A. Armulik, G. Genove, and C. Betsholtz, Pericytes: Developmental, Physiological, and Pathological Perspectives, Problems, and Promises, Developmental Cell, vol.21, issue.2, pp.193-215, 2011.
DOI : 10.1016/j.devcel.2011.07.001

. Tuberin, the tuberous sclerosis complex 2 tumor suppressor gene product, regulates Rho activation, cell adhesion and migration, Oncogene, vol.21, pp.8470-8476

E. N. Atochina-vasserman, D. A. Goncharov, A. V. Volgina, M. Milavec, M. L. James et al., Statins in lymphangioleiomyomatosis. Simvastatin and atorvastatin induce differential effects on tuberous sclerosis complex 2-null cell growth and signaling. American journal of respiratory cell, pp.704-709, 2013.

B. C. Grabiner, E. D. Spear, S. L. Carter, M. Meyerson, and D. M. Sabatini, A Tumor suppressor complex with GAP activity for the Rag GTPases that signal amino acid sufficiency to mTORC1, Science, vol.340, pp.1100-1106, 2013.

L. Bar-peled, L. D. Schweitzer, R. Zoncu, and D. M. Sabatini, Ragulator Is a GEF for the Rag GTPases that Signal Amino Acid Levels to mTORC1, Cell, vol.150, issue.6, 2012.
DOI : 10.1016/j.cell.2012.07.032

G. Benvenuto, S. Li, S. J. Brown, R. Braverman, W. C. Vass et al., The tuberous, J.E, 2000.

J. Brugarolas, K. Lei, R. L. Hurley, B. D. Manning, J. H. Reiling et al., Regulation of mTOR function in response to hypoxia by REDD1 and the TSC1, 2004.

M. P. Byfield, J. T. Murray, and J. M. Backer, hVps34 Is a Nutrient-regulated Lipid Kinase Required for Activation of p70 S6 Kinase, Journal of Biological Chemistry, vol.280, issue.38, pp.33076-33082, 2005.
DOI : 10.1074/jbc.M507201200

S. L. Cai, A. R. Tee, J. D. Short, J. M. Bergeron, J. Kim et al., Activity of TSC2 is inhibited by AKT-mediated phosphorylation and membrane partitioning, The Journal of Cell Biology, vol.58, issue.2, pp.279-289, 2006.
DOI : 10.1038/ncb999

F. D. Camargo, S. Gokhale, J. B. Johnnidis, D. Fu, G. W. Bell et al., YAP1 increases organ size and expands undifferentiated progenitor cells, pp.2054-2060, 2007.
DOI : 10.1016/j.cub.2007.11.016

URL : http://doi.org/10.1016/j.cub.2007.11.016

T. Carsillo, A. Astrinidis, and E. P. Henske, Mutations in the tuberous sclerosis complex gene TSC2 are a cause of sporadic pulmonary lymphangioleiomyomatosis, Proceedings of the National Academy of Sciences, vol.97, issue.11, pp.6085-6090, 2000.
DOI : 10.1073/pnas.97.11.6085

H. I. Chen, A. Einbond, S. J. Kwak, H. Linn, E. Koepf et al., Characterization of the WW Domain of Human Yes-associated Protein and Its Polyproline-containing Ligands, Journal of Biological Chemistry, vol.272, issue.27, pp.17070-17077, 1997.
DOI : 10.1074/jbc.272.27.17070

P. B. Crino, K. L. Nathanson, and E. P. Henske, The Tuberous Sclerosis Complex, New England Journal of Medicine, vol.355, issue.13, pp.1345-1356, 2006.
DOI : 10.1056/NEJMra055323

C. Demetriades, N. Doumpas, and A. A. Teleman, Regulation of TORC1 in Response to Amino Acid Starvation via Lysosomal Recruitment of TSC2, Cell, vol.156, issue.4, pp.786-799, 2014.
DOI : 10.1016/j.cell.2014.01.024

C. C. Dibble, W. Elis, S. Menon, W. Qin, J. Klekota et al., TBC1D7 Is a Third Subunit of the TSC1-TSC2 Complex Upstream of mTORC1, Molecular Cell, vol.47, issue.4, pp.535-546, 2012.
DOI : 10.1016/j.molcel.2012.06.009

M. F. Anders, R. A. Maitra, A. Pan, and D. , Elucidation of a universal sizecontrol mechanism in Drosophila and mammals, Cell, vol.130, pp.1120-1133, 2007.

S. Dupont, L. Morsut, M. Aragona, E. Enzo, S. Giulitti et al., Role of YAP/TAZ in mechanotransduction, Nature, vol.13, issue.7350, pp.179-183, 2011.
DOI : 10.1038/nature10137

N. El-hashemite, H. Zhang, E. P. Henske, and D. J. Kwiatkowski, Mutation in TSC2 and activation of mammalian target of rapamycin signalling pathway in renal angiomyolipoma, The Lancet, vol.361, issue.9366, pp.1348-1349, 2003.
DOI : 10.1016/S0140-6736(03)13044-9

R. Fan, N. G. Kim, and B. M. Gumbiner, Regulation of Hippo pathway by mitogenic growth factors via phosphoinositide 3-kinase and phosphoinositidedependent kinase-1, Proceedings of the National Academy of Sciences of the United States of America, pp.2569-2574, 2013.

B. G. Fernandez, P. Gaspar, C. Bras-pereira, B. Jezowska, S. R. Rebelo et al., Actin-Capping Protein and the Hippo pathway regulate F-actin and tissue growth in Drosophila, Development, vol.138, issue.11, pp.2337-2346, 2011.
DOI : 10.1242/dev.063545

G. M. Findlay, L. Yan, J. Procter, V. Mieulet, and R. F. Lamb, is a nutrient-sensitive regulator of mTOR signalling, Biochemical Journal, vol.403, issue.1, pp.13-20, 2007.
DOI : 10.1042/BJ20061881

URL : https://hal.archives-ouvertes.fr/hal-00478723

D. N. Franz, Everolimus in the treatment of subependymal giant cell astrocytomas, angiomyolipomas, and pulmonary and skin lesions associated with tuberous sclerosis complex, Biologics: Targets and Therapy, vol.7, pp.211-221, 2013.
DOI : 10.2147/BTT.S25095

N. J. Ganem, H. Cornils, S. Y. Chiu, K. P. O-'rourke, J. Arnaud et al., Cytokinesis Failure Triggers Hippo Tumor Suppressor Pathway Activation, Cell, vol.158, issue.4, pp.833-848, 2014.
DOI : 10.1016/j.cell.2014.06.029

URL : https://hal.archives-ouvertes.fr/hal-01085073

X. Gao, T. P. Neufeld, and D. Pan, Drosophila PTEN Regulates Cell Growth and Proliferation through PI3K-Dependent and -Independent Pathways, Developmental Biology, vol.221, issue.2, pp.404-418, 2000.
DOI : 10.1006/dbio.2000.9680

URL : http://doi.org/10.1006/dbio.2000.9680

X. Gao and D. Pan, TSC1 and TSC2 tumor suppressors antagonize insulin signaling in cell growth, Genes & Development, vol.15, issue.11, pp.1383-1392, 2001.
DOI : 10.1101/gad.901101

X. Gao, Y. Zhang, P. Arrazola, O. Hino, T. Kobayashi et al., Tsc tumour suppressor proteins antagonize amino-acid???TOR signalling, Nature Cell Biology, vol.4, issue.9, pp.699-704, 2002.
DOI : 10.1038/ncb847

D. Nardo, A. Bronson, R. T. Chan, and J. A. , Regulable neural progenitorspecific Tsc1 loss yields giant cells with organellar dysfunction in a model of tuberous sclerosis complex, Proceedings of the National Academy of Sciences of the United States of America, pp.1070-1079, 2011.

G. Halder, J. , and R. L. , Hippo signaling: growth control and beyond, Development, vol.138, issue.1, pp.9-22, 2011.
DOI : 10.1242/dev.045500

URL : http://www.ncbi.nlm.nih.gov/pmc/articles/PMC2998162

Z. Hossain, S. M. Ali, H. L. Ko, J. Xu, C. P. Ng et al., Glomerulocystic kidney disease in mice with a targeted inactivation of Wwtr1, Proceedings of the National Academy of Sciences of the United States of America, pp.1631-1636, 2007.
DOI : 10.1073/pnas.0605266104

H. Huang, C. J. Potter, W. Tao, D. M. Li, W. Brogiolo et al., PTEN affects cell size, cell proliferation and apoptosis during Drosophila eye development, Development, vol.126, pp.5365-5372, 1999.

J. Huang, C. C. Dibble, M. Matsuzaki, and B. D. Manning, The TSC1-TSC2 Complex Is Required for Proper Activation of mTOR Complex 2, Molecular and Cellular Biology, vol.28, issue.12, pp.4104-4115, 2008.
DOI : 10.1128/MCB.00289-08

J. Huang and B. D. Manning, The TSC1???TSC2 complex: a molecular switchboard controlling cell growth, Biochemical Journal, vol.412, issue.2, pp.179-190, 2008.
DOI : 10.1042/BJ20080281

X. Huang and J. P. Saint-jeannet, Induction of the neural crest and the opportunities of life on the edge, Developmental Biology, vol.275, issue.1, pp.1-11, 2004.
DOI : 10.1016/j.ydbio.2004.07.033

K. Inoki, Y. Li, T. Xu, and K. L. Guan, Rheb GTPase is a direct target of TSC2 GAP activity and regulates mTOR signaling, Genes & Development, vol.17, issue.15, pp.1829-1834, 2003.
DOI : 10.1101/gad.1110003

K. Inoki, Y. Li, T. Zhu, J. Wu, and K. L. Guan, TSC2 is phosphorylated and inhibited by Akt and suppresses mTOR signalling, Nature Cell Biology, vol.4, issue.9, pp.648-657, 2002.
DOI : 10.1038/ncb839

K. Inoki, H. Ouyang, T. Zhu, C. Lindvall, Y. Wang et al., TSC2 Integrates Wnt and Energy Signals via a Coordinated Phosphorylation by AMPK and GSK3 to Regulate Cell Growth, Cell, vol.126, issue.5, pp.955-968, 2006.
DOI : 10.1016/j.cell.2006.06.055

S. Jiao, H. Wang, Z. Shi, A. Dong, W. Zhang et al., A Peptide Mimicking VGLL4 Function Acts as a YAP Antagonist Therapy against Gastric Cancer, Cancer Cell, vol.25, issue.2, pp.166-180, 2014.
DOI : 10.1016/j.ccr.2014.01.010

R. S. Kandt, J. L. Haines, M. Smith, H. Northrup, R. J. Gardner et al., Linkage of an important gene locus for tuberous sclerosis to a chromosome 16 marker for polycystic kidney disease, Nature Genetics, vol.58, issue.1, pp.37-41, 1992.
DOI : 10.1038/352077a0

M. Karbowniczek, J. Yu, and E. P. Henske, Renal Angiomyolipomas from Patients with Sporadic Lymphangiomyomatosis Contain Both Neoplastic and Non-Neoplastic Vascular Structures, The American Journal of Pathology, vol.162, issue.2, pp.491-500, 2003.
DOI : 10.1016/S0002-9440(10)63843-6

H. L. Kenerson, L. D. Aicher, L. D. True, and R. S. Yeung, Activated mammalian target of rapamycin pathway in the pathogenesis of tuberous sclerosis complex renal tumors, Cancer research, vol.62, pp.5645-5650, 2002.

S. Kim, S. F. Kim, D. Maag, M. J. Maxwell, A. C. Resnick et al., Amino Acid Signaling to mTOR Mediated by Inositol Polyphosphate Multikinase, Cell Metabolism, vol.13, issue.2, pp.215-221, 2011.
DOI : 10.1016/j.cmet.2011.01.007

A. G. Knudson and . Jr, Mutation and Cancer: Statistical Study of Retinoblastoma, Proceedings of the National Academy of Sciences, vol.68, issue.4, 1971.
DOI : 10.1073/pnas.68.4.820

T. Kobayashi, O. Minowa, J. Kuno, H. Mitani, O. Hino et al., Renal carcinogenesis, hepatic hemangiomatosis, and embryonic lethality caused by a germ-line Tsc2 mutation in mice, Cancer research, vol.59, pp.1206-1211, 1999.

T. Kobayashi, O. Minowa, Y. Sugitani, S. Takai, H. Mitani et al., A germ-line Tsc1 mutation causes tumor development and embryonic lethality that are similar, but not identical to, those caused by Tsc2 mutation in mice, Proceedings of the National Academy of Sciences of the United States of America 98, pp.8762-8767, 2001.
DOI : 10.1073/pnas.151033798

N. Hashemite and H. Onda, A mouse model of TSC1 reveals sexdependent lethality from liver hemangiomas, and up-regulation of p70S6 kinase activity in Tsc1 null cells, Human molecular genetics, vol.11, pp.525-534, 2002.

Y. Kwon, A. Vinayagam, X. Sun, N. Dephoure, S. P. Gygi et al., The Hippo Signaling Pathway Interactome, Science, vol.342, issue.6159, pp.737-740, 2013.
DOI : 10.1126/science.1243971

R. F. Lamb, C. Roy, T. J. Diefenbach, H. V. Vinters, M. W. Johnson et al., The TSC1 tumour suppressor hamartin regulates cell adhesion through ERM proteins and the GTPase Rho, Nature Cell Biology, vol.2, issue.5, pp.281-287, 2000.
DOI : 10.1038/35010550

M. Laplante and D. M. Sabatini, mTOR Signaling in Growth Control and Disease, Cell, vol.149, issue.2, pp.274-293, 2012.
DOI : 10.1016/j.cell.2012.03.017

D. F. Lee, H. P. Kuo, C. T. Chen, J. M. Hsu, C. K. Chou et al., IKK?? Suppression of TSC1 Links Inflammation and Tumor Angiogenesis via the mTOR Pathway, Cell, vol.130, issue.3, pp.440-455, 2007.
DOI : 10.1016/j.cell.2007.05.058

S. D. Lim, W. Stallcup, B. Lefkove, B. Govindarajan, K. S. Au et al., Expression of the neural stem cell markers NG2 and L1 in human angiomyolipoma: are angiomyolipomas neoplasms of stem cells? Molecular medicine 13, pp.160-165, 2007.

Y. Liu-chittenden, B. Huang, J. S. Shim, Q. Chen, S. J. Lee et al., Genetic and pharmacological disruption of the TEAD-YAP complex suppresses the oncogenic activity of YAP, Genes & Development, vol.26, issue.12, pp.1300-1305, 2012.
DOI : 10.1101/gad.192856.112

X. Long, Y. Lin, S. Ortiz-vega, K. Yonezawa, and J. Avruch, Rheb Binds and Regulates the mTOR Kinase, Current Biology, vol.15, issue.8, pp.702-713, 2005.
DOI : 10.1016/j.cub.2005.02.053

A. Bulfone, J. M. Garcia-verdugo, L. Leocani, and F. Minicucci, Sustained activation of mTOR pathway in embryonic neural stem cells leads to development of tuberous sclerosis complex-associated lesions, Cell stem cell, vol.9, pp.447-462, 2011.

A. J. Malhowski, H. Hira, S. Bashiruddin, R. Warburton, J. Goto et al., Smooth muscle protein-22-mediated deletion of Tsc1 results in cardiac hypertrophy that is mTORC1-mediated and reversed by rapamycin, Human Molecular Genetics, vol.20, issue.7, pp.1290-1305, 2011.
DOI : 10.1093/hmg/ddq570

L. Meikle, D. M. Talos, H. Onda, K. Pollizzi, A. Rotenberg et al., A Mouse Model of Tuberous Sclerosis: Neuronal Loss of Tsc1 Causes Dysplastic and Ectopic Neurons, Reduced Myelination, Seizure Activity, and Limited Survival, Journal of Neuroscience, vol.27, issue.21, pp.5546-5558, 2007.
DOI : 10.1523/JNEUROSCI.5540-06.2007

S. Menon, C. C. Dibble, G. Talbott, G. Hoxhaj, A. J. Valvezan et al., Spatial Control of the TSC Complex Integrates Insulin and Nutrient Regulation of mTORC1 at the Lysosome, Cell, vol.156, issue.4, pp.771-785, 2014.
DOI : 10.1016/j.cell.2013.11.049

E. Miller, J. Yang, M. Deran, C. Wu, A. I. Su et al., Identification of Serum-Derived Sphingosine-1-Phosphate as a Small Molecule Regulator of YAP, Chemistry & Biology, vol.19, issue.8, pp.955-962, 2012.
DOI : 10.1016/j.chembiol.2012.07.005

E. M. Morin-kensicki, B. N. Boone, M. Howell, J. R. Stonebraker, J. Teed et al., Defects in Yolk Sac Vasculogenesis, Chorioallantoic Fusion, and Embryonic Axis Elongation in Mice with Targeted Disruption of Yap65, Molecular and Cellular Biology, vol.26, issue.1, pp.77-87, 2006.
DOI : 10.1128/MCB.26.1.77-87.2006

T. Oka, E. Remue, K. Meerschaert, B. Vanloo, C. Boucherie et al., Functional complexes between YAP2 and ZO-2 are PDZ domain-dependent, and regulate YAP2 nuclear localization and signalling, Biochemical Journal, vol.9, issue.3, pp.461-472, 2010.
DOI : 10.1074/jbc.M602901200

M. Ota and H. Sasaki, Mammalian Tead proteins regulate cell proliferation and contact inhibition as transcriptional mediators of Hippo signaling, Development, vol.135, issue.24, pp.4059-4069, 2008.
DOI : 10.1242/dev.027151

D. Pan, The Hippo Signaling Pathway in Development and Cancer, Developmental Cell, vol.19, issue.4, pp.491-505, 2010.
DOI : 10.1016/j.devcel.2010.09.011

A. Parkhitko, F. Myachina, T. A. Morrison, K. M. Hindi, N. Auricchio et al., Tumorigenesis in tuberous sclerosis complex is autophagy and p62/sequestosome 1 (SQSTM1)-dependent, Proceedings of the National Academy of Sciences of the United States of America, pp.12455-12460, 2011.
DOI : 10.1073/pnas.1104361108

M. Pea, G. Martignoni, G. Zamboni, and F. Bonetti, Perivascular Epithelioid Cell, The American Journal of Surgical Pathology, vol.20, issue.9, pp.1149-1153, 1996.
DOI : 10.1097/00000478-199609000-00012

T. R. Peterson, S. S. Sengupta, T. E. Harris, A. E. Carmack, S. A. Kang et al., mTOR Complex 1 Regulates Lipin 1 Localization to Control the SREBP Pathway, Cell, vol.146, issue.3, pp.408-420, 2011.
DOI : 10.1016/j.cell.2011.06.034

T. L. Plank, R. S. Yeung, and E. P. Henske, Hamartin, the product of the tuberous sclerosis 1 (TSC1) gene, interacts with tuberin and appears to be localized to cytoplasmic vesicles, Cancer research, vol.58, pp.4766-4770, 1998.

