Y. C. Hsu, S. L. Chen, D. Y. Wang, and I. M. Chiu, Stem cell-based therapy in neural repair, Biomed J, vol.36, issue.3, pp.98-105, 2013.

F. J. Muller, E. Y. Snyder, and J. F. Loring, Gene therapy: can neural stem cells deliver?, Nat Rev Neurosci, vol.7, issue.1, pp.75-84, 2006.

L. Mazzini, M. Gelati, D. C. Profico, G. Sgaravizzi, P. Pensi et al., Human neural stem cell transplantation in ALS: initial results from a phase I trial, J Transl Med, vol.13, p.4359401, 2015.

A. Nkan, Stem cells in neurological disorders, Stem Cells in clinic and research, 2011.

A. Pino, G. Fumagalli, F. Bifari, and I. Decimo, New neurons in adult brain: distribution, molecular mechanisms and therapies, Biochemical pharmacology, vol.141, pp.4-22, 2017.

S. Grade and M. Gotz, Neuronal replacement therapy: previous achievements and challenges ahead, NPJ Regen Med, vol.2, p.5677983, 2017.

A. R. Maldonado-soto, D. H. Oakley, H. Wichterle, J. Stein, F. K. Doetsch et al., Stem cells in the nervous system, Am J Phys Med Rehabil, vol.93, issue.11, p.4197112, 2014.

D. J. Guillaume, S. L. Huhn, N. R. Selden, and R. D. Steiner, Cellular therapy for childhood neurodegenerative disease. Part I: rationale and preclinical studies, Neurosurg Focus, vol.24, issue.3-4, 2008.

A. Bocquet, R. Berges, R. Frank, P. Robert, A. C. Peterson et al., Neurofilaments bind tubulin and modulate its polymerization, J Neurosci, vol.29, issue.35, pp.11043-54, 2009.

R. Berges, J. Balzeau, M. Takahashi, C. Prevost, and J. Eyer, Structure-function analysis of the glioma targeting NFL-TBS.40-63 peptide corresponding to the tubulin-binding site on the light neurofilament subunit, PLoS One, vol.7, issue.11, p.49436, 2012.

P. Central and P. , , p.3494675

C. Fressinaud and J. Eyer, Neurofilament-tubulin binding site peptide NFL-TBS.40-63 increases the differentiation of oligodendrocytes in vitro and partially prevents them from lysophosphatidyl choline toxiciy, J Neurosci Res, vol.92, issue.2, pp.243-53, 2014.

C. Lepinoux-chambaud, K. Barreau, and J. Eyer, The Neurofilament-Derived Peptide NFL-TBS.40-63 Targets Neural Stem Cells and Affects Their Properties, Stem Cells Transl Med, vol.5, issue.7, p.4922845, 2016.
URL : https://hal.archives-ouvertes.fr/hal-01412546

D. Brnic, V. Stevanovic, M. Cochet, C. Agier, J. Richardson et al., Borna disease virus infects human neural progenitor cells and impairs neurogenesis, J Virol, vol.86, issue.5, p.3302287, 2012.

Y. Hu, A. C. Hung, H. Cui, E. Dawkins, M. Bolos et al., Role of cystatin C in amyloid precursor protein-induced proliferation of neural stem/progenitor cells, J Biol Chem, vol.288, issue.26, p.3696661, 2013.

G. Narayanan, Y. H. Yu, M. Tham, H. T. Gan, S. Ramasamy et al., Enumeration of Neural Stem Cells Using Clonal Assays, J Vis Exp, issue.116, 2016.

P. Central and P. , , p.5092163

K. J. Livak and T. D. Schmittgen, Analysis of relative gene expression data using real-time quantitative PCR and the 2(-Delta Delta C(T)) Method, Methods, vol.25, issue.4, pp.402-410, 2001.

Z. Guo, H. Peng, J. Kang, and D. Sun, Cell-penetrating peptides: Possible transduction mechanisms and therapeutic applications, Biomed Rep, vol.4, issue.5, p.4840506, 2016.

G. Drin, S. Cottin, E. Blanc, A. R. Rees, and J. Temsamani, Studies on the internalization mechanism of cationic cell-penetrating peptides, J Biol Chem, vol.278, issue.33, pp.31192-201, 2003.

