Facteur de liaison à la séquence CCCTC/métabolisme , Différenciation cellulaire , Cellules cultivées , Méthylation de l'ADN , Épigenèse génétique , Humains , Isocitrate dehydrogenases/métabolisme , Souris , Souris SCID , Grading des tumeurs , Invasion tumorale , Cellules souches neurales/cytologie , Cellules souches neurales/métabolisme , Interférence par ARN , Protéine p53 suppresseur de tumeur/antagonistes et inhibiteurs , Protéine p53 suppresseur de tumeur/métabolisme , Protéine nucléaire liée à l'X/antagonistes et inhibiteurs , Protéine nucléaire liée à l'X/métabolisme
16.
Oncotarget ; 8(39): 64932-64953, 2017 Sep 12.
Article de Anglais | MEDLINE | ID: mdl-29029402

RÉSUMÉ

Glioblastoma (GBM) stem cells (GSCs) reside in both hypoxic and vascular microenvironments within tumors. The molecular mechanisms that allow GSCs to occupy such contrasting niches are not understood. We used patient-derived GBM cultures to identify GSC subtypes with differential activation of Notch signaling, which co-exist in tumors but occupy distinct niches and match their metabolism accordingly. Multipotent GSCs with Notch pathway activation reside in perivascular niches, and are unable to entrain anaerobic glycolysis during hypoxia. In contrast, most CD133-expressing GSCs do not depend on canonical Notch signaling, populate tumors regardless of local vascularity and selectively utilize anaerobic glycolysis to expand in hypoxia. Ectopic activation of Notch signaling in CD133-expressing GSCs is sufficient to suppress anaerobic glycolysis and resistance to hypoxia. These findings demonstrate a novel role for Notch signaling in regulating GSC metabolism and suggest intratumoral GSC heterogeneity ensures metabolic adaptations to support tumor growth in diverse tumor microenvironments.

SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE