. Landthaler, They were introduced in GSCs using Neon electroporation system according to manufacturer's instructions, pRluc-LC3wt and pRluc-LC3BG120A were a gift from Marja Jaattela, 2008.

. Pma) and . Sigma-aldrich, Z-VRPR-FMK was purchased from Enzo Life Sciences. Q-VD-OPh and tumor necrosis factor-alpha (TNFa) were obtained from R&D Systems. Ionomycin was purchased from Calbiochem. The following primary antibodies were used: NESTIN (Millipore MAB5326), SOX2 (Millipore AB5603), GAPDH (Santa Cruz SC-25778 and SC-32233), TUBULIN (Santa Cruz SC-8035), MALT1 (Santa Cruz SC-46677), EEA1 (BD Bioscience 610456), CTSD (BD Bioscience 610800), PEX1 (BD Bioscience 611719), PECAM (BD Bioscience 557355), TFEB (Bethyl A303-673A), PDI (Abcam ab2792), GM130 (Abcam ab52649), QKI (Atlas HPA019123), CTSD (Atlas HPA063001), ULK1 (Sigma A7481), and FLAG (Sigma F1804). HRP-conjugated secondary antibodies, vol.2, pp.4-5

S. Aits and M. Jaattela, Lysosomal cell death at a glance, J Cell Sci, vol.126, pp.1905-1912, 2013.

E. Arias, H. Koga, A. Diaz, E. Mocholi, B. Patel et al., Lysosomal mTORC2/PHLPP1/Akt regulate chaperone-mediated autophagy, Mol Cell, vol.59, pp.270-284, 2015.

S. Bao, Q. Wu, R. E. Mclendon, Y. Hao, Q. Shi et al., Glioma stem cells promote radioresistance by preferential activation of the DNA damage response, Nature, vol.444, pp.756-760, 2006.

R. C. Bargou, C. Leng, D. Krappmann, F. Emmerich, M. Y. Mapara et al., High-level nuclear NF-kappa B and Oct-2 is a common feature of cultured Hodgkin/Reed-Sternberg cells, Blood, vol.87, pp.4340-4347, 1996.

R. L. Bowman, Q. Wang, A. Carro, R. Verhaak, and M. Squatrito, GlioVis data portal for visualization and analysis of brain tumor expression datasets, Neuro Oncol, vol.19, pp.139-141, 2007.

C. E. Brown, D. Alizadeh, R. Starr, L. Weng, J. R. Wagner et al., Regression of glioblastoma after chimeric antigen receptor T-cell therapy, N Engl J Med, vol.375, pp.2561-2569, 2016.

C. Calabrese, H. Poppleton, M. Kocak, T. L. Hogg, C. Fuller et al., A perivascular niche for brain tumor stem cells, Cancer Cell, vol.11, pp.69-82, 2007.

J. Chen, Y. Li, T. S. Yu, R. M. Mckay, D. K. Burns et al., A restricted cell population propagates glioblastoma growth after chemotherapy, Nature, vol.488, pp.522-526, 2012.

O. L. Chinot, W. Wick, W. Mason, R. Henriksson, F. Saran et al., Bevacizumab plus radiotherapy-temozolomide for newly diagnosed glioblastoma, N Engl J Med, vol.370, pp.709-722, 2014.

R. E. Davis, K. D. Brown, U. Siebenlist, and L. M. Staudt, Constitutive nuclear factor kappaB activity is required for survival of activated B cell-like diffuse large B cell lymphoma cells, J Exp Med, vol.194, pp.1861-1874, 2001.

D. Pilato, M. Kim, E. Y. Cadilha, B. L. Prüßmann, J. N. Nasrallah et al., Targeting the CBM complex causes Treg cells to prime tumours for immune checkpoint therapy, Nature, vol.570, pp.112-116, 2019.