C. J. Potter, H. Huang, and T. Xu, Drosophila Tsc1 Functions with Tsc2 to Antagonize Insulin Signaling in Regulating Cell Growth, Cell Proliferation, and Organ Size, Cell, vol.105, issue.3, pp.357-368, 2001.
DOI : 10.1016/S0092-8674(01)00333-6

P. P. Roux, B. A. Ballif, R. Anjum, S. P. Gygi, and J. Blenis, Tumor-promoting phorbol esters and activated Ras inactivate the tuberous sclerosis tumor suppressor complex via p90 ribosomal S6 kinase, Proceedings of the National Academy of Sciences, vol.101, issue.37, pp.13489-13494, 2004.
DOI : 10.1073/pnas.0405659101

Y. Sancak, T. R. Peterson, Y. D. Shaul, R. A. Lindquist, C. C. Thoreen et al., The Rag GTPases Bind Raptor and Mediate Amino Acid Signaling to mTORC1, Science, vol.320, issue.5882, pp.1496-1501, 2008.
DOI : 10.1126/science.1157535

G. Halder, Modulating F-actin organization induces organ growth by affecting the Hippo pathway, The EMBO journal, vol.30, pp.2325-2335, 2011.

H. B. Sarnat and L. Flores-sarnat, Embryology of the Neural Crest: Its Inductive Role in the Neurocutaneous Syndromes, Journal of Child Neurology, vol.126, issue.8, 2005.
DOI : 10.1177/08830738050200080101

B. T. Kreger, V. Vasioukhin, J. Avruch, and T. R. Brummelkamp, Yap1 acts downstream of alpha-catenin to control epidermal proliferation, Cell, vol.144, pp.782-795, 2011.

T. Sepp, J. R. Yates, and A. J. Green, Loss of heterozygosity in tuberous sclerosis hamartomas., Journal of Medical Genetics, vol.33, issue.11, pp.962-964, 1996.
DOI : 10.1136/jmg.33.11.962

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, The EMBO Journal, vol.12, issue.5, pp.1095-1108, 2012.
DOI : 10.1038/emboj.2012.32

R. J. Shaw, N. Bardeesy, B. D. Manning, L. Lopez, M. Kosmatka et al., The LKB1 tumor suppressor negatively regulates mTOR signaling, Cancer Cell, vol.6, issue.1, pp.91-99, 2004.
DOI : 10.1016/j.ccr.2004.06.007

C. M. Sidor, R. Brain, and B. J. Thompson, Mask proteins are cofactors of, 2013.

E. M. Smith, S. G. Finn, A. R. Tee, G. J. Browne, and C. G. Proud, The Tuberous Sclerosis Protein TSC2 Is Not Required for the Regulation of the Mammalian Target of Rapamycin by Amino Acids and Certain Cellular Stresses, Journal of Biological Chemistry, vol.280, issue.19, pp.18717-18727, 2005.
DOI : 10.1074/jbc.M414499200

H. Stocker and E. Hafen, Genetic control of cell size. Current opinion in genetics & development 10, pp.529-535, 2000.

H. Stocker, T. Radimerski, B. Schindelholz, F. Wittwer, P. Belawat et al., Rheb is an essential regulator of S6K in controlling cell growth in Drosophila, Nature Cell Biology, vol.5, issue.6, pp.559-565, 2003.
DOI : 10.1038/ncb995

A. Stolz, A. Ernst, and I. Dikic, Cargo recognition and trafficking in selective autophagy, Nature Cell Biology, vol.115, issue.6, pp.495-501, 2014.
DOI : 10.1038/16264

S. Strano, E. Munarriz, M. Rossi, L. Castagnoli, Y. Shaul et al., Physical Interaction with Yes-associated Protein Enhances p73 Transcriptional Activity, Journal of Biological Chemistry, vol.276, issue.18, pp.15164-15173, 2001.
DOI : 10.1074/jbc.M010484200

N. Tapon, H. , and K. F. , The Hippo pathway???From top to bottom and everything in between, Seminars in cell & developmental biology, pp.768-769, 2012.
DOI : 10.1016/j.semcdb.2012.08.007

N. Tapon, N. Ito, B. J. Dickson, J. E. Treisman, and I. K. Hariharan, The Drosophila Tuberous Sclerosis Complex Gene Homologs Restrict Cell Growth and Cell Proliferation, Cell, vol.105, issue.3, pp.345-355, 2001.
DOI : 10.1016/S0092-8674(01)00332-4

P. T. Tsai, C. Hull, Y. Chu, E. Greene-colozzi, A. R. Sadowski et al., Autistic-like behaviour and cerebellar dysfunction in Purkinje cell Tsc1 mutant mice, Nature, vol.73, issue.7413, pp.647-651, 2012.
DOI : 10.1038/nature11310

K. Tumaneng, R. C. Russell, and K. L. Guan, Organ Size Control by Hippo and TOR Pathways, Current Biology, vol.22, issue.9, pp.368-379, 2012.
DOI : 10.1016/j.cub.2012.03.003

URL : http://doi.org/10.1016/j.cub.2012.03.003

E. J. Uhlmann, M. Wong, R. L. Baldwin, M. L. Bajenaru, H. Onda et al., Astrocyte-specificTSC1 conditional knockout mice exhibit abnormal neuronal organization and seizures, Annals of Neurology, vol.17, issue.3, pp.285-296, 2002.
DOI : 10.1002/ana.10283

K. Wada, K. Itoga, T. Okano, S. Yonemura, and H. Sasaki, Hippo pathway regulation by cell morphology and stress fibers, Development, vol.138, issue.18, pp.3907-3914, 2011.
DOI : 10.1242/dev.070987

Y. Wang, Q. Dong, Q. Zhang, Z. Li, E. Wang et al., Overexpression of yes-associated protein contributes to progression and poor prognosis of non-small-cell lung cancer, Cancer Science, vol.9, issue.5, pp.1279-1285, 2010.
DOI : 10.1111/j.1349-7006.2010.01511.x

R. Wienecke, A. Konig, and J. E. Declue, Identification of Tuberin, the Tuberous Sclerosis-2 Product. TUBERIN POSSESSES SPECIFIC Rap1GAP ACTIVITY, Journal of Biological Chemistry, vol.270, issue.27, pp.16409-16414, 1995.
DOI : 10.1074/jbc.270.27.16409

G. H. Xiao, F. Shoarinejad, F. Jin, E. A. Golemis, and R. S. Yeung, The Tuberous Sclerosis 2 Gene Product, Tuberin, Functions as a Rab5 GTPase Activating Protein (GAP) in Modulating Endocytosis, Journal of Biological Chemistry, vol.272, issue.10, pp.6097-6100, 1997.
DOI : 10.1074/jbc.272.10.6097

Z. Xie and D. J. Klionsky, Autophagosome formation: core machinery and adaptations, Nature Cell Biology, vol.2, issue.10, pp.1102-1109, 2007.
DOI : 10.1016/j.bbrc.2005.10.163

M. Xin, Y. Kim, L. B. Sutherland, X. Qi, J. Mcanally et al., Regulation of Insulin-Like Growth Factor Signaling by Yap Governs Cardiomyocyte Proliferation and Embryonic Heart Size, Science Signaling, vol.4, issue.196, 2011.
DOI : 10.1126/scisignal.2002278

M. Z. Xu, T. J. Yao, N. P. Lee, I. O. Ng, Y. T. Chan et al., Yes-associated protein is an independent prognostic marker in hepatocellular carcinoma, Cancer, vol.120, issue.2 pt 1, pp.4576-4585, 2009.
DOI : 10.1002/cncr.24495

R. Yagi, L. F. Chen, K. Shigesada, Y. Murakami, and Y. Ito, A WW domaincontaining yes-associated protein (YAP) is a novel transcriptional co-activator. The EMBO journal 18, pp.2551-2562, 1999.

