R. A. Saxton and D. M. Sabatini, mTOR signaling in growth, metabolism, and disease, Cell, vol.169, pp.361-371, 2017.

J. M. Shillingford, The mTOR pathway is regulated by polycystin-1, and its inhibition reverses renal cystogenesis in polycystic kidney disease, Proc. Natl Acad. Sci. USA, vol.103, pp.5466-5471, 2006.

A. Zullo, Kidney-specific inactivation of Ofd1 leads to renal cystic disease associated with upregulation of the mTOR pathway, Hum. Mol. Genet, vol.19, pp.2792-2803, 2010.

T. R. Hartman, The role of the Birt-Hogg-Dube protein in mTOR activation and renal tumorigenesis, Oncogene, vol.28, pp.1594-1604, 2009.

L. S. Schmidt and W. M. Linehan, Genetic predisposition to kidney cancer, Semin. Oncol, vol.43, pp.566-574, 2016.

T. Weimbs, J. M. Shillingford, J. Torres, S. L. Kruger, and B. C. Bourgeois, Emerging targeted strategies for the treatment of autosomal dominant polycystic kidney disease, Clin. Kidney J, vol.11, pp.27-38, 2018.

S. J. Holditch, A study of sirolimus and mTOR kinase inhibitor in a hypomorphic Pkd1 mouse model of autosomal dominant polycystic kidney disease, Am. J. Physiol. Ren. Physiol, vol.317, pp.187-196, 2019.

C. Bergmann, Polycystic kidney disease, Nat. Rev. Dis. Primers, vol.4, p.50, 2018.

E. P. Henske, S. Jozwiak, J. C. Kingswood, J. R. Sampson, and E. A. Thiele, Tuberous sclerosis complex, Nat. Rev. Dis. Primers, vol.2, p.16035, 2016.

S. Menon, Spatial control of the TSC complex integrates insulin and nutrient regulation of mTORC1 at the lysosome, Cell, vol.156, pp.771-785, 2014.

P. P. Hsu, The mTOR-regulated phosphoproteome reveals a mechanism of mTORC1-mediated inhibition of growth factor signaling, Science, vol.332, pp.1317-1322, 2011.

Y. Yu, Phosphoproteomic analysis identifies Grb10 as an mTORC1 substrate that negatively regulates insulin signaling, Science, vol.332, pp.1322-1326, 2011.

S. A. Kang, mTORC1 phosphorylation sites encode their sensitivity to starvation and rapamycin, Science, vol.341, p.1236566, 2013.

X. Gao, Tsc tumour suppressor proteins antagonize amino-acid-TOR signalling, Nat. Cell Biol, vol.4, pp.699-704, 2002.

M. Pende, S6K2(-/-) mice exhibit perinatal lethality and rapamycin-sensitive 5'-terminal oligopyrimidine mRNA translation and reveal a mitogen-activated protein kinase-dependent S6 kinase pathway, Mol. Cell. Biol, vol.24, pp.3112-3124, 2004.

H. Shima, Disruption of the p70(s6k)/p85(s6k) gene reveals a small mouse phenotype and a new functional S6 kinase, EMBO J, vol.17, pp.6649-6659, 1998.

L. R. Pearce, D. Komander, and D. R. Alessi, The nuts and bolts of AGC protein kinases, Nat. Rev. Mol. Cell Biol, vol.11, pp.9-22, 2010.

D. D. Sarbassov, D. A. Guertin, S. M. Ali, and D. M. Sabatini, Phosphorylation and regulation of Akt/PKB by the rictor-mTOR complex, Science, vol.307, pp.1098-1101, 2005.

J. Huang and B. D. Manning, A complex interplay between Akt, TSC2 and the two mTOR complexes, Biochem. Soc. Trans, vol.37, pp.217-222, 2009.

M. Pema, mTORC1-mediated inhibition of polycystin-1 expression drives renal cyst formation in tuberous sclerosis complex, Nat. Commun, vol.7, p.10786, 2016.

X. Shao, S. Somlo, and P. Igarashi, Epithelial-specific Cre/lox recombination in the developing kidney and genitourinary tract, J. Am. Soc. Nephrol, vol.13, pp.1837-1846, 2002.

E. A. Armour, R. P. Carson, and K. C. Ess, Cystogenesis and elongated primary cilia in Tsc1-deficient distal convoluted tubules, Am. J. Physiol. Ren. Physiol, vol.303, pp.584-592, 2012.

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, Hum. Mol. Genet, vol.18, pp.4428-4441, 2009.

N. Liang, Regulation of YAP by mTOR and autophagy reveals a therapeutic target of tuberous sclerosis complex, J. Exp. Med, vol.211, pp.2249-2263, 2014.
URL : https://hal.archives-ouvertes.fr/tel-01126889

I. Ben-sahra, J. J. Howell, J. M. Asara, and B. D. Manning, Stimulation of de novo pyrimidine synthesis by growth signaling through mTOR and S6K1, Science, vol.339, pp.1323-1328, 2013.

C. Treins, P. H. Warne, M. A. Magnuson, M. Pende, and J. Downward, Rictor is a novel target of p70 S6 kinase-1, Oncogene, vol.29, pp.1003-1016, 2010.

M. Pende, Hypoinsulinaemia, glucose intolerance and diminished betacell size in S6K1-deficient mice, Nature, vol.408, pp.994-997, 2000.

M. Ohanna, Atrophy of S6K1(?/?) skeletal muscle cells reveals distinct mTOR effectors for cell cycle and size control, Nat. Cell Biol, vol.7, pp.286-294, 2005.

E. Fischer, Defective planar cell polarity in polycystic kidney disease, Nat. Genet, vol.38, pp.21-23, 2006.
URL : https://hal.archives-ouvertes.fr/hal-01973787

S. Saburi, Loss of Fat4 disrupts PCP signaling and oriented cell division and leads to cystic kidney disease, Nat. Genet, vol.40, pp.1010-1015, 2008.

V. Patel, Acute kidney injury and aberrant planar cell polarity induce cyst formation in mice lacking renal cilia, Hum. Mol. Genet, vol.17, pp.1578-1590, 2008.

C. S. Bonnet, Defects in cell polarity underlie TSC and ADPKDassociated cystogenesis, Hum. Mol. Genet, vol.18, pp.2166-2176, 2009.

S. Nishio, Loss of oriented cell division does not initiate cyst formation, J. Am. Soc. Nephrol, vol.21, pp.295-302, 2010.

K. Kunimoto, Disruption of core planar cell polarity signaling regulates renal tubule morphogenesis but is not cystogenic, Curr. Biol, vol.27, pp.3120-3131, 2017.

C. Cadart, E. Zlotek-zlotkiewicz, M. Le-berre, M. Piel, and H. K. Matthews, Exploring the function of cell shape and size during mitosis, Dev. Cell, vol.29, pp.159-169, 2014.

R. H. Giles, H. Ajzenberg, and P. K. Jackson, 3D spheroid model of mIMCD3 cells for studying ciliopathies and renal epithelial disorders, Nat. Protoc, vol.9, pp.2725-2731, 2014.

F. Di-pietro, A. Echard, and X. Morin, Regulation of mitotic spindle orientation: an integrated view, EMBO Rep, vol.17, pp.1106-1130, 2016.

M. Thery, The extracellular matrix guides the orientation of the cell division axis, Nat. Cell Biol, vol.7, pp.947-953, 2005.

J. Fink, External forces control mitotic spindle positioning, Nat. Cell Biol, vol.13, pp.771-778, 2011.

I. Dupin, E. Camand, and S. Etienne-manneville, Classical cadherins control nucleus and centrosome position and cell polarity, J. Cell Biol, vol.185, pp.779-786, 2009.

J. Rush, Immunoaffinity profiling of tyrosine phosphorylation in cancer cells, Nat. Biotechnol, vol.23, pp.94-101, 2005.

K. Mizutani and Y. Takai, Nectin spot: a novel type of nectin-mediated cell adhesion apparatus, Biochem. J, vol.473, pp.2691-2715, 2016.

