X. Teng, M. Dayhoff-brannigan, W. C. Cheng, C. E. Gilbert, C. N. Sing et al., Genome-wide consequences of deleting any single gene, Mol Cell, vol.52, issue.4, pp.485-94, 2013.

D. Marcotullio, L. Ferretti, E. De-smaele, E. Argenti, B. Mincione et al., REN(KCTD11) is a suppressor of Hedgehog signaling and is deleted in human medulloblastoma, Proc Natl Acad Sci U S A, vol.101, issue.29, pp.10833-10841, 2004.

R. Cui, W. Meng, H. L. Sun, T. Kim, Z. Ye et al., MicroRNA-224 promotes tumor progression in nonsmall cell lung cancer, Proc Natl Acad Sci, vol.112, issue.31, pp.4288-97, 2015.

P. Van-bogaert, R. Azizieh, J. Desir, A. Aeby, L. De-meirleir et al., Mutation of a potassium channel-related gene in progressive myoclonic epilepsy, Ann Neurol, vol.61, issue.6, pp.579-86, 2007.

K. A. Metz, X. Teng, I. Coppens, T. S. Wang, H. M. Lamb et al., KCTD7 mutations define a distinct neurodegenerative disorder with defective autophagy (in submission), 2018.

B. L. Carter and P. E. Sudbery, Small-sized mutants of Saccharomyces cerevisiae, Genetics, vol.96, issue.3, pp.561-567, 1980.

D. J. Saul and P. E. Sudbery, Molecular cloning of WHI2, a gene involved in the regulation of cell proliferation in Saccharomyces cerevisiae, J Gen Microbiol, vol.131, issue.7, pp.1797-806, 1985.

P. A. Radcliffe, K. M. Binley, J. Trevethick, M. Hall, and P. E. Sudbery, Filamentous growth of the budding yeast Saccharomyces cerevisiae induced by overexpression of the WHi2 gene. Microbiology, pp.1867-76, 1997.

P. Radcliffe, J. Trevethick, M. Tyers, and P. Sudbery, Deregulation of CLN1 and CLN2 in the Saccharomyces cerevisiae whi2 mutant, vol.13, p.9219335, 1997.

D. Kaida, H. Yashiroda, A. Toh-e, and Y. Kikuchi, Yeast Whi2 and Psr1-phosphatase form a complex and regulate STRE-mediated gene expression, Genes Cells, vol.7, issue.6, pp.543-52, 2002.

M. Boeckstaens, E. Llinares, P. Van-vooren, and A. M. Marini, The TORC1 effector kinase Npr1 fine tunes the inherent activity of the Mep2 ammonium transport protein, Nat Commun, vol.5, p.3101, 2014.

I. Gladwyn-ng, L. Huang, L. Ngo, S. S. Li, Z. Qu et al., Bacurd1/Kctd13 and Bacurd2/Tnfaip1 are interacting partners to Rnd proteins which influence the longterm positioning and dendritic maturation of cerebral cortical neurons, Neural Dev, vol.11, 2016.

Y. Wang, Y. Zheng, F. Luo, X. Fan, J. Chen et al., KCTD10 interacts with proliferating cell nuclear antigen and its down-regulation could inhibit cell proliferation, J Cell Biochem, vol.106, issue.3, pp.409-422, 2009.

N. Stavropoulos and M. W. Young, Cullin-3 regulate sleep and wakefulness in Drosophila, Neuron, vol.72, issue.6, pp.964-76, 2011.

J. Schwenk, M. Metz, G. Zolles, R. Turecek, T. Fritzius et al., Native GABA(B) receptors are heteromultimers with a family of auxiliary subunits, Nature, vol.465, issue.7295, pp.231-236, 2010.

C. Scuoppo, C. Miething, L. Lindqvist, J. Reyes, C. Ruse et al., A tumour suppressor network relying on the polyamine-hypusine axis, Nature, vol.487, issue.7406, pp.244-252, 2012.

R. Tong, B. Yang, H. Xiao, C. Peng, W. Hu et al., KCTD11 inhibits growth and metastasis of hepatocellular carcinoma through activating Hippo signaling, Oncotarget, vol.8, issue.23, pp.37717-37729, 2017.

