S. Cruz, anti-pCdk2 polyclonal (Santa Cruz) au 1/1000, anti-PARP clivée polyclonal, 1000.

. La-membrane-est-ensuite-lavée, × 5 min) avant d'être marquée pendant 1 h avec un anticorps secondaire couplé à la peroxydase de Raifort (HRP) : anti-IgG de lapin préparé chez la chèvre

L. Après-traitement, P. Totalité-des-cellules-est-récupérée-et-lavée-au, and . 1x, Les cellules sont ensuite fixées et perméabilisées dans l'éthanol à 80 % puis sont stockées à -20°C jusqu'au moment de l'analyse. Le marquage à l'iodure de propidium

B. B. Aggarwal and S. Shishodia, Molecular targets of dietary agents for prevention and therapy of cancer, Biochemical Pharmacology, vol.71, issue.10
DOI : 10.1016/j.bcp.2006.02.009

N. Ahmad, V. M. Adhami, F. Afaq, D. K. Feyes, and H. Mukhtar, Resveratrol causes WAF-1/p21-mediated G(1)- phase arrest of cell cycle and induction of apoptosis in human epidermoid carcinoma A431 cells

G. M. Arantes, The Catalytic Acid in the Dephosphorylation of the Cdk2-pTpY/CycA Protein Complex by Cdc25B Phosphatase, The Journal of Physical Chemistry B, vol.112, issue.47, pp.15244-15247, 2008.
DOI : 10.1021/jp8070019

G. M. Arantes, Flexibility and inhibitor binding in cdc25 phosphatases, Proteins: Structure, Function, and Bioinformatics, vol.465, issue.14, 2010.
DOI : 10.1002/cmdc.200500092

B. Aressy, B. Bugler, A. Valette, D. Biard, and B. Ducommun, Moderate variations in CDC25B protein levels modulate the response to DNA damaging agents, Cell Cycle, vol.7, issue.14, pp.2234-2240, 2008.
DOI : 10.4161/cc.7.14.6305

URL : https://hal.archives-ouvertes.fr/hal-00317265

B. Aressy and B. Ducommun, Cell Cycle Control by the CDC25 Phosphatases, Anti-Cancer Agents in Medicinal Chemistry, vol.8, issue.8
DOI : 10.2174/187152008786847756

A. Arora, K. Seth, and Y. Shukla, Reversal of P-glycoprotein-mediated multidrug resistance by diallyl sulfide in K562 leukemic cells and in mouse liver, Carcinogenesis, vol.25, issue.6, pp.941-949, 2004.
DOI : 10.1093/carcin/bgh060

M. Athar, J. H. Back, X. Tang, K. H. Kim, L. Kopelovich et al., Resveratrol: A Review of Pre-clinical Studies for Human Cancer Prevention, Toxicol. Appl. Pharmacol, pp.224-274, 2007.

M. Bahassi, R. F. Hennigan, D. L. Myer, and P. J. Stambrook, Cdc25C phosphorylation on serine 191 by Plk3 promotes its nuclear translocation, Oncogene, vol.23, issue.15, pp.2658-2663, 2004.
DOI : 10.1074/jbc.M008151200

V. Baldin, C. Cans, M. Knibiehler, and B. Ducommun, Phosphorylation of Human CDC25B Phosphatase by CDK1-Cyclin A Triggers Its Proteasome-dependent Degradation, Journal of Biological Chemistry, vol.10, issue.52, pp.272-32731, 1997.
DOI : 10.1101/gad.7.5.812

P. Bansal and J. S. Lazo, Induction of Cdc25B Regulates Cell Cycle Resumption after Genotoxic Stress, Cancer Research, vol.67, issue.7
DOI : 10.1158/0008-5472.CAN-06-3685

S. Baürle, T. Blume, J. Günther, D. Henschel, R. C. Hillig et al., Design and Synthesis of Macrocyclic Inhibitors of Phosphatase Cdc25B., ChemInform, vol.14, issue.34
DOI : 10.1002/chin.200434105

A. Begouin, S. Hesse, M. R. Queiroz, and G. Kirsch, Synthesis of Diarylamines in the Thiophene Series by Buchwald???Hartwig Coupling., ChemInform, vol.37, issue.4, pp.2373-2378, 2005.
DOI : 10.1002/chin.200604102

A. Begouin, S. Hesse, M. R. Queiroz, and G. Kirsch, Reactivity of substituted bromothiophenecarboxylates in palladium-catalyzed N-arylation of hetarylamines, Synthesis, vol.16, pp.2794-2798, 2006.

A. Begouin, S. Hesse, M. R. Queiroz, and G. Kirsch, Palladium-catalyzed Buchwald-Hartwig coupling of deactivated aminothiophenes with substituted halopyridines, Eur. J. Org. Chem, vol.10, pp.1678-1682, 2007.

A. Bélot, M. Velten, P. Grosclaude, N. Bossard, G. Launoy et al., Estimation nationale de l'incidence et de la mortalité par cancer en France entre, 1980.

B. A. Bhat, K. L. Dhar, S. C. Puri, A. K. Saxena, M. Shanmugavel et al., Synthesis and biological evaluation of chalcones and their derived pyrazoles as potential cytotoxic agents, Bioorganic & Medicinal Chemistry Letters, vol.15, issue.12
DOI : 10.1016/j.bmcl.2005.03.121

J. L. Blanchard, D. M. Epstein, M. D. Boisclair, J. Rudolph, and K. Pal, Dysidiolide and related ??-hydroxy butenolide compounds as inhibitors of the protein tyrosine phosphatase, CDC25, Bioorganic & Medicinal Chemistry Letters, vol.9, issue.17, pp.2537-2538, 1999.
DOI : 10.1016/S0960-894X(99)00431-X

A. Blasina, I. Van-de-weyer, M. C. Laus, W. H. Luyten, A. E. Parker et al., A human homologue of the checkpoint kinase Cds1 directly inhibits Cdc25 phosphatase, Current Biology, vol.9, issue.1, pp.1-10, 1999.
DOI : 10.1016/S0960-9822(99)80041-4

I. Blomberg and I. Hoffmann, /S Transition and Leads to Premature Activation of Cyclin E- and Cyclin A-Dependent Kinases, Molecular and Cellular Biology, vol.19, issue.9, pp.6183-6194, 1999.
DOI : 10.1128/MCB.19.9.6183

J. Bonnet, P. Coopman, and M. C. Morris, Characterization of centrosomal localization and dynamics of Cdc25C phosphatase in mitosis, Cell Cycle, vol.7, issue.13, 1991.
DOI : 10.4161/cc.7.13.6095

URL : https://hal.archives-ouvertes.fr/hal-00350866

R. Boutros, C. Dozier, and B. Ducommun, The when and wheres of CDC25 phosphatases, Current Opinion in Cell Biology, vol.18, issue.2, pp.185-191, 2006.
DOI : 10.1016/j.ceb.2006.02.003

URL : https://hal.archives-ouvertes.fr/hal-00317392

R. Boutros, V. Lobjois, and B. Ducommun, CDC25B Involvement in the Centrosome Duplication Cycle and in Microtubule Nucleation, Cancer Research, vol.67, issue.24, pp.11557-11564, 2007.
DOI : 10.1158/0008-5472.CAN-07-2415

URL : https://hal.archives-ouvertes.fr/hal-00317336

R. Boutros, V. Lobjois, and B. Ducommun, CDC25 phosphatases in cancer cells: key players? Good targets?, Nature Reviews Cancer, vol.20, issue.7, pp.495-507, 2007.
DOI : 10.1158/0008-5472.CAN-05-1054

URL : https://hal.archives-ouvertes.fr/hal-00317350

E. Braud, M. L. Goddard, S. Kolb, M. P. Brun, O. Mondésert et al., Novel naphthoquinone and quinolinedione inhibitors of CDC25 phosphatase activity with antiproliferative properties, Bioorganic & Medicinal Chemistry, vol.16, issue.19, pp.16-9040, 2008.
DOI : 10.1016/j.bmc.2008.08.009

L. Brault and D. Bagrel, Activity of novel Cdc25 inhibitors and preliminary evaluation of their potentiation of chemotherapeutic drugs in human breast cancer cells, Life Sciences, vol.82, issue.5-6, pp.315-323, 2008.
DOI : 10.1016/j.lfs.2007.12.001

L. Brault, M. Denancé, E. Banaszak, S. Maadidi, E. Battaglia et al., Synthesis and biological evaluation of dialkylsubstituted maleic anhydrides as novel inhibitors of CDC25 dual specificity phosphatases, Eur. J. Med. Chem, pp.42-243, 2007.

