N. Oakes, S. Camilleri, S. Furler, D. Chisholm, and E. Kraegen, The insulin sensitizer, BRL 49653, reduces systemic fatty acid supply and utilization and tissue lipid availability in the rat, Metabolism, vol.46, issue.8, pp.935-942, 1997.
DOI : 10.1016/S0026-0495(97)90083-4

L. Michalik, B. Desvergne, and W. Wahli, Peroxisome-proliferator-activated receptors and cancers: complex stories, Nature Reviews Cancer, vol.63, issue.1, pp.61-70, 2004.
DOI : 10.1172/JCI1235

H. Koeffler, Peroxisome proliferator-activated receptor gamma and cancers, Clin Cancer Res, vol.9, pp.1-9, 2003.

A. Castrillo, M. Diaz-guerra, S. Hortelano, P. Martin-sanz, and L. Bosca, Inhibition of I?B kinase and I?B phosphorylation by 15-deoxy? 12-14 -prostaglandin J 2 in activated murine macrophages

D. Feinstein, A. Spagnolo, C. Akar, G. Weinberg, P. Murphy et al., Receptor-independent actions of PPAR thiazolidinedione agonists: Is mitochondrial function the key?, Biochemical Pharmacology, vol.70, issue.2, pp.177-188, 2005.
DOI : 10.1016/j.bcp.2005.03.033

S. Wei, Y. J. Lee, S. Kulp, S. Chen, and C. , PPAR??-independent antitumor effects of thiazolidinediones, Cancer Letters, vol.276, issue.2, pp.119-124, 2009.
DOI : 10.1016/j.canlet.2008.08.008

S. Niture, J. Kaspar, J. Shen, and A. Jaiswal, Nrf2 signaling and cell survival, Toxicology and Applied Pharmacology, vol.244, issue.1, pp.37-42, 2009.
DOI : 10.1016/j.taap.2009.06.009

URL : http://europepmc.org/articles/pmc2837794?pdf=render

P. Gong, D. Stewart, B. Hu, N. Li, J. Cook et al., Is Mediated by the Stress Response Elements and Transcription Factor Nrf2, Antioxidants & Redox Signaling, vol.4, issue.2, pp.249-257, 2002.
DOI : 10.1089/152308602753666307

S. Palakurthi, H. Aktas, L. Grubissich, R. Mortensen, and J. Halperin, Anticancer effects of thiazolidinediones are independent of peroxisome proliferator-activated receptor gamma and mediated by inhibition of translation initiation, Cancer Res, vol.61, pp.6213-6218, 2001.

C. Shiau, C. Yang, S. Kulp, K. Chen, C. Chen et al., Thiazolidenediones Mediate Apoptosis in Prostate Cancer Cells in Part through Inhibition of Bcl-xL/Bcl-2 Functions Independently of PPAR??, Cancer Research, vol.65, issue.4, pp.1561-1569, 2005.
DOI : 10.1158/0008-5472.CAN-04-1677

T. Sohda, Y. Momose, K. Meguro, Y. Kawamatsu, Y. Sugiyama et al., Studies on antidiabetic agents. Synthesis and hypoglycemic activity of 5-[4-(pyridylalkoxy)-benzyl]-2,4-thia- zolidinediones, Arzneim Forsch, vol.40, pp.37-42, 1990.

G. Deguest, L. Bischoff, L. Fruit, and F. Marsais, Anionic, in Situ Generation of Formaldehyde:?? A Very Useful and Versatile Tool in Synthesis, Organic Letters, vol.9, issue.6, pp.1165-1167, 2007.
DOI : 10.1021/ol070145b

K. Lenton, H. Therriault, and J. Wagner, Analysis of Glutathione and Glutathione Disulfide in Whole Cells and Mitochondria by Postcolumn Derivatization High-Performance Liquid Chromatography with ortho-Phthalaldehyde, Analytical Biochemistry, vol.274, issue.1, pp.125-130, 1999.
DOI : 10.1006/abio.1999.4258

E. Atarod and J. Kehrer, Dissociation of oxidant production by peroxisome proliferator-activated receptor ligands from cell death in human cell lines, Free Radical Biology and Medicine, vol.37, issue.1, pp.36-47, 2004.
DOI : 10.1016/j.freeradbiomed.2004.04.015

M. Liu, N. Wikonkal, and D. Brash, Induction of cyclin-dependent kinase inhibitors and G1 prolongation by the chemopreventive agent N-acetylcysteine, Carcinogenesis, vol.20, issue.9, pp.1869-1872, 1999.
DOI : 10.1093/carcin/20.9.1869

K. Uchida and T. Shibata, : An Electrophilic Trigger of Cellular Responses, Chemical Research in Toxicology, vol.21, issue.1, pp.138-144, 2008.
DOI : 10.1021/tx700177j

S. Kliewer, A prostaglandin J2 metabolite binds peroxisome proliferator-activated receptor ?? and promotes adipocyte differentiation, Cell, vol.83, issue.5, pp.813-819, 1995.
DOI : 10.1016/0092-8674(95)90194-9