F. Zazzeroni, D. Nicosia, A. Tessitore, R. Gallo, D. Verzella et al., KCTD11 tumor suppressor gene expression is reduced in prostate adenocarcinoma, Biomed Res Int, p.380398, 2014.

W. C. Cheng, X. Teng, H. K. Park, C. M. Tucker, M. J. Dunham et al., Fis1 deficiency selects for compensatory mutations responsible for cell death and growth control defects, Cell Death Differ, vol.15, issue.12, pp.1838-1884, 2008.

Y. Fannjiang, W. C. Cheng, S. J. Lee, B. Qi, J. Pevsner et al., Mitochondrial fission proteins regulate programmed cell death in yeast, Genes Dev, vol.18, issue.22, pp.2785-97, 2004.

I. Ivanovska and J. M. Hardwick, Viruses activate a genetically conserved cell death pathway in a unicellular organism, J Cell Biol, vol.170, issue.3, pp.391-400, 2005.

J. E. Leadsham, K. Miller, K. R. Ayscough, S. Colombo, E. Martegani et al., Whi2p links nutritional sensing to actin-dependent Ras-cAMP-PKA regulation and apoptosis in yeast, J Cell Sci, issue.122, pp.706-721, 2009.

L. Bar-peled, L. Chantranupong, A. D. Cherniack, W. W. Chen, K. A. Ottina et al., A Tumor suppressor complex with GAP activity for the Rag GTPases that signal amino acid sufficiency to mTORC1, Science, vol.340, issue.6136, pp.1100-1106, 2013.

N. Panchaud, M. P. Peli-gulli, D. Virgilio, and C. , Amino acid deprivation inhibits TORC1 through a GTPase-activating protein complex for the Rag family GTPase Gtr1, Sci Signal, issue.6, p.42, 2013.

A. Gonzalez and M. N. Hall, Nutrient sensing and TOR signaling in yeast and mammals, EMBO J, vol.36, issue.4, pp.397-408, 2017.

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.1496-501, 2008.

R. L. Wolfson, L. Chantranupong, R. A. Saxton, K. Shen, S. M. Scaria et al., Sestrin2 is a leucine sensor for the mTORC1 pathway, Science, vol.351, issue.6268, pp.43-51, 2016.

R. L. Wolfson, L. Chantranupong, G. A. Wyant, X. Gu, J. M. Orozco et al., KICSTOR recruits GATOR1 to the lysosome and is necessary for nutrients to regulate mTORC1, Nature, vol.543, issue.7645, pp.438-442, 2017.

L. Chantranupong, S. M. Scaria, R. A. Saxton, M. P. Gygi, K. Shen et al., The CASTOR Proteins Are Arginine Sensors for the mTORC1 Pathway, Cell, vol.165, issue.1, pp.153-164, 2016.

R. L. Wolfson and D. M. Sabatini, The Dawn of the Age of Amino Acid Sensors for the mTORC1 Pathway, Cell Metab, vol.26, issue.2, pp.301-309, 2017.

X. Gao, Y. Zhang, P. Arrazola, O. Hino, T. Kobayashi et al., Tsc tumour suppressor proteins antagonize amino-acid-TOR signalling, Nat Cell Biol, vol.4, issue.9, pp.699-704, 2002.

S. Yerlikaya, M. Meusburger, R. Kumari, A. Huber, D. Anrather et al., TORC1 and TORC2 work together to regulate ribosomal protein S6 phosphorylation in Saccharomyces cerevisiae, Mol Biol Cell, vol.27, issue.2, pp.397-409, 2016.

J. Urban, A. Soulard, A. Huber, S. Lippman, D. Mukhopadhyay et al., Sch9 is a major target of TORC1 in Saccharomyces cerevisiae, Mol Cell, vol.26, issue.5, pp.663-74, 2007.

A. Gonzalez, M. Shimobayashi, T. Eisenberg, D. A. Merle, T. Pendl et al., TORC1 promotes phosphorylation of ribosomal protein S6 via the AGC kinase Ypk3 in Saccharomyces cerevisiae, PLoS One, vol.10, issue.3, p.120250, 2015.