M. C. Brezak, M. Quaranta, O. Mondésert, M. O. Galcera, O. Lavergne et al., A Novel Synthetic Inhibitor of CDC25 Phosphatases, Cancer Research, vol.64, issue.9, pp.3320-3325, 2004.
DOI : 10.1158/0008-5472.CAN-03-3984

URL : https://hal.archives-ouvertes.fr/hal-00317444

M. C. Brezak, M. Quaranta, M. O. Contour-galcera, O. Lavergne, O. Mondesert et al., Inhibition of human tumor cell growth in vivo by an orally bioavailable inhibitor of CDC25 phosphatases, Molecular Cancer Therapeutics, vol.4, issue.9, pp.1378-1387, 2005.
DOI : 10.1158/1535-7163.MCT-05-0168

URL : https://hal.archives-ouvertes.fr/hal-00317405

M. C. Brezak, P. G. Kasprzyk, M. O. Galcera, and O. Lavergne, CDC25 Inhibitors as Anticancer Agents Are Moving Forward, Prévost G. P. CDC25 Inhibitors as Anticancer Agents Are Moving Forward, pp.857-862, 2008.
DOI : 10.2174/187152008786847701

M. C. Brezak, A. Valette, M. Quaranta, M. O. Contour-galcera, D. Jullien et al., IRC-083864, a novel bis quinone inhibitor of CDC25 phosphatases active against human cancer cells, International Journal of Cancer, vol.29, issue.6, pp.1449-1456, 2009.
DOI : 10.4161/cc.7.2.5306

M. Brisson, C. Foster, P. Wipf, B. Joo, R. J. Tomko et al., Independent Mechanistic Inhibition of Cdc25 Phosphatases by a Natural Product Caulibugulone, Molecular Pharmacology, vol.71, issue.1, pp.71-185, 2007.
DOI : 10.1124/mol.106.028589

M. Brisson, T. Nguyen, A. Vogt, J. Yalowich, A. Giorgianni et al., Discovery and Characterization of Novel Small Molecule Inhibitors of Human Cdc25B Dual Specificity Phosphatase, Molecular Pharmacology, vol.66, issue.4, pp.66-824, 2004.
DOI : 10.1124/mol.104.001784

M. P. Brun, E. Braud, D. Angotti, O. Mondésert, M. Quaranta et al., Design, synthesis, and biological evaluation of novel naphthoquinone derivatives with CDC25 phosphatase inhibitory activity, Bioorganic & Medicinal Chemistry, vol.13, issue.16, pp.13-4871, 2005.
DOI : 10.1016/j.bmc.2005.05.005

URL : https://hal.archives-ouvertes.fr/hal-00317419

B. Bugler, E. Schmitt, B. Aressy, and B. Ducommun, Unscheduled expression of CDC25B in S-phase leads to replicative stress and DNA damage, Molecular Cancer, vol.9, issue.1, pp.29-31, 2010.
DOI : 10.1186/1476-4598-9-29

. Jr, Initiation of a G2/M checkpoint after ultraviolet radiation requires p38 kinase, Nature, vol.411, pp.102-107, 2001.

D. V. Bulavin, Y. Higashimoto, Z. N. Demidenko, S. Meek, P. Graves et al., Dual phosphorylation controls Cdc25 phosphatases and mitotic entry, Nature Cell Biology, vol.36, issue.6, pp.545-551, 2003.
DOI : 10.1016/S0091-679X(08)60298-8

C. Busch, O. Barton, E. Morgenstern, C. Götz, J. Günther et al., The G2/M checkpoint phosphatase cdc25C is located within centrosomes, The International Journal of Biochemistry & Cell Biology, vol.39, issue.9, pp.1707-1713, 2007.
DOI : 10.1016/j.biocel.2007.04.022

C. Busch, C. Jacob, A. Anwar, T. Burkholz, L. A. Ba et al., Diallylpolysulfides induce growth arrest and apoptosis, Int. J. Oncol, pp.36-743, 2010.

L. Busino, M. Chiesa, G. F. Draetta, and M. Donzelli, Cdc25A phosphatase: combinatorial phosphorylation, ubiquitylation and proteolysis, Oncogene, vol.23, issue.11, pp.2050-2056, 2004.
DOI : 10.1126/science.280.5363.593

URL : http://www.nature.com/onc/journal/v23/n11/pdf/1207394a.pdf

M. G. Cangi, B. Cukor, P. Soung, S. Signoretti, G. Moreira et al., Role of the Cdc25A phosphatase in human breast cancer, Journal of Clinical Investigation, vol.106, issue.6, pp.763-761, 2000.
DOI : 10.1172/JCI9174

S. Cao, C. Foster, M. Brisson, J. S. Lazo, and D. G. Kingston, Halenaquinone and xestoquinone derivatives, inhibitors of CDC25B phosphatase from a Xestospongia sp, Bioorg. Med. Chem, pp.13-999, 2005.

S. Cao, B. T. Murphy, C. Foster, J. S. Lazo, and D. G. Kingston, Bioactivities of simplified adociaquinone B and naphthoquinone derivatives against Cdc25B, MKP-1, and MKP-3 phosphatases, Bioorganic & Medicinal Chemistry, vol.17, issue.6, pp.17-2276, 2009.
DOI : 10.1016/j.bmc.2008.10.090

Z. Cao, J. Fang, C. Xia, X. Shi, and B. H. Jiang, Trans-3,4',5-trihydroxystibene inhibits hypoxia-inducible factor 1alpha and vascular endothelial growth factor expression in human ovarian cancer cells, Clin. Cancer Res, pp.10-5253, 2004.

M. Cazales, R. Boutros, M. C. Brezak, S. Chaumeron, G. Prevost et al., Pharmacologic inhibition of CDC25 phosphatases impairs interphase microtubule dynamics and mitotic spindle assembly, Molecular Cancer Therapeutics, vol.6, issue.1, pp.318-325, 2007.
DOI : 10.1158/1535-7163.MCT-06-0299

URL : https://hal.archives-ouvertes.fr/hal-00317361

R. E. Cebula, J. L. Blanchard, M. D. Boisclair, K. Pal, and N. J. Bockovich, Synthesis and phosphatase inhibitory activity of analogs of sulfircin, Bioorganic & Medicinal Chemistry Letters, vol.7, issue.15, pp.2015-2020, 1997.
DOI : 10.1016/S0960-894X(97)00357-0

C. Cerella, C. Scherer, S. Cristofanon, E. Henry, A. Anwar et al., Cell cycle arrest in early mitosis and induction of caspase-dependent apoptosis in U937 cells by diallyltetrasulfide (Al2S4), Apoptosis, vol.16, issue.Suppl 9B, pp.641-654, 2009.
DOI : 10.4161/cc.1.3.126

M. Chen and C. E. Ryan, Chk1 Kinase Negatively Regulates Mitotic Function of Cdc25A Phosphatase through 14-3-3 Binding, Molecular and Cellular Biology, vol.23, issue.21, pp.7488-7497, 2003.
DOI : 10.1128/MCB.23.21.7488-7497.2003

H. G. Cheon, S. M. Kim, S. D. Yang, J. D. Ha, and J. K. Choi, Discovery of a novel protein tyrosine phosphatase-1B inhibitor, KR61639: potential development as an antihyperglycemic agent, European Journal of Pharmacology, vol.485, issue.1-3, pp.485-333, 2004.
DOI : 10.1016/j.ejphar.2003.11.070

P. Chomczynski and N. Sacchi, Single-step method of RNA isolation by acid guanidinium thiocyanate-phenol-chloroform extraction, Analytical Biochemistry, vol.162, issue.1, pp.156-159, 1987.
DOI : 10.1016/0003-2697(87)90021-2

S. T. Chou, Y. C. Yen, C. M. Lee, and M. S. Chen, Pro-apoptotic Role of Cdc25A, Journal of Biological Chemistry, vol.55, issue.23, pp.17833-17845, 2010.
DOI : 10.1002/cyto.a.10072

D. S. Conklin, K. Galaktionov, and D. Beach, 14-3-3 proteins associate with cdc25 phosphatases., Proceedings of the National Academy of Sciences, vol.92, issue.17, pp.7892-7896, 1995.
DOI : 10.1073/pnas.92.17.7892

M. Contour-galcera, A. Sidhu, G. Prévost, D. Bigg, and B. Ducommun, What's new on CDC25 phosphatase inhibitors, Pharmacology & Therapeutics, vol.115, issue.1, pp.1-12, 2007.
DOI : 10.1016/j.pharmthera.2007.03.009

N. Davezac, V. Baldin, J. Blot, B. Ducommun, and J. P. Tassan, Human pEg3 kinase associates with and phosphorylates CDC25B phosphatase: a potential role for pEg3 in cell cycle regulation, Oncogene, vol.21, issue.50, pp.7630-7641, 2002.
DOI : 10.1093/emboj/18.8.2174

G. Deep and R. Agarwal, New combination therapies with cell cycle agents, Curr. Opin. Investig. Drugs, vol.9, pp.591-604, 2008.

A. R. Demidova, M. Y. Aau, L. Zhuang, and Q. Yu, Dual Regulation of Cdc25A by Chk1 and p53-ATF3 in DNA Replication Checkpoint Control, Journal of Biological Chemistry, vol.269, issue.7, pp.284-4132, 2009.
DOI : 10.1073/pnas.0505585102

G. Derbesy and D. N. Harpp, A simple method to prepare unsymmetrical di- tri- and tetrasulfides, Tetrahedron Letters, vol.35, issue.30
DOI : 10.1016/S0040-4039(00)73505-2

K. Dodo, M. Takahashi, Y. Yamada, Y. Sugimoto, Y. Hashimoto et al., Synthesis of a novel class of cdc25A inhibitors from vitamin D 3, Bioorganic & Medicinal Chemistry Letters, vol.10, issue.7, pp.615-617, 2000.
DOI : 10.1016/S0960-894X(00)00065-2

M. Donzelli, M. Squatrito, D. Ganoth, A. Hershko, M. Pagano et al., Dual mode of degradation of Cdc25 A phosphatase, The EMBO Journal, vol.21, issue.18, pp.4875-4884, 2002.
DOI : 10.1093/emboj/cdf491

N. Druesne, A. Pagniez, C. Mayeur, M. Thomas, C. Cherbuy et al., Diallyl disulfide (DADS) increases histone acetylation and p21waf1/cip1 expression in human colon tumor cell lines, Carcinogenesis, vol.25, issue.7, pp.1227-1236, 2004.
DOI : 10.1093/carcin/bgh123

A. P. Ducruet and J. S. Lazo, Regulation of Cdc25A Half-life in Interphase by Cyclin-dependent Kinase 2 Activity, Journal of Biological Chemistry, vol.9, issue.34, pp.31838-31842, 2003.
DOI : 10.1073/pnas.95.19.11324

S. Dutertre, M. Cazales, M. Quaranta, C. Froment, V. Trabut et al., Phosphorylation of CDC25B by Aurora-A at the centrosome contributes to the G2-M transition, Journal of Cell Science, vol.117, issue.12, pp.2523-2531, 2004.
DOI : 10.1242/jcs.01108

URL : https://hal.archives-ouvertes.fr/hal-00317440

I. Erdogan-orhan, B. Sener, D. Rosa, S. Perez-baz, J. Lozach et al., Polyprenyl-hydroquinones and -furans from three marine sponges inhibit the cell cycle regulating phosphatase CDC25A, Natural Product Research, vol.22, issue.1, 2004.
DOI : 10.1007/BF01957811

URL : https://hal.archives-ouvertes.fr/hal-00020132

P. Fantes, Epistatic gene interactions in the control of division in fission yeast, Nature, vol.115, issue.5712, pp.428-430, 1979.
DOI : 10.1042/bst0051191

E. B. Fauman, J. P. Cogswell, B. Lovejoy, W. J. Rocque, W. Holmes et al., Crystal Structure of the Catalytic Domain of the Human Cell Cycle Control Phosphatase, Cdc25A, Crystal structure of the catalytic domain of the human cell cycle control phosphatase, pp.617-625, 1998.
DOI : 10.1016/S0092-8674(00)81190-3

X. Feng, L. N. Wang, Y. Y. Zhou, H. P. Yu, Q. Shen et al., Discovery and characterization of a novel inhibitor of CDC25B, LGH00045, Acta Pharmacol. Sin, vol.10, pp.1268-1274, 2008.

A. M. Ferguson, L. S. White, and P. J. Donovan, Piwnica-Worms H. Normal Cell cycle and checkpoint responses in mice and cells lacking CDC25B and CDC25C protein phosphatases, Mol. Cell. Biol, pp.25-2853, 2005.

J. Ferlay, D. M. Parkin, and E. Steliarova-foucher, Estimates of cancer incidence and mortality in Europe, 2008.

B. G. Gabrielli, D. Souza, C. P. Tonks, I. D. Clark, J. M. Hayward et al., Cytoplasmic accumulation of CDC25B phosphatase in mitosis triggers centrosomal microtubule nucleation in HeLa cells, J. Cell Sci, vol.109, pp.1081-1093, 1996.

B. G. Gabrielli, J. M. Clark, A. K. Mccormack, and K. A. Ellem, Hyperphosphorylation of the N-terminal Domain of Cdc25 Regulates Activity toward Cyclin B1/Cdc2 But Not Cyclin A/Cdk2, Journal of Biological Chemistry, vol.101, issue.45, pp.272-28607, 1997.
DOI : 10.1038/sj.onc.1201254

K. Galaktionov, A. K. Lee, J. Eckstein, G. Draetta, J. Meckler et al., CDC25 phosphatases as potential human oncogenes, CDC25 phosphatases as potential human oncogenes, pp.1575-1577, 1995.
DOI : 10.1126/science.7667636

K. Galaktionov, X. Chen, and D. Beach, Cdc25 cell-cycle phosphatase as a target of c-myc, Nature, vol.382, issue.6591, pp.511-517, 1996.
DOI : 10.1038/382511a0

P. A. Garner-hamrick and C. Fisher, Antisense phosphorothioate oligonucleotides specifically down-regulate cdc25B causing S-phase delay and persistent antiproliferative effects, International Journal of Cancer, vol.271, issue.5, pp.720-728, 1998.
DOI : 10.1074/jbc.271.9.5118

URL : http://onlinelibrary.wiley.com/doi/10.1002/(SICI)1097-0215(19980529)76:5<720::AID-IJC18>3.0.CO;2-7/pdf

S. P. Gunasekera, P. J. Mccarthy, M. Kelly-borges, E. Lobkovsky, and J. Clardy, de Laubenfels, Journal of the American Chemical Society, vol.118, issue.36, pp.8759-8760, 1996.
DOI : 10.1021/ja961961+

J. Guo, J. Kleeff, J. Li, J. Ding, J. Hammer et al., Expression and functional significance of CDC25B in human pancreatic ductal adenocarcinoma, Oncogene, vol.23, issue.1, pp.71-81, 2004.
DOI : 10.1006/dbio.1995.1207

J. Guo, R. A. Parise, E. Joseph, J. Lan, S. S. Pan et al., Pharmacology and antitumor activity of a quinolinedione CDC25 phosphatase inhibitor DA3003-1 (NSC 663284), Anticancer Res, vol.27, pp.3067-3074, 2007.

S. W. Ham, H. J. Park, and D. H. Lim, Studies on Menadione as an Inhibitor of the cdc25 Phosphatase, Bioorganic Chemistry, vol.25, issue.1
DOI : 10.1006/bioo.1996.1052

A. Herman-antosiewicz and S. V. Singh, Checkpoint Kinase 1 Regulates Diallyl Trisulfide-induced Mitotic Arrest in Human Prostate Cancer Cells, Journal of Biological Chemistry, vol.1602, issue.31, pp.28519-28528, 2005.
DOI : 10.1054/drup.2001.0213

A. Herman-antosiewicz, A. A. Powolny, and S. V. Singh, Molecular targets of cancer chemoprevention by garlic-derived organosulfides, Acta Pharmacologica Sinica, vol.117, issue.9, pp.1355-1364, 2007.
DOI : 10.2174/1566524033479465

H. Hochegger, S. Takeda, and T. Hunt, Cyclin-dependent kinases and cell-cycle transitions: does one fit all?, Nature Reviews Molecular Cell Biology, vol.10, issue.11
DOI : 10.1128/MCB.19.10.7011

I. Hoffmann, P. R. Clarke, M. J. Marcote, E. Karsenti, and G. Draetta, Phosphorylation and activation of human CDC25-C by cdc2 -cyclin B and its involvement in the self-amplification of MPF at mitosis, EMBO J, vol.12, pp.53-63, 1993.

I. Hoffmann, G. Draetta, and E. Karsenti, Activation of the phosphatase activity of human CDC25A by a cdk2- cyclin E dependent phosphorylation at the G1/S transition, EMBO J, vol.13, pp.4302-4310, 1994.

Y. Honaker and H. Piwnica-worms, Casein kinase 1 functions as both penultimate and ultimate kinase in regulating Cdc25A destruction, Oncogene, vol.6, issue.23, pp.3324-3334, 2010.
DOI : 10.1128/MCB.19.9.6379

T. Horiguchi, K. Nishi, S. Hakoda, S. Tanida, A. Nagata et al., Dnacin A1 and dnacin B1 are antitumor antibiotics that inhibit cdc25B phosphatase activity, Biochemical Pharmacology, vol.48, issue.11, pp.48-2139, 1994.
DOI : 10.1016/0006-2952(94)90516-9

T. Hosono, T. Fukao, J. Ogihara, Y. Ito, H. Shiba et al., Diallyl trisulfide suppresses the proliferation and induces apoptosis of human colon cancer cells through oxidative modification of betatubulin, J. Biol. Chem, pp.280-41487, 2005.

E. W. Howard, M. T. Ling, C. W. Chua, H. W. Cheung, X. Wang et al., Garlic-derived Sallylmercaptocysteine is a novel in vivo antimetastatic agent for androgen-independent prostate cancer

A. W. Hsing, A. P. Chokkalingam, Y. T. Gao, M. P. Madigan, J. Deng et al., Allium vegetables and risk of prostate cancer:a population-based study, J. Natl. Cancer Inst, pp.94-1648, 2002.

W. G. Huang, Y. Y. Jiang, Q. Li, J. Li, J. Y. Li et al., Synthesis and biological evaluation of (??)-cryptotanshinone and its simplified analogues as potent CDC25 inhibitors, Tetrahedron, vol.61, issue.7, pp.1863-1870, 2005.
DOI : 10.1016/j.tet.2004.12.033

W. G. Huang, J. Y. Li, Y. Luo, J. Li, and W. Lu, Synthesis of tanshinone IIA analogues and their inhibitory activities against Cdc25 phosphatases, Chinese Chemical Letters, vol.20, issue.12, pp.1461-1464, 2009.
DOI : 10.1016/j.cclet.2009.07.008

M. Iciek, I. Kwiecien, and L. W?odek, Biological properties of garlic and garlic-derived organosulfur compounds, Environmental and Molecular Mutagenesis, vol.10, issue.3, pp.247-265, 2009.
DOI : 10.3748/wjg.v10.i18.2731

C. Jacob, A. Anwar, and T. Burkholz, Perspective on Recent Developments on Sulfur-Containing Agents and Hydrogen Sulfide Signaling, Planta Medica, vol.74, issue.13, pp.1580-1592, 2008.
DOI : 10.1055/s-0028-1088299

C. Jacob and A. Anwar, Sulfides in Allium Vegetables In: Chemoprevention of cancer and DNA damage by dietary factors, pp.663-684, 2009.

M. Jang, L. Cai, G. O. Udeani, K. V. Slowing, C. F. Thomas et al., Cancer Chemopreventive Activity of Resveratrol, a Natural Product Derived from Grapes, Science, vol.275, issue.5297, pp.218-220, 1997.
DOI : 10.1126/science.275.5297.218

S. Jinno, K. Suto, A. Nagata, M. Igarashi, Y. Kanaoka et al., CDC25A is a novel phosphatase functioning early in the cell cycle, EMBO J, vol.13, pp.1549-1556, 1994.

P. A. Johnston, C. A. Foster, M. Brisson-tierno, T. Y. Shun, S. N. Shinde et al., Cdc25B Dual-Specificity Phosphatase Inhibitors Identified in a High-Throughput Screen of the NIH Compound Library, Lazo J. S. CDC25B Dual-Specificity Phosphatase Inhibitors Identified in a High-Throughput Screen of the NIH Compound Library, pp.250-265, 2009.
DOI : 10.1089/adt.2008.186

J. Ju, G. Lu, J. D. Lambert, and C. S. Yang, Inhibition of carcinogenesis by tea constituents, Seminars in Cancer Biology, vol.17, issue.5, pp.395-402, 2007.
DOI : 10.1016/j.semcancer.2007.06.013

C. C. Juan and F. Y. Wu, Vitamin K3 inhibits growth of human hepatoma HepG2 cells by decreasing activities of both p34 cdc2 kinase and phosphatase, Biochem. Biophys. Res. Commun, pp.190-907, 1993.

H. Källström, A. Lindqvist, V. Pospisil, A. Lundgren, and C. Karlsson-rosenthal, Cdc25A localisation and shuttling: characterisation of sequences mediating nuclear export and import, Experimental Cell Research, vol.303, issue.1, pp.89-100, 2005.
DOI : 10.1016/j.yexcr.2004.09.012

Y. Kanemori, K. Uto, and N. Sagata, ??-TrCP recognizes a previously undescribed nonphosphorylated destruction motif in Cdc25A and Cdc25B phosphatases, Proceedings of the National Academy of Sciences, vol.101, issue.13, pp.6279-6284, 2005.
DOI : 10.1073/pnas.0307700101

S. Kar, M. Wang, Z. Ren, X. Chen, and B. I. Carr, Antitumor and anticarcinogenic actions of Cpd 5: a new class of protein phosphatase inhibitor, Carcinogenesis, vol.24, issue.3, pp.411-416, 2003.
DOI : 10.1093/carcin/24.3.411

S. Kar, M. Wang, S. W. Ham, and B. I. Carr, H32, a non-quinone sulfone analog of vitamin K3, inhibits human hepatoma cell growth by inhibiting Cdc25 and activating ERK, Cancer Biology & Therapy, vol.5, issue.10, pp.1340-1347, 2006.
DOI : 10.4161/cbt.5.10.3223

S. Kar, M. Wang, W. Yao, C. J. Michejda, and B. I. Carr, PM-20, a novel inhibitor of Cdc25A, induces extracellular signal-regulated kinase 1/2 phosphorylation and inhibits hepatocellular carcinoma growth in vitro and in vivo, Molecular Cancer Therapeutics, vol.5, issue.6, pp.1511-1519, 2006.
DOI : 10.1158/1535-7163.MCT-05-0485

S. Kar, M. Wang, S. W. Ham, and B. I. Carr, Fluorinated Cpd 5, a pure arylating K-vitamin derivative, inhibits human hepatoma cell growth by inhibiting Cdc25 and activating MAPK, Biochemical Pharmacology, vol.72, issue.10, pp.72-1217, 2006.
DOI : 10.1016/j.bcp.2006.07.024

S. Kar, M. Wang, and B. Carr, 2-Methoxyestradiol inhibits hepatocellular carcinoma cell growth by inhibiting Cdc25 and inducing cell cycle arrest and apoptosis, Cancer Chemotherapy and Pharmacology, vol.24, issue.2080, pp.62-831, 2007.
DOI : 10.4161/cbt.5.10.3223

N. Khan, F. Afaq, M. Saleem, N. Ahmad, and H. Mukhtar, Targeting Multiple Signaling Pathways by Green Tea Polyphenol (???)-Epigallocatechin-3-Gallate: Figure 1., Cancer Research, vol.66, issue.5, pp.2500-2505, 2006.
DOI : 10.1158/0008-5472.CAN-05-3636

F. Khanum, K. R. Anilakumar, and K. R. Viswanathan, Anticarcinogenic Properties of Garlic: A Review, Critical Reviews in Food Science and Nutrition, vol.58, issue.6
DOI : 10.1021/jf00015a002

I. Kieffer, C. Lorenzo, C. Dozier, E. Schmitt, and B. Ducommun, Differential mitotic degradation of the CDC25B phosphatase variants, Oncogene, vol.14, issue.57, pp.7847-7858, 2007.
DOI : 10.1038/sj.onc.1208617

URL : https://hal.archives-ouvertes.fr/hal-00317345

K. S. Kim, I. Kawasaki, Y. Chong, and Y. H. Shim, Inhibition of overexpressed CDC-25.1 phosphatase activity by flavone in Caenorhabditis elegans, Molecules and Cells, vol.46, issue.3, pp.345-350, 2009.
DOI : 10.1007/s10059-009-0044-9

Y. A. Kim, B. T. Choi, Y. T. Lee, D. I. Park, S. H. Rhee et al., Resveratrol inhibits cell proliferation and induces apoptosis of human breast carcinoma MCF-7 cells, Oncology Reports, pp.11-441, 2004.
DOI : 10.3892/or.11.2.441

S. Kolb, O. Mondésert, M. L. Goddard, D. Jullien, B. O. Villoutreix et al., Development of Novel Thiazolopyrimidines as CDC25B Phosphatase Inhibitors, ChemMedChem, vol.49, issue.4, pp.633-648, 2009.
DOI : 10.4161/cc.7.2.5306

K. Kristjansdottir and J. Rudolph, Cdc25 Phosphatases and Cancer, Chemistry & Biology, vol.11, issue.8, pp.1043-1051, 2004.
DOI : 10.1016/j.chembiol.2004.07.007

J. K. Kundu and Y. J. Surh, Cancer chemopreventive and therapeutic potential of resveratrol: Mechanistic perspectives, Cancer Letters, vol.269, issue.2, pp.243-261, 2008.
DOI : 10.1016/j.canlet.2008.03.057

C. Lammer, S. Wagerer, R. Saffrich, D. Mertens, W. Ansorge et al., The CDC25B phosphatase is essential for the G2/M phase transition in human cells, J. Cell Sci, vol.111, pp.2445-2453, 1998.

A. Lavecchia, A. Coluccia, D. Giovanni, C. Novellino, and E. , Cdc25B Phosphatase Inhibitors in Cancer Therapy: Latest Developments, Trends and Medicinal Chemistry Perspective, Anti-Cancer Agents in Medicinal Chemistry, vol.8, issue.8, pp.843-856, 2008.
DOI : 10.2174/187152008786847783

A. Lavecchia, D. Giovanni, C. Novellino, and E. , CDC25A and B Dual-Specificity Phosphatase Inhibitors: Potential Agents for Cancer Therapy, Current Medicinal Chemistry, vol.16, issue.15, pp.1831-1849, 2009.
DOI : 10.2174/092986709788186084

A. Lavecchia, D. Giovanni, C. Novellino, and E. , Inhibitors of Cdc25 phosphatases as anticancer agents: a patent review, Expert Opinion on Therapeutic Patents, vol.303, issue.3, pp.405-425, 2010.
DOI : 10.1126/science.1092472

J. S. Lazo, D. C. Aslan, E. C. Southwick, K. A. Cooley, A. P. Ducruet et al., Discovery and Biological Evaluation of a New Family of Potent Inhibitors of the Dual Specificity Protein Phosphatase Cdc25, Journal of Medicinal Chemistry, vol.44, issue.24, pp.44-4042, 2001.
DOI : 10.1021/jm0102046

J. Lazo, K. Nemoto, K. E. Pestell, K. Cooley, E. C. Southwick et al., Identification of a Potent and Selective Pharmacophore for Cdc25 Dual Specificity Phosphatase Inhibitors., Molecular Pharmacology, vol.61, issue.4, pp.61-720, 2002.
DOI : 10.1124/mol.61.4.720

J. S. Lazo and P. Wipf, Is Cdc25 a Druggable Target?, Anti-Cancer Agents in Medicinal Chemistry, vol.8, issue.8, pp.837-842, 2008.
DOI : 10.2174/187152008786847738

M. Lemaire, C. Froment, R. Boutros, O. Mondesert, A. R. Nebreda et al., CDC25B Phosphorylation by p38 and MK-2, Cell Cycle, vol.5, issue.15, pp.1649-1653, 2006.
DOI : 10.4161/cc.5.15.3006

URL : https://hal.archives-ouvertes.fr/hal-00317373

M. Lemaire, B. Ducommun, and A. R. Nebreda, UV-induced downregulation of the CDC25B protein in human cells, FEBS Letters, vol.8, issue.6, pp.1199-1204, 2010.
DOI : 10.4161/cc.8.2.7595

A. Lindqvist, H. Källström, A. Lundgren, E. Barsoum, K. Rosenthal et al., Cdc25B cooperates with Cdc25A to induce mitosis but has a unique role in activating cyclin B1???Cdk1 at the centrosome, The Journal of Cell Biology, vol.269, issue.1, pp.171-206, 2005.
DOI : 10.1074/jbc.M305178200

V. Lisowski, S. Léonce, L. Kraus-berthier, J. Sopkova-de-oliveira-santos, A. Pierré et al., Design, Synthesis, and Evaluation of Novel Thienopyrrolizinones as Antitubulin Agents, Journal of Medicinal Chemistry, vol.47, issue.6
DOI : 10.1021/jm030961z

H. Löffler, R. G. Syljuåsen, J. Bartkova, J. Worm, J. Lukas et al., Distinct modes of deregulation of the proto-oncogenic Cdc25A phosphatase in human breast cancer cell lines, Oncogene, vol.22, issue.50, pp.8063-8071, 2003.
DOI : 10.1038/35044005

G. Lolli, L. N. Johnson, and . Cak?-?-?-?cyclin-dependent-activating-kinase, A key kinase in cell cycle control and a target for drugs ?, Cell cycle, vol.4, pp.572-577, 2005.

A. Loukaci, L. Saout, I. Samadi, M. Leclerc, S. Damiens et al., Coscinosulfate, a CDC25 phosphatase inhibitor from the sponge Coscinoderma mathewsi, Bioorganic & Medicinal Chemistry, vol.9, issue.11, pp.3049-3054, 2001.
DOI : 10.1016/S0968-0896(01)00208-5

URL : https://hal.archives-ouvertes.fr/hal-00086659

Z. Q. Ma, S. S. Chua, F. J. Demayo, and S. Y. Tsai, Induction of mammary gland hyperplasia in transgenic mice over-expressing human Cdc25B, Oncogene, vol.18, issue.32, pp.4564-4576, 1999.
DOI : 10.1038/351194b0

J. P. Mackeigan, L. O. Murphy, and J. Blenis, Sensitized RNAi screen of human kinases and phosphatases identifies new regulators of apoptosis and chemoresistance, Nature Cell Biology, vol.7, issue.6, pp.591-600, 2005.
DOI : 10.1016/j.ccr.2005.01.010

N. Mailand, A. V. Podtelejnikov, A. Groth, M. Mann, J. Bartek et al., Regulation of G2/M events by Cdc25A through phosphorylation-dependent modulation of its stability, The EMBO Journal, vol.21, issue.21, pp.5911-5920, 2002.
DOI : 10.1093/emboj/cdf567

I. A. Manke, A. Nguyen, D. Lim, M. Q. Stewart, A. E. Elia et al., MAPKAP Kinase-2 Is a Cell Cycle Checkpoint Kinase that Regulates the G2/M Transition and S Phase Progression in Response to UV Irradiation, Molecular Cell, vol.17, issue.1, pp.17-37, 2005.
DOI : 10.1016/j.molcel.2004.11.021

S. Matsuoka, M. Huang, and S. J. Elledge, Linkage of ATM to Cell Cycle Regulation by the Chk2 Protein Kinase, Science, vol.282, issue.5395, pp.1893-1897, 1998.
DOI : 10.1126/science.282.5395.1893

A. Maul, Application of generalized linear models to the analysis of toxicity tests data, Environ. Monit. Assess, vol.23, pp.153-163, 1992.

P. Mehdipour, S. Pirouzpanah, A. Sarafnejad, M. Atri, S. T. Shahrestani et al., Prognostic implication of CDC25A and cyclin E expression on primary breast cancer patients, Cell Biology International, vol.33, issue.10, pp.33-1050, 2009.
DOI : 10.1016/j.cellbi.2009.06.016

L. Meijer, Le cycle de division cellulaire et sa régulation, pp.311-326, 2003.

M. Melixetian, D. K. Klein, C. S. Sørensen, and K. Helin, NEK11 regulates CDC25A degradation and the IR-induced G2/M checkpoint, NEK11 regulates CDC25A degradation and the IRinduced G2/M checkpoint, pp.1247-1253, 2009.
DOI : 10.1128/MCB.21.10.3445-3450.2001

B. Milligan, B. Saville, and J. M. Swan, New syntheses of trisulphides, J. Chem. Soc, pp.4850-4853, 1961.

J. Mimoto, K. Kiura, K. Matsuo, T. Yoshino, I. Takata et al., (-)-Epigallocatechin gallate can prevent cisplatin-induced lung tumorigenesis in A/J mice, Carcinogenesis, vol.21, issue.5, pp.915-919, 2000.
DOI : 10.1093/carcin/21.5.915

G. Mirey, I. Chartrain, C. Froment, M. Quaranta, J. P. Bouché et al., CDC25B Phosphorylated by pEg3 Localizes to the Centrosome and the Spindle Poles at Mitosis, Cell Cycle, vol.4, issue.6, pp.806-811, 2005.
DOI : 10.4161/cc.4.6.1716

URL : https://hal.archives-ouvertes.fr/hal-00317422

M. Montes, E. Braud, M. A. Miteva, M. L. Goddard, O. Mondésert et al., Receptor-Based Virtual Ligand Screening for the Identification of Novel CDC25 Phosphatase Inhibitors, Journal of Chemical Information and Modeling, vol.48, issue.1, pp.48-157, 2008.
DOI : 10.1021/ci700313e

T. Mosmann, Rapid colorimetric assay for cellular growth and survival: Application to proliferation and cytotoxicity assays, Journal of Immunological Methods, vol.65, issue.1-2, pp.55-63, 1983.
DOI : 10.1016/0022-1759(83)90303-4

U. Münchberg, A. Anwar, S. Mecklenburg, and C. Jacob, Polysulfides as Biologically Active Ingredients of Garlic, ChemInform, vol.5, issue.35
DOI : 10.1002/chin.200735239

I. Nilsson and I. Hoffmann, Cell cycle regulation by the Cdc25 phosphatase family, Prog. Cell Cycle Res, vol.4, pp.107-114, 2000.
DOI : 10.1007/978-1-4615-4253-7_10

H. Park, B. I. Carr, M. Li, and S. W. Ham, Fluorinated NSC as a Cdc25 inhibitor, Bioorganic & Medicinal Chemistry Letters, vol.17, issue.8, pp.2351-2354, 2007.
DOI : 10.1016/j.bmcl.2006.12.046

H. Park, Y. J. Bahn, S. K. Jung, D. G. Jeong, S. H. Lee et al., Discovery of Novel Cdc25 Phosphatase Inhibitors with Micromolar Activity Based on the Structure-Based Virtual Screening, Journal of Medicinal Chemistry, vol.51, issue.18, pp.51-5533, 2008.
DOI : 10.1021/jm701157g

H. Park, Y. J. Bahn, and S. E. Ryu, Structure-based de novo design and biochemical evaluation of novel Cdc25 phosphatase inhibitors, Bioorganic & Medicinal Chemistry Letters, vol.19, issue.15, pp.4330-4334, 2009.
DOI : 10.1016/j.bmcl.2009.05.084

H. Park, M. Li, J. Choi, H. Cho, and S. W. Ham, Structure-based virtual screening approach to identify novel classes of Cdc25B phosphatase inhibitors, Bioorganic & Medicinal Chemistry Letters, vol.19, issue.15, pp.4372-4375, 2009.
DOI : 10.1016/j.bmcl.2009.05.078

J. M. Parks, H. Hu, J. Rudolph, and W. Yang, -Nitrophenyl Phosphate from QM/MM-MFEP Calculations, The Journal of Physical Chemistry B, vol.113, issue.15, pp.5217-5224, 2009.
DOI : 10.1021/jp805137x

URL : http://europepmc.org/articles/pmc2712249?pdf=render

Y. Pereg, B. Y. Liu, K. M. O-'rourke, M. Sagolla, A. Dey et al., Ubiquitin hydrolase Dub3 promotes oncogenic transformation by stabilizing Cdc25A, Nature Cell Biology, vol.7, issue.4, pp.12-400, 2010.
DOI : 10.1038/ncb2041

V. P. Peyregne, S. Kar, S. W. Ham, M. Wang, Z. Wang et al., Novel hydroxyl naphthoquinones with potent Cdc25 antagonizing and growth inhibitory properties, Molecular Cancer Therapeutics, vol.4, issue.4, pp.595-602, 2005.
DOI : 10.1158/1535-7163.MCT-04-0274

L. Pu, A. A. Amoscato, M. E. Bier, and J. S. Lazo, Dual G1 and G2 phase inhibition by a novel, selective CDC25 inhibitor 7-Chloro-6-(2-morpholin-4-ylethylamino)-quinoline-5,8-dione, J. Biol. Chem, pp.277-46877, 2002.

D. Ray, Y. Terao, D. Nimbalkar, H. Hirai, E. C. Osmundson et al., Hemizygous Disruption of Cdc25A Inhibits Cellular Transformation and Mammary Tumorigenesis in Mice, Cancer Research, vol.67, issue.14, pp.6605-6611, 2007.
DOI : 10.1158/0008-5472.CAN-06-4815

R. A. Reynolds, A. W. Yem, C. L. Wolfe, M. R. Jr, C. G. Chidester et al., Crystal structure of the catalytic subunit of CDC25B required for G2/M phase transition of the cell cycle, J. Mol

J. Rudolph, Redox Regulation of the Cdc25 Phosphatases, Antioxidants & Redox Signaling, vol.7, issue.5-6, pp.761-767, 2005.
DOI : 10.1089/ars.2005.7.761

K. Sadhu, S. Reed, H. Richardson, and P. Russell, Human homolog of fission yeast cdc25 mitotic inducer is predominantly expressed in G2., Proceedings of the National Academy of Sciences, vol.87, issue.13, pp.5139-5143, 1990.
DOI : 10.1073/pnas.87.13.5139

A. Sancar, L. A. Lindsey-boltz, K. Ünsal-kaçmaz, and S. Linn, Molecular Mechanisms of Mammalian DNA Repair and the DNA Damage Checkpoints, Annual Review of Biochemistry, vol.73, issue.1, pp.73-112, 2004.
DOI : 10.1146/annurev.biochem.73.011303.073723

P. A. Savitsky and T. Finkel, Redox Regulation of Cdc25C, Journal of Biological Chemistry, vol.1176, issue.23, pp.20535-20540, 2002.
DOI : 10.1002/(SICI)1097-4652(199810)177:1<148::AID-JCP16>3.0.CO;2-9

E. M. Schaffer, J. Z. Liu, J. Green, C. A. Dangler, and J. A. Milner, Garlic and associated allyl sulfur components inhibit N-methyl-N-nitrosourea induced rat mammary carcinogenesis, Cancer Letters, vol.102, issue.1-2, pp.199-204, 1996.
DOI : 10.1016/0304-3835(96)04160-2

C. Scherer, C. Jacob, M. Dicato, and M. Diederich, Potential role of organic sulfur compounds from Allium species in cancer prevention and therapy, Phytochemistry Reviews, vol.46, issue.2, pp.349-368, 2009.
DOI : 10.1016/B978-0-12-642760-8.50005-3

G. K. Schwartz and M. A. Shah, Targeting the Cell Cycle: A New Approach to Cancer Therapy, Journal of Clinical Oncology, vol.23, issue.36, pp.9408-9421, 2005.
DOI : 10.1200/JCO.2005.01.5594

R. Shimazawa, T. Suzuki, K. Dodo, and R. Shirai, Design and synthesis of dysidiolide analogs from vitamin D3: novel class of Cdc25A inhibitors, Bioorganic & Medicinal Chemistry Letters, vol.14, issue.12, pp.3291-3294, 2004.
DOI : 10.1016/j.bmcl.2004.03.100

R. Shimazawa, M. Gochomori, and R. Shirai, Design and synthesis of novel Cdc25A-inhibitors having phosphate group as a hydrophilic residue, Bioorganic & Medicinal Chemistry Letters, vol.14, issue.16, pp.4339-4342, 2004.
DOI : 10.1016/j.bmcl.2004.05.083

R. Shimazawa, M. Kuriyama, and R. Shirai, Design and synthesis of N-alkyl oxindolylidene acetic acids as a new class of potent Cdc25A inhibitors, Bioorganic & Medicinal Chemistry Letters, vol.18, issue.11, pp.3350-3353, 2008.
DOI : 10.1016/j.bmcl.2008.04.027

S. Shreeram, W. K. Hee, and D. V. Bulavin, Cdc25A Serine 123 Phosphorylation Couples Centrosome Duplication with DNA Replication and Regulates Tumorigenesis, Molecular and Cellular Biology, vol.28, issue.24, pp.7442-7450, 2008.
DOI : 10.1128/MCB.00138-08

J. Sohn, K. Kristjánsdóttir, A. Safi, B. Parker, B. Kiburz et al., Remote hot spots mediate protein substrate recognition for the Cdc25 phosphatase, Proceedings of the National Academy of Sciences, vol.14, issue.1, pp.16437-16441, 2004.
DOI : 10.1016/0263-7855(96)00018-5

D. Spitkovsky, P. Jansen-dürr, E. Karsenti, and I. Hoffman, S-phase induction by adenovirus E1A requires activation of CDC25A tyrosine phosphatase, Oncogene, vol.12, pp.2549-2554, 1996.

S. D. Stan, S. Kar, G. D. Stoner, and S. V. Singh, Bioactive food components and cancer risk reduction, Journal of Cellular Biochemistry, vol.2, issue.1, pp.339-356, 2008.
DOI : 10.1111/j.1349-7006.1997.tb00440.x

E. C. Stuart, M. J. Scandlyn, and R. J. Rosengren, Role of epigallocatechin gallate (EGCG) in the treatment of breast and prostate cancer, Life Sciences, vol.79, issue.25, pp.2329-2336, 2006.
DOI : 10.1016/j.lfs.2006.07.036

Y. J. Surh, Cancer chemoprevention with dietary phytochemicals, Nature Reviews Cancer, vol.3, issue.10, pp.768-780, 2003.
DOI : 10.1038/nrc1189

M. Takahashi, K. Dodo, Y. Sugimoto, Y. Aoyagi, Y. Yamada et al., Synthesis of the novel analogues of dysidiolide and their structure???activity relationship, Bioorganic & Medicinal Chemistry Letters, vol.10, issue.22, pp.2571-2574, 2000.
DOI : 10.1016/S0960-894X(00)00527-8

D. Thomae, G. Kirsch, P. Seck, and T. Kaminskia, One-Pot Synthesis of 7-Hydroxythieno[3,2-b]pyridin-5(4H)-ones., ChemInform, vol.38, issue.48, pp.2153-2156, 2007.
DOI : 10.1002/chin.200748135

O. Timofeev, O. Cizmecioglu, E. Hu, T. Orlik, and I. Hoffmann, Human Cdc25A phosphatase has a non-redundant function in G2 phase by activating Cyclin A-dependent kinases, FEBS Letters, vol.5, issue.4, pp.841-847, 2009.
DOI : 10.1038/ncb918

O. Timofeev, O. Cizmecioglu, F. Settele, T. Kempf, and I. Hoffmann, /M Transition, Journal of Biological Chemistry, vol.19, issue.22, pp.16978-16990, 2010.
DOI : 10.1038/sj.emboj.7601136

F. Toyoshima-morimoto, E. Taniguchi, and E. Nishida, Plk1 promotes nuclear translocation of human Cdc25C during prophase, EMBO Reports, vol.3, issue.4, pp.341-348, 2002.
DOI : 10.1093/embo-reports/kvf069

P. Turowski, C. Franckhauser, M. C. Morris, P. Vaglio, A. Fernandez et al., Functional cdc25C Dual-Specificity Phosphatase Is Required for S-Phase Entry in Human Cells, Molecular Biology of the Cell, vol.14, issue.7, pp.2984-2998, 2003.
DOI : 10.1091/mbc.E02-08-0515

S. Uchida, M. Ohtsubo, M. Shimura, M. Hirata, H. Nakagama et al., Nuclear export signal in CDC25B, Biochemical and Biophysical Research Communications, vol.316, issue.1, pp.316-226, 2004.
DOI : 10.1016/j.bbrc.2004.02.039

S. Uchida, K. Yoshioka, R. Kizu, H. Nakagama, T. Matsunaga et al., Stressactivated mitogen-activated protein kinases c-Jun NH2-terminal kinase and p38 target CDC25B for degradation, Cancer Res, pp.69-6438, 2009.

S. Valente, E. Bana, E. Viry, D. Bagrel, and G. Kirsch, Synthesis and biological evaluation of novel coumarin-based inhibitors of Cdc25 phosphatases, Bioorganic & Medicinal Chemistry Letters, vol.20, issue.19, pp.5827-5830, 2010.
DOI : 10.1016/j.bmcl.2010.07.130

V. Vugt, M. A. Bràs, A. Medema, and R. H. , Restarting the cell cycle when the checkpoint comes to a halt

K. Vermeulen, D. R. Van-bockstaele, and Z. N. Berneman, The cell cycle: a review of regulation, deregulation and therapeutic targets in cancer, Cell Proliferation, vol.59, issue.3, pp.131-149, 2003.
DOI : 10.1101/gad.827700

Z. Wang, C. G. Trope, V. A. Flørenes, Z. Suo, J. M. Nesland et al., Overexpression of CDC25B, CDC25C and phospho-CDC25C (Ser216) in vulvar squamous cell carcinomas are associated with malignant features and aggressive cancer phenotypes, BMC Cancer, vol.5, issue.1, pp.233-244, 2010.
DOI : 10.1038/ncb994

R. D. Whelan, C. J. Waring, C. R. Wolf, J. D. Hayes, L. K. Hosking et al., Over-expression of P-glycoprotein and glutathione S-transferase PI in MCF-7 cells selected for vincristine resistancein vitro, International Journal of Cancer, vol.1, issue.2, pp.241-246, 1992.
DOI : 10.1128/MCB.5.8.2114

P. Wipf, B. Joo, T. Nguyen, J. S. Lazo, and ?. E. Org, Synthesis and biological evaluation of caulibugulones A

C. C. Wu, J. G. Chung, S. J. Tsai, J. H. Yang, and L. Y. Sheen, Differential effects of allyl sulfides from garlic essential oil on cell cycle regulation in human liver tumor cells, Food and Chemical Toxicology, vol.42, issue.12, pp.42-1937, 2004.
DOI : 10.1016/j.fct.2004.07.008

F. Y. Wu and T. Sun, Vitamin K3 induces cell cycle arrest and cell death by inhibiting Cdc25 phosphatase, European Journal of Cancer, vol.35, issue.9, pp.1388-1393, 1999.
DOI : 10.1016/S0959-8049(99)00156-2

C. C. Wu, L. Y. Sheen, H. W. Chen, W. W. Kuo, S. J. Tsai et al., Differential Effects of Garlic Oil and Its Three Major Organosulfur Components on the Hepatic Detoxification System in Rats, Journal of Agricultural and Food Chemistry, vol.50, issue.2, pp.378-383, 2002.
DOI : 10.1021/jf010937z

X. Wu, F. Kassie, and V. Mersch-sundermann, Induction of apoptosis in tumor cells by naturally occurring sulfur-containing compounds, Mutation Research/Reviews in Mutation Research, vol.589, issue.2, pp.81-102, 2005.
DOI : 10.1016/j.mrrev.2004.11.001

D. Xiao, S. Choi, D. E. Johnson, V. G. Vogel, C. S. Johnson et al., Diallyl trisulfide-induced apoptosis in human prostate cancer cells involves c-Jun N-terminal kinase and extracellular-signal regulated kinase-mediated phosphorylation of Bcl-2, Oncogene, vol.23, issue.33, pp.5594-5606, 2004.
DOI : 10.1093/jnci/81.2.162

D. Xiao, A. Herman-antosiewicz, J. Antosiewicz, H. Xiao, M. Brisson et al., Diallyl trisulfide-induced G2???M???phase cell cycle arrest in human prostate cancer cells is caused by reactive oxygen species-dependent destruction and hyperphosphorylation of Cdc25C, Oncogene, vol.24, issue.41, pp.6256-6268, 2005.
DOI : 10.1093/jnci/81.2.162

D. Xiao, K. L. Lew, Y. A. Kim, Y. Zeng, E. R. Hahm et al., Diallyl Trisulfide Suppresses Growth of PC-3 Human Prostate Cancer Xenograft In vivo in Association with Bax and Bak Induction, Clinical Cancer Research, vol.12, issue.22
DOI : 10.1158/1078-0432.CCR-06-1273

D. Xiao, Y. Zeng, and S. V. Singh, Diallyl trisulfide-induced apoptosis in human cancer cells is linked to checkpoint kinase 1-mediated mitotic arrest, Molecular Carcinogenesis, vol.117, issue.11, pp.48-1018, 2009.
DOI : 10.1091/mbc.6.2.185

Y. Yamashita, I. Kasugai, M. Sato, N. Tanuma, I. Sato et al., CDC25A mRNA levels significantly correlate with Ki-67 expression in human glioma samples, Journal of Neuro-Oncology, vol.59, issue.1, 2010.
DOI : 10.2174/187152008786847738

H. Yang, S. Zheng, L. Meijer, S. Li, S. Leclerc et al., kinase, Journal of Zhejiang University SCIENCE, vol.6, issue.7, pp.6-656, 2005.
DOI : 10.1631/jzus.2005.B0656

URL : https://hal.archives-ouvertes.fr/hal-00700472

J. P. Yuan, G. H. Wang, H. Ling, Q. Su, Y. H. Yang et al., Diallyl disulfide-induced G2/M arrest of human gastric cancer MGC803 cells involves activation of p38 MAP kinase pathways, World Journal of Gastroenterology, vol.10, issue.18, pp.2731-2734, 2004.
DOI : 10.1128/jvi.76.19.9763-9772.2002

O. D. Zakharova, L. P. Ovchinnikova, L. I. Goryunov, N. M. Troshkova, V. D. Shteingarts et al., Cytotoxicity of new alkylamino- and phenylamino-containing polyfluorinated derivatives of 1,4-naphthoquinone, European Journal of Medicinal Chemistry, vol.45, issue.6, pp.45-2321, 2010.
DOI : 10.1016/j.ejmech.2010.02.009

Q. Zhang, X. Tang, Q. Y. Lu, Z. F. Zhang, J. Brown et al., Resveratrol inhibits hypoxia-induced accumulation of hypoxia-inducible factor-1?? and VEGF expression in human tongue squamous cell carcinoma and hepatoma cells, Molecular Cancer Therapeutics, vol.4, issue.10, pp.1465-1474, 2005.
DOI : 10.1158/1535-7163.MCT-05-0198

Y. B. Zhou, X. Feng, L. N. Wang, J. Q. Du, Y. Y. Zhou et al., LGH00031, a novel ortho-quinonoid inhibitor of cell division cycle 25B, inhibits human cancer cells via ROS generation, Acta Pharmacologica Sinica, vol.68, issue.9, pp.30-1359, 2009.
DOI : 10.1074/jbc.M104889200