T. Douanne, J. Gavard, and N. Bidere, The paracaspase MALT1 cleaves the LUBAC subunit HOIL1 during antigen receptor signaling, J Cell Sci, vol.129, pp.1775-1780, 2016.
URL : https://hal.archives-ouvertes.fr/inserm-01311283

S. M. Dubois, A. C. Wu, Y. Leclair, H. M. Leveau, C. Schol et al., A catalytic-independent role for the LUBAC in NF-kappaB activation upon antigen receptor engagement and in lymphoma cells, Blood, vol.123, pp.2199-2203, 2014.

, The Authors The EMBO Journal, vol.102030, 2019.

P. Ebner, I. Poetsch, L. Deszcz, T. Hoffman, J. Zuber et al., The IAP family member BRUCE regulates autophagosome-lysosome fusion, Nat Commun, vol.9, p.599, 2018.

A. C. Eitelhuber, O. Vosyka, D. Nagel, M. Bognar, D. Lenze et al., Activity-based probes for detection of active MALT1 paracaspase in immune cells and lymphomas, Chem Biol, vol.22, pp.129-138, 2015.

M. J. Elrick and A. P. Lieberman, Autophagic dysfunction in a lysosomal storage disorder due to impaired proteolysis, Autophagy, vol.9, pp.234-235, 2013.

T. Farkas and M. Jaattela, Chapter one -renilla luciferase-LC3B based reporter assay for measuring autophagic flux, Methods Enzymol, vol.588, pp.1-13, 2017.

C. Fennelly and R. K. Amaravadi, Lysosomal biology in cancer, Methods Mol Biol, vol.1594, pp.293-308, 2017.

E. M. Galan-moya, L. Guelte, A. , L. Fernandes, E. Thirant et al., Secreted factors from brain endothelial cells maintain glioblastoma stem-like cell expansion through the mTOR pathway, EMBO Rep, vol.12, pp.470-476, 2011.

A. C. Gingras, S. G. Kennedy, M. A. O'leary, N. Sonenberg, and N. Hay, 4EBP1, a repressor of mRNA translation, is phosphorylated and inactivated by the AKT (PKB) signaling pathway, Genes Dev, vol.12, pp.502-513, 1998.

K. S. Hamilton, B. Phong, C. Corey, J. Cheng, B. Gorentla et al., T cell receptor-dependent activation of mTOR signaling in T cells is mediated by Carma1 and MALT1, but not Bcl10, Sci Signal, vol.7, p.55, 2014.

S. Hailfinger, G. Lenz, V. Ngo, A. Posvitz-fejfar, F. Rebeaud et al., Essential role of MALT1 protease activity in activated B cell-like diffuse large B-cell lymphoma, Proc Natl Acad Sci, vol.106, pp.19946-19951, 2009.

E. Harford-wright, G. Andre-gregoire, K. A. Jacobs, L. Treps, L. Gonidec et al., Pharmacological targeting of apelin impairs glioblastoma growth, Brain, vol.140, pp.2939-2954, 2017.
URL : https://hal.archives-ouvertes.fr/inserm-01611131

Y. Hu and G. K. Smyth, ELDA: extreme limiting dilution analysis for comparing depleted and enriched populations in stem cell and other assays, J Immunol Methods, vol.347, pp.70-78, 2009.

K. A. Jacobs, E. Harford-wright, and J. Gavard, Neutralizing gp130 interferes with endothelial-mediated effects on glioblastoma stem-like cells, Cell Death Differ, vol.24, p.384, 2017.
URL : https://hal.archives-ouvertes.fr/inserm-01444543

K. M. Jeltsch, D. Hu, S. Brenner, J. Zöller, G. A. Heinz et al., Cleavage of roquin and regnase-1 by the paracaspase MALT1 releases their cooperatively repressed targets to promote TH17 differentiation, Nat Immunol, vol.15, pp.1079-1089, 2014.

R. Jia and J. S. Bonifacino, Lysosome positioning influences mTORC2 and AKT signaling, Mol Cell, vol.75, pp.26-38, 2019.

X. Jin, L. Kim, Q. Wu, L. C. Wallace, B. C. Prager et al., Targeting glioma stem cells through combined BMI1 and EZH2 inhibition, Nat Med, vol.23, pp.1352-1361, 2017.