D. H. Kim, D. D. Sarbassov, S. M. Ali, J. E. King, R. R. Latek et al., mTOR interacts with raptor to form a nutrient-sensitive complex that signals to the cell growth machinery, Cell, vol.110, issue.2, pp.163-75, 2002.

I. Ruvinsky and O. Meyuhas, Ribosomal protein S6 phosphorylation: from protein synthesis to cell size, Trends Biochem Sci, vol.31, issue.6, pp.342-350, 2006.

H. Hallett, J. E. Luo, X. Capaldi, and A. P. , State transitions in the TORC1 signaling pathway and information processing in Saccharomyces cerevisiae, Genetics, vol.198, issue.2, pp.773-86, 2014.

H. Hallett, J. E. Luo, X. Capaldi, and A. P. , Snf1/AMPK promotes the formation of Kog1/Raptorbodies to increase the activation threshold of TORC1 in budding yeast

K. Inoki, T. Zhu, and K. L. Guan, TSC2 mediates cellular energy response to control cell growth and survival, Cell, vol.115, issue.5, pp.577-90, 2003.

D. M. Gwinn, D. B. Shackelford, D. F. Egan, M. M. Mihaylova, A. Mery et al., AMPK phosphorylation of raptor mediates a metabolic checkpoint, Mol Cell, vol.30, issue.2, pp.214-240, 2008.

J. Hong and D. Gresham, Molecular specificity, convergence and constraint shape adaptive evolution in nutrient-poor environments, PLoS Genet, vol.10, issue.1, p.1004041, 2014.

S. Ghaemmaghami, W. K. Huh, K. Bower, R. W. Howson, A. Belle et al., Global analysis of protein expression in yeast, Nature, vol.425, issue.6959, pp.737-778, 2003.

T. K. Neklesa and R. W. Davis, A genome-wide screen for regulators of TORC1 in response to amino acid starvation reveals a conserved Npr2/3 complex, PLoS Genet, vol.5, issue.6, p.1000515, 2009.

M. Stanbrough, D. W. Rowen, and M. B. , Role of the GATA factors Gln3p and Nil1p of Saccharomyces cerevisiae in the expression of nitrogen-regulated genes, Proc Natl Acad Sci, vol.92, issue.21, pp.9450-9454, 1995.

A. Schmidt, T. Beck, A. Koller, J. Kunz, and M. N. Hall, The TOR nutrient signalling pathway phosphorylates NPR1 and inhibits turnover of the tryptophan permease, EMBO J, vol.17, issue.23, pp.6924-6955, 1998.

M. Gao and C. A. Kaiser, A conserved GTPase-containing complex is required for intracellular sorting of the general amino-acid permease in yeast, Nat Cell Biol, vol.8, issue.7, pp.657-67, 2006.

T. Ito, T. Chiba, R. Ozawa, M. Yoshida, M. Hattori et al., A comprehensive two-hybrid analysis to explore the yeast protein interactome, Proc Natl Acad Sci U S A, vol.98, issue.8, pp.4569-74, 2001.

N. J. Krogan, G. Cagney, H. Yu, G. Zhong, X. Guo et al., Global landscape of protein complexes in the yeast Saccharomyces cerevisiae, Nature, vol.440, issue.7084, pp.637-680, 2006.

H. Yu, P. Braun, M. A. Yildirim, I. Lemmens, K. Venkatesan et al., Highquality binary protein interaction map of the yeast interactome network, Science, vol.322, issue.5898, pp.104-114, 2008.

Y. Ho, A. Gruhler, A. Heilbut, G. D. Bader, L. Moore et al., Systematic identification of protein complexes in Saccharomyces cerevisiae by mass spectrometry, Nature, vol.415, issue.6868, pp.180-183, 2002.

T. Toda, S. Cameron, P. Sass, M. Zoller, and M. Wigler, Three different genes in S. cerevisiae encode the catalytic subunits of the cAMP-dependent protein kinase, Cell, vol.50, issue.2, pp.277-87, 1987.