D. Vercauteren, R. E. Vandenbroucke, A. T. Jones, J. Rejman, J. Demeester et al., The use of inhibitors to study endocytic pathways of gene carriers: optimization and pitfalls, Mol Ther, vol.18, issue.3, p.2839427, 2010.

C. L. Chen, W. H. Hou, I. H. Liu, G. Hsiao, S. S. Huang et al., Inhibitors of clathrin-dependent endocytosis enhance TGFbeta signaling and responses, J Cell Sci, vol.122, p.2684837, 2009.

A. C. Boquest, B. N. Day, and R. S. Prather, Flow cytometric cell cycle analysis of cultured porcine fetal fibroblast cells, Biology of reproduction, vol.60, issue.4, pp.1013-1022, 1999.

R. Berges, J. Balzeau, A. C. Peterson, and J. Eyer, A tubulin binding peptide targets glioma cells disrupting their microtubules, blocking migration, and inducing apoptosis, Mol Ther, vol.20, issue.7, p.3392973, 2012.

C. Lepinoux-chambaud and J. Eyer, The NFL-TBS.40-63 anti-glioblastoma peptide enters selectively in glioma cells by endocytosis, Int J Pharm, vol.454, issue.2, pp.738-785, 2013.
URL : https://hal.archives-ouvertes.fr/hal-01412546

C. Fressinaud, J. Eyer, and N. Neurofilaments, 40-63 peptide penetrate oligodendrocytes through clathrin-dependent endocytosis to promote their growth and survival in vitro, Neuroscience, vol.298, pp.42-51, 2015.
URL : https://hal.archives-ouvertes.fr/hal-01392420

C. Bechara and S. Sagan, Cell-penetrating peptides: 20 years later, where do we stand?, FEBS Lett, vol.587, issue.12, pp.1693-702, 2013.

G. C. , Cell proliferation in development and differentiation, Sinauer Asociates, 2000.

I. Ajioka, Coordination of proliferation and neuronal differentiation by the retinoblastoma protein family, Dev Growth Differ, vol.56, issue.5, pp.324-358, 2014.

M. Guizzetti, T. J. Kavanagh, and L. G. Costa, Measurements of astrocyte proliferation, Methods Mol Biol, vol.758, pp.349-59, 2011.

G. Margolis, E. Hertzberg-bigelman, R. Levy, J. Ben-shoshan, K. G. Entin-meer et al., Differential Effects of Colchicine on Cardiac Cell Viability in an in vitro Model Simulating Myocardial Infarction, Cardiology, vol.134, issue.1, pp.57-64, 2016.

D. B. Bosco, R. Kenworthy, D. A. Zorio, and Q. X. Sang, Human mesenchymal stem cells are resistant to Paclitaxel by adopting a non-proliferative fibroblastic state, PLoS One, vol.10, issue.6, p.4452335, 2015.

Y. Laurin, P. Savarin, C. H. Robert, M. Takahashi, J. Eyer et al., Investigating the Structural Variability and Binding Modes of the Glioma Targeting NFL-TBS.40-63 Peptide on Tubulin, Biochemistry, vol.54, issue.23, pp.3660-3669, 2015.
URL : https://hal.archives-ouvertes.fr/hal-01451181

Y. Laurin, J. Eyer, C. H. Robert, C. Prevost, and S. Sacquin-mora, Mobility and Core-Protein Binding Patterns of Disordered C-Terminal Tails in beta-Tubulin Isotypes, Biochemistry, vol.56, issue.12, pp.1746-56, 2017.

L. Leskela, S. Landa, I. Montero-conde, C. Lopez-jimenez, E. Leton et al., Tumoral and tissue-specific expression of the major human beta-tubulin isotypes, Cytoskeleton (Hoboken), vol.67, issue.4, pp.214-237, 2010.

Y. Nakamura, M. Yamamoto, O. E. Yamamoto, A. Kanemura, Y. Hara et al., Expression of tubulin beta II in neural stem/progenitor cells and radial fibers during human fetal brain development, Lab Invest, vol.83, issue.4, pp.479-89, 2003.