T. Y. Huang, C. Dermardirossian, and G. M. Bokoch, Cofilin phosphatases and regulation of actin dynamics, Curr. Opin. Cell Biol, vol.18, pp.26-31, 2006.

W. Ning, The CAMSAP3-ACF7 complex couples noncentrosomal microtubules with actin filaments to coordinate their dynamics, Dev. Cell, vol.39, pp.61-74, 2016.

T. R. Graham and C. G. Burd, Coordination of Golgi functions by phosphatidylinositol 4-kinases, Trends Cell Biol, vol.21, pp.113-121, 2011.

M. Ito, T. Nakano, F. Erdodi, and D. J. Hartshorne, Myosin phosphatase: structure, regulation and function, Mol. Cell Biochem, vol.259, pp.197-209, 2004.

M. Bershteyn, S. X. Atwood, W. M. Woo, M. Li, and A. E. Oro, MIM and cortactin antagonism regulates ciliogenesis and hedgehog signaling, Dev. Cell, vol.19, pp.270-283, 2010.

L. Gao, Afadin orients cell division to position the tubule lumen in developing renal tubules, Development, vol.144, pp.3511-3520, 2017.

M. Barilari, ZRF1 is a novel S6 kinase substrate that drives the senescence programme, EMBO J, vol.36, pp.736-750, 2017.

S. Elloul, D. Kedrin, N. W. Knoblauch, A. H. Beck, and A. Toker, The adherens junction protein afadin is an AKT substrate that regulates breast cancer cell migration, Mol. Cancer Res, vol.12, pp.464-476, 2014.

T. Sato, Regulation of the assembly and adhesion activity of E-cadherin by nectin and afadin for the formation of adherens junctions in Madin-Darby canine kidney cells, J. Biol. Chem, vol.281, pp.5288-5299, 2006.

I. Orhon, Primary-cilium-dependent autophagy controls epithelial cell volume in response to fluid flow, Nat. Cell Biol, vol.18, pp.657-667, 2016.

L. Bacquer and O. , 4E-BP1 and 4E-BP2 double knockout mice are protected from aging-associated sarcopenia, J. Cachexia Sarcopenia Muscle, vol.10, pp.696-709, 2019.
URL : https://hal.archives-ouvertes.fr/hal-02101567

M. Carminati, Concomitant binding of Afadin to LGN and F-actin directs planar spindle orientation, Nat. Struct. Mol. Biol, vol.23, pp.155-163, 2016.

M. Lorger and K. Moelling, Regulation of epithelial wound closure and intercellular adhesion by interaction of AF6 with actin cytoskeleton, J. Cell Sci, vol.119, pp.3385-3398, 2006.

R. F. Lamb, The TSC1 tumour suppressor hamartin regulates cell adhesion through ERM proteins and the GTPase Rho, Nat. Cell Biol, vol.2, pp.281-287, 2000.

A. Astrinidis, Tuberin, the tuberous sclerosis complex 2 tumor suppressor gene product, regulates Rho activation, cell adhesion and migration, Oncogene, vol.21, pp.8470-8476, 2002.

H. Kugoh, E. Kleymenova, and C. L. Walker, Retention of membrane-localized beta-catenin in cells lacking functional polycystin-1 and tuberin, Mol. Carcinog, vol.33, pp.131-136, 2002.

E. A. Barnes, H. L. Kenerson, X. Jiang, and R. S. Yeung, Tuberin regulates Ecadherin localization: implications in epithelial-mesenchymal transition, Am. J. Pathol, vol.177, pp.1765-1778, 2010.

E. A. Barnes, H. L. Kenerson, B. C. Mak, and R. S. Yeung, The loss of tuberin promotes cell invasion through the ss-catenin pathway, Am. J. Respir. Cell Mol. Biol, vol.43, pp.617-627, 2010.

F. Verdeguer, A mitotic transcriptional switch in polycystic kidney disease, Nat. Med, vol.16, pp.106-110, 2010.

A. Luyten, Aberrant regulation of planar cell polarity in polycystic kidney disease, J. Am. Soc. Nephrol, vol.21, pp.1521-1532, 2010.