M. Skoblov, A. Marakhonov, E. Marakasova, A. Guskova, V. Chandhoke et al., Protein partners of KCTD proteins provide insights about their functional roles in cell differentiation and vertebrate development, Bioessays, vol.35, issue.7, pp.586-96, 2013.

M. Faryna, C. Konermann, S. Aulmann, J. L. Bermejo, M. Brugger et al., Genome-wide methylation screen in low-grade breast cancer identifies novel epigenetically altered genes as potential biomarkers for tumor diagnosis, FASEB J, vol.26, issue.12, pp.4937-50, 2012.

T. Hasegawa, H. Asanuma, J. Ogino, Y. Hirohashi, Y. Shinomura et al., Use of potassium channel tetramerization domain-containing 12 as a biomarker for diagnosis and prognosis of gastrointestinal stromal tumor, Hum Pathol, vol.44, issue.7, pp.1271-1278, 2013.

G. Canettieri, D. Marcotullio, L. Greco, A. Coni, S. Antonucci et al., Histone deacetylase and Cullin3-REN(KCTD11) ubiquitin ligase interplay regulates Hedgehog signalling through Gli acetylation, Nat Cell Biol, vol.12, issue.2, pp.132-174, 2010.

M. Binda, M. P. Peli-gulli, G. Bonfils, N. Panchaud, J. Urban et al., The Vam6 GEF controls TORC1 by activating the EGO complex, Mol Cell, vol.35, issue.5, pp.563-73, 2009.

D. Stracka, S. Jozefczuk, F. Rudroff, U. Sauer, and M. N. Hall, Nitrogen source activates TOR (target of rapamycin) complex 1 via glutamine and independently of Gtr/Rag proteins, J Biol Chem, vol.289, issue.36, pp.25010-25030, 2014.

B. M. Sutter, X. Wu, S. Laxman, and T. B. , Methionine inhibits autophagy and promotes growth by inducing the SAM-responsive methylation of PP2A, Cell, vol.154, issue.2, pp.403-418, 2013.

J. L. Jewell, Y. C. Kim, R. C. Russell, F. X. Yu, H. W. Park et al., Differential regulation of mTORC1 by leucine and glutamine, Science, vol.347, issue.6218, pp.194-202, 2015.

S. Wang, Z. Y. Tsun, R. L. Wolfson, K. Shen, G. A. Wyant et al., Lysosomal amino acid transporter SLC38A9 signals arginine sufficiency to mTORC1, Science, vol.347, issue.6218, pp.188-94, 2015.

R. Dechant, S. Saad, A. J. Ibanez, and P. M. , Cytosolic pH regulates cell growth through distinct GTPases, Arf1 and Gtr1, to promote Ras/PKA and TORC1 activity, Mol Cell, vol.55, issue.3, pp.409-430, 2014.

P. E. Sudbery, A. R. Goodey, and B. L. Carter, Genes which control cell proliferation in the yeast Saccharomyces cerevisiae, Nature, vol.288, issue.5789, pp.401-405, 1980.

T. Beck and M. N. Hall, The TOR signalling pathway controls nuclear localization of nutrient-regulated transcription factors, Nature, vol.402, issue.6762, pp.689-92, 1999.

E. Saliba, M. Evangelinos, C. Gournas, F. Corrillon, I. Georis et al., The yeast H(+)-ATPase Pma1 promotes Rag/Gtr-dependent TORC1 activation in response to H(+)-coupled nutrient uptake. Elife, 2018.

N. Mendl, A. Occhipinti, M. Muller, P. Wild, I. Dikic et al., Mitophagy in yeast is independent of mitochondrial fission and requires the stress response gene WHI2, J Cell Sci, issue.124, pp.1339-50, 2011.

K. Mao, K. Wang, X. Liu, and D. J. Klionsky, The scaffold protein Atg11 recruits fission machinery to drive selective mitochondria degradation by autophagy, Dev Cell, vol.26, issue.1, pp.9-18, 2013.

