A. Aitken, 14-3-3 and its possible role in co-ordinating multiple signalling pathways, Trends in Cell Biology, vol.6, issue.9, pp.341-347, 1996.
DOI : 10.1016/0962-8924(96)10029-5

J. Birkenfeld, P. Nalbant, B. P. Bohl, O. Pertz, K. M. Hahn et al., GEF-H1 Modulates Localized RhoA Activation during Cytokinesis under the Control of Mitotic Kinases, Developmental Cell, vol.12, issue.5, pp.699-712, 2007.
DOI : 10.1016/j.devcel.2007.03.014

M. Chircop, Rho GTPases as regulators of mitosis and cytokinesis in mammalian cells, Small GTPases, vol.108, issue.2, 2014.
DOI : 10.1126/science.1163086

M. David, D. Petit, and J. Bertoglio, Cell cycle regulation of Rho signaling pathways, Cell Cycle, vol.3, issue.16, pp.3003-3010, 2012.
DOI : 10.1126/scisignal.2000475

M. D. David, D. Petit, and J. Bertoglio, The RhoGAP ARHGAP19 controls cytokinesis and chromosome segregation in T lymphocytes, Journal of Cell Science, vol.127, issue.2, pp.400-410, 2014.
DOI : 10.1242/jcs.135079

Y. Feng, P. V. Lograsso, O. Defert, L. , and R. , Rho Kinase (ROCK) Inhibitors and Their Therapeutic Potential, Journal of Medicinal Chemistry, vol.59, issue.6, pp.2269-2300, 2016.
DOI : 10.1021/acs.jmedchem.5b00683

A. Gohla and G. M. Bokoch, 14-3-3 Regulates Actin Dynamics by Stabilizing Phosphorylated Cofilin, Current Biology, vol.12, issue.19, pp.1704-1710, 2002.
DOI : 10.1016/S0960-9822(02)01184-3

URL : https://doi.org/10.1016/s0960-9822(02)01184-3

A. Hall, Rho family GTPases, Biochemical Society Transactions, vol.29, issue.6, pp.1378-1382, 2012.
DOI : 10.1016/j.tips.2010.12.006

A. Hausser, G. Link, M. Hoene, C. Russo, O. Selchow et al., Phospho-specific binding of 14-3-3 proteins to phosphatidylinositol 4-kinase III ?? protects from dephosphorylation and stabilizes lipid kinase activity, Journal of Cell Science, vol.119, issue.17, pp.3613-3621, 2006.
DOI : 10.1242/jcs.03104

K. Hirose, T. Kawashima, I. Iwamoto, T. Nosaka, and T. Kitamura, MgcRacGAP Is Involved in Cytokinesis through Associating with Mitotic Spindle and Midbody, Journal of Biological Chemistry, vol.111, issue.8, pp.5821-5828, 2001.
DOI : 10.1083/jcb.129.6.1617

K. Kamijo, N. Ohara, M. Abe, T. Uchimura, H. Hosoya et al., Dissecting the Role of Rho-mediated Signaling in Contractile Ring Formation, Molecular Biology of the Cell, vol.17, issue.1, pp.43-55, 2006.
DOI : 10.1091/mbc.E05-06-0569

C. Lindon, Control of mitotic exit and cytokinesis by the APC/C, Biochemical Society Transactions, vol.36, issue.3, pp.405-410, 2008.
DOI : 10.1042/BST0360405

C. Liot, L. Seguin, A. Siret, C. Crouin, S. Schmidt et al., APC(cdh1) mediates degradation of the oncogenic Rho-GEF Ect2 after mitosis, PLoS One, vol.6, 2011.

A. S. Maddox and K. Burridge, RhoA is required for cortical retraction and rigidity during mitotic cell rounding, The Journal of Cell Biology, vol.57, issue.6, pp.255-265, 2003.
DOI : 10.1091/mbc.8.8.1415

P. Mertins, J. W. Qiao, J. Patel, N. D. Udeshi, K. R. Clauser et al., Integrated proteomic analysis of post-translational modifications by serial enrichment, Nature Methods, vol.11, issue.7, pp.634-637, 2013.
DOI : 10.1021/pr101065j

C. Ramachandran, R. V. Patil, K. Combrink, N. A. Sharif, S. et al., Rho-Rho kinase pathway in the actomyosin contraction and cell-matrix adhesion in immortalized human trabecular meshwork cells, Mol Vis, vol.17, pp.1877-1890, 2011.

M. Sebbagh, J. Hamelin, J. Bertoglio, E. Solary, and J. Breard, Direct cleavage of ROCK II by granzyme B induces target cell membrane blebbing in a caspase-independent manner, The Journal of Experimental Medicine, vol.16, issue.3, pp.465-471, 2005.
DOI : 10.1128/MCB.16.10.5313

L. Seguin, C. Liot, R. Mzali, R. Harada, A. Siret et al., CUX1 and E2F1 Regulate Coordinated Expression of the Mitotic Complex Genes Ect2, MgcRacGAP, and MKLP1 in S Phase, Molecular and Cellular Biology, vol.29, issue.2, pp.570-581, 2009.
DOI : 10.1128/MCB.01275-08

L. Su, J. M. Agati, and S. J. Parsons, p190RhoGAP is cell cycle regulated and affects cytokinesis, The Journal of Cell Biology, vol.7, issue.3, pp.571-582, 2003.
DOI : 10.1016/S0960-9822(01)00307-4

URL : http://jcb.rupress.org/content/jcb/163/3/571.full.pdf

C. G. Takizawa, M. , and D. O. , Control of mitosis by changes in the subcellular location of cyclin-B1???Cdk1 and Cdc25C, Current Opinion in Cell Biology, vol.12, issue.6, pp.658-665, 2000.
DOI : 10.1016/S0955-0674(00)00149-6

T. Tatsumoto, X. Xie, R. Blumenthal, I. Okamoto, M. et al., Human Ect2 Is an Exchange Factor for Rho Gtpases, Phosphorylated in G2/M Phases, and Involved in Cytokinesis, The Journal of Cell Biology, vol.11, issue.5, pp.921-928, 1999.
DOI : 10.1093/emboj/17.21.6404

B. K. Tripathi, X. Qian, P. Mertins, D. Wang, A. G. Papageorge et al., CDK5 is a major regulator of the tumor suppressor DLC1, The Journal of Cell Biology, vol.63, issue.5, pp.627-642, 2014.
DOI : 10.1016/S0896-6273(00)81200-3

D. Wu, M. Asiedu, R. S. Adelstein, W. , and Q. , A Novel Guanine Nucleotide Exchange Factor, MYOGEF, is Required for Cytokinesis, Cell Cycle, vol.5, issue.11, pp.1234-1239, 2006.
DOI : 10.4161/cc.5.11.2815

M. Matsuda, Cell type-specific regulation of RhoA activity during cytokinesis, J Biol Chem, vol.279, pp.44756-44762, 2004.

E. Zanin, A. Desai, I. Poser, Y. Toyoda, C. Andree et al., A Conserved RhoGAP Limits M Phase Contractility and Coordinates with Microtubule Asters to Confine RhoA during Cytokinesis, Developmental Cell, vol.26, issue.5, pp.496-510, 2013.
DOI : 10.1016/j.devcel.2013.08.005

A. Bergman, H. Abdollahi, J. Friess, and . Kleeff, Consensus transcriptome signature of perineural invasion in pancreatic carcinoma, Mol Cancer Ther, vol.8, issue.6, pp.1494-1504, 2009.

A. Abo, E. Pick, A. Hall, N. Totty, C. G. Teahan et al., Activation of the NADPH oxidase involves the small GTP-binding protein p21rac1, Nature, vol.353, issue.6345, pp.668-670, 1991.
DOI : 10.1038/353668a0

S. Abramson, R. G. Miller, and R. A. Phillips, The identification in adult bone marrow of pluripotent and restricted stem cells of the myeloid and lymphoid systems, Journal of Experimental Medicine, vol.145, issue.6, pp.1567-1579, 1977.
DOI : 10.1084/jem.145.6.1567

. Jacobsen, Identification of Flt3+ lympho-myeloid stem cells lacking erythromegakaryocytic potential a revised road map for adult blood lineage commitment, Cell, vol.121, issue.2, pp.295-306, 2005.