C. M. Karner, Wnt9b signaling regulates planar cell polarity and kidney tubule morphogenesis, Nat. Genet, vol.41, pp.793-799, 2009.

L. Li, Aberrant planar cell polarity induced by urinary tract obstruction, Am. J. Physiol. Ren. Physiol, vol.297, pp.1526-1533, 2009.

N. Sharma, Proximal tubule proliferation is insufficient to induce rapid cyst formation after cilia disruption, J. Am. Soc. Nephrol, vol.24, pp.456-464, 2013.

K. R. Kipp, Comparison of folate-conjugated rapamycin versus unconjugated rapamycin in an orthologous mouse model of polycystic kidney disease, Am. J. Physiol. Ren. Physiol, vol.315, pp.395-405, 2018.

J. J. Bissler, Everolimus for angiomyolipoma associated with tuberous sclerosis complex or sporadic lymphangioleiomyomatosis (EXIST-2): a multicentre, randomised, double-blind, placebo-controlled trial, Lancet, vol.381, pp.817-824, 2013.

G. Walz, Everolimus in patients with autosomal dominant polycystic kidney disease, N. Engl. J. Med, vol.363, pp.830-840, 2010.

A. L. Serra, Sirolimus and kidney growth in autosomal dominant polycystic kidney disease, N. Engl. J. Med, vol.363, pp.820-829, 2010.

N. Perico, Sirolimus therapy to halt the progression of ADPKD, J. Am. Soc. Nephrol, vol.21, pp.1031-1040, 2010.

D. W. Lamming, Rapamycin-induced insulin resistance is mediated by mTORC2 loss and uncoupled from longevity, Science, vol.335, pp.1638-1643, 2012.

E. A. Susaki, Advanced CUBIC protocols for whole-brain and wholebody clearing and imaging, Nat. Protoc, vol.10, pp.1709-1727, 2015.

A. B. Jaffe, N. Kaji, J. Durgan, and A. Hall, Cdc42 controls spindle orientation to position the apical surface during epithelial morphogenesis, J. Cell Biol, vol.183, pp.625-633, 2008.

Z. Zheng, LGN regulates mitotic spindle orientation during epithelial morphogenesis, J. Cell Biol, vol.189, pp.275-288, 2010.

S. Shibazaki, Cyst formation and activation of the extracellular regulated kinase pathway after kidney specific inactivation of Pkd1, Hum. Mol. Genet, vol.17, pp.1505-1516, 2008.

E. Zlotek-zlotkiewicz, S. Monnier, G. Cappello, M. Le-berre, and M. Piel, Optical volume and mass measurements show that mammalian cells swell during mitosis, J. Cell Biol, vol.211, pp.765-774, 2015.

M. P. Stokes, Complementary PTM profiling of drug response in human gastric carcinoma by immunoaffinity and IMAC methods with total proteome analysis, Proteomes, vol.3, pp.160-183, 2015.

J. Rappsilber, Y. Ishihama, and M. Mann, Stop and go extraction tips for matrix-assisted laser desorption/ionization, nanoelectrospray, and LC/MS sample pretreatment in proteomics, Anal. Chem, vol.75, pp.663-670, 2003.

J. V. Olsen, Parts per million mass accuracy on an Orbitrap mass spectrometer via lock mass injection into a C-trap, Mol. Cell. Proteom, vol.4, pp.2010-2021, 2005.

J. K. Eng, A. L. Mccormack, and J. R. Yates, An approach to correlate tandem mass spectral data of peptides with amino acid sequences in a protein database, J. Am. Soc. Mass Spectrom, vol.5, pp.976-989, 1994.

E. L. Huttlin, A tissue-specific atlas of mouse protein phosphorylation and expression, Cell, vol.143, pp.1174-1189, 2010.

J. Villen, S. A. Beausoleil, S. A. Gerber, and S. P. Gygi, Large-scale phosphorylation analysis of mouse liver, Proc. Natl Acad. Sci. USA, vol.104, pp.1488-1493, 2007.