M. Juilland and M. Thome, Holding all the CARDs: how MALT1 controls CARMA/CARD-dependent signaling, Front Immunol, vol.9, p.1927, 2018.

M. Karin and F. R. Greten, NF-kappaB: linking inflammation and immunity to cancer development and progression, Nat Rev Immunol, vol.5, pp.749-759, 2005.

E. Kip, J. Staal, L. Verstrepen, H. G. Tima, S. Terryn et al., MALT1 controls attenuated rabies virus by inducing early inflammation and T cell activation in the brain, J Virol, vol.92, pp.2029-2046, 2018.

V. I. Korolchuk, S. Saiki, M. Lichtenberg, F. H. Siddiqi, E. A. Roberts et al., Lysosomal positioning coordinates cellular nutrient responses, Nat Cell Biol, vol.13, pp.453-460, 2011.

C. Korth, B. C. May, F. E. Cohen, and S. B. Prusiner, Acridine and phenothiazine derivatives as pharmacotherapeutics for prion disease, Proc Natl Acad Sci, vol.98, pp.9836-9841, 2001.

M. Landthaler, D. Gaidatzis, A. Rothballer, P. Y. Chen, S. J. Soll et al., Molecular characterization of human Argonaute-containing ribonucleoprotein complexes and their bound target mRNAs, RNA, vol.14, pp.2580-2596, 2008.

J. D. Lathia, S. C. Mack, E. E. Mulkearns-hubert, C. L. Valentim, and J. N. Rich, Cancer stem cells in glioblastoma, Genes Dev, vol.29, pp.1203-1217, 2015.

L. Joncour, V. Filppu, P. Hyvonen, M. Holopainen, M. Turunen et al., Vulnerability of invasive glioblastoma cells to lysosomal membrane destabilization, EMBO Mol Med, vol.11, p.9034, 2019.

J. Li, H. Jia, L. Xie, X. Wang, X. Wang et al., Association of constitutive nuclear factor-kappaB activation with aggressive aspects and poor prognosis in cervical cancer, Int J Gynecol Cancer, vol.19, pp.1421-1426, 2009.

W. Li, C. Wu, N. Chen, H. Gu, Y. A. Cao et al., PI3K/Akt/ mTOR signaling pathway and targeted therapy for glioblastoma, Oncotarget, vol.7, pp.33440-33450, 2016.

J. L. Lomas, Treatment of schizophrenia, Br Med J, vol.2, pp.78-80, 1957.

B. Loos, A. Du-toit, and J. Hofmeyr, Defining and measuring autophagosome flux-concept and reality, Autophagy, vol.10, pp.2087-2096, 2014.

J. Luo, N. L. Solimini, and S. J. Elledge, Principles of cancer therapy: oncogene and non-oncogene addiction, Cell, vol.138, p.807, 2009.

T. T. Mai, A. Hamai, A. Hienzsch, T. Caneque, S. Müller et al., Salinomycin kills cancer stem cells by sequestering iron in lysosomes, Nat Chem, vol.10, pp.1025-1033, 2017.
URL : https://hal.archives-ouvertes.fr/hal-01787681

J. Man, X. Yu, H. Huang, W. Zhou, C. Xiang et al., Hypoxic induction of vasorin regulates Notch1 turnover to maintain glioma stem-like cells, Cell Stem Cell, vol.22, pp.104-118, 2017.

J. R. Mcauley, K. M. Bailey, P. Ekambaram, L. R. Klei, H. Kang et al., MALT1 is a critical mediator of PAR1-driven NF-jB activation and metastasis in multiple tumor types, 2019.

, Oncogene

C. Mcguire, L. Elton, P. Wieghofer, J. Staal, S. Voet et al., Pharmacological inhibition of MALT1 protease activity protects mice in a mouse model of multiple sclerosis, J Neuroinflammation, vol.11, p.124, 2014.

L. Meloni, L. Verstrepen, M. Kreike, J. Staal, Y. Driege et al., Mepazine inhibits RANK-induced osteoclastogenesis independent of its MALT1 inhibitory function, Beyaert R, vol.23, p.3144, 2018.

D. Nagel, S. Spranger, M. Vincendeau, M. Grau, S. Raffegerst et al., Pharmacologic inhibition of MALT1 protease by phenothiazines as a therapeutic approach for the treatment of aggressive ABC-DLBCL, Cancer Cell, vol.22, pp.825-837, 2012.

M. Nakaya, Y. Xiao, X. Zhou, J. H. Chang, M. Chang et al., Inflammatory T cell responses rely on amino acid transporter ASCT2 facilitation of glutamine uptake and mTORC1 kinase activation, Immunity, vol.40, pp.692-705, 2014.

V. N. Ngo, R. E. Davis, L. Lamy, X. Yu, H. Zhao et al., A loss-of-function RNA interference screen for molecular targets in cancer, Nature, vol.441, pp.106-110, 2006.

C. R. Oliva, W. Zhang, C. Langford, M. J. Suto, and C. E. Griguer, Repositioning chlorpromazine for treating chemoresistant glioma through the inhibition of cytochrome c oxidase bearing the COX4-1 regulatory subunit, Oncotarget, vol.8, pp.37568-37583, 2017.

A. Pietras, A. M. Katz, E. J. Ekström, B. Wee, J. J. Halliday et al., Osteopontin-CD44 signaling in the glioma perivascular niche enhances cancer stem cell phenotypes and promotes aggressive tumor growth, Cell Stem Cell, vol.14, pp.357-369, 2014.

X. Qin, B. Jiang, and Y. Zhang, 4E-BP1, a multifactor regulated multifunctional protein, Cell Cycle, vol.15, pp.781-786, 2016.

S. Rahim, A. Dirkse, A. Oudin, A. Schuster, J. Bohler et al., Regulation of hypoxiainduced autophagy in glioblastoma involves ATG9A, Br J Cancer, vol.117, pp.813-825, 2017.

F. Rebeaud, S. Hailfinger, A. Posevitz-fejfar, M. Tapernoux, R. Moser et al., The proteolytic activity of the paracaspase MALT1 is key in T cell activation, Nat Immunol, vol.9, pp.272-281, 2008.

M. Rosenbaum, A. Gewies, K. Pechloff, C. Heuser, T. Engleitner et al., Bcl10-controlled Malt1 paracaspase activity is key for the immune suppressive function of regulatory T cells, Nat Commun, vol.10, p.2352, 2019.

M. Sardiello, M. Palmieri, A. Di-ronza, D. L. Medina, M. Valenza et al., A gene network regulating lysosomal biogenesis and function, Science, vol.325, pp.473-477, 2009.

F. Schlauderer, K. Lammens, D. Nagel, M. Vincendeau, A. C. Eitelhuber et al., Structural analysis of phenothiazine derivatives as allosteric inhibitors of the MALT1 paracaspase, Angew Chem Int Ed Engl, vol.52, pp.10384-10387, 2013.

C. Settembre, R. Zoncu, D. L. Medina, F. Vetrini, S. Erdin et al., A lysosome-to-nucleus signalling mechanism senses and regulates the lysosome via mTOR and TFEB, EMBO J, vol.31, pp.1095-1108, 2012.

K. Shchors, A. Massaras, and D. Hanahan, Dual targeting of autophagic regulatory circuitry in gliomas with repurposed drugs elicits cell-lethal autophagy and therapeutic benefit, Cancer Cell, vol.28, pp.456-471, 2015.

T. Shingu, A. L. Ho, L. Yuan, X. Zhou, C. Dai et al., Qki deficiency maintains stemness of glioma stem cells in suboptimal environment by downregulating endolysosomal degradation, Nat Genet, vol.49, pp.75-86, 2016.

S. K. Singh, C. Hawkins, I. D. Clarke, J. A. Squire, J. Bayani et al., Identification of human brain tumour initiating cells, Nature, vol.432, pp.396-401, 2004.

J. Staal, Y. Driege, T. Bekaert, A. Demeyer, D. Muyllaert et al., T-cell receptor-induced JNK activation requires proteolytic inactivation of CYLD by MALT1: CYLD is cleaved by MALT1, 2011.

, EMBO J, vol.30, pp.1742-1752

L. M. Staudt, Oncogenic activation of NF-kappaB, Cold Spring Harb Perspect Biol, vol.2, p.109, 2010.

R. Stupp, M. E. Hegi, W. P. Mason, M. J. Van-den-bent, M. J. Taphoorn et al., Effects of radiotherapy with concomitant and adjuvant temozolomide versus radiotherapy alone on survival in glioblastoma in a randomised phase III study: 5-year analysis of the EORTC-NCIC trial, Lancet Oncol, vol.10, pp.459-466, 2009.

R. Stupp, S. Taillibert, A. A. Kanner, S. Kesari, D. M. Steinberg et al., Maintenance therapy with tumor-treating fields plus temozolomide vs temozolomide alone for glioblastoma: a randomized clinical trial, JAMA, vol.314, pp.2535-2543, 2015.

S. K. Tan, A. Jermakowicz, K. Mookhtiar-adnan, C. B. Nemeroff, S. C. Schurer et al., Drug repositioning in glioblastoma: a pathway perspective, Front Pharmacol, vol.9, p.218, 2018.

A. Thys, T. Douanne, and N. Bidere, Post-translational modifications of the CARMA1-BCL10-MALT1 complex in lymphocytes and activated B-cell like subtype of diffuse large B-cell lymphoma, Front Oncol, vol.8, p.498, 2018.
URL : https://hal.archives-ouvertes.fr/inserm-01908802

L. Treps, S. Edmond, E. Harford-wright, E. M. Galan-moya, A. Schmitt et al., Extracellular vesicletransported Semaphorin3A promotes vascular permeability in glioblastoma, Oncogene, vol.35, pp.2615-2623, 2016.
URL : https://hal.archives-ouvertes.fr/inserm-01406334

V. Tropepe, M. Sibilia, B. G. Ciruna, J. Rossant, E. F. Wagner et al., Distinct neural stem cells proliferate in response to EGF and FGF in the developing mouse telencephalon, Dev Biol, vol.208, pp.166-188, 1999.

T. Uehata, H. Iwasaki, A. Vandenbon, K. Matsushita, E. Hernandez-cuellar et al., Malt1-induced cleavage of Regnase-1 in CD4 + helper T cells regulates immune activation, Cell, vol.153, pp.1036-1049, 2013.

F. Wang, A. Salvati, and P. Boya, Lysosome-dependent cell death and deregulated autophagy induced by amine-modified polystyrene nanoparticles, Open Biol, vol.8, p.170271, 2018.

X. Wang, B. C. Prager, Q. Wu, L. Kim, R. C. Gimple et al., Reciprocal signaling between glioblastoma stem cells and differentiated tumor cells promotes malignant progression, Cell Stem Cell, vol.22, pp.514-528, 2018.

P. Weyerhäuser, S. R. Kantelhardt, and E. L. Kim, Re-purposing chloroquine for glioblastoma: potential merits and confounding variables, Front Oncol, vol.8, p.335, 2018.

J. Wu, L. Zhou, K. Tonissen, R. Tee, and K. Artzt, The quaking 1-5 protein (QKI-5) has a novel nuclear localization signal and shuttles between the nucleus and the cytoplasm, J Biol Chem, vol.274, pp.29202-29210, 1999.

K. Yan, K. Yang, and J. N. Rich, The evolving landscape of glioblastoma stem cells, Curr Opin Neurol, vol.26, pp.701-707, 2013.

L. Yu, C. K. Mcphee, L. Zheng, G. A. Mardones, Y. Rong et al., Termination of autophagy and reformation of lysosomes regulated by mTOR, Nature, vol.465, pp.942-946, 2010.

S. Zielke, N. Meyer, M. Mari, K. Abou-el-ardat, F. Reggiori et al., Loperamide, pimozide, and STF-62247 trigger autophagy-dependent cell death in glioblastoma cells, The Authors The EMBO Journal, vol.9, 2018.