B. Aghazadeh, K. Zhu, T. J. Kubiseski, G. A. Liu, T. Pawson et al., Structure and mutagenesis of the Dbl homology domain, Nature Structural Biology, vol.11, issue.12, pp.1098-1107, 1998.
DOI : 10.1002/prot.340110407

S. Ahmed, J. Lee, R. Kozma, A. Best, C. Monfries et al., A novel functional target for tumor-promoting phorbol esters and lysophosphatidic acid. The p21rac-GTPase activating protein n-chimaerin, J Biol Chem, vol.268, issue.15, pp.10709-10712, 1993.

A. Aitken, 14-3-3 and its possible role in co-ordinating multiple signalling pathways, Trends in Cell Biology, vol.6, issue.9, pp.341-347, 1996.
DOI : 10.1016/0962-8924(96)10029-5

K. Akashi, D. Traver, T. Miyamoto, and I. L. Weissman, A clonogenic common myeloid progenitor that gives rise to all myeloid lineages, Nature, vol.362, issue.6774, pp.193-197, 2000.
DOI : 10.1038/362722a0

O. Alekhina, E. Burstein, and D. D. Billadeau, Cellular functions of WASP family proteins at a glance, Journal of Cell Science, vol.130, issue.14, pp.2235-2241, 2017.
DOI : 10.1091/mbc.E11-12-0992

R. Alfaro-aco and S. Petry, Building the Microtubule Cytoskeleton Piece by Piece, Journal of Biological Chemistry, vol.3, issue.28, pp.17154-17162, 2015.
DOI : 10.1126/science.1243147

M. Amano, Y. Fukata, and K. Kaibuchi, Regulation and Functions of Rho-Associated Kinase, Experimental Cell Research, vol.261, issue.1, pp.44-51, 2000.
DOI : 10.1006/excr.2000.5046

. Kaibuchi, Phosphorylation and activation of myosin by Rho-associated kinase (Rhokinase ), J Biol Chem, vol.271, issue.34, pp.20246-20249, 1996.

V. Archambault and D. M. Glover, Polo-like kinases: conservation and divergence in their functions and regulation, Nature Reviews Molecular Cell Biology, vol.99, issue.4, pp.265-275, 2009.
DOI : 10.4161/cc.6.20.5079

P. Aspenstrom, Effectors for the Rho GTPases, Current Opinion in Cell Biology, vol.11, issue.1, pp.95-102, 1999.
DOI : 10.1016/S0955-0674(99)80011-8

C. Baarlink, H. Wang, and R. Grosse, Nuclear Actin Network Assembly by Formins Regulates the SRF Coactivator MAL, Science, vol.43, issue.51, pp.864-867, 2013.
DOI : 10.1021/bi048092i

D. P. Bartel, MicroRNAs, Cell, vol.116, issue.2, pp.281-297, 2004.
DOI : 10.1016/S0092-8674(04)00045-5

URL : https://hal.archives-ouvertes.fr/hal-00369966

L. D. Belmont and T. J. Mitchison, Identification of a Protein That Interacts with Tubulin Dimers and Increases the Catastrophe Rate of Microtubules, Cell, vol.84, issue.4, pp.623-631, 1996.
DOI : 10.1016/S0092-8674(00)81037-5

J. Birkenfeld, P. Nalbant, B. P. Bohl, O. Pertz, K. M. Hahn et al., GEF-H1 Modulates Localized RhoA Activation during Cytokinesis under the Control of Mitotic Kinases, Developmental Cell, vol.12, issue.5, pp.699-712, 2007.
DOI : 10.1016/j.devcel.2007.03.014

A. L. Bishop and A. Hall, Rho GTPases and their effector proteins, Biochem J, p.348, 2000.

P. Boquet and E. Lemichez, Bacterial virulence factors targeting Rho GTPases: parasitism or symbiosis?, Trends in Cell Biology, vol.13, issue.5, pp.238-246, 2003.
DOI : 10.1016/S0962-8924(03)00037-0

J. L. Bos, H. Rehmann, and A. Wittinghofer, GEFs and GAPs: Critical Elements in the Control of Small G Proteins, Cell, vol.129, issue.5, pp.865-877, 2007.
DOI : 10.1016/j.cell.2007.05.018

E. Boulter and R. Garcia-mata, Analysis of the Role of RhoGDI1 and Isoprenylation in the Degradation of RhoGTPases, Methods Mol Biol, vol.827, pp.97-105, 2012.
DOI : 10.1007/978-1-61779-442-1_7

A. Boureux, E. Vignal, S. Faure, and P. Fort, Evolution of the Rho Family of Ras-Like GTPases in Eukaryotes, Molecular Biology and Evolution, vol.90, issue.1, pp.203-216, 2007.
DOI : 10.1016/S0092-8674(00)80353-0

URL : https://hal.archives-ouvertes.fr/hal-00203003

H. R. Bourne, D. A. Sanders, and F. Mccormick, The GTPase superfamily: conserved structure and molecular mechanism, Nature, vol.349, issue.6305, pp.117-127, 1991.
DOI : 10.1038/349117a0

H. Brzeska, J. Szczepanowska, F. Matsumura, and E. D. Korn, Rac-induced increase of phosphorylation of myosin regulatory light chain in HeLa cells, Cell Motility and the Cytoskeleton, vol.18, issue.128, pp.186-199, 2004.
DOI : 10.1128/MCB.18.4.2153

F. Buss, C. Temm-grove, S. Henning, and B. M. Jockusch, Distribution of profilin in fibroblasts correlates with the presence of highly dynamic actin filaments, Cell Motility and the Cytoskeleton, vol.1, issue.Suppl. 30, pp.51-61, 1992.
DOI : 10.1016/0960-9822(91)90093-C

J. Plass, A. Krijgsveld, and . Trumpp, Identification of regulatory networks in HSCs and their immediate progeny via integrated proteome, transcriptome, and DNA methylome analysis, Cell Stem Cell, vol.15, issue.4, pp.507-522, 2014.

R. Carsetti, G. Kohler, and M. C. Lamers, Transitional B cells are the target of negative selection in the B cell compartment, Journal of Experimental Medicine, vol.181, issue.6, pp.2129-2140, 1995.
DOI : 10.1084/jem.181.6.2129

R. Ceredig and A. G. Rolink, The key role of IL-7 in lymphopoiesis, Seminars in Immunology, vol.24, issue.3, pp.159-164, 2012.
DOI : 10.1016/j.smim.2012.02.004

Y. Chang, F. Aurade, F. Larbret, Y. Zhang, J. P. Le-couedic et al., Proplatelet formation is regulated by the Rho/ROCK pathway, Blood, vol.109, issue.10, pp.4229-4236, 2007.
DOI : 10.1182/blood-2006-04-020024

Y. Chang, D. Bluteau, N. Debili, and W. Vainchenker, From hematopoietic stem cells to platelets, Journal of Thrombosis and Haemostasis, vol.105, issue.Suppl. 1, pp.318-327, 2007.
DOI : 10.1172/JCI21197

P. Chardin, The ras superfamily proteins, Biochimie, vol.70, issue.7, pp.865-868, 1988.
DOI : 10.1016/0300-9084(88)90226-X

K. Chen, J. Liu, S. Heck, J. A. Chasis, X. An et al., Resolving the distinct stages in erythroid differentiation based on dynamic changes in membrane protein expression during erythropoiesis, Proceedings of the National Academy of Sciences, vol.61, issue.4, pp.17413-17418, 2009.
DOI : 10.1182/blood.V98.12.3261

M. Chircop, Rho GTPases as regulators of mitosis and cytokinesis in mammalian cells, Small GTPases, vol.108, issue.2, p.29770, 2014.
DOI : 10.1126/science.1163086

M. Chircop, Rho GTPases as regulators of mitosis and cytokinesis in mammalian cells, Small GTPases, vol.108, issue.2, 2014.
DOI : 10.1126/science.1163086

M. M. Chou, J. M. Masuda-robens, and M. L. Gupta, Progression through p70 S6 Kinase-mediated Induction of Cyclin E Expression, Journal of Biological Chemistry, vol.16, issue.37, pp.35241-35247, 2003.
DOI : 10.1101/gad.835000

M. Ciofani and J. C. Zuniga-pflucker, Determining ???? versus ???? T cell development, Nature Reviews Immunology, vol.106, issue.9, pp.657-663, 2010.
DOI : 10.4049/jimmunol.168.4.1649

J. Colicelli, Human RAS Superfamily Proteins and Related GTPases, Science Signaling, vol.277, issue.41, p.13, 2004.
DOI : 10.1074/jbc.M204132200

URL : http://europepmc.org/articles/pmc2828947?pdf=render

D. R. Cook, K. L. Rossman, and C. J. Der, Rho guanine nucleotide exchange factors: regulators of Rho GTPase activity in development and disease, Oncogene, vol.128, issue.31, pp.4021-4035, 2014.
DOI : 10.1038/374474a0

I. Corre, M. Gomez, S. Vielkind, and D. A. Cantrell, Analysis of Thymocyte Development Reveals That the Gtpase Rhoa Is a Positive Regulator of T Cell Receptor Responses in Vivo, The Journal of Experimental Medicine, vol.18, issue.7, pp.903-914, 2001.
DOI : 10.1038/35020093

O. A. Coso, M. Chiariello, J. C. Yu, H. Teramoto, P. Crespo et al., The small GTP-binding proteins Rac1 and Cdc42regulate the activity of the JNK/SAPK signaling pathway, Cell, vol.81, issue.7, pp.1137-1146, 1995.
DOI : 10.1016/S0092-8674(05)80018-2

J. F. Cote, Identification of an evolutionarily conserved superfamily of DOCK180-related proteins with guanine nucleotide exchange activity, Journal of Cell Science, vol.115, issue.24, pp.4901-4913, 2002.
DOI : 10.1242/jcs.00219

A. D. Cox and C. J. Der, Ras history, Small GTPases, vol.14, issue.1, pp.2-27, 2010.
DOI : 10.1016/j.tcb.2004.09.014

M. Crisan and E. Dzierzak, The many faces of hematopoietic stem cell heterogeneity, Development, vol.124, issue.24, pp.4571-4581, 2016.
DOI : 10.1016/j.stem.2008.09.018

D. R. Croft and M. F. Olson, Transcriptional regulation of Rho GTPase signaling, Transcription, vol.2, issue.5, pp.211-215, 2011.
DOI : 10.1038/nature09459

H. Daub, K. Gevaert, J. Vandekerckhove, A. Sobel, and A. Hall, Rac/Cdc42 and p65PAK Regulate the Microtubule-destabilizing Protein Stathmin through Phosphorylation at Serine 16, Journal of Biological Chemistry, vol.161, issue.3, pp.1677-1680, 2001.
DOI : 10.1002/(SICI)1097-0169(1999)42:1<48::AID-CM5>3.0.CO;2-8

M. David, D. Petit, and J. Bertoglio, Cell cycle regulation of Rho signaling pathways, Cell Cycle, vol.3, issue.16, pp.3003-3010, 2012.
DOI : 10.1126/scisignal.2000475

M. D. David, D. Petit, and J. Bertoglio, The RhoGAP ARHGAP19 controls cytokinesis and chromosome segregation in T lymphocytes, Journal of Cell Science, vol.127, issue.2, pp.400-410, 2014.
DOI : 10.1242/jcs.135079

J. De-boer, A. Williams, G. Skavdis, N. Harker, M. Coles et al., Transgenic mice with hematopoietic and lymphoid specific expression of Cre, European Journal of Immunology, vol.33, issue.2, pp.314-325, 2003.
DOI : 10.1002/immu.200310005

C. Dermardirossian and G. M. Bokoch, GDIs: central regulatory molecules in Rho GTPase activation, Trends in Cell Biology, vol.15, issue.7, pp.356-363, 2005.
DOI : 10.1016/j.tcb.2005.05.001

D. Diviani, L. Abuin, S. Cotecchia, and L. Pansier, Anchoring of both PKA and 14-3-3 inhibits the Rho-GEF activity of the AKAP-Lbc signaling complex, The EMBO Journal, vol.18, issue.14, pp.2811-2820, 2004.
DOI : 10.1074/jbc.270.16.9031

D. C. Edwards, L. C. Sanders, G. M. Bokoch, and G. N. Gill, Activation of LIM-kinase by Pak1 couples Rac/Cdc42 GTPase signalling to actin cytoskeletal dynamics, Nature Cell Biology, vol.224, issue.5, pp.253-259, 1999.
DOI : 10.1006/abio.1995.1070

S. Etienne-manneville and A. Hall, Rho GTPases in cell biology, Nature, vol.92, issue.6916, pp.629-635, 2002.
DOI : 10.1074/jbc.M108297200

S. Etienne-manneville and A. Hall, Cdc42 regulates GSK-3?? and adenomatous polyposis coli to control cell polarity, Nature, vol.9, issue.6924, pp.753-756, 2003.
DOI : 10.1016/S0960-9822(99)80190-0

A. Eva and S. A. Aaronson, Isolation of a new human oncogene from a diffuse B-cell lymphoma, Nature, vol.312, issue.6025, pp.273-275, 1985.
DOI : 10.1128/MCB.4.9.1695

F. Famili, A. S. Wiekmeijer, and F. J. Staal, The development of T cells from stem cells in mice and humans, Future Science OA, vol.93, issue.9, p.186, 2017.
DOI : 10.4049/jimmunol.174.10.6477

H. J. Fehling and H. Von-boehmer, Early ???? T cell development in the thymus of normal and genetically altered mice, Current Opinion in Immunology, vol.9, issue.2, pp.263-275, 1997.
DOI : 10.1016/S0952-7915(97)80146-X

Y. Feng, P. V. Lograsso, O. Defert, and R. Li, Rho Kinase (ROCK) Inhibitors and Their Therapeutic Potential, Journal of Medicinal Chemistry, vol.59, issue.6, pp.2269-2300, 2016.
DOI : 10.1021/acs.jmedchem.5b00683

J. L. Freeman, A. Abo, and J. D. Lambeth, Rac ???Insert Region??? Is a Novel Effector Region That Is Implicated in the Activation of NADPH Oxidase, but Not PAK65, Journal of Biological Chemistry, vol.15, issue.33, pp.19794-19801, 1996.
DOI : 10.1074/jbc.271.1.83

F. Iwamatsu, K. Perez, and . Kaibuchi, Rac1 and Cdc42 capture microtubules through IQGAP1 and CLIP-170, Cell, vol.109, issue.7, pp.873-885, 2002.

J. L. Galloway and L. I. Zon, 3 Ontogeny of hematopoiesis: Examining the emergence of hematopoietic cells in the vertebrate embryo, Curr Top Dev Biol, vol.53, pp.139-158, 2003.
DOI : 10.1016/S0070-2153(03)53004-6

S. J. Gamblin and S. J. Smerdon, GTPase-activating proteins and their complexes, Current Opinion in Structural Biology, vol.8, issue.2, pp.195-201, 1998.
DOI : 10.1016/S0959-440X(98)80038-9

Y. Gao, E. Smith, P. Ker, E. C. Campbell, S. Cheng et al., Role of RhoA-Specific Guanine Exchange Factors in Regulation of Endomitosis in Megakaryocytes, Developmental Cell, vol.22, issue.3, pp.573-584, 2012.
DOI : 10.1016/j.devcel.2011.12.019

M. D. Garrett, A. J. Self, C. Van-oers, and A. Hall, Identification of distinct cytoplasmic targets for ras/R-ras and rho regulatory proteins, J Biol Chem, vol.264, issue.1, pp.10-13, 1989.

G. Ghiaur, A. Lee, J. Bailey, J. A. Cancelas, Y. Zheng et al., Inhibition of RhoA GTPase activity enhances hematopoietic stem and progenitor cell proliferation and engraftment, Blood, vol.108, issue.6, pp.2087-2094, 2006.
DOI : 10.1182/blood-2006-02-001560

O. Gjoerup, J. Lukas, J. Bartek, and B. M. Willumsen, Rac and Cdc42 Are Potent Stimulators of E2F-dependent Transcription Capable of Promoting Retinoblastoma Susceptibility Gene Product Hyperphosphorylation, Journal of Biological Chemistry, vol.17, issue.30, pp.18812-18818, 1998.
DOI : 10.1016/0076-6879(95)50078-2

D. I. Godfrey, J. Kennedy, T. Suda, and A. Zlotnik, A developmental pathway involving four phenotypically and functionally distinct subsets of CD3-CD4-CD8-triple- negative adult mouse thymocytes defined by CD44 and CD25 expression, J Immunol, vol.150, issue.10, pp.4244-4252, 1993.

A. Gohla and G. M. Bokoch, 14-3-3 Regulates Actin Dynamics by Stabilizing Phosphorylated Cofilin, Current Biology, vol.12, issue.19, pp.1704-1710, 2002.
DOI : 10.1016/S0960-9822(02)01184-3

URL : https://doi.org/10.1016/s0960-9822(02)01184-3

C. J. Gregory and A. C. Eaves, Three stages of erythropoietic progenitor cell differentiation distinguished by a number of physical and biologic properties, Blood, vol.51, issue.3, pp.527-537, 1978.

H. Gu, J. D. Marth, P. C. Orban, H. Mossmann, and K. Rajewsky, Deletion of a DNA polymerase beta gene segment in T cells using cell type-specific gene targeting, Science, vol.265, issue.5168, pp.103-106, 1994.
DOI : 10.1126/science.8016642

F. Guo, J. A. Cancelas, D. Hildeman, D. A. Williams, and Y. Zheng, Rac GTPase isoforms Rac1 and Rac2 play a redundant and crucial role in T-cell development, Blood, vol.112, issue.5, pp.1767-1775, 2008.
DOI : 10.1182/blood-2008-01-132068

F. Guo, C. S. Velu, H. L. Grimes, and Y. Zheng, Rho GTPase Cdc42 is essential for B-lymphocyte development and activation, Blood, vol.114, issue.14, pp.2909-2916, 2009.
DOI : 10.1182/blood-2009-04-214676

T. Hakoshima, T. Shimizu, and R. Maesaki, Structural Basis of the Rho GTPase Signaling, Journal of Biochemistry, vol.134, issue.3, pp.327-331, 2003.
DOI : 10.1093/jb/mvg149

A. Hall, Rho family GTPases, Biochemical Society Transactions, vol.29, issue.6, pp.1378-1382, 2012.
DOI : 10.1016/j.tips.2010.12.006

T. Hara, M. Abe, H. Inoue, L. R. Yu, T. D. Veenstra et al., Cytokinesis regulator ECT2 changes its conformation through phosphorylation at Thr-341 in G2/M phase, Oncogene, vol.25, issue.4, pp.566-578, 2006.
DOI : 10.1038/sj.onc.1209078

M. J. Hart, A. Eva, T. Evans, S. A. Aaronson, and R. A. Cerione, Catalysis of guanine nucleotide exchange on the CDC42Hs protein by the dbloncogene product, Nature, vol.354, issue.6351, pp.311-314, 1991.
DOI : 10.1038/354311a0

L. Aumailley and . Bruckner-tuderman, Kindlin-1 Is required for RhoGTPase-mediated lamellipodia formation in keratinocytes, Am J Pathol, vol.175, issue.4, pp.1442-1452, 2009.

C. S. Hill, J. Wynne, and R. Treisman, The Rho family GTPases RhoA, Racl , and CDC42Hsregulate transcriptional activation by SRF, Cell, vol.81, issue.7, pp.1159-1170, 1995.
DOI : 10.1016/S0092-8674(05)80020-0

K. Hirose, T. Kawashima, I. Iwamoto, T. Nosaka, and T. Kitamura, MgcRacGAP Is Involved in Cytokinesis through Associating with Mitotic Spindle and Midbody, Journal of Biological Chemistry, vol.111, issue.8, pp.5821-5828, 2001.
DOI : 10.1083/jcb.129.6.1617

R. G. Hodge and A. J. Ridley, Regulating Rho GTPases and their regulators, Nature Reviews Molecular Cell Biology, vol.15, issue.8, pp.496-510, 2016.
DOI : 10.1038/onc.2012.124

J. Wagle, K. Wargo, D. L. Chong, K. Morton, G. Stemke-hale et al., A landscape of driver mutations in melanoma, Cell, vol.150, issue.2, pp.251-263, 2012.

A. B. Jaffe and A. Hall, RHO GTPASES: Biochemistry and Biology, Annual Review of Cell and Developmental Biology, vol.21, issue.1, pp.247-269, 2005.
DOI : 10.1146/annurev.cellbio.21.020604.150721

M. Jagannathan-bogdan and L. I. Zon, Hematopoiesis, Development, vol.140, issue.12, pp.2463-2467, 2013.
DOI : 10.1242/dev.083147

S. Jenna, N. K. Hussain, E. I. Danek, I. Triki, S. Wasiak et al., The Activity of the GTPase-activating Protein CdGAP Is Regulated by the Endocytic Protein Intersectin, Journal of Biological Chemistry, vol.114, issue.8, pp.6366-6373, 2002.
DOI : 10.1038/ncb1001-927

J. Ji, X. Feng, M. Shi, Q. Cai, Y. Yu et al., Rac1 is correlated with aggressiveness and a potential therapeutic target for gastric cancer, International Journal of Oncology, vol.46, issue.3, pp.1343-1353, 2015.
DOI : 10.3892/ijo.2015.2836

C. N. Johnstone, S. Castellvi-bel, L. M. Chang, X. Bessa, H. Nakagawa et al., ARHGAP8 is a novel member of the RHOGAP family related to ARHGAP1/CDC42GAP/p50RHOGAP: mutation and expression analyses in colorectal and breast cancers, Gene, vol.336, issue.1, pp.59-71, 2004.
DOI : 10.1016/j.gene.2004.01.025

J. K. Segall, C. J. Westwick, and R. G. Der, Integration of Rac-dependent regulation of cyclin D1 transcription through a nuclear factor-kappaB-dependent pathway, J Biol Chem, vol.274, issue.36, pp.25245-25249, 1999.

H. Jumaa, R. W. Hendriks, and M. Reth, B Cell Signaling and Tumorigenesis, Annual Review of Immunology, vol.23, issue.1, pp.415-445, 2005.
DOI : 10.1146/annurev.immunol.23.021704.115606

H. Tatsuno, Y. Katoh, T. Watanabe, T. Ichimura, S. Ushiku et al., Recurrent gain-of-function mutations of RHOA in diffuse-type gastric carcinoma, Nat Genet, vol.46, issue.6, pp.583-587, 2014.

H. Karasuyama, A. Kudo, and F. Melchers, The proteins encoded by the VpreB and lambda 5 pre-B cell-specific genes can associate with each other and with mu heavy chain, Journal of Experimental Medicine, vol.172, issue.3, pp.969-972, 1990.
DOI : 10.1084/jem.172.3.969

K. Nakano, A. Okawa, K. Iwamatsu, and . Kaibuchi, Regulation of myosin phosphatase by Rho and Rho-associated kinase (Rho-kinase), Science, vol.273, issue.5272, pp.245-248, 1996.

W. B. Kiosses, R. H. Daniels, C. Otey, G. M. Bokoch, and M. A. Schwartz, A Role for P21-Activated Kinase in Endothelial Cell Migration, The Journal of Cell Biology, vol.18, issue.4, pp.831-844, 1999.
DOI : 10.1128/MCB.18.4.2153

M. Kondo, I. L. Weissman, and K. Akashi, Identification of Clonogenic Common Lymphoid Progenitors in Mouse Bone Marrow, Cell, vol.91, issue.5, pp.661-672, 1997.
DOI : 10.1016/S0092-8674(00)80453-5

D. G. Konstantinidis, A. George, and T. A. Kalfa, Rac GTPases in erythroid biology, Transfusion Clinique et Biologique, vol.17, issue.3, pp.126-130, 2010.
DOI : 10.1016/j.tracli.2010.05.002

P. A. Konstantinopoulos, M. V. Karamouzis, and A. G. Papavassiliou, Post-translational modifications and regulation of the RAS superfamily of GTPases as anticancer targets, Nature Reviews Drug Discovery, vol.49, issue.7, pp.541-555, 2007.
DOI : 10.4049/jimmunol.176.1.640

S. Lo, S. Mane, K. K. Ma, N. K. Kidd, R. P. Hayward et al., Exome sequencing identifies recurrent somatic RAC1 mutations in melanoma, Nat Genet, vol.44, issue.9, pp.1006-1014, 2012.

R. Kuhn, F. Schwenk, M. Aguet, and K. Rajewsky, Inducible gene targeting in mice, Science, vol.200, issue.1, pp.1427-1429, 1995.
DOI : 10.1111/j.1432-1033.1991.tb21041.x

S. Kuroda, M. Fukata, K. Kobayashi, M. Nakafuku, N. Nomura et al., Identification of IQGAP as a Putative Target for the Small GTPases, Cdc42 and Rac1, Journal of Biological Chemistry, vol.79, issue.38, pp.23363-23367, 1996.
DOI : 10.1016/0092-8674(91)90410-Z

D. A. Lauffenburger and A. F. Horwitz, Cell Migration: A Physically Integrated Molecular Process, Cell, vol.84, issue.3, pp.359-369, 1996.
DOI : 10.1016/S0092-8674(00)81280-5

URL : https://doi.org/10.1016/s0092-8674(00)81280-5

T. W. Lebien, B-cell lymphopoiesis in mouse and man, Current Opinion in Immunology, vol.10, issue.2, pp.188-195, 1998.
DOI : 10.1016/S0952-7915(98)80248-3

G. Letort, H. Ennomani, L. Gressin, M. Thery, and L. Blanchoin, Dynamic reorganization of the actin cytoskeleton, F1000Research, 2015.
DOI : 10.12688/f1000research.6374.1

URL : https://hal.archives-ouvertes.fr/hal-01600852

T. Leung, X. Q. Chen, I. Tan, E. Manser, and L. Lim, Myotonic Dystrophy Kinase-Related Cdc42-Binding Kinase Acts as a Cdc42 Effector in Promoting Cytoskeletal Reorganization, Molecular and Cellular Biology, vol.18, issue.1, pp.130-140, 1998.
DOI : 10.1128/MCB.18.1.130

M. Levay, J. Settleman, and E. Ligeti, Regulation of the Substrate Preference of p190RhoGAP by Protein Kinase C-Mediated Phosphorylation of a Phospholipid Binding Site, Biochemistry, vol.48, issue.36, pp.8615-8623, 2009.
DOI : 10.1021/bi900667y

C. Lindon, Control of mitotic exit and cytokinesis by the APC/C, Biochemical Society Transactions, vol.36, issue.3, pp.405-410, 2008.
DOI : 10.1042/BST0360405

C. Liot, L. Seguin, A. Siret, C. Crouin, S. Schmidt et al., APCcdh1 Mediates Degradation of the Oncogenic Rho-GEF Ect2 after Mitosis, PLoS ONE, vol.382, issue.8, p.23676, 2011.
DOI : 10.1371/journal.pone.0023676.s005

J. Liu, J. Zhang, Y. Ginzburg, H. Li, F. Xue et al., Quantitative analysis of murine terminal erythroid differentiation in vivo: novel method to study normal and disordered erythropoiesis, Blood, vol.121, issue.8, pp.43-49, 2013.
DOI : 10.1182/blood-2012-09-456079

M. Liu, F. Bi, X. Zhou, and Y. Zheng, Rho GTPase regulation by miRNAs and covalent modifications, Trends in Cell Biology, vol.22, issue.7, pp.365-373, 2012.
DOI : 10.1016/j.tcb.2012.04.004

F. Livak, M. Tourigny, D. G. Schatz, and H. T. Petrie, Characterization of TCR gene rearrangements during adult murine T cell development, J Immunol, vol.162, issue.5, pp.2575-2580, 1999.

M. Lu, H. Kawamoto, Y. Katsube, T. Ikawa, and Y. Katsura, The Common Myelolymphoid Progenitor: A Key Intermediate Stage in Hemopoiesis Generating T and B Cells, The Journal of Immunology, vol.169, issue.7, pp.3519-3525, 2002.
DOI : 10.4049/jimmunol.169.7.3519

P. Madaule and R. Axel, A novel ras-related gene family, Cell, vol.41, issue.1, pp.31-40, 1985.
DOI : 10.1016/0092-8674(85)90058-3

A. S. Maddox and K. Burridge, RhoA is required for cortical retraction and rigidity during mitotic cell rounding, The Journal of Cell Biology, vol.57, issue.6, pp.255-265, 2003.
DOI : 10.1091/mbc.8.8.1415

P. Mali, D. Wirtz, and P. C. Searson, Interplay of RhoA and Motility in the Programmed Spreading of Daughter Cells Postmitosis, Biophysical Journal, vol.99, issue.11, pp.3526-3534, 2010.
DOI : 10.1016/j.bpj.2010.10.006

I. L. Martensson and R. Ceredig, Role of the surrogate light chain and the pre-B-cell receptor in mouse B-cell development, Immunology, vol.14, issue.4, pp.435-441, 2000.
DOI : 10.1016/S0092-8674(00)80472-9

H. K. Matthews, U. Delabre, J. L. Rohn, J. Guck, P. Kunda et al., Changes in Ect2 Localization Couple Actomyosin-Dependent Cell Shape Changes to Mitotic Progression, Developmental Cell, vol.23, issue.2, pp.371-383, 2012.
DOI : 10.1016/j.devcel.2012.06.003

URL : https://hal.archives-ouvertes.fr/hal-01556637

N. Meller, S. Merlot, and C. Guda, CZH proteins: a new family of Rho-GEFs, Journal of Cell Science, vol.118, issue.21, pp.4937-4946, 2005.
DOI : 10.1242/jcs.02671

J. D. Gillette, S. A. Jaffe, and . Carr, Integrated proteomic analysis of posttranslational modifications by serial enrichment, Nat Methods, vol.10, issue.7, pp.634-637, 2013.

A. Minden, A. Lin, F. X. Claret, A. Abo, and M. Karin, Selective activation of the JNK signaling cascadeand c-Jun transcriptional activity by the small GTPases Rac and Cdc42Hs, Cell, vol.81, issue.7, pp.1147-1157, 1995.
DOI : 10.1016/S0092-8674(05)80019-4

S. Tatsuka, W. S. Narumiya, . May, T. Jr, K. Nosaka et al., Phosphorylation by aurora B converts MgcRacGAP to a RhoGAP during cytokinesis, Dev Cell, vol.4, issue.4, pp.549-560, 2003.

F. Miralles, G. Posern, A. I. Zaromytidou, and R. Treisman, Actin Dynamics Control SRF Activity by Regulation of Its Coactivator MAL, Cell, vol.113, issue.3, pp.329-342, 2003.
DOI : 10.1016/S0092-8674(03)00278-2

K. Kato, T. Takenaka, T. Miyamoto, K. Maeda, and . Akashi, Identification of unipotent megakaryocyte progenitors in human hematopoiesis, Blood, vol.129, issue.25, pp.3332-3343, 2017.

S. Y. Moon and Y. Zheng, Rho GTPase-activating proteins in cell regulation, Trends in Cell Biology, vol.13, issue.1, pp.13-22, 2003.
DOI : 10.1016/S0962-8924(02)00004-1

K. Mori, M. Amano, M. Takefuji, K. Kato, Y. Morita et al., Rho-kinase Contributes to Sustained RhoA Activation through Phosphorylation of p190A RhoGAP, Journal of Biological Chemistry, vol.87, issue.8, pp.5067-5076, 2009.
DOI : 10.3317/jraas.2002.001

Y. Morishita, K. Tsutsumi, and Y. Ohta, Phosphorylation of Serine 402 Regulates RacGAP Protein Activity of FilGAP Protein, Journal of Biological Chemistry, vol.5, issue.43, pp.26328-26338, 2015.
DOI : 10.1074/jbc.M109.059592

S. J. Morrison, A. M. Wandycz, H. D. Hemmati, D. E. Wright, and I. L. Weissman, Identification of a lineage of multipotent hematopoietic progenitors, Development, vol.124, issue.10, pp.1929-1939, 1997.

S. Na, I. S. Li, H. Grewal, R. J. Enslen, J. H. Davis et al., Expression of activated CDC42 induces T cell apoptosis in thymus and peripheral lymph organs via different pathways, Oncogene, vol.18, issue.56, pp.7966-7974, 1999.
DOI : 10.1016/0022-1759(95)00124-S

M. Nethe and P. L. Hordijk, The role of ubiquitylation and degradation in RhoGTPase signalling, Journal of Cell Science, vol.123, issue.23, pp.4011-4018, 2010.
DOI : 10.1242/jcs.078360

P. A. Ney, Normal and disordered reticulocyte maturation, Current Opinion in Hematology, vol.18, issue.3, pp.152-157, 2011.
DOI : 10.1097/MOH.0b013e328345213e

URL : http://europepmc.org/articles/pmc3157046?pdf=render

C. D. Nobes and A. Hall, Rho, Rac, and Cdc42 GTPases regulate the assembly of multimolecular focal complexes associated with actin stress fibers, lamellipodia, and filopodia, Cell, vol.81, issue.1, pp.53-62, 1995.
DOI : 10.1016/0092-8674(95)90370-4

M. Ogawa, E. Boekel, and F. Melchers, Identification of CD19???B220+c-Kit+Flt3/Flk-2+cells as early B lymphoid precursors before pre-B-I cells in juvenile mouse bone marrow, International Immunology, vol.12, issue.3, pp.313-324, 2000.
DOI : 10.1016/0092-8674(92)90293-L

S. Okada, H. Nakauchi, K. Nagayoshi, S. Nishikawa, Y. Miura et al., In vivo and in vitro stem cell function of c-kit-and Sca-1-positive murine hematopoietic cells, Blood, vol.80, issue.12, pp.3044-3050, 1992.

M. F. Olson, A. Ashworth, and A. Hall, An essential role for Rho, Rac, and Cdc42 GTPases in cell cycle progression through G1, Science, vol.372, issue.6508, pp.1270-1272, 1995.
DOI : 10.1038/372798a0

J. L. Orgaz, C. Herraiz, and V. Sanz-moreno, Rho GTPases modulate malignant transformation of tumor cells, Small GTPases, vol.90, issue.4, p.29019, 2014.
DOI : 10.1677/ERC-06-0036

S. H. Orkin and L. I. Zon, Hematopoiesis: An Evolving Paradigm for Stem Cell Biology, Cell, vol.132, issue.4, pp.631-644, 2008.
DOI : 10.1016/j.cell.2008.01.025

URL : https://doi.org/10.1016/j.cell.2008.01.025

M. Osawa, K. Hanada, H. Hamada, and H. Nakauchi, Long-Term Lymphohematopoietic Reconstitution by a Single CD34-Low/Negative Hematopoietic Stem Cell, Science, vol.273, issue.5272, pp.242-245, 1996.
DOI : 10.1126/science.273.5272.242

S. K. Pandit, B. Westendorp, and A. De-bruin, Physiological significance of polyploidization in mammalian cells, Trends in Cell Biology, vol.23, issue.11, pp.556-566, 2013.
DOI : 10.1016/j.tcb.2013.06.002

R. Perona, S. Montaner, L. Saniger, I. Sanchez-perez, R. Bravo et al., Activation of the nuclear factor-kappaB by Rho, CDC42, and Rac-1 proteins., Genes & Development, vol.11, issue.4, pp.463-475, 1997.
DOI : 10.1101/gad.11.4.463

J. Pines and C. L. Rieder, Re-staging mitosis: a contemporary view of mitotic progression, Nature Cell Biology, vol.282, issue.1, pp.3-6, 2001.
DOI : 10.1126/science.282.5394.1721

I. L. Sigvardsson, D. Weissman, and . Bryder, Elucidation of the phenotypic, functional, and molecular topography of a myeloerythroid progenitor cell hierarchy, Cell Stem Cell, vol.1, issue.4, pp.428-442, 2007.

C. Ramachandran, R. V. Patil, K. Combrink, N. A. Sharif, and S. P. Srinivas, Rho- Rho kinase pathway in the actomyosin contraction and cell-matrix adhesion in immortalized human trabecular meshwork cells, Mol Vis, vol.17, pp.1877-1890, 2011.

L. Reibel, O. Dorseuil, R. Stancou, J. Bertoglio, and G. Gacon, A hemopoietic specific gene encoding a small GTP binding protein is overexpressed during T cell activation, Biochemical and Biophysical Research Communications, vol.175, issue.2, pp.451-458, 1991.
DOI : 10.1016/0006-291X(91)91585-Z

T. Reya, S. J. Morrison, M. F. Clarke, and I. L. Weissman, Stem cells, cancer, and cancer stem cells, Nature, vol.414, issue.6859, pp.105-111, 2001.
DOI : 10.1038/35102167

A. J. Ridley and A. Hall, The small GTP-binding protein rho regulates the assembly of focal adhesions and actin stress fibers in response to growth factors, Cell, vol.70, issue.3, pp.389-399, 1992.
DOI : 10.1016/0092-8674(92)90163-7

A. J. Ridley, H. F. Paterson, C. L. Johnston, D. Diekmann, and A. Hall, The small GTP-binding protein rac regulates growth factor-induced membrane ruffling, Cell, vol.70, issue.3, pp.401-410, 1992.
DOI : 10.1016/0092-8674(92)90164-8

A. J. Ridley, M. A. Schwartz, K. Burridge, R. A. Firtel, M. H. Ginsberg et al., Cell Migration: Integrating Signals from Front to Back, Science, vol.302, issue.5651, pp.1704-1709, 2003.
DOI : 10.1126/science.1092053

C. L. Rieder, Mitosis in vertebrates: the G2/M and M/A transitions and their associated checkpoints, Chromosome Research, vol.12, issue.Suppl 1, pp.291-306, 2011.
DOI : 10.1016/j.devcel.2007.01.003

K. Riento and A. J. Ridley, ROCKs: multifunctional kinases in cell behaviour, Nature Reviews Molecular Cell Biology, vol.277, issue.6, pp.446-456, 2003.
DOI : 10.1074/jbc.M110508200

A. E. Rodriguez-fraticelli, S. Vergarajauregui, D. J. Eastburn, A. Datta, M. A. Alonso et al., The Cdc42 GEF Intersectin 2 controls mitotic spindle orientation to form the lumen during epithelial morphogenesis, The Journal of Cell Biology, vol.114, issue.4, pp.725-738, 2010.
DOI : 10.1083/jcb.201002047.dv

K. L. Rossman, C. J. Der, and J. Sondek, GEF means go: turning on RHO GTPases with guanine nucleotide-exchange factors, Nature Reviews Molecular Cell Biology, vol.374, issue.2, pp.167-180, 2005.
DOI : 10.1038/374474a0

M. Rothkegel, O. Mayboroda, M. Rohde, C. Wucherpfennig, R. Valenta et al., Plant and animal profilins are functionally equivalent and stabilize microfilaments in living animal cells, J Cell Sci, vol.109, pp.83-90, 1996.

O. Hasegawa, S. Nureki, N. Miyano, K. Nakamura, S. Takeuchi et al., Somatic RHOA mutation in angioimmunoblastic T cell lymphoma, Nat Genet, vol.46, issue.2, pp.171-175, 2014.

. Marshall, Rac activation and inactivation control plasticity of tumor cell movement, Cell, vol.135, issue.3, pp.510-523, 2008.

A. Schaefer, N. R. Reinhard, and P. L. Hordijk, Toward understanding RhoGTPase specificity: structure, function and local activation, Small GTPases, vol.269, issue.2, p.6, 2014.
DOI : 10.1021/bi010288k

URL : http://www.tandfonline.com/doi/pdf/10.4161/21541248.2014.968004?needAccess=true

A. Schmidt and A. Hall, Guanine nucleotide exchange factors for Rho GTPases: turning on the switch, Genes & Development, vol.16, issue.13, pp.1587-1609, 2002.
DOI : 10.1101/gad.1003302

M. Schmidt, A. Rohe, C. Platzer, A. Najjar, F. Erdmann et al., Regulation of G2/M Transition by Inhibition of WEE1 and PKMYT1 Kinases, Molecules, vol.22, issue.12, 2017.

A. Schnelzer, D. Prechtel, U. Knaus, K. Dehne, M. Gerhard et al., Rac1 in human breast cancer: overexpression, mutation analysis and characterization of a new isoform, Rac1b, Oncogene, vol.19, issue.26, pp.3013-3020, 2000.
DOI : 10.1038/sj.onc.1202181

R. P. Scholz, J. Regner, A. Theil, P. Erlmann, G. Holeiter et al., DLC1 interacts with 14-3-3 proteins to inhibit RhoGAP activity and block nucleocytoplasmic shuttling, Journal of Cell Science, vol.122, issue.1, pp.92-102, 2009.
DOI : 10.1242/jcs.036251

M. Sebbagh, J. Hamelin, J. Bertoglio, E. Solary, and J. Breard, Direct cleavage of ROCK II by granzyme B induces target cell membrane blebbing in a caspase-independent manner, The Journal of Experimental Medicine, vol.16, issue.3, pp.465-471, 2005.
DOI : 10.1128/MCB.16.10.5313

L. Seguin, C. Liot, R. Mzali, R. Harada, A. Siret et al., CUX1 and E2F1 Regulate Coordinated Expression of the Mitotic Complex Genes Ect2, MgcRacGAP, and MKLP1 in S Phase, Molecular and Cellular Biology, vol.29, issue.2, pp.570-581, 2009.
DOI : 10.1128/MCB.01275-08

M. A. Sells, J. T. Boyd, and J. Chernoff, p21-Activated Kinase 1 (Pak1) Regulates Cell Motility in Mammalian Fibroblasts, The Journal of Cell Biology, vol.17, issue.4, pp.837-849, 1999.
DOI : 10.1074/jbc.273.29.18481

M. A. Sells, U. G. Knaus, S. Bagrodia, D. M. Ambrose, G. M. Bokoch et al., Human p21-activated kinase (Pak1) regulates actin organization in mammalian cells, Current Biology, vol.7, issue.3, pp.202-210, 1997.
DOI : 10.1016/S0960-9822(97)70091-5

M. Socolovsky, H. Nam, M. D. Fleming, V. H. Haase, C. Brugnara et al., Ineffective erythropoiesis in Stat5a-/-5b-/- mice due to decreased survival of early erythroblasts, Blood, vol.98, issue.12, pp.3261-3273, 2001.
DOI : 10.1182/blood.V98.12.3261

B. Song, X. S. Liu, and X. Liu, Polo-like kinase 1 (Plk1): an Unexpected Player in DNA Replication, Cell Division, vol.7, issue.1, 2012.
DOI : 10.1186/1747-1028-7-3

G. J. Spangrude, S. Heimfeld, and I. L. Weissman, Purification and characterization of mouse hematopoietic stem cells, Science, vol.241, issue.4861, pp.58-62, 1988.
DOI : 10.1126/science.2898810

N. Sternberg and D. Hamilton, Bacteriophage P1 site-specific recombination, Journal of Molecular Biology, vol.150, issue.4, pp.467-486, 1981.
DOI : 10.1016/0022-2836(81)90375-2

L. Su, J. M. Agati, and S. J. Parsons, p190RhoGAP is cell cycle regulated and affects cytokinesis, The Journal of Cell Biology, vol.7, issue.3, pp.571-582, 2003.
DOI : 10.1016/S0960-9822(01)00307-4

URL : http://jcb.rupress.org/content/jcb/163/3/571.full.pdf

A. Sudarov, F. Gooden, D. Tseng, W. B. Gan, and M. E. Ross, Lis1 controls dynamics of neuronal filopodia and spines to impact synaptogenesis and social behaviour, EMBO Molecular Medicine, vol.46, issue.4, pp.591-607, 2013.
DOI : 10.1016/j.neuron.2005.04.001

T. Sumi, K. Matsumoto, A. Shibuya, and T. Nakamura, Activation of LIM Kinases by Myotonic Dystrophy Kinase-related Cdc42-binding Kinase ??, Journal of Biological Chemistry, vol.249, issue.25, pp.23092-23096, 2001.
DOI : 10.1016/S0092-8674(00)81281-7

A. Suzuki, J. W. Shin, Y. Wang, S. H. Min, M. Poncz et al., RhoA Is Essential for Maintaining Normal Megakaryocyte Ploidy and Platelet Generation, PLoS ONE, vol.91, issue.7, p.69315, 2013.
DOI : 10.1371/journal.pone.0069315.g006

G. Szalai, A. C. Larue, and D. K. Watson, Molecular mechanisms of megakaryopoiesis, Cellular and Molecular Life Sciences, vol.63, issue.21, pp.2460-2476, 2006.
DOI : 10.1007/s00018-006-6190-8

T. Takenawa and S. Suetsugu, The WASP???WAVE protein network: connecting the membrane to the cytoskeleton, Nature Reviews Molecular Cell Biology, vol.45, issue.1, pp.37-48, 2007.
DOI : 10.1091/mbc.10.10.3521

C. G. Takizawa and D. O. Morgan, Control of mitosis by changes in the subcellular location of cyclin-B1???Cdk1 and Cdc25C, Current Opinion in Cell Biology, vol.12, issue.6, pp.658-665, 2000.
DOI : 10.1016/S0955-0674(00)00149-6

T. Tatsumoto, X. Xie, R. Blumenthal, I. Okamoto, and T. Miki, Human Ect2 Is an Exchange Factor for Rho Gtpases, Phosphorylated in G2/M Phases, and Involved in Cytokinesis, The Journal of Cell Biology, vol.11, issue.5, pp.921-928, 1999.
DOI : 10.1093/emboj/17.21.6404

J. Tcherkezian, E. I. Danek, S. Jenna, I. Triki, and N. Lamarche-vane, Extracellular Signal-Regulated Kinase 1 Interacts with and Phosphorylates CdGAP at an Important Regulatory Site, Molecular and Cellular Biology, vol.25, issue.15, pp.6314-6329, 2005.
DOI : 10.1128/MCB.25.15.6314-6329.2005

G. Terszowski, C. Waskow, P. Conradt, D. Lenze, J. Koenigsmann et al., Prospective isolation and global gene expression analysis of the erythrocyte colony-forming unit (CFU-E), Blood, vol.105, issue.5, pp.1937-1945, 2005.
DOI : 10.1182/blood-2004-09-3459

J. Frierson, Reduced expression of metastasis suppressor RhoGDI2 is associated with decreased survival for patients with bladder cancer, Clin Cancer Res, vol.10, issue.11, pp.3800-3806, 2004.

K. F. Tolias, L. C. Cantley, and C. L. Carpenter, Rho Family GTPases Bind to Phosphoinositide Kinases, Journal of Biological Chemistry, vol.264, issue.30, pp.17656-17659, 1995.
DOI : 10.1042/bj2880637

M. Trahey and F. Mccormick, A cytoplasmic protein stimulates normal N-ras p21, 1987.
DOI : 10.1126/science.2821624

. Gtpase, but does not affect oncogenic mutants, Science, vol.238, issue.4826, pp.542-545

B. K. Tripathi, X. Qian, P. Mertins, D. Wang, A. G. Papageorge et al., CDK5 is a major regulator of the tumor suppressor DLC1, The Journal of Cell Biology, vol.63, issue.5, pp.627-642, 2014.
DOI : 10.1016/S0896-6273(00)81200-3

M. G. Tucci, G. Lucarini, D. Brancorsini, A. Zizzi, A. Pugnaloni et al., Involvement of E-cadherin, ??-catenin, Cdc42 and CXCR4 in the progression and prognosis of cutaneous melanoma, British Journal of Dermatology, vol.113, issue.6, pp.1212-1216, 2007.
DOI : 10.1158/0008-5472.CAN-03-1757

N. Uchida and I. L. Weissman, Searching for hematopoietic stem cells: evidence that Thy-1.1lo Lin- Sca-1+ cells are the only stem cells in C57BL/Ka-Thy-1.1 bone marrow, Journal of Experimental Medicine, vol.175, issue.1, pp.175-184, 1992.
DOI : 10.1084/jem.175.1.175

F. M. Vega and A. J. Ridley, Rho GTPases in cancer cell biology, FEBS Letters, vol.6, issue.14, pp.2093-2101, 2008.
DOI : 10.2174/156802606777812095

O. Visvikis, M. P. Maddugoda, and E. Lemichez, Direct modifications of Rho proteins: deconstructing GTPase regulation, Biology of the Cell, vol.313, issue.7, pp.377-389, 2010.
DOI : 10.1126/science.1129551

M. P. Williams, V. L. Cancro, and . Tybulewicz, Critical roles for Rac1 and Rac2 GTPases in B cell development and signaling, Science, vol.302, issue.5644, pp.459-462, 2003.

. Wang, Activation of Rac1 GTPase promotes leukemia cell chemotherapy resistance, quiescence and niche interaction, Molecular Oncology, vol.24, issue.2, pp.907-916, 2013.
DOI : 10.1128/MCB.24.14.6205-6214.2004

L. Wang, L. Yang, M. D. Filippi, D. A. Williams, and Y. Zheng, Genetic deletion of Cdc42GAP reveals a role of Cdc42 in erythropoiesis and hematopoietic stem/progenitor cell survival, adhesion, and engraftment, Blood, vol.107, issue.1, pp.98-105, 2006.
DOI : 10.1182/blood-2005-05-2171

N. Watanabe, P. Madaule, T. Reid, T. Ishizaki, G. Watanabe et al., p140mDia, a mammalian homolog of Drosophila diaphanous, is a target protein for Rho small GTPase and is a ligand for profilin, The EMBO Journal, vol.16, issue.11, pp.3044-3056, 1997.
DOI : 10.1093/emboj/16.11.3044

S. Watanabe, T. De-zan, T. Ishizaki, S. Yasuda, H. Kamijo et al., Loss of a Rho-Regulated Actin Nucleator, mDia2, Impairs Cytokinesis during Mouse Fetal Erythropoiesis, Cell Reports, vol.5, issue.4, pp.926-932, 2013.
DOI : 10.1016/j.celrep.2013.10.021

I. L. Weissman, D. J. Anderson, and F. Gage, Stem and Progenitor Cells: Origins, Phenotypes, Lineage Commitments, and Transdifferentiations, Annual Review of Cell and Developmental Biology, vol.17, issue.1, pp.387-403, 2001.
DOI : 10.1146/annurev.cellbio.17.1.387

Y. Wen, C. H. Eng, J. Schmoranzer, N. Cabrera-poch, E. J. Morris et al., EB1 and APC bind to mDia to stabilize microtubules downstream of Rho and promote cell migration, Nature Cell Biology, vol.26, issue.9, pp.820-830, 2004.
DOI : 10.1016/S1046-2023(02)00023-3

K. Wennerberg, K. L. Rossman, and C. J. Der, The Ras superfamily at a glance, Journal of Cell Science, vol.118, issue.5, pp.843-846, 2005.
DOI : 10.1242/jcs.01660

. Der, Rac regulation of transformation, gene expression, and actin organization by multiple, PAK-independent pathways, Mol Cell Biol, vol.17, issue.3, pp.1324-1335, 1997.

M. Wherlock and H. Mellor, The Rho GTPase family: a Racs to Wrchs story, J Cell Sci, vol.115, issue.2, pp.239-240, 2002.

B. Wickstead and K. Gull, The evolution of the cytoskeleton, The Journal of Cell Biology, vol.8, issue.4, pp.513-525, 2011.
DOI : 10.1186/1745-6150-4-9

C. F. Offner, L. Dunant, E. Eshkind, P. Bockamp, H. R. Lio et al., Hematopoietic stem cells reversibly switch from dormancy to self-renewal during homeostasis and repair, Cell, vol.135, issue.6, pp.1118-1129, 2008.

A. Wilson and A. Trumpp, Bone-marrow haematopoietic-stem-cell niches, Nature Reviews Immunology, vol.89, issue.2, pp.93-106, 2006.
DOI : 10.1038/nature04247

R. M. Wolf, N. Draghi, X. Liang, C. Dai, L. Uhrbom et al., p190RhoGAP can act to inhibit PDGF-induced gliomas in mice: a putative tumor suppressor encoded on human Chromosome 19q13.3, Genes & Development, vol.17, issue.4, pp.476-487, 2003.
DOI : 10.1101/gad.1040003

D. Wu, M. Asiedu, R. S. Adelstein, and Q. Wei, A Novel Guanine Nucleotide Exchange Factor, MYOGEF, is Required for Cytokinesis, Cell Cycle, vol.5, issue.11, pp.1234-1239, 2006.
DOI : 10.4161/cc.5.11.2815

J. Xavier-ferrucio, L. Ricon, K. Vieira, A. L. Longhini, M. Lazarini et al., Hematopoietic defects in response to reduced Arhgap21, Stem Cell Research, vol.26, pp.17-27, 2017.
DOI : 10.1016/j.scr.2017.11.014

H. Xu, S. Eleswarapu, H. Geiger, K. Szczur, D. Daria et al., Loss of the Rho GTPase activating protein p190-B enhances hematopoietic stem cell engraftment potential, Blood, vol.114, issue.17, pp.3557-3566, 2009.
DOI : 10.1182/blood-2009-02-205815

M. Kuehnel, S. Wigler, L. Powers, S. W. Zender, and . Lowe, DLC1 is a chromosome 8p tumor suppressor whose loss promotes hepatocellular carcinoma, Genes Dev, vol.22, issue.11, pp.1439-1444, 2008.

M. B. Yaffe, K. Rittinger, S. Volinia, P. R. Caron, A. Aitken et al., The Structural Basis for 14-3-3:Phosphopeptide Binding Specificity, Cell, vol.91, issue.7, pp.961-971, 1997.
DOI : 10.1016/S0092-8674(00)80487-0

M. Yamamoto, N. Marui, T. Sakai, N. Morii, S. Kozaki et al., ADP-ribosylation of the rhoA gene product by botulinum C3 exoenzyme causes Swiss 3T3 cells to accumulate in the G1 phase of the cell cycle, Oncogene, vol.8, issue.6, pp.1449-1455, 1993.

F. C. Yang, S. J. Atkinson, Y. Gu, J. B. Borneo, A. W. Roberts et al., Rac and Cdc42 GTPases control hematopoietic stem cell shape, adhesion, migration, and mobilization, Proceedings of the National Academy of Sciences, vol.10, issue.7, pp.98-5614, 2001.
DOI : 10.1016/S0960-9822(00)00417-6

L. Yang, L. Wang, H. Geiger, J. A. Cancelas, J. Mo et al., Rho GTPase Cdc42 coordinates hematopoietic stem cell quiescence and niche interaction in the bone marrow, Proceedings of the National Academy of Sciences, vol.17, issue.11, pp.5091-5096, 2007.
DOI : 10.1091/mbc.E06-05-0466

L. Yang, L. Wang, T. A. Kalfa, J. A. Cancelas, X. Shang et al., Cdc42 critically regulates the balance between myelopoiesis and erythropoiesis, Blood, vol.110, issue.12, pp.3853-3861, 2007.
DOI : 10.1182/blood-2007-03-079582

S. Yasuda, F. Oceguera-yanez, T. Kato, M. Okamoto, S. Yonemura et al., Cdc42 and mDia3 regulate microtubule attachment to kinetochores, Nature, vol.113, issue.6984, pp.767-771, 2004.
DOI : 10.1016/S0092-8674(03)00465-3

K. H. Lee, W. Cho, H. Kim, J. Ju, S. J. Kim et al., A recurrent inactivating mutation in RHOA GTPase in angioimmunoblastic T cell lymphoma, Nat Genet, vol.46, issue.4, pp.371-375, 2014.

Y. S. Yoon, J. H. Choo, T. Yoo, K. Kang, and J. H. Chung, RhoB is epigenetically regulated in an age- and tissue-specific manner, Biochemical and Biophysical Research Communications, vol.362, issue.1, pp.164-169, 2007.
DOI : 10.1016/j.bbrc.2007.08.002

H. Yoshizaki, Y. Ohba, M. C. Parrini, N. G. Dulyaninova, A. R. Bresnick et al., Cell Type-specific Regulation of RhoA Activity during Cytokinesis, Journal of Biological Chemistry, vol.72, issue.43, pp.44756-44762, 2004.
DOI : 10.1242/jcs.00993

H. Yu, D. Leitenberg, B. Li, and R. A. , Deficiency of Small Gtpase Rac2 Affects T Cell Activation, The Journal of Experimental Medicine, vol.16, issue.7, pp.915-926, 2001.
DOI : 10.1016/S0092-8674(00)81371-9

O. Yuce, A. Piekny, and M. Glotzer, An ECT2???centralspindlin complex regulates the localization and function of RhoA, The Journal of Cell Biology, vol.16, issue.4, pp.571-582, 2005.
DOI : 10.1016/j.yexcr.2004.01.005

E. Zanin, A. Desai, I. Poser, Y. Toyoda, C. Andree et al., A Conserved RhoGAP Limits M Phase Contractility and Coordinates with Microtubule Asters to Confine RhoA during Cytokinesis, Developmental Cell, vol.26, issue.5, pp.496-510, 2013.
DOI : 10.1016/j.devcel.2013.08.005

J. Zhang, M. Socolovsky, A. W. Gross, and H. F. Lodish, Role of Ras signaling in erythroid differentiation of mouse fetal liver cells: functional analysis by a flow cytometry-based novel culture system, Blood, vol.102, issue.12, pp.3938-3946, 2003.
DOI : 10.1182/blood-2003-05-1479

Y. Zheng, Dbl family guanine nucleotide exchange factors, Trends in Biochemical Sciences, vol.26, issue.12, pp.724-732, 2001.
DOI : 10.1016/S0968-0004(01)01973-9

Q. Showe, A. Liu, S. M. Vachani, and J. L. Albelda, The Rac1 splice form Rac1b promotes K-ras-induced lung tumorigenesis, Oncogene, vol.32, issue.7, pp.903-909, 2013.

X. Zhou, M. C. Florian, P. Arumugam, X. Chen, J. A. Cancelas et al., RhoA GTPase controls cytokinesis and programmed necrosis of hematopoietic progenitors, The Journal of Experimental Medicine, vol.23, issue.11, pp.2371-2385, 2013.
DOI : 10.1016/j.stem.2011.07.003

S. H. Zigmond, Formin-induced nucleation of actin filaments, Current Opinion in Cell Biology, vol.16, issue.1, pp.99-105, 2004.
DOI : 10.1016/j.ceb.2003.10.019