I. Versteege, Truncating mutations of hSNF5/INI1 in aggressive paediatric cancer, Nature, vol.394, pp.203-206, 1998.

B. G. Wilson, Epigenetic antagonism between polycomb and SWI/SNF complexes during oncogenic transformation, Cancer Cell, vol.18, pp.316-328, 2010.

O. Warburg, On the origin of cancer cells, Science, vol.123, pp.309-314, 1956.

N. N. Pavlova and C. B. Thompson, The Emerging Hallmarks of Cancer Metabolism, Cell Metab, vol.23, pp.27-47, 2016.

J. W. Locasale, Phosphoglycerate dehydrogenase diverts glycolytic flux and contributes to oncogenesis, Nat. Genet, vol.43, pp.869-874, 2011.

R. Possemato, Functional genomics reveal that the serine synthesis pathway is essential in breast cancer, Nature, vol.476, pp.346-350, 2011.

K. R. Mattaini, M. R. Sullivan, and M. G. Vander-heiden, The importance of serine metabolism in cancer, J. Cell Biol, vol.214, pp.249-257, 2016.

E. Mullarky, K. R. Mattaini, M. G. Vander-heiden, L. C. Cantley, and J. W. Locasale, PHGDH amplification and altered glucose metabolism in human melanoma, Pigment Cell Melanoma Res, vol.24, pp.1112-1115, 2011.

S. Yoon, Clinical Implication of Serine Metabolism-Associated Enzymes in Colon Cancer, Oncology, vol.89, pp.351-359, 2015.

Y. Ye, EBV-miR-BART1 is involved in regulating metabolism-associated genes in nasopharyngeal carcinoma, Biochem. Biophys. Res. Commun, vol.436, pp.19-24, 2013.

Z. Jing, Downregulation of phosphoglycerate dehydrogenase inhibits proliferation and enhances cisplatin sensitivity in cervical adenocarcinoma cells by regulating Bcl-2 and caspase-3, Cancer Biol. Ther, vol.16, pp.541-548, 2015.

J. Liu, Phosphoglycerate dehydrogenase induces glioma cells proliferation and invasion by stabilizing forkhead box M1, J. Neurooncol, vol.111, pp.245-255, 2013.

J. M. Tanner, EWS/FLI is a Master Regulator of Metabolic Reprogramming in Ewing Sarcoma, Mol. Cancer Res. MCR, vol.15, pp.1517-1530, 2017.

J. W. Locasale and . Serine, glycine and one-carbon units: cancer metabolism in full circle, Nat. Rev. Cancer, vol.13, pp.572-583, 2013.

G. S. Ducker and J. D. Rabinowitz, One-Carbon Metabolism in Health and Disease, Cell Metab, vol.25, pp.27-42, 2017.

S. Medjkane, E. Novikov, I. Versteege, and O. Delattre, The tumor suppressor hSNF5/INI1 modulates cell growth and actin cytoskeleton organization, Cancer Res, vol.64, pp.3406-3413, 2004.

A. Subramanian, Gene set enrichment analysis: a knowledge-based approach for interpreting genome-wide expression profiles, PNAS, vol.102, pp.15545-15550, 2005.

R. T. Nakayama, SMARCB1 is required for widespread BAF complex-mediated activation of enhancers and bivalent promoters, Nat. Genet, 2017.

P. D. Johann, Atypical Teratoid/Rhabdoid Tumors Are Comprised of Three Epigenetic Subgroups with Distinct Enhancer Landscapes, Cancer Cell, vol.29, pp.379-393, 2016.

M. Somel, Transcriptional neoteny in the human brain, PNAS, vol.106, pp.5743-5748, 2009.

J. Xia and D. S. Wishart, MSEA: a web-based tool to identify biologically meaningful patterns in quantitative metabolomic data, Nucleic Acids Res, vol.38, pp.71-77, 2010.

M. E. Pacold, A PHGDH inhibitor reveals coordination of serine synthesis and onecarbon unit fate, Nat. Chem. Biol, vol.12, pp.452-458, 2016.

C. Kadoch, Dynamics of BAF-Polycomb complex opposition on heterochromatin in normal and oncogenic states, Nat. Genet, vol.49, pp.213-222, 2017.

X. Wang, SMARCB1-mediated SWI/SNF complex function is essential for enhancer regulation, Nat. Genet, vol.49, pp.289-295, 2017.

S. S. Sabharwal and P. T. Schumacker, Mitochondrial ROS in cancer: initiators, amplifiers or an Achilles' heel?, Nat. Rev. Cancer, vol.14, pp.709-721, 2014.

X. Sun, Arid1a Has Context-Dependent Oncogenic and Tumor Suppressor Functions in Liver Cancer, Cancer Cell, vol.32, p.6, 2017.

H. E. Chun, Genome-Wide Profiles of Extra-cranial Malignant Rhabdoid Tumors Reveal Heterogeneity and Dysregulated Developmental Pathways, Cancer Cell, vol.29, pp.394-406, 2016.

C. F. Labuschagne, N. J. Van-den-broek, G. M. Mackay, K. H. Vousden, and O. Maddocks,

K. Serine, but not glycine, supports one-carbon metabolism and proliferation of cancer cells, Cell Rep, vol.7, pp.1248-1258, 2014.

E. Abrams, L. Neigeborn, and M. Carlson, Molecular analysis of SNF2 and SNF5, genes required for expression of glucose-repressible genes in Saccharomyces cerevisiae, Mol. Cell. Biol, vol.6, pp.3643-3651, 1986.

A. Agaimy, M. Koch, M. Lell, S. Semrau, W. Dudek et al., SMARCB1(INI1)-deficient sinonasal basaloid carcinoma: a novel member of the expanding family of SMARCB1-deficient neoplasms, Am. J. Surg. Pathol, vol.38, pp.1274-1281, 2014.

A. Agaimy, A. Hartmann, C. R. Antonescu, S. I. Chiosea, S. K. El-mofty et al., SMARCB1 (INI-1)-deficient Sinonasal Carcinoma: A Series of 39 Cases Expanding the Morphologic and Clinicopathologic Spectrum of a Recently Described Entity, Am. J. Surg. Pathol, vol.41, pp.458-471, 2017.

B. Akhtar-zaidi, R. Cowper-sal-lari, O. Corradin, A. Saiakhova, C. F. Bartels et al., Epigenomic enhancer profiling defines a signature of colon cancer, Science, vol.336, pp.736-739, 2012.

B. C. Altenburg, D. P. Via, and S. H. Steiner, Modification of the phenotype of murine sarcoma virus-transformed cells by sodium butyrate. Effects on morphology and cytoskeletal elements, Exp. Cell Res, vol.102, pp.223-231, 1976.

M. Antonelli, A. Raso, S. Mascelli, M. Gessi, P. Nozza et al., SMARCB1/INI1 Involvement in Pediatric Chordoma: A Mutational and Immunohistochemical Analysis, Am. J. Surg. Pathol, vol.41, pp.56-61, 2017.

