N. J. Andersen, B. J. Nickoloff, K. J. Dykema, E. A. Boguslawski, R. I. Krivochenitser et al., Pharmacologic inhibition of MEK signaling prevents growth of canine hemangiosarcoma, Molecular Cancer Therapeutics, vol.12, pp.1701-1714, 2013.

S. Berghmans, R. D. Murphey, E. Wienholds, D. Neuberg, J. L. Kutok et al., Look AT. 2005. tp53 mutant zebrafish develop malignant peripheral nerve sheath tumors, PNAS, vol.102, pp.407-412

E. Y. Chen, M. T. Deran, M. S. Ignatius, K. B. Grandinetti, R. Clagg et al., Glycogen synthase kinase 3 inhibitors induce the canonical WNT/b-catenin pathway to suppress growth and self-renewal in embryonal Rhabdomyosarcoma, PNAS, vol.111, pp.5349-5354, 2014.

X. Chen, E. Stewart, A. A. Shelat, C. Qu, A. Bahrami et al., Targeting oxidative stress in embryonal Rhabdomyosarcoma, Cancer Cell, vol.24, pp.710-724, 2013.

S. Choorapoikayil, R. V. Kuiper, A. De-bruin, and J. Den-hertog, Haploinsufficiency of the genes encoding the tumor suppressor pten predisposes zebrafish to hemangiosarcoma, Disease Models & Mechanisms, vol.5, pp.241-247, 2012.

A. Cicalese, G. Bonizzi, C. E. Pasi, M. Faretta, S. Ronzoni et al., The tumor suppressor p53 regulates polarity of self-renewing divisions in mammary stem cells, Cell, vol.138, pp.1083-1095, 2009.

A. Dobin, C. A. Davis, F. Schlesinger, J. Drenkow, C. Zaleski et al., STAR: ultrafast universal RNA-seq aligner, Bioinformatics, vol.29, pp.15-21, 2013.

L. A. Donehower, M. Harvey, B. L. Slagle, M. J. Mcarthur, C. A. Montgomery et al., Mice deficient for p53 are developmentally normal but susceptible to spontaneous tumours, Nature, vol.356, pp.215-221, 1992.

M. Harvey, M. J. Mcarthur, C. A. Montgomery, J. S. Butel, A. Bradley et al., Spontaneous and carcinogen-induced tumorigenesis in p53-deficient mice, Nature Genetics, vol.5, pp.225-229, 1993.

M. N. Hayes, K. M. Mccarthy, J. A. Oliveira, M. L. Iyer, S. Garcia et al., Vangl2 regulates stem cell self-renewal programs and growth in Rhabdomyosarcoma through a wnt/PCP/RhoA signaling Axis, Cell Stem Cell, vol.22, pp.414-427, 2018.

M. S. Ignatius, E. Chen, N. M. Elpek, A. Z. Fuller, I. M. Tenente et al., In vivo imaging of tumor-propagating cells, regional tumor heterogeneity, and dynamic cell movements in embryonal Rhabdomyosarcoma, Cancer Cell, vol.21, pp.680-693, 2012.

M. S. Ignatius, M. N. Hayes, R. Lobbardi, E. Y. Chen, K. M. Mccarthy et al., The NOTCH1/SNAIL1/MEF2C pathway regulates growth and Self-Renewal in embryonal Rhabdomyosarcoma, Cell Reports, vol.19, pp.2304-2318, 2017.

T. Jacks, R. L. Williams, B. O. Schmitt, E. M. Halachmi, S. Bronson et al., Tumor spectrum analysis in p53-mutant mice, Current Biology, vol.4, pp.1-7, 1994.

E. R. Kastenhuber and S. W. Lowe, Putting p53 in Context, Cell, vol.170, pp.1062-1078, 2017.

K. Kawakami, A. Shima, and N. Kawakami, Identification of a functional transposase of the Tol2 element, an Aclike element from the japanese medaka fish, and its transposition in the zebrafish germ lineage, PNAS, vol.97, pp.11403-11408, 2000.

M. Kolberg, M. Høland, G. E. Lind, T. H. Å-gesen, R. I. Skotheim et al., Protein expression of BIRC5, TK1, and TOP2A in malignant peripheral nerve sheath tumours-A prognostic test after surgical resection, Molecular Oncology, vol.9, pp.1129-1139, 2015.

G. A. Lang, T. Iwakuma, Y. A. Suh, G. Liu, V. A. Rao et al., Gain of function of a p53 hot spot mutation in a mouse model of Li-Fraumeni syndrome, Cell, vol.119, pp.861-872, 2004.

D. M. Langenau, M. D. Keefe, N. Y. Storer, J. R. Guyon, J. L. Kutok et al., Effects of RAS on the genesis of embryonal Rhabdomyosarcoma, Genes & Development, vol.21, pp.1382-1395, 2007.

D. M. Langenau, M. D. Keefe, N. Y. Storer, C. A. Jette, A. C. Smith et al., Coinjection strategies to modify radiation sensitivity and tumor initiation in transgenic zebrafish, Oncogene, vol.27, pp.4242-4248, 2008.

A. Lavigueur, V. Maltby, D. Mock, J. Rossant, T. Pawson et al., High incidence of lung, bone, and lymphoid tumors in transgenic mice overexpressing mutant alleles of the p53 oncogene, Molecular and Cellular Biology, vol.9, pp.3982-3991, 1989.

J. M. Lee, J. L. Abrahamson, R. Kandel, L. A. Donehower, and A. Bernstein, Susceptibility to radiation-carcinogenesis and accumulation of chromosomal breakage in p53 deficient mice, Oncogene, vol.9, pp.3731-3736, 1994.

X. Liang and D. K. Graham, Natural killer cell neoplasms, Cancer, vol.112, pp.1425-1436, 2008.

Y. Liao, G. K. Smyth, and W. Shi, featureCounts: an efficient general purpose program for assigning sequence reads to genomic features, Bioinformatics, vol.30, pp.923-930, 2014.

M. I. Love, W. Huber, and S. Anders, Moderated estimation of fold change and dispersion for RNA-seq data with DESeq2, Genome Biology, vol.15, p.550, 2014.

A. C. Ma, H. B. Lee, K. J. Clark, and S. C. Ekker, High efficiency in vivo genome engineering with a simplified 15-RVD GoldyTALEN design, PloS One, vol.8, p.65259, 2013.

D. Malkin, Li-fraumeni syndrome, Genes & Cancer, vol.2, pp.475-484, 2011.
URL : https://hal.archives-ouvertes.fr/hal-01262929

K. Meletis, V. Wirta, S. M. Hede, . Nisté-r-m, J. Lundeberg et al., p53 suppresses the self-renewal of adult neural stem cells, Development, vol.133, pp.363-369, 2006.

I. V. Mizgireuv and S. Y. Revskoy, Transplantable tumor lines generated in clonal zebrafish, Cancer Research, vol.66, pp.3120-3125, 2006.

V. K. Mootha, C. M. Lindgren, K. Eriksson, A. Subramanian, S. Sihag et al., PGC-1a-responsive genes involved in oxidative phosphorylation are coordinately downregulated in human diabetes, Nature Genetics, vol.34, pp.267-273, 2003.

C. Mosimann, C. K. Kaufman, P. Li, E. K. Pugach, O. J. Tamplin et al., Ubiquitous transgene expression and Crebased recombination driven by the ubiquitin promoter in zebrafish, Development, vol.138, pp.169-177, 2011.

P. A. Muller and K. H. Vousden, Mutant p53 in Cancer: new functions and therapeutic opportunities, Cancer Cell, vol.25, pp.304-317, 2014.

K. L. Neff, D. P. Argue, A. C. Ma, H. B. Lee, K. J. Clark et al., Mojo hand, a TALEN design tool for genome editing applications, BMC Bioinformatics, vol.14, p.1, 2013.

J. C. Neumann, K. Lillard, V. Damoulis, and J. F. Amatruda, Zebrafish models of germ cell tumor, Methods in Cell Biology, vol.105, pp.1-24, 2011.

K. P. Olive, D. A. Tuveson, Z. C. Ruhe, B. Yin, N. A. Willis et al., Mutant p53 gain of function in two mouse models of Li-Fraumeni syndrome, Cell, vol.119, pp.847-860, 2004.

J. M. Parant, S. A. George, J. A. Holden, and H. J. Yost, Genetic modeling of Li-Fraumeni syndrome in zebrafish, Disease Models & Mechanisms, vol.3, pp.45-56, 2010.

M. Seki, R. Nishimura, K. Yoshida, T. Shimamura, Y. Shiraishi et al., Integrated genetic and epigenetic analysis defines novel molecular subgroups in Rhabdomyosarcoma, Nature Communications, vol.6, p.8557, 2015.

J. Shin, A. Padmanabhan, E. D. De-groh, J. S. Lee, S. Haidar et al., Zebrafish neurofibromatosis type 1 genes have redundant functions in tumorigenesis and embryonic development, Disease Models & Mechanisms, vol.5, pp.881-894, 2012.

A. C. Smith, A. R. Raimondi, C. D. Salthouse, M. S. Ignatius, J. S. Blackburn et al., High-throughput cell transplantation establishes that tumorinitiating cells are abundant in zebrafish T-cell acute lymphoblastic leukemia, Blood, vol.115, pp.3296-3303, 2010.

D. S. Soliman, A. A. Sabbagh, H. E. Omri, F. A. Ibrahim, A. M. Amer et al., Rare aggressive natural killer cell leukemia presented with bone marrow fibrosis -a diagnostic challenge, SpringerPlus, vol.3, p.390, 2014.

A. Subramanian, P. Tamayo, V. K. Mootha, S. Mukherjee, B. L. Ebert et al., Gene set enrichment analysis: a knowledge-based approach for interpreting genome-wide expression profiles, PNAS, vol.102, pp.15545-15550, 2005.

R. Suzuki and S. Nakamura, Malignancies of natural killer (NK) cell precursor: myeloid/NK cell precursor acute leukemia and blastic NK cell lymphoma/leukemia, Leukemia Research, vol.23, issue.98, pp.194-199, 1999.

Q. Tang, N. S. Abdelfattah, J. S. Blackburn, J. C. Moore, S. A. Martinez et al., Optimized cell transplantation using adult rag2 mutant zebrafish, Nature Methods, vol.11, pp.821-824, 2014.

Q. Tang, S. Iyer, R. Lobbardi, J. C. Moore, H. Chen et al., Dissecting hematopoietic and renal cell heterogeneity in adult zebrafish at single-cell resolution using RNA sequencing, The Journal of Experimental Medicine, vol.214, pp.2875-2887, 2017.

Q. Tang, J. C. Moore, M. S. Ignatius, I. M. Tenente, M. N. Hayes et al., Imaging tumour cell heterogeneity following cell transplantation into optically clear immune-deficient zebrafish, Nature Communications, vol.7, p.10358, 2016.

H. Taubert, A. Meye, and P. Wü, Prognosis is correlated with p53 mutation type for soft tissue sarcoma patients, Cancer Research, vol.56, pp.4134-4136, 1996.

A. C. Taylor, L. Shu, M. K. Danks, C. A. Poquette, S. Shetty et al., P53 mutation and MDM2 amplification frequency in pediatric rhabdomyosarcoma tumors and cell lines, Medical and Pediatric Oncology, vol.35, pp.96-103, 2000.

M. Tekippe, H. De, and C. J. , Expansion of hematopoietic stem cell phenotype and activity in Trp53-null mice, Experimental Hematology, vol.31, pp.521-527, 2003.

I. M. Tenente, M. N. Hayes, M. S. Ignatius, K. Mccarthy, M. Yohe et al., Myogenic regulatory transcription factors regulate growth in Rhabdomyosarcoma, vol.6, p.19214, 2017.

L. Wang, S. Wang, L. W. Wei, and L. , RSeQC: quality control of RNA-seq experiments, Bioinformatics, vol.28, pp.2184-2185, 2012.

M. Yagita, C. L. Huang, H. Umehara, Y. Matsuo, R. Tabata et al., A novel natural killer cell line (KHYG-1) from a patient with aggressive natural killer cell leukemia carrying a p53 point mutation, Leukemia, vol.14, pp.922-930, 2000.

Z. Zhao, J. Zuber, E. Diaz-flores, L. Lintault, S. C. Kogan et al., p53 loss promotes acute myeloid leukemia by enabling aberrant self-renewal, Genes & Development, vol.24, pp.1389-1402, 2010.