R. Bojsen, B. Regenberg, and F. A. , Persistence and drug tolerance in pathogenic yeast, Curr Genet, vol.63, issue.1, p.27193346, 2017.

K. Harata, T. Nishiuchi, and Y. Kubo, Colletotrichum orbiculare WHI2, a Yeast Stress-Response Regulator Homolog, Controls the Biotrophic Stage of Hemibiotrophic Infection Through TOR Signaling, Mol Plant Microbe Interact, vol.29, issue.6, p.27018615, 2016.

F. Dubouloz, O. Deloche, V. Wanke, E. Cameroni, D. Virgilio et al., The TOR and EGO protein complexes orchestrate microautophagy in yeast, Mol Cell, vol.19, issue.1, pp.15-26, 2005.

D. Gresham, V. M. Boer, A. Caudy, N. Ziv, N. J. Brandt et al., System-level analysis of genes and functions affecting survival during nutrient starvation in Saccharomyces cerevisiae, Genetics, vol.187, issue.1, pp.299-317, 2011.

S. A. Comyn, S. Flibotte, and M. T. , Recurrent background mutations in WHI2 impair proteostasis and degradation of misfolded cytosolic proteins in Saccharomyces cerevisiae, vol.7, p.28646136, 2017.

J. D. Thomas, Y. J. Zhang, Y. H. Wei, J. H. Cho, L. E. Morris et al., Rab1A is an mTORC1 activator and a colorectal oncogene, Cancer Cell, vol.26, issue.5, pp.754-69, 2014.

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, pp.678-83, 2011.

D. C. Goberdhan, C. Wilson, and H. A. , Amino Acid Sensing by mTORC1: Intracellular Transporters Mark the Spot, Cell Metab, vol.23, issue.4, pp.580-589, 2016.

I. Yofe, U. Weill, M. Meurer, S. Chuartzman, E. Zalckvar et al., One library to make them all: streamlining the creation of yeast libraries via a SWAp-Tag strategy, Nat Methods, vol.13, issue.4, pp.371-378, 2016.

M. Kousi, V. Anttila, A. Schulz, S. Calafato, E. Jakkula et al., Novel mutations consolidate KCTD7 as a progressive myoclonus epilepsy gene, J Med Genet, vol.49, issue.6, pp.391-400, 2012.

J. Brechbiel, K. Miller-moslin, and A. A. Adjei, Crosstalk between hedgehog and other signaling pathways as a basis for combination therapies in cancer, Cancer Treat Rev, vol.40, issue.6, pp.750-759, 2014.

D. D. Shoemaker, D. A. Lashkari, D. Morris, M. Mittmann, and D. R. , Quantitative phenotypic analysis of yeast deletion mutants using a highly parallel molecular bar-coding strategy, Nat Genet, vol.14, issue.4, pp.450-456, 1996.

R. D. Gietz and A. Sugino, New yeast-Escherichia coli shuttle vectors constructed with in vitro mutagenized yeast genes lacking six-base pair restriction sites, Gene, vol.74, issue.2, pp.527-561, 1988.

C. B. Brachmann, A. Davies, G. J. Cost, E. Caputo, J. Li et al., Designer deletion strains derived from Saccharomyces cerevisiae S288C: a useful set of strains and plasmids for PCRmediated gene disruption and other applications, Yeast, vol.14, issue.2, pp.1097-0061, 1998.

R. S. Sikorski and P. Hieter, A system of shuttle vectors and yeast host strains designed for efficient manipulation of DNA in Saccharomyces cerevisiae, Genetics, vol.122, issue.1, pp.19-27, 1989.

H. Benedetti, S. Raths, F. Crausaz, and H. Riezman, The END3 gene encodes a protein that is required for the internalization step of endocytosis and for actin cytoskeleton organization in yeast, Mol Biol Cell, vol.5, issue.9, pp.1023-1060, 1994.
URL : https://hal.archives-ouvertes.fr/hal-02130204

S. Correale, L. Pirone, D. Marcotullio, L. De-smaele, E. Greco et al., Molecular organization of the cullin E3 ligase adaptor KCTD11, Biochimie, vol.93, issue.4, pp.715-739, 2011.