N. Attar and S. K. Kurdistani, Exploitation of EP300 and CREBBP Lysine Acetyltransferases by Cancer. Cold Spring Harb, Perspect. Med, vol.7, 2017.

R. Banerjee, S. J. Bultman, D. Holley, C. Hillhouse, J. R. Bain et al., Non-targeted metabolomics of Brg1/Brm double-mutant cardiomyocytes reveals a novel role for SWI/SNF complexes in metabolic homeostasis, Metabolomics Off. J. Metabolomic Soc, vol.11, pp.1287-1301, 2015.

J. Banerji, S. Rusconi, and W. Schaffner, Expression of a beta-globin gene is enhanced by remote SV40 DNA sequences, Cell, vol.27, pp.299-308, 1981.

A. Barski, S. Cuddapah, K. Cui, T. Roh, D. E. Schones et al., High-resolution profiling of histone methylations in the human genome, Cell, vol.129, pp.823-837, 2007.

S. L. Berger and P. Sassone-corsi, Metabolic Signaling to Chromatin, Cold Spring Harb. Perspect. Biol, vol.8, 2016.

B. L. Betz, M. W. Strobeck, D. N. Reisman, E. S. Knudsen, and B. E. Weissman, Reexpression of hSNF5/INI1/BAF47 in pediatric tumor cells leads to G1 arrest associated with induction of p16ink4a and activation of RB, Oncogene, vol.21, pp.5193-5203, 2002.

J. A. Bishop, C. R. Antonescu, and W. H. Westra, SMARCB1 (INI-1)-deficient carcinomas of the sinonasal tract, Am. J. Surg. Pathol, vol.38, pp.1282-1289, 2014.

V. Boeva, C. Louis-brennetot, A. Peltier, S. Durand, C. Pierre-eugène et al., Heterogeneity of neuroblastoma cell identity defined by transcriptional circuitries, Nat. Genet, vol.49, pp.1408-1413, 2017.
URL : https://hal.archives-ouvertes.fr/hal-01741718

L. C. Boffa, G. Vidali, R. S. Mann, A. , and V. G. , Suppression of histone deacetylation in vivo and in vitro by sodium butyrate, J. Biol. Chem, vol.253, pp.3364-3366, 1978.

C. Bossen, C. S. Murre, A. N. Chang, R. Mansson, H. Rodewald et al., The chromatin remodeler Brg1 activates enhancer repertoires to establish B cell identity and modulate cell growth, Nat. Immunol, vol.16, pp.775-784, 2015.

F. Bourdeaut, D. Lequin, L. Brugières, S. Reynaud, C. Dufour et al., Frequent hSNF5/INI1 germline mutations in patients with rhabdoid tumor, Clin. Cancer Res. Off. J. Am. Assoc. Cancer Res, vol.17, pp.31-38, 2011.

B. Brennan, C. Stiller, and F. Bourdeaut, Extracranial rhabdoid tumours: what we have learned so far and future directions, Lancet Oncol, vol.14, pp.329-336, 2013.

C. S. Bruggers, S. B. Bleyl, T. Pysher, P. Barnette, Z. Afify et al., , 2011.

, Clinicopathologic comparison of familial versus sporadic atypical teratoid/rhabdoid tumors (AT/RT) of the central nervous system. Pediatr, Blood Cancer, vol.56, pp.1026-1031

M. Bulger, G. , and M. , Functional and mechanistic diversity of distal transcription enhancers, Cell, vol.144, pp.327-339, 2011.

J. Calderaro, J. Masliah-planchon, W. Richer, L. Maillot, P. Maille et al., Balanced Translocations Disrupting SMARCB1, 2016.

, Are Hallmark Recurrent Genetic Alterations in Renal Medullary Carcinomas, Eur. Urol, vol.69, pp.1055-1061

M. I. Carlo, J. Chaim, S. Patil, Y. Kemel, A. M. Schram et al., Genomic Characterization of Renal Medullary Carcinoma and Treatment Outcomes, Clin. Genitourin. Cancer, 2017.

M. Carlson and D. Botstein, Two differentially regulated mRNAs with different 5' ends encode secreted with intracellular forms of yeast invertase, Cell, vol.28, pp.145-154, 1982.

M. Carlson, B. C. Osmond, and D. Botstein, Mutants of yeast defective in sucrose utilization, Genetics, vol.98, pp.25-40, 1981.

E. Ceccacci and S. Minucci, Inhibition of histone deacetylases in cancer therapy: lessons from leukaemia, Br. J. Cancer, vol.114, pp.605-611, 2016.

B. A. Chabner and T. G. Roberts, Timeline: Chemotherapy and the war on cancer, Nat. Rev. Cancer, vol.5, pp.65-72, 2005.

B. Chaneton, P. Hillmann, L. Zheng, A. C. Martin, O. D. Maddocks et al., Serine is a natural ligand and allosteric activator of pyruvate kinase M2, Nature, vol.491, pp.458-462, 2012.

J. X. Cheng, M. Tretiakova, C. Gong, S. Mandal, T. Krausz et al., Renal medullary carcinoma: rhabdoid features and the absence of INI1 expression as markers of aggressive behavior, Mod. Pathol. Off. J. U. S. Can. Acad. Pathol. Inc, vol.21, pp.647-652, 2008.

S. W. Cheng, K. P. Davies, E. Yung, R. J. Beltran, J. Yu et al., c-MYC interacts with INI1/hSNF5 and requires the SWI/SNF complex for transactivation function, Nat. Genet, vol.22, pp.102-105, 1999.

H. R. Christofk, M. G. Vander-heiden, M. H. Harris, A. Ramanathan, R. E. Gerszten et al., The M2 splice isoform of pyruvate kinase is important for cancer metabolism and tumour growth, Nature, vol.452, pp.230-233, 2008.

H. E. Chun, E. L. Lim, A. Heravi-moussavi, S. Saberi, K. L. Mungall et al., , 2016.

, Genome-Wide Profiles of Extra-cranial Malignant Rhabdoid Tumors Reveal Heterogeneity and Dysregulated Developmental Pathways, Cancer Cell, vol.29, pp.394-406

H. E. Chun, E. L. Lim, A. Heravi-moussavi, S. Saberi, K. L. Mungall et al., , 2016.

, Genome-Wide Profiles of Extra-cranial Malignant Rhabdoid Tumors Reveal Heterogeneity and Dysregulated Developmental Pathways, Cancer Cell, vol.29, pp.394-406

C. R. Clapier and B. R. Cairns, The biology of chromatin remodeling complexes, Annu. Rev. Biochem, vol.78, pp.273-304, 2009.

I. Comet, E. M. Riising, B. Leblanc, and K. Helin, Maintaining cell identity: PRC2-mediated regulation of transcription and cancer, Nat. Rev. Cancer, vol.16, pp.803-810, 2016.

E. A. Cossins, C. , and L. , Folates and one-carbon metabolism in plants and fungi, Phytochemistry, vol.45, pp.437-452, 1997.

J. Côté, J. Quinn, J. L. Workman, and C. L. Peterson, Stimulation of GAL4 derivative binding to nucleosomal DNA by the yeast SWI/SNF complex, Science, vol.265, pp.53-60, 1994.

M. P. Creyghton, A. W. Cheng, G. G. Welstead, T. Kooistra, B. W. Carey et al., Histone H3K27ac separates active from poised enhancers and predicts developmental state, Proc. Natl. Acad. Sci. U. S. A, vol.107, pp.21931-21936, 2010.

S. R. Daigle, E. J. Olhava, C. A. Therkelsen, A. Basavapathruni, L. Jin et al., Potent inhibition of DOT1L as treatment of MLL-fusion leukemia, Blood, vol.122, pp.1017-1025, 2013.

L. Dang, D. W. White, S. Gross, B. D. Bennett, M. A. Bittinger et al., Cancer-associated IDH1 mutations produce 2hydroxyglutarate, Nature, vol.462, pp.739-744, 2009.

S. M. Davidson, T. Papagiannakopoulos, B. A. Olenchock, J. E. Heyman, M. A. Keibler et al., Environment Impacts the Metabolic Dependencies of Ras-Driven Non, Small Cell Lung Cancer. Cell Metab, vol.23, pp.517-528, 2016.

M. A. Dawson and T. Kouzarides, Cancer epigenetics: from mechanism to therapy, Cell, vol.150, pp.12-27, 2012.

O. Delattre, J. Zucman, B. Plougastel, C. Desmaze, T. Melot et al., Gene fusion with an ETS DNA-binding domain caused by chromosome translocation in human tumours, Nature, vol.359, pp.162-165, 1992.

J. E. Delmore, G. C. Issa, M. E. Lemieux, P. B. Rahl, J. Shi et al., BET bromodomain inhibition as a therapeutic strategy to target c-Myc, Cell, vol.146, pp.904-917, 2011.

C. Dhalluin, J. E. Carlson, L. Zeng, C. He, A. K. Aggarwal et al., Structure and ligand of a histone acetyltransferase bromodomain, Nature, vol.399, pp.491-496, 1999.

Y. Drier, M. J. Cotton, K. E. Williamson, S. M. Gillespie, R. J. Ryan et al., An oncogenic MYB feedback loop drives alternate cell fates in adenoid cystic carcinoma, Nat. Genet, vol.48, pp.265-272, 2016.

A. Dutta, M. Sardiu, M. Gogol, J. Gilmore, D. Zhang et al., Composition and Function of Mutant Swi/Snf Complexes, Cell Rep, vol.18, pp.2124-2134, 2017.

J. Ernst, P. Kheradpour, T. S. Mikkelsen, N. Shoresh, L. D. Ward et al., Mapping and analysis of chromatin state dynamics in nine human cell types, Nature, vol.473, pp.43-49, 2011.

J. Fan, X. Teng, L. Liu, K. R. Mattaini, R. E. Looper et al., Human phosphoglycerate dehydrogenase produces the oncometabolite D-2-hydroxyglutarate, ACS Chem. Biol, vol.10, pp.510-516, 2015.

A. P. Feinberg and B. Tycko, The history of cancer epigenetics, Nat. Rev. Cancer, vol.4, pp.143-153, 2004.

M. E. Figueroa, O. Abdel-wahab, C. Lu, P. S. Ward, J. Patel et al., Leukemic IDH1 and IDH2 mutations result in a hypermethylation phenotype, disrupt TET2 function, and impair hematopoietic differentiation, Cancer Cell, vol.18, pp.553-567, 2010.

W. A. Flavahan, E. Gaskell, and B. E. Bernstein, Epigenetic plasticity and the hallmarks of cancer, Science, vol.357, 2017.

L. F. Gamwell, K. Gambaro, M. Merziotis, C. Crane, S. L. Arcand et al., Small cell ovarian carcinoma: genomic stability and responsiveness to therapeutics, Orphanet J. Rare Dis, vol.8, p.33, 2013.

W. Gao, W. Li, T. Xiao, X. S. Liu, and W. G. Kaelin, Inactivation of the PBRM1 tumor suppressor gene amplifies the HIF-response in VHL-/-clear cell renal carcinoma, Proc. Natl. Acad. Sci. U. S. A, vol.114, pp.1027-1032, 2017.

T. Van-groningen, J. Koster, L. J. Valentijn, D. A. Zwijnenburg, N. Akogul et al., Neuroblastoma is composed of two super-enhancer-associated differentiation states, Nat. Genet, vol.49, pp.1261-1266, 2017.

W. Gu and R. G. Roeder, Activation of p53 sequence-specific DNA binding by acetylation of the p53 C-terminal domain, Cell, vol.90, pp.595-606, 1997.

D. A. Guertin and D. M. Sabatini, Defining the role of mTOR in cancer, Cancer Cell, vol.12, pp.9-22, 2007.

C. J. Guidi, A. T. Sands, B. P. Zambrowicz, T. K. Turner, D. A. Demers et al., Disruption of Ini1 leads to peri-implantation lethality and tumorigenesis in mice, Mol. Cell. Biol, vol.21, pp.3598-3603, 2001.

K. D. Hadfield, W. G. Newman, N. L. Bowers, A. Wallace, C. Bolger et al., Molecular characterisation of SMARCB1 and NF2 in familial and sporadic schwannomatosis, J. Med. Genet, vol.45, pp.332-339, 2008.

Z. Y. Han, W. Richer, P. Fréneaux, C. Chauvin, C. Lucchesi et al., The occurrence of intracranial rhabdoid tumours in mice depends on temporal control of Smarcb1 inactivation, Nat. Commun, vol.7, p.10421, 2016.

D. Hanahan and R. A. Weinberg, The hallmarks of cancer, Cell, vol.100, pp.57-70, 2000.

D. Hanahan and R. A. Weinberg, Hallmarks of cancer: the next generation, Cell, vol.144, pp.646-674, 2011.

D. C. Hargreaves and G. R. Crabtree, ATP-dependent chromatin remodeling: genetics, genomics and mechanisms, Cell Res, vol.21, pp.396-420, 2011.
DOI : 10.1038/cr.2011.32

URL : https://www.nature.com/articles/cr201132.pdf

M. Hasselblatt, S. Gesk, F. Oyen, S. Rossi, E. Viscardi et al., Nonsense mutation and inactivation of SMARCA4 (BRG1) in an atypical teratoid/rhabdoid tumor showing retained SMARCB1 (INI1) expression, Am. J. Surg. Pathol, vol.35, pp.933-935, 2011.

M. Hasselblatt, I. Nagel, F. Oyen, K. Bartelheim, R. B. Russell et al., SMARCA4-mutated atypical teratoid/rhabdoid tumors are associated with inherited germline alterations and poor prognosis, Acta Neuropathol, vol.128, pp.453-456, 2014.
DOI : 10.1007/s00401-014-1323-x

M. Hasselblatt, C. Thomas, V. Hovestadt, D. Schrimpf, P. Johann et al., Poorly differentiated chordoma with SMARCB1/INI1 loss: a distinct molecular entity with dismal prognosis, Acta Neuropathol, vol.132, pp.149-151, 2016.

K. Hata, M. Okano, H. Lei, L. , and E. , Dnmt3L cooperates with the Dnmt3 family of de novo DNA methyltransferases to establish maternal imprints in mice, Dev. Camb. Engl, vol.129, pp.1983-1993, 2002.

Y. He, B. Li, Z. Li, P. Liu, Y. Wang et al., , 2011.

, Tet-mediated formation of 5-carboxylcytosine and its excision by TDG in mammalian DNA, Science, vol.333, pp.1303-1307

N. D. Heintzman, R. K. Stuart, G. Hon, Y. Fu, C. W. Ching et al., Distinct and predictive chromatin signatures of transcriptional promoters and enhancers in the human genome, Nat. Genet, vol.39, pp.311-318, 2007.

N. D. Heintzman, G. C. Hon, R. D. Hawkins, P. Kheradpour, A. Stark et al., Histone modifications at human enhancers reflect global celltype-specific gene expression, Nature, vol.459, pp.108-112, 2009.
DOI : 10.1038/nature07829

URL : http://europepmc.org/articles/pmc2910248?pdf=render

N. Heisterkamp, K. Stam, J. Groffen, A. De-klein, G. et al., Structural organization of the bcr gene and its role in the Ph' translocation, Nature, vol.315, pp.758-761, 1985.

J. G. Herman, F. Latif, Y. Weng, M. I. Lerman, B. Zbar et al., Silencing of the VHL tumor-suppressor gene by DNA methylation in renal carcinoma, Proc. Natl. Acad. Sci. U. S. A, vol.91, pp.9700-9704, 1994.

D. Hnisz, B. J. Abraham, T. I. Lee, A. Lau, V. Saint-andré et al., Super-enhancers in the control of cell identity and disease, Cell, vol.155, pp.934-947, 2013.

K. Von-hoff, B. Hinkes, E. Dannenmann-stern, A. O. Von-bueren, M. Warmuth-metz et al., Frequency, risk-factors and survival of children with atypical teratoid rhabdoid tumors (AT/RT) of the CNS diagnosed between 1988 and 2004, and registered to the German HIT database, Pediatr. Blood Cancer, vol.57, pp.978-985, 2011.

T. J. Hollmann and J. L. Hornick, INI1-deficient tumors: diagnostic features and molecular genetics, Am. J. Surg. Pathol, vol.35, pp.47-63, 2011.
DOI : 10.1097/pas.0b013e31822b325b

J. L. Hornick, P. Dal-cin, and C. D. Fletcher, Loss of INI1 expression is characteristic of both conventional and proximal-type epithelioid sarcoma, Am. J. Surg. Pathol, vol.33, pp.542-550, 2009.

H. A. Hou, Y. Y. Kuo, C. Y. Liu, W. C. Chou, M. C. Lee et al., DNMT3A mutations in acute myeloid leukemia: stability during disease evolution and clinical implications, Blood, vol.119, pp.559-568, 2012.

B. X. Huang, M. Akbar, K. Kevala, K. , and H. Y. , Phosphatidylserine is a critical modulator for Akt activation, J. Cell Biol, vol.192, pp.979-992, 2011.

H. Huang, B. R. Sabari, B. A. Garcia, C. D. Allis, and Y. Zhao, SnapShot: histone modifications, Cell, vol.159, pp.458-458, 2014.

N. G. Iyer, H. Ozdag, C. , and C. , p300/CBP and cancer, vol.23, pp.4225-4231, 2004.

Z. Jagani, E. L. Mora-blanco, C. G. Sansam, E. S. Mckenna, B. Wilson et al., Loss of the tumor suppressor Snf5 leads to aberrant activation of the Hedgehog-Gli pathway, Nat. Med, vol.16, pp.1429-1433, 2010.

A. K. Jain and M. C. Barton, Bromodomain Histone Readers and Cancer, J. Mol. Biol, vol.429, 2003.

A. Jambhekar, J. N. Anastas, and Y. Shi, Histone Lysine Demethylase Inhibitors. Cold Spring Harb, Perspect. Med, vol.7, 2017.

P. Jelinic, J. J. Mueller, N. Olvera, F. Dao, S. N. Scott et al., Recurrent SMARCA4 mutations in small cell carcinoma of the ovary, Nat. Genet, vol.46, pp.424-426, 2014.

C. H. Jeong, J. , and S. H. , Downregulation of Reactive Oxygen Species in Apoptosis, J. Cancer Prev, vol.21, pp.13-20, 2016.

D. Jia, R. Z. Jurkowska, X. Zhang, A. Jeltsch, and X. Cheng, Structure of Dnmt3a bound to Dnmt3L suggests a model for de novo DNA methylation, Nature, vol.449, pp.248-251, 2007.

V. Y. Jo and C. D. Fletcher, Epithelioid malignant peripheral nerve sheath tumor: clinicopathologic analysis of 63 cases, Am. J. Surg. Pathol, vol.39, pp.673-682, 2015.

P. D. Johann, S. Erkek, M. Zapatka, K. Kerl, I. Buchhalter et al., Atypical Teratoid/Rhabdoid Tumors Are Comprised of Three Epigenetic Subgroups with Distinct Enhancer Landscapes, Cancer Cell, vol.29, pp.379-393, 2016.

D. T. Jones, N. Jäger, M. Kool, T. Zichner, B. Hutter et al., Dissecting the genomic complexity underlying medulloblastoma, Nature, vol.488, pp.100-105, 2012.

M. Kaneda, M. Okano, K. Hata, T. Sado, N. Tsujimoto et al., Essential role for de novo DNA methyltransferase Dnmt3a in paternal and maternal imprinting, Nature, vol.429, pp.900-903, 2004.

Y. Kato, M. Kaneda, K. Hata, K. Kumaki, M. Hisano et al., Role of the Dnmt3 family in de novo methylation of imprinted and repetitive sequences during male germ cell development in the mouse, Hum. Mol. Genet, vol.16, pp.2272-2280, 2007.

A. Kinnaird, S. Zhao, K. E. Wellen, and E. D. Michelakis, Metabolic control of epigenetics in cancer, Nat. Rev. Cancer, vol.16, pp.694-707, 2016.

A. Klochendler-yeivin, L. Fiette, J. Barra, C. Muchardt, C. Babinet et al., The murine SNF5/INI1 chromatin remodeling factor is essential for embryonic development and tumor suppression, EMBO Rep, vol.1, pp.500-506, 2000.

C. M. Koch, R. M. Andrews, P. Flicek, S. C. Dillon, U. Karaöz et al., The landscape of histone modifications across 1% of the human genome in five human cell lines, Genome Res, vol.17, pp.691-707, 2007.

R. D. Kornberg, Chromatin structure: a repeating unit of histones and DNA, Science, vol.184, pp.868-871, 1974.

R. D. Kornberg, T. , and J. O. , Chromatin structure; oligomers of the histones, Science, vol.184, pp.865-868, 1974.

M. J. Koster, B. Snel, and H. T. Timmers, Genesis of chromatin and transcription dynamics in the origin of species, Cell, vol.161, pp.724-736, 2015.

S. Kudithipudi, J. , and A. , Role of somatic cancer mutations in human protein lysine methyltransferases, Biochim. Biophys. Acta, vol.1846, pp.366-379, 2014.

M. Kulis and M. Esteller, DNA methylation and cancer, Adv. Genet, vol.70, pp.27-56, 2010.

C. Kung, J. Hixon, S. Choe, K. Marks, S. Gross et al., Small molecule activation of PKM2 in cancer cells induces serine auxotrophy, Chem. Biol, vol.19, pp.1187-1198, 2012.

S. Kuwamoto, M. Matsushita, K. Takeda, N. Tanaka, Y. Endo et al., SMARCA4-deficient thoracic sarcoma: report of a case and insights into how to reach the diagnosis using limited samples and resources, Hum. Pathol, 2017.

M. S. Lawrence, P. Stojanov, P. Polak, G. V. Kryukov, K. Cibulskis et al., Mutational heterogeneity in cancer and the search for new cancer-associated genes, Nature, vol.499, pp.214-218, 2013.

L. Loarer, F. Watson, S. Pierron, G. De-montpreville, V. T. Ballet et al., SMARCA4 inactivation defines a group of undifferentiated thoracic malignancies transcriptionally related to BAF-deficient sarcomas, Nat. Genet, vol.47, pp.1200-1205, 2015.
URL : https://hal.archives-ouvertes.fr/hal-01684775

T. J. Ley, L. Ding, M. J. Walter, M. D. Mclellan, T. Lamprecht et al., DNMT3A mutations in acute myeloid leukemia, N. Engl. J. Med, vol.363, pp.2424-2433, 2010.

M. V. Liberti and J. W. Locasale, The Warburg Effect: How Does it Benefit Cancer Cells?, Trends Biochem. Sci, vol.41, pp.211-218, 2016.

G. Liou and P. Storz, Reactive oxygen species in cancer, Free Radic. Res, vol.44, pp.479-496, 2010.

J. Liu, S. Guo, Q. Li, L. Yang, Z. Xia et al., , 2013.

, Phosphoglycerate dehydrogenase induces glioma cells proliferation and invasion by stabilizing forkhead box M1, J. Neurooncol, vol.111, pp.245-255

J. W. Locasale, A. R. Grassian, T. Melman, C. A. Lyssiotis, K. R. Mattaini et al., Phosphoglycerate dehydrogenase diverts glycolytic flux and contributes to oncogenesis, Nat. Genet, vol.43, pp.869-874, 2011.

J. Lovén, H. A. Hoke, C. Y. Lin, A. Lau, D. A. Orlando et al., Selective inhibition of tumor oncogenes by disruption of super-enhancers, Cell, vol.153, pp.320-334, 2013.

K. Luger, A. W. Mäder, R. K. Richmond, D. F. Sargent, R. et al., Crystal structure of the nucleosome core particle at 2.8 A resolution, Nature, vol.389, pp.251-260, 1997.

C. Lussey-lepoutre, A. Buffet, A. P. Gimenez-roqueplo, and J. Favier, Mitochondrial Deficiencies in the Predisposition to Paraganglioma, 2017.

D. N. Mancini, D. I. Rodenhiser, P. J. Ainsworth, F. P. O'malley, S. M. Singh et al., CpG methylation within the 5' regulatory region of the BRCA1 gene is tumor specific and includes a putative CREB binding site, Oncogene, vol.16, pp.1161-1169, 1998.

B. Manoranjan, C. Venugopal, N. Mcfarlane, B. W. Doble, S. E. Dunn et al., Medulloblastoma stem cells: where development and cancer cross pathways, Pediatr. Res, vol.71, pp.516-522, 2012.

E. R. Mardis, L. Ding, D. J. Dooling, D. E. Larson, M. D. Mclellan et al., Recurring mutations found by sequencing an acute myeloid leukemia genome, N. Engl. J. Med, vol.361, pp.1058-1066, 2009.

U. E. Martinez-outschoorn, M. Peiris-pagés, R. G. Pestell, F. Sotgia, and M. P. Lisanti, Cancer metabolism: a therapeutic perspective, Nat. Rev. Clin. Oncol, vol.14, pp.11-31, 2017.

J. Masliah-planchon, I. Bièche, J. Guinebretière, F. Bourdeaut, D. et al., , 2015.

, SWI/SNF chromatin remodeling and human malignancies, Annu. Rev. Pathol, vol.10, pp.145-171

J. Masliah-planchon, M. Machet, P. Fréneaux, A. Jourdain, I. Mortemousque et al., SMARCA4-Mutated Atypical Teratoid/Rhabdoid Tumor with Retained BRG1 Expression. Pediatr, Blood Cancer, vol.63, pp.568-569, 2016.

J. M. Mato, F. J. Corrales, S. C. Lu, and M. A. Avila, S-Adenosylmethionine: a control switch that regulates liver function, FASEB J. Off. Publ. Fed. Am. Soc. Exp. Biol, vol.16, pp.15-26, 2002.

J. M. Mato, M. L. Martínez-chantar, and S. C. Lu, Methionine metabolism and liver disease, Annu. Rev. Nutr, vol.28, pp.273-293, 2008.

S. Medjkane, E. Novikov, I. Versteege, D. , and O. , The tumor suppressor hSNF5/INI1 modulates cell growth and actin cytoskeleton organization, Cancer Res, vol.64, pp.3406-3413, 2004.

A. Merlo, J. G. Herman, L. Mao, D. J. Lee, E. Gabrielson et al., 5' CpG island methylation is associated with transcriptional silencing of the tumour suppressor p16/CDKN2/MTS1 in human cancers, Nat. Med, vol.1, pp.686-692, 1995.

D. Milani, F. M. Manzoni, L. Pezzani, P. Ajmone, C. Gervasini et al., Rubinstein-Taybi syndrome: clinical features, genetic basis, diagnosis, and management, Ital. J. Pediatr, vol.41, p.4, 2015.

J. Min, Q. Feng, Z. Li, Y. Zhang, and R. M. Xu, Structure of the catalytic domain of human DOT1L, a non-SET domain nucleosomal histone methyltransferase, Cell, vol.112, pp.711-723, 2003.

B. C. Mobley, J. K. Mckenney, C. D. Bangs, K. Callahan, K. W. Yeom et al., Loss of SMARCB1/INI1 expression in poorly differentiated chordomas, Acta Neuropathol, vol.120, pp.745-753, 2010.

P. Modena, E. Lualdi, F. Facchinetti, L. Galli, M. R. Teixeira et al., , 2005.

, SMARCB1/INI1 tumor suppressor gene is frequently inactivated in epithelioid sarcomas, Cancer Res, vol.65, pp.4012-4019

E. L. Mora-blanco, Y. Mishina, E. J. Tillman, Y. Cho, C. S. Thom et al., Activation of ?-catenin/TCF targets following loss of the tumor suppressor SNF5, Oncogene, vol.33, pp.933-938, 2014.

L. Morera, M. Lübbert, J. , and M. , Targeting histone methyltransferases and demethylases in clinical trials for cancer therapy, Clin. Epigenetics, vol.8, p.57, 2016.

M. A. Morgan and A. Shilatifard, Drosophila SETs its sights on cancer: Trr/MLL3/4 COMPASS-like complexes in development and disease, Mol. Cell. Biol, vol.33, pp.1698-1701, 2013.

M. A. Morgan and A. Shilatifard, Chromatin signatures of cancer, Genes Dev, vol.29, pp.238-249, 2015.

I. M. Morison, C. J. Paton, and S. D. Cleverley, The imprinted gene and parent-of-origin effect database, Nucleic Acids Res, vol.29, pp.275-276, 2001.

E. Mullarky, N. C. Lucki, R. Beheshti-zavareh, J. L. Anglin, A. P. Gomes et al., Identification of a small molecule inhibitor of 3-phosphoglycerate dehydrogenase to target serine biosynthesis in cancers, Proc. Natl. Acad. Sci. U. S. A, vol.113, pp.1778-1783, 2016.

R. T. Nakayama, J. L. Pulice, A. M. Valencia, M. J. Mcbride, Z. M. Mckenzie et al., SMARCB1 is required for widespread BAF complex-mediated activation of enhancers and bivalent promoters, Nat. Genet, 2017.

A. M. Nargund, C. G. Pham, Y. Dong, P. I. Wang, H. U. Osmangeyoglu et al., The SWI/SNF Protein PBRM1 Restrains VHL-Loss-Driven Clear Cell Renal Cell Carcinoma, Cell Rep, vol.18, pp.2893-2906, 2017.

B. Ning, W. Li, W. Zhao, W. , and R. , Targeting epigenetic regulations in cancer, Acta Biochim. Biophys. Sin, vol.48, pp.97-109, 2016.

P. A. Northcott, D. T. Jones, M. Kool, G. W. Robinson, R. J. Gilbertson et al., Medulloblastomics: the end of the beginning, Nat. Rev. Cancer, vol.12, pp.818-834, 2012.

P. A. Northcott, I. Buchhalter, A. S. Morrissy, V. Hovestadt, J. Weischenfeldt et al., The whole-genome landscape of medulloblastoma subtypes, Nature, vol.547, pp.311-317, 2017.

M. Okano, D. W. Bell, D. A. Haber, L. , and E. , DNA methyltransferases Dnmt3a and Dnmt3b are essential for de novo methylation and mammalian development, Cell, vol.99, pp.247-257, 1999.

J. W. Orr, N. , and A. C. , Interaction of protein kinase C with phosphatidylserine. 2. Specificity and regulation, Biochemistry (Mosc.), vol.31, pp.4667-4673, 1992.

M. E. Pacold, K. R. Brimacombe, S. H. Chan, J. M. Rohde, C. A. Lewis et al., A PHGDH inhibitor reveals coordination of serine synthesis and one-carbon unit fate, Nat. Chem. Biol, vol.12, pp.452-458, 2016.

H. E. Pelish, B. B. Liau, I. I. Nitulescu, A. Tangpeerachaikul, Z. C. Poss et al., Mediator kinase inhibition further activates super-enhancer-associated genes in AML, Nature, vol.526, pp.273-276, 2015.

J. Peters, The role of genomic imprinting in biology and disease: an expanding view, Nat. Rev. Genet, vol.15, pp.517-530, 2014.

R. Popovic and J. D. Licht, Emerging epigenetic targets and therapies in cancer medicine, Cancer Discov, vol.2, pp.405-413, 2012.

R. Possemato, K. M. Marks, Y. D. Shaul, M. E. Pacold, D. Kim et al., Functional genomics reveal that the serine synthesis pathway is essential in breast cancer, Nature, vol.476, pp.346-350, 2011.

T. J. Pugh, S. D. Weeraratne, T. C. Archer, D. A. Pomeranz-krummel, D. Auclair et al., Medulloblastoma exome sequencing uncovers subtype-specific somatic mutations, Nature, vol.488, pp.106-110, 2012.

A. Puissant, S. M. Frumm, G. Alexe, C. F. Bassil, J. Qi et al., Targeting MYCN in neuroblastoma by BET bromodomain inhibition, Cancer Discov, vol.3, pp.308-323, 2013.

P. Ramos, A. N. Karnezis, D. W. Craig, A. Sekulic, M. L. Russell et al., Small cell carcinoma of the ovary, hypercalcemic type, displays frequent inactivating germline and somatic mutations in SMARCA4, Nat. Genet, vol.46, pp.427-429, 2014.

R. C. Rao and Y. Dou, Hijacked in cancer: the KMT2 (MLL) family of methyltransferases, Nat. Rev. Cancer, vol.15, pp.334-346, 2015.

K. D. Rasmussen, G. Jia, J. V. Johansen, M. T. Pedersen, N. Rapin et al., Loss of TET2 in hematopoietic cells leads to DNA hypermethylation of active enhancers and induction of leukemogenesis, Genes Dev, vol.29, pp.910-922, 2015.

W. Richer, J. Masliah-planchon, N. Clement, I. Jimenez, L. Maillot et al., Embryonic signature distinguishes pediatric and adult rhabdoid tumors from other SMARCB1-deficient cancers, Oncotarget, vol.8, pp.34245-34257, 2017.

M. G. Riggs, R. G. Whittaker, J. R. Neumann, and V. M. Ingram, n-Butyrate causes histone modification in HeLa and Friend erythroleukaemia cells, Nature, vol.268, pp.462-464, 1977.

C. W. Roberts, S. A. Galusha, M. E. Mcmenamin, C. D. Fletcher, and S. H. Orkin, , 2000.

, Haploinsufficiency of Snf5 (integrase interactor 1) predisposes to malignant rhabdoid tumors in mice, Proc. Natl. Acad. Sci. U. S. A, vol.97, pp.13796-13800

G. Robinson, M. Parker, T. A. Kranenburg, C. Lu, X. Chen et al., Novel mutations target distinct subgroups of medulloblastoma, Nature, vol.488, pp.43-48, 2012.

D. Rohle, J. Popovici-muller, N. Palaskas, S. Turcan, C. Grommes et al., An inhibitor of mutant IDH1 delays growth and promotes differentiation of glioma cells, Science, vol.340, pp.626-630, 2013.

S. Y. Roth, J. M. Denu, A. , and C. D. , Histone acetyltransferases, Annu. Rev. Biochem, vol.70, pp.81-120, 2001.

J. L. Sauter, R. P. Graham, B. T. Larsen, S. M. Jenkins, A. C. Roden et al., , 2017.

, SMARCA4-deficient thoracic sarcoma: a distinctive clinicopathological entity with undifferentiated rhabdoid morphology and aggressive behavior

R. Schneppenheim, M. C. Frühwald, S. Gesk, M. Hasselblatt, A. Jeibmann et al., Germline nonsense mutation and somatic inactivation of SMARCA4/BRG1 in a family with rhabdoid tumor predisposition syndrome, Am. J. Hum. Genet, vol.86, pp.279-284, 2010.

L. Scourzic, E. Mouly, and O. A. Bernard, TET proteins and the control of cytosine demethylation in cancer, Genome Med, vol.7, p.9, 2015.

G. L. Semenza, HIF-1: upstream and downstream of cancer metabolism, Curr. Opin. Genet. Dev, vol.20, pp.51-56, 2010.

P. Sen, J. Luo, A. Hada, S. G. Hailu, M. L. Dechassa et al., Loss of Snf5 Induces Formation of an Aberrant SWI/SNF Complex, Cell Rep, vol.18, pp.2135-2147, 2017.

J. Shi, E. Wang, J. P. Milazzo, Z. Wang, J. B. Kinney et al., Discovery of cancer drug targets by CRISPR-Cas9 screening of protein domains, Nat. Biotechnol, vol.33, pp.661-667, 2015.

Y. Shi, F. Lan, C. Matson, P. Mulligan, J. R. Whetstine et al., Histone demethylation mediated by the nuclear amine oxidase homolog LSD1, Cell, vol.119, pp.941-953, 2004.

H. U. Simon, A. Haj-yehia, L. , and F. , Role of reactive oxygen species (ROS) in apoptosis induction, Apoptosis Int. J. Program. Cell Death, vol.5, pp.415-418, 2000.

D. J. Simpson, N. A. Hibberts, A. M. Mcnicol, R. N. Clayton, and W. E. Farrell, Loss of pRb expression in pituitary adenomas is associated with methylation of the RB1 CpG island, Cancer Res, vol.60, pp.1211-1216, 2000.

M. J. Smith, A. J. Wallace, N. L. Bowers, H. Eaton, and D. G. Evans, SMARCB1 mutations in schwannomatosis and genotype correlations with rhabdoid tumors, Cancer Genet, vol.207, pp.373-378, 2014.

B. Z. Stanton, C. Hodges, J. P. Calarco, S. M. Braun, W. L. Ku et al., Smarca4 ATPase mutations disrupt direct eviction of PRC1 from chromatin, Nat. Genet, vol.49, pp.282-288, 2017.

D. J. Steger, M. I. Lefterova, L. Ying, A. J. Stonestrom, M. Schupp et al., DOT1L/KMT4 recruitment and H3K79 methylation are ubiquitously coupled with gene transcription in mammalian cells, Mol. Cell. Biol, vol.28, pp.2825-2839, 2008.

X. Su, K. E. Wellen, and J. D. Rabinowitz, Metabolic control of methylation and acetylation, Curr. Opin. Chem. Biol, vol.30, pp.52-60, 2016.

X. Sun, S. C. Wang, Y. Wei, X. Luo, Y. Jia et al., Arid1a Has Context-Dependent Oncogenic and Tumor Suppressor Functions in Liver Cancer, Cancer Cell, vol.32, pp.574-589, 2017.

M. Tahiliani, K. P. Koh, Y. Shen, W. A. Pastor, H. Bandukwala et al., Conversion of 5-methylcytosine to 5hydroxymethylcytosine in mammalian DNA by MLL partner TET1, Science, vol.324, pp.930-935, 2009.

J. M. Tanner, C. Bensard, P. Wei, N. M. Krah, J. C. Schell et al., EWS/FLI is a Master Regulator of Metabolic Reprogramming in Ewing Sarcoma, Mol. Cancer Res. MCR, 2017.

M. D. Taylor, P. A. Northcott, A. Korshunov, M. Remke, Y. J. Cho et al., Molecular subgroups of medulloblastoma: the current consensus, Acta Neuropathol, vol.123, pp.465-472, 2012.

N. Traverso, R. Ricciarelli, M. Nitti, B. Marengo, A. L. Furfaro et al., Role of glutathione in cancer progression and chemoresistance, Oxid. Med. Cell. Longev, p.972913, 2013.

J. E. Unterlass, A. Baslé, T. J. Blackburn, J. Tucker, C. Cano et al., Validating and enabling phosphoglycerate dehydrogenase (PHGDH) as a target for fragmentbased drug discovery in PHGDH-amplified breast cancer, Oncotarget, 2016.
DOI : 10.18632/oncotarget.11487

URL : http://www.oncotarget.com/index.php?journal=oncotarget&page=article&op=view&path%5B%5D=11487&path%5B%5D=36386

N. L. Valtz, T. E. Hayes, T. Norregaard, S. M. Liu, and R. D. Mckay, An embryonic origin for medulloblastoma, New Biol, vol.3, pp.364-371, 1991.

M. G. Vander-heiden, L. C. Cantley, and C. B. Thompson, Understanding the Warburg effect: the metabolic requirements of cell proliferation, Science, vol.324, pp.1029-1033, 2009.

I. Versteege, N. Sévenet, J. Lange, M. F. Rousseau-merck, P. Ambros et al., Truncating mutations of hSNF5/INI1 in aggressive paediatric cancer, Nature, vol.394, pp.203-206, 1998.
DOI : 10.1038/28212

N. Vié, V. Copois, C. Bascoul-mollevi, V. Denis, N. Bec et al., Overexpression of phosphoserine aminotransferase PSAT1 stimulates cell growth and increases chemoresistance of colon cancer cells, Mol. Cancer, vol.7, p.14, 2008.

A. Visel, E. M. Rubin, and L. A. Pennacchio, Genomic views of distant-acting enhancers, Nature, vol.461, pp.199-205, 2009.
DOI : 10.1038/nature08451

URL : http://europepmc.org/articles/pmc2923221?pdf=render

J. Vitte, F. Gao, G. Coppola, A. R. Judkins, G. et al., Transcriptional repression mediated by the human polycombgroup protein EED involves histone deacetylation, Nat. Commun, vol.23, pp.474-478, 1999.

P. Voigt, W. Tee, R. , and D. , A double take on bivalent promoters, Genes Dev, vol.27, pp.1318-1338, 2013.
DOI : 10.1101/gad.219626.113

URL : http://genesdev.cshlp.org/content/27/12/1318.full.pdf

F. Wang, J. Travins, B. Delabarre, V. Penard-lacronique, S. Schalm et al., Targeted inhibition of mutant IDH2 in leukemia cells induces cellular differentiation, Science, vol.340, pp.622-626, 2013.

Q. Wang, M. V. Liberti, P. Liu, X. Deng, Y. Liu et al., Rational Design of Selective Allosteric Inhibitors of PHGDH and Serine Synthesis with Anti-tumor Activity, Cell Chem. Biol, vol.24, pp.55-65, 2017.

X. Wang, R. S. Lee, B. H. Alver, J. R. Haswell, S. Wang et al., SMARCB1-mediated SWI/SNF complex function is essential for enhancer regulation, Nat. Genet, vol.49, pp.289-295, 2017.
DOI : 10.1038/ng.3746

URL : http://europepmc.org/articles/pmc5285474?pdf=render

O. Warburg, On the origin of cancer cells, Science, vol.123, pp.309-314, 1956.

O. Warburg, F. Wind, and E. Negelein, THE METABOLISM OF TUMORS IN THE BODY, J. Gen. Physiol, vol.8, pp.519-530, 1927.

D. A. Weeks, J. B. Beckwith, G. W. Mierau, and D. W. Luckey, Rhabdoid tumor of kidney. A report of 111 cases from the National Wilms' Tumor Study Pathology Center, Am. J. Surg. Pathol, vol.13, pp.439-458, 1989.
DOI : 10.1097/00000478-198906000-00001

W. A. Whyte, D. A. Orlando, D. Hnisz, B. J. Abraham, C. Y. Lin et al., Master transcription factors and mediator establish super-enhancers at key cell identity genes, Cell, vol.153, pp.307-319, 2013.
DOI : 10.1016/j.cell.2013.03.035

URL : https://doi.org/10.1016/j.cell.2013.03.035

B. G. Wilson, X. Wang, X. Shen, E. S. Mckenna, M. E. Lemieux et al., Epigenetic antagonism between polycomb and SWI/SNF complexes during oncogenic transformation, Cancer Cell, vol.18, pp.316-328, 2010.
DOI : 10.1016/j.ccr.2011.01.016

URL : https://doi.org/10.1016/j.ccr.2011.01.016

F. Winston and M. Carlson, Yeast SNF/SWI transcriptional activators and the SPT/SIN chromatin connection, Trends Genet. TIG, vol.8, pp.387-391, 1992.
DOI : 10.1016/0168-9525(92)90167-3

D. R. Wise, R. J. Deberardinis, A. Mancuso, N. Sayed, X. Zhang et al., Myc regulates a transcriptional program that stimulates mitochondrial glutaminolysis and leads to glutamine addiction, Proc. Natl. Acad. Sci. U. S. A, vol.105, pp.18782-18787, 2008.
DOI : 10.1073/pnas.0810199105

URL : http://www.pnas.org/content/105/48/18782.full.pdf

L. Witkowski, E. Lalonde, J. Zhang, S. Albrecht, N. Hamel et al., Familial rhabdoid tumour "avant la lettre"-from pathology review to exome sequencing and back again, J. Pathol, vol.231, pp.35-43, 2013.
DOI : 10.1002/path.4225

L. Witkowski, J. Carrot-zhang, S. Albrecht, S. Fahiminiya, N. Hamel et al., Germline and somatic SMARCA4 mutations characterize small cell carcinoma of the ovary, hypercalcemic type, Nat. Genet, vol.46, pp.438-443, 2014.

Z. Wu, J. Connolly, and K. K. Biggar, Beyond histones-the expanding roles of protein lysine methylation, FEBS J, vol.284, pp.2732-2744, 2017.

S. Yamaguchi, L. Shen, Y. Liu, D. Sendler, and Y. Zhang, Role of Tet1 in erasure of genomic imprinting, Nature, vol.504, pp.460-464, 2013.

J. Yamazaki, J. Jelinek, Y. Lu, M. Cesaroni, J. Madzo et al., TET2 Mutations Affect Non-CpG Island DNA Methylation at Enhancers and Transcription Factor-Binding Sites in Chronic Myelomonocytic Leukemia, Cancer Res, vol.75, pp.2833-2843, 2015.

H. Yan, D. W. Parsons, G. Jin, R. Mclendon, B. A. Rasheed et al., IDH1 and IDH2 mutations in gliomas, N. Engl. J. Med, vol.360, pp.765-773, 2009.

W. Yang, L. , and Z. , Pyruvate kinase M2 at a glance, J. Cell Sci, vol.128, pp.1655-1660, 2015.
DOI : 10.1242/jcs.166629

URL : http://jcs.biologists.org/content/128/9/1655.full.pdf

Y. Yang, J. Wu, J. Cai, Z. He, J. Yuan et al., PSAT1 regulates cyclin D1 degradation and sustains proliferation of non-small cell lung cancer cells, Int. J. Cancer, vol.136, pp.39-50, 2015.

X. Yao, J. Tan, K. J. Lim, J. Koh, W. F. Ooi et al., VHL deficiency drives enhancer activation of oncogenes in clear cell renal cell carcinoma, 2017.

J. Ye, A. Mancuso, X. Tong, P. S. Ward, J. Fan et al., , 2012.

, Pyruvate kinase M2 promotes de novo serine synthesis to sustain mTORC1 activity and cell proliferation, Proc. Natl. Acad. Sci. U. S. A, vol.109, pp.6904-6909

A. Yoshida, E. Kobayashi, T. Kubo, M. Kodaira, T. Motoi et al., Clinicopathological and molecular characterization of SMARCA4-deficient thoracic sarcomas with comparison to potentially related entities, Mod. Pathol. Off. J. U. S. Can. Acad. Pathol. Inc, vol.30, pp.797-809, 2017.

R. H. Young, E. Oliva, and R. E. Scully, Small cell carcinoma of the ovary, hypercalcemic type. A clinicopathological analysis of 150 cases, Am. J. Surg. Pathol, vol.18, pp.1102-1116, 1994.

E. A. Zaal, W. Wu, G. Jansen, S. Zweegman, J. Cloos et al., Bortezomib resistance in multiple myeloma is associated with increased serine synthesis, Cancer Metab, vol.5, p.7, 2017.

N. Zaware and M. Zhou, Chemical modulators for epigenome reader domains as emerging epigenetic therapies for cancer and inflammation, Curr. Opin. Chem. Biol, vol.39, pp.116-125, 2017.

J. Zhang, S. Vlasevska, V. A. Wells, S. Nataraj, A. B. Holmes et al., The CREBBP Acetyltransferase Is a Haploinsufficient Tumor Suppressor in B-cell Lymphoma, Cancer Discov, vol.7, pp.322-337, 2017.

L. Zhang, L. Deng, F. Chen, Y. Yao, B. Wu et al., Inhibition of histone H3K79 methylation selectively inhibits proliferation, self-renewal and metastatic potential of breast cancer, Oncotarget, vol.5, pp.10665-10677, 2014.

Z. Zhang, K. P. Davies, J. Allen, L. Zhu, R. G. Pestell et al., , 2002.

, Cell cycle arrest and repression of cyclin D1 transcription by INI1/hSNF5, Mol. Cell. Biol, vol.22, pp.5975-5988