C. D. Katsetos, E. Draberova, B. Smejkalova, G. Reddy, L. Bertrand et al., Class III beta-tubulin and gamma-tubulin are co-expressed and form complexes in human glioblastoma cells, Neurochemical research, vol.32, issue.8, p.17406983, 2007.

P. O. Guichet, S. Guelfi, M. Teigell, L. Hoppe, N. Bakalara et al., Notch1 stimulation induces a vascularization switch with pericyte-like cell differentiation of glioblastoma stem cells, Stem Cells, vol.33, issue.1, pp.21-34, 2015.
URL : https://hal.archives-ouvertes.fr/hal-02278893

E. Pastrana, V. Silva-vargas, and F. Doetsch, Eyes wide open: a critical review of sphere-formation as an assay for stem cells, Cell Stem Cell, vol.8, issue.5, p.3633588, 2011.

R. Rivalin, C. Lepinoux-chambaud, J. Eyer, and F. Savagner, The NFL-TBS.40-63 anti-glioblastoma peptide disrupts microtubule and mitochondrial networks in the T98G glioma cell line, PLoS One, vol.9, issue.6, p.4045719, 2014.

K. Birsoy, T. Wang, W. W. Chen, E. Freinkman, M. Abu-remaileh et al., An Essential Role of the Mitochondrial Electron Transport Chain in Cell Proliferation Is to Enable Aspartate Synthesis, Cell, vol.162, issue.3, p.4522279, 2015.

D. Wang, A. Villasante, S. A. Lewis, and N. J. Cowan, The mammalian beta-tubulin repertoire: hematopoietic expression of a novel, heterologous beta-tubulin isotype, The Journal of Cell Biology, vol.103, issue.5, pp.1903-1913, 1986.

M. C. Geloso, V. Corvino, D. Maria, V. Marchese, E. Michetti et al., Cellular targets for neuropeptide Y-mediated control of adult neurogenesis, Front Cell Neurosci, vol.9, p.4360818, 2015.

Y. Itokazu and R. K. Yu, Amyloid beta-peptide 1-42 modulates the proliferation of mouse neural stem cells: upregulation of fucosyltransferase IX and notch signaling, Mol Neurobiol, vol.50, issue.1, p.4102671, 2014.

S. Remaud, S. A. Lopez-juarez, A. L. Bolcato-bellemin, P. Neuberg, F. Stock et al., Inhibition of Sox2 Expression in the Adult Neural Stem Cell Niche In Vivo by Monocationic-based siRNA Delivery

, Mol Ther Nucleic Acids, vol.2, p.3650249, 2013.

J. Zhang and J. Jiao, Molecular Biomarkers for Embryonic and Adult Neural Stem Cell and Neurogenesis, Biomed Res Int, p.4569757, 2015.

L. E. Alvarez-palazuelos, M. S. Robles-cervantes, G. Castillo-velazquez, M. Rivas-souza, J. Guzman-muniz et al., Regulation of neural stem cell in the human SVZ by trophic and morphogenic factors, Curr Signal Transduct Ther, vol.6, issue.3, p.3204663, 2011.

M. Moreno-estelles, P. Gonzalez-gomez, R. Hortiguela, M. Diaz-moreno, S. Emeterio et al., Symmetric expansion of neural stem cells from the adult olfactory bulb is driven by astrocytes via WNT7A, Stem Cells, vol.30, issue.12, pp.2796-809, 2012.

L. E. Oikari, R. K. Okolicsanyi, L. R. Griffiths, and L. M. Haupt, Data defining markers of human neural stem cell lineage potential, Data Brief, vol.7, p.4773572, 2016.

J. Balzeau, M. Pinier, R. Berges, P. Saulnier, J. P. Benoit et al., The effect of functionalizing lipid nanocapsules with NFL-TBS.40-63 peptide on their uptake by glioblastoma cells, Biomaterials, vol.34, issue.13, pp.3381-3390, 2013.

D. Carradori, P. Saulnier, V. Preat, A. Des-rieux, and J. Eyer, NFL-lipid nanocapsules for brain neural stem cell targeting in vitro and in vivo, J Control Release, vol.238, pp.253-62, 2016.
URL : https://hal.archives-ouvertes.fr/hal-01392425