T. Shimada, K. Kojima, K. Yoshiura, H. Hiraishi, and A. Terano, Characteristics of the peroxisome proliferator activated receptor gamma (PPARgamma) ligand induced apoptosis in colon cancer cells, Gut, vol.50, issue.5, pp.658-664, 2002.
DOI : 10.1136/gut.50.5.658

A. Levonen, D. Dickinson, D. Moellering, R. Mulcahy, H. Forman et al., Biphasic Effects of 15-Deoxy-??12,14-Prostaglandin J2 on Glutathione Induction and Apoptosis in Human Endothelial Cells, Arteriosclerosis, Thrombosis, and Vascular Biology, vol.21, issue.11, pp.1846-1851, 2001.
DOI : 10.1161/hq1101.098488

S. Polvani, M. Tarocchi, and A. Galli, PPARgamma and oxidative stress: Con(beta) catenating NRF2 and FOXO, PPAR Res, p.641087, 2012.

K. Schaefer, H. Takahashi, V. Morales, G. Harris, S. Barton et al., PPAR?? inhibitors reduce tubulin protein levels by a PPAR??, PPAR?? and proteasome-independent mechanism, resulting in cell cycle arrest, apoptosis and reduced metastasis of colorectal carcinoma cells, International Journal of Cancer, vol.22, issue.3, pp.702-713, 2007.
DOI : 10.1091/mbc.8.6.973

E. Mueller, P. Sarraf, P. Tontonoz, R. Evans, K. Martin et al., Terminal Differentiation of Human Breast Cancer through PPAR??, Molecular Cell, vol.1, issue.3, pp.465-470, 1998.
DOI : 10.1016/S1097-2765(00)80047-7

L. Cerquetti, C. Sampaoli, D. Amendola, B. Bucci, L. Masuelli et al., Rosiglitazone induces autophagy in H295R and cell cycle deregulation in SW13 adrenocortical cancer cells, Experimental Cell Research, vol.317, issue.10, pp.1397-1410, 2011.
DOI : 10.1016/j.yexcr.2011.02.014

J. Zhou, W. Zhang, B. Liang, M. Casimiro, D. Whitaker-menezes et al., PPAR?? activation induces autophagy in breast cancer cells, The International Journal of Biochemistry & Cell Biology, vol.41, issue.11, pp.2334-2342, 2009.
DOI : 10.1016/j.biocel.2009.06.007

H. Rodway, A. Hunt, J. Kohler, A. Postle, and K. Lillycrop, in neuroblastoma cells, Biochemical Journal, vol.382, issue.1, pp.83-91, 2004.
DOI : 10.1042/BJ20040107

V. Rao, S. Klein, S. Bonar, J. Zielonka, N. Mizuno et al., The Antioxidant Transcription Factor Nrf2 Negatively Regulates Autophagy and Growth Arrest Induced by the Anticancer Redox Agent Mitoquinone, Journal of Biological Chemistry, vol.4, issue.45, pp.34447-34459, 2010.
DOI : 10.1016/j.bpj.2008.10.042

J. Perez-ortiz, P. Tranque, M. Burgos, C. Vaquero, and J. Llopis, Glitazones Induce Astroglioma Cell Death by Releasing Reactive Oxygen Species from Mitochondria: Modulation of Cytotoxicity by Nitric Oxide, Molecular Pharmacology, vol.72, issue.2, pp.407-417, 2007.
DOI : 10.1124/mol.106.032458

M. Kondo, T. Oya-ito, T. Kumagai, T. Osawa, and K. Uchida, Cyclopentenone Prostaglandins as Potential Inducers of Intracellular Oxidative Stress, Journal of Biological Chemistry, vol.3, issue.15, pp.12076-12083, 2001.
DOI : 10.1038/9550

S. Ghosh, N. Patel, D. Rahn, J. Mcallister, S. Sadeghi et al., The Thiazolidinedione Pioglitazone Alters Mitochondrial Function in Human Neuron-Like Cells, Molecular Pharmacology, vol.71, issue.6, pp.1695-1702, 2007.
DOI : 10.1124/mol.106.033845

E. Brunoldi, G. Zanoni, G. Vidari, S. Sasi, M. Freeman et al., , Is Metabolized by HepG2 Cells via Conjugation with Glutathione, Chemical Research in Toxicology, vol.20, issue.10, pp.1528-1535, 2007.
DOI : 10.1021/tx700231a

G. Wu, Y. Fang, S. Yang, J. Lupton, and N. Turner, Glutathione Metabolism and Its Implications for Health, The Journal of Nutrition, vol.552, issue.3, pp.489-492, 2004.
DOI : 10.1113/jphysiol.2003.049478

R. Alvarez-sanchez, F. Montavon, T. Hartung, A. Pähler, M. Uchiyama et al., Thazolididedione bioactivation: a comparison of the bioactivation potentials of troglitazone, rosiglitazone, and pioglitazone using stable isotope-labeled analogues and liquid chromatography tandem mass spectrophometry Identification of novel metabolic pathways of pioglitazone in hepatocytes: N-glucuronidation of thiazolidinedione ring and sequential ring-opening pathway, Chem Res Toxicol Drug Metab Dispos, vol.16, issue.38, pp.1106-1116, 2006.

H. Ferguson, T. Thