C. , D. Verrecchia, D. Heymann, L. W. Ellisen, B. Ory et al., Ory Acquisition of data (provided animals, acquired and managed patients, provided facilities Ory Analysis and interpretation of data (e.g., statistical analysis, biostatistics, computational analysis) Ory Writing, review, and/or revision of the manuscript, Ory Administrative, technical, or material support (i.e., reporting or organizing data, constructing databases): L.R. Calleja, J. Amiaud, R. Brion, B. Ory Study supervision: L.R. Calleja, B. Ory Other (contributed with samples and other clinical material and providing expertise on animal models): F. Lecanda References 1. Vousden KH, Prives C. Blinded by the light: the growing complexity of p53, pp.413-444, 2009.

U. Moll and N. Slade, p63 and p73: roles in development and tumor formation, Mol Cancer Res, vol.2, pp.371-86, 2004.

M. Deyoung and L. Ellisen, p63 and p73 in human cancer: defining the network, Oncogene, vol.58, issue.36, pp.5169-83, 2007.
DOI : 10.1128/MCB.19.5.3257

R. Tomasini, K. Tsuchihara, M. Wilhelm, M. Fujitani, A. Rufini et al., TAp73 knockout shows genomic instability with infertility and tumor suppressor functions, Genes & Development, vol.22, issue.19, pp.2677-91, 2008.
DOI : 10.1101/gad.1695308

URL : http://genesdev.cshlp.org/content/22/19/2677.full.pdf

A. Yang, R. Schweitzer, D. Sun, M. Kaghad, N. Walker et al., p63 is essential for regenerative proliferation in limb, craniofacial and epithelial development, Nature, vol.4, issue.6729, pp.714-722, 1999.
DOI : 10.1016/S0960-9822(00)00002-6

C. Leong, N. Vidnovic, M. Deyoung, D. Sgroi, and L. Ellisen, The p63/p73 network mediates chemosensitivity to cisplatin in a biologically defined subset of primary breast cancers, Journal of Clinical Investigation, vol.117, issue.5, pp.1370-80, 2007.
DOI : 10.1172/JCI30866

J. Sniezek, K. Matheny, M. Westfall, and J. Pietenpol, Dominant Negative p63 Isoform Expression in Head and Neck Squamous Cell Carcinoma, The Laryngoscope, vol.23, issue.12, pp.2063-72, 2004.
DOI : 10.1111/j.1600-0714.1999.tb01994.x

J. Rocco, C. Leong, N. Kuperwasser, M. Deyoung, and L. Ellisen, p63 mediates survival in squamous cell carcinoma by suppression of p73-dependent apoptosis, Cancer Cell, vol.9, issue.1, pp.45-56, 2006.
DOI : 10.1016/j.ccr.2005.12.013

E. Candi, A. Rufini, A. Terrinoni, D. Dinsdale, M. Ranalli et al., Differential roles of p63 isoforms in epidermal development: selective genetic complementation in p63 null mice, Cell Death & Differentiation, vol.48, issue.6, pp.1037-1084, 2006.
DOI : 10.1016/j.neuron.2005.10.027

R. Romano, K. Smalley, C. Magraw, V. Serna, T. Kurita et al., ??Np63 knockout mice reveal its indispensable role as a master regulator of epithelial development and differentiation, Development, vol.139, issue.4, pp.772-82, 2012.
DOI : 10.1242/dev.071191

X. Su, M. Paris, Y. Gi, K. Tsai, M. Cho et al., TAp63 Prevents Premature Aging by Promoting Adult Stem Cell Maintenance, Cell Stem Cell, vol.5, issue.1, pp.64-75, 2009.
DOI : 10.1016/j.stem.2009.04.003

URL : https://doi.org/10.1016/j.stem.2009.04.003

L. He, X. He, L. Lim, E. De-stanchina, Z. Xuan et al., A microRNA component of the p53 tumour suppressor network, Nature, vol.296, issue.7148, pp.1130-1134, 2007.
DOI : 10.1038/nature05939

B. Ory, M. Ramsey, C. Wilson, D. Vadysirisack, N. Forster et al., A microRNA-dependent program controls p53-independent survival and chemosensitivity in human and murine squamous cell carcinoma, Journal of Clinical Investigation, vol.121, issue.2, pp.809-829, 2011.
DOI : 10.1172/JCI43897DS1

N. Pencheva and S. Tavazoie, Control of metastatic progression by microRNA regulatory networks, Nature Cell Biology, vol.25, issue.6, pp.546-54, 2013.
DOI : 10.1128/MCB.25.5.1737-1748.2005

K. Png, N. Halberg, M. Yoshida, and S. Tavazoie, A microRNA regulon that mediates endothelial recruitment and metastasis by cancer cells, Nature, vol.365, issue.7380, pp.190-194, 2012.
DOI : 10.1016/S0140-6736(05)70933-8

J. Massague, TGF?? in Cancer, Cell, vol.134, issue.2, pp.215-245, 2008.
DOI : 10.1016/j.cell.2008.07.001

M. Taylor, J. Parvani, and W. Schiemann, The Pathophysiology of Epithelial-Mesenchymal Transition Induced by Transforming Growth Factor-?? in Normal and Malignant Mammary Epithelial Cells, Journal of Mammary Gland Biology and Neoplasia, vol.138, issue.4, pp.169-90, 2010.
DOI : 10.1023/A:1026404227624

X. Su, D. Chakravarti, M. Cho, L. Liu, Y. Gi et al., TAp63 suppresses metastasis through coordinate regulation of Dicer and miRNAs, Nature, vol.5, issue.7318, pp.986-90, 2010.
DOI : 10.1016/S0002-9440(10)64396-9

M. Adorno, M. Cordenonsi, M. Montagner, S. Dupont, C. Wong et al., A Mutant-p53/Smad Complex Opposes p63 to Empower TGF??-Induced Metastasis, Cell, vol.137, issue.1, pp.87-98, 2009.
DOI : 10.1016/j.cell.2009.01.039

URL : https://doi.org/10.1016/j.cell.2009.01.039

M. Cho, I. Chan, and E. Flores, , a gene with diverse roles in limb development, tumorigenesis and metastasis, Cell Cycle, vol.9, issue.12, pp.2434-2475, 2010.
DOI : 10.4161/cc.9.12.12051

A. Danilov, D. Neupane, A. Nagaraja, E. Feofanova, L. Humphries et al., DeltaNp63alpha-Mediated Induction of Epidermal Growth Factor Receptor Promotes Pancreatic Cancer Cell Growth and Chemoresistance, PLoS ONE, vol.6, issue.10, p.26815, 2011.
DOI : 10.1371/journal.pone.0026815.s004

J. Oh, R. Kim, K. Shin, N. Park, and M. Kang, ??Np63?? Protein Triggers Epithelial-Mesenchymal Transition and Confers Stem Cell Properties in Normal Human Keratinocytes, Journal of Biological Chemistry, vol.63, issue.44, pp.38757-67, 2011.
DOI : 10.1126/science.1151526

A. Pati~-no-garcía, M. Zalacain, C. Folio, C. Zandueta, L. Sierrases-umaga et al., Profiling of Chemonaive Osteosarcoma and Paired-Normal Cells Identifies EBF2 as a Mediator of Osteoprotegerin Inhibition to Tumor Necrosis Factor-Related Apoptosis-Inducing Ligand-Induced Apoptosis, Clinical Cancer Research, vol.15, issue.16, pp.5082-91, 2009.
DOI : 10.1158/1078-0432.CCR-09-0300

G. Melino, p63 is a suppressor of tumorigenesis and metastasis interacting with mutant p53, Cell Death & Differentiation, vol.60, issue.9, pp.1487-99, 2011.
DOI : 10.1038/onc.2010.215

X. Gu, P. Coates, L. Boldrup, and K. Nylander, p63 contributes to cell invasion and migration in squamous cell carcinoma of the head and neck, Cancer Letters, vol.263, issue.1, pp.26-34, 2008.
DOI : 10.1016/j.canlet.2007.12.011

C. Barbieri, L. Tang, K. Brown, and J. Pietenpol, Loss of p63 Leads to Increased Cell Migration and Up-regulation of Genes Involved in Invasion and Metastasis, Cancer Research, vol.66, issue.15, pp.7589-97, 2006.
DOI : 10.1158/0008-5472.CAN-06-2020

X. Yang, H. Lu, Y. B. Romano, R. Bian, Y. Friedman et al., ??Np63 Versatilely Regulates a Broad NF-??B Gene Program and Promotes Squamous Epithelial Proliferation, Migration, and Inflammation, Cancer Research, vol.71, issue.10, pp.3688-700, 2011.
DOI : 10.1158/0008-5472.CAN-10-3445

P. Tucci, M. Agostini, F. Grespi, E. Markert, A. Terrinoni et al., Loss of p63 and its microRNA-205 target results in enhanced cell migration and metastasis in prostate cancer, Proceedings of the National Academy of Sciences, vol.68, issue.4, pp.15312-15319, 2012.
DOI : 10.1158/0008-5472.CAN-07-2017

M. Tran, W. Choi, M. Wszolek, N. Navai, I. Lee et al., The p63 Protein Isoform ??Np63?? Inhibits Epithelial-Mesenchymal Transition in Human Bladder Cancer Cells, Journal of Biological Chemistry, vol.3, issue.5, pp.3275-88, 2013.
DOI : 10.1002/(SICI)1098-2388(199709/10)13:5<291::AID-SSU2>3.0.CO;2-8

J. Bergholz, Y. Zhang, J. Wu, L. Meng, E. Walsh et al., ??Np63?? regulates Erk signaling via MKP3 to inhibit cancer metastasis, Oncogene, vol.65, issue.2, pp.212-236, 2014.
DOI : 10.1128/MCB.25.14.6154-6164.2005

URL : https://www.ncbi.nlm.nih.gov/pmc/articles/PMC3962654/pdf

N. Thurfjell, P. Coates, B. Vojtesek, P. Benham-motlagh, M. Eisold et al., Endogenous p63 acts as a survival factor for tumour cells of SCCHN origin, International Journal of Molecular Medicine, vol.16, pp.1065-70, 2005.
DOI : 10.3892/ijmm.16.6.1065

M. Westfall, D. Mays, J. Sniezek, and J. Pietenpol, The Delta Np63 alpha phosphoprotein binds the p21 and 14?3-3 sigma promoters in vivo and Cancer Res Cancer Research has transcriptional repressor activity that is reduced by Hay-Wells syndrome-derived mutations, Mol Cell Biol, vol.7623, issue.11, pp.2264-76, 2003.

D. Betel, M. Wilson, A. Gabow, D. Marks, and C. Sander, The microRNA.org resource: targets and expression, Nucleic Acids Research, vol.35, issue.suppl_1, pp.149-53, 2008.
DOI : 10.1093/nar/gkl928

S. Griffiths-jones, H. Saini, S. Van-dongen, and A. Enright, miRBase: tools for microRNA genomics, Nucleic Acids Research, vol.21, issue.5, pp.154-162, 2008.
DOI : 10.1093/bioinformatics/bth437

URL : https://academic.oup.com/nar/article-pdf/36/suppl_1/D154/7635053/gkm952.pdf

K. Harms, S. Nozell, and X. Chen, The common and distinct target genes of the p53 family transcription factors, Cellular and Molecular Life Sciences (CMLS), vol.61, issue.7-8, pp.822-864, 2004.
DOI : 10.1007/s00018-003-3304-4

M. Deyoung, C. Johannessen, C. Leong, W. Faquin, J. Rocco et al., Tumor-Specific p73 Up-regulation Mediates p63 Dependence in Squamous Cell Carcinoma, Cancer Research, vol.66, issue.19, pp.9362-9370, 2006.
DOI : 10.1158/0008-5472.CAN-06-1619

S. Dennler, S. Itoh, D. Vivien, P. Ten-dijke, S. Huet et al., Direct binding of Smad3 and Smad4 to critical TGFbeta -inducible elements in the promoter of human plasminogen activator inhibitor-type 1gene, The EMBO Journal, vol.17, issue.11, pp.3091-100, 1998.
DOI : 10.1093/emboj/17.11.3091

J. Celli, P. Duijf, B. Hamel, M. Bamshad, B. Kramer et al., Heterozygous Germline Mutations in the p53 Homolog p63 Are the Cause of EEC Syndrome, Cell, vol.99, issue.2, pp.143-53, 1999.
DOI : 10.1016/S0092-8674(00)81646-3

W. Kozlow and T. Guise, Breast Cancer Metastasis to Bone: Mechanisms of Osteolysis and Implications for Therapy, Journal of Mammary Gland Biology and Neoplasia, vol.97, issue.3 Suppl, pp.169-80, 2005.
DOI : 10.1054/bjoc.1999.1131

A. Calon, E. Espinet, S. Palomo-ponce, D. Tauriello, M. Iglesias et al., Dependency of Colorectal Cancer on a TGF-??-Driven Program in Stromal Cells for Metastasis Initiation, Cancer Cell, vol.22, issue.5, pp.571-84, 2012.
DOI : 10.1016/j.ccr.2012.08.013

A. Lamora, J. Talbot, G. Bougras, J. Amiaud, M. Leduc et al., Overexpression of Smad7 Blocks Primary Tumor Growth and Lung Metastasis Development in Osteosarcoma, Clinical Cancer Research, vol.20, issue.19, pp.5097-112, 2014.
DOI : 10.1158/1078-0432.CCR-13-3191

J. Rousseau, V. Escriou, P. Perrot, G. Picarda, C. Charrier et al., Advantages of bioluminescence imaging to follow siRNA or chemotherapeutic treatments in osteosarcoma preclinical models, Cancer Gene Therapy, vol.8, issue.6, pp.387-97, 2010.
DOI : 10.1002/jcb.21556

G. Gurtner, S. Werner, Y. Barrandon, and M. Longaker, Wound repair and regeneration, Nature, vol.23, issue.7193, pp.314-335, 2008.
DOI : 10.1038/nm1504

A. Hameedaldeen, J. Liu, A. Batres, G. Graves, and D. Graves, FOXO1, TGF-?? Regulation and Wound Healing, International Journal of Molecular Sciences, vol.51, issue.12, pp.16257-69, 2014.
DOI : 10.1002/jbmr.59

URL : http://www.mdpi.com/1422-0067/15/9/16257/pdf

T. Ichikawa, Y. Suenaga, T. Koda, T. Ozaki, and A. Nakagawara, ??Np63/BMP-7-dependent expression of matrilin-2 is involved in keratinocyte migration in response to wounding, Biochemical and Biophysical Research Communications, vol.369, issue.4, pp.994-1000, 2008.
DOI : 10.1016/j.bbrc.2008.02.128

B. Noszczyk and S. Majewski, p63 Expression during Normal Cutaneous Wound Healing in Humans, Plastic and Reconstructive Surgery, vol.108, issue.5, pp.1242-1249, 2001.
DOI : 10.1097/00006534-200110000-00022

G. Sundaram, J. Common, F. Gopal, S. Srikanta, K. Lakshman et al., ???See-saw??? expression of microRNA-198 and FSTL1 from a single transcript in wound healing, Nature, vol.495, issue.7439, pp.103-109, 2013.
DOI : 10.1172/JCI32169

M. Trabucchi, P. Briata, M. Garcia-mayoral, A. Haase, W. Filipowicz et al., The RNA-binding protein KSRP promotes the biogenesis of a subset of microRNAs, Nature, vol.5, issue.5, pp.1010-1014, 2009.
DOI : 10.1007/BF00197809

M. Ramsey, C. Wilson, B. Ory, S. Rothenberg, W. Faquin et al., FGFR2 signaling underlies p63 oncogenic function in squamous cell carcinoma, Journal of Clinical Investigation, vol.123, issue.8, pp.3525-3563, 2013.
DOI : 10.1172/JCI68899DS1

I. Celardo, F. Grespi, A. Antonov, F. Bernassola, A. Garabadgiu et al., Caspase-1 is a novel target of p63 in tumor suppression, Cell Death & Disease, vol.60, issue.5, p.645, 2013.
DOI : 10.1093/nar/gkr372

J. Wu, S. Liang, J. Bergholz, H. He, E. Walsh et al., ??Np63?? activates CD82 metastasis suppressor to inhibit cancer cell invasion, Cell Death & Disease, vol.6, issue.6, p.1280, 2014.
DOI : 10.1093/nar/gkn923

URL : http://www.nature.com/cddis/journal/v5/n6/pdf/cddis2014239a.pdf

S. Dallas, J. Rosser, G. Mundy, and L. Bonewald, Proteolysis of Latent Transforming Growth Factor-?? (TGF-??)-binding Protein-1 by Osteoclasts, Journal of Biological Chemistry, vol.14, issue.24, pp.21352-60, 2002.
DOI : 10.1042/bj3020527