F. X. Yu and K. L. Guan, The Hippo pathway: regulators and regulations, Genes & Development, vol.27, issue.4, 2013.
DOI : 10.1101/gad.210773.112

F. X. Yu, B. Zhao, N. Panupinthu, J. L. Jewell, I. Lian et al., Regulation of the Hippo-YAP Pathway by G-Protein-Coupled Receptor Signaling, Cell, vol.150, issue.4, pp.780-791, 2012.
DOI : 10.1016/j.cell.2012.06.037

H. Zhang, G. Cicchetti, H. Onda, H. B. Koon, K. Asrican et al., Loss of Tsc1, 2003.

Z. Y. Zhang, Yes-associated protein promotes cell proliferation by activating Fos Related Activator-1 in oral squamous cell carcinoma, Oral Oncology, vol.47, issue.8, pp.693-697, 2011.
DOI : 10.1016/j.oraloncology.2011.06.003

W. Zhang, Y. Gao, P. Li, Z. Shi, T. Guo et al., VGLL4 functions as a new tumor suppressor in lung cancer by negatively regulating the YAP-TEAD transcriptional complex, Cell Research, vol.126, issue.3, pp.331-343, 2014.
DOI : 10.1073/pnas.1004952107

X. Zhang, C. C. Milton, P. O. Humbert, H. , and K. F. , Transcriptional Output of the Salvador/Warts/Hippo Pathway Is Controlled in Distinct Fashions in Drosophila melanogaster and Mammalian Cell Lines, Cancer Research, vol.69, issue.15, pp.6033-6041, 2009.
DOI : 10.1158/0008-5472.CAN-08-4592

Y. Zhang, J. Nicholatos, J. R. Dreier, S. J. Ricoult, S. B. Widenmaier et al., Coordinated regulation of protein synthesis and degradation by mTORC1, Nature, vol.23, issue.7518, 2014.
DOI : 10.1038/nature13492

B. Zhao, L. Li, K. Tumaneng, C. Y. Wang, and K. L. Guan, A coordinated phosphorylation by Lats and CK1 regulates YAP stability through SCF(beta-TRCP), 2010.

B. Zhao, X. Wei, W. Li, R. S. Udan, Q. Yang et al., Inactivation of YAP oncoprotein by the Hippo pathway is involved in cell contact inhibition and tissue growth control, Genes & Development, vol.21, issue.21, pp.2747-2761, 2007.
DOI : 10.1101/gad.1602907

B. Zhao, X. Ye, J. Yu, L. Li, W. Li et al., TEAD mediates YAP-dependent gene induction and growth control, Genes & Development, vol.22, issue.14, pp.1962-1971, 2008.
DOI : 10.1101/gad.1664408

J. Zhou, J. Brugarolas, and L. F. Parada, Loss of Tsc1, but not Pten, in renal tubular cells causes polycystic kidney disease by activating mTORC1, Human Molecular Genetics, vol.18, issue.22, pp.4428-4441, 2009.
DOI : 10.1093/hmg/ddp398

J. Zhou, G. Shrikhande, J. Xu, R. M. Mckay, D. K. Burns et al., Tsc1 mutant neural stem/progenitor cells exhibit migration deficits and give rise to subependymal lesions in the lateral ventricle, Genes & Development, vol.25, issue.15, pp.1595-1600, 2011.
DOI : 10.1101/gad.16750211

R. Zoncu, L. Bar-peled, A. Efeyan, S. Wang, Y. Sancak et al., mTORC1 Senses Lysosomal Amino Acids Through an Inside-Out Mechanism That Requires the Vacuolar H+-ATPase, Science, vol.334, issue.6056, pp.678-683, 2011.
DOI : 10.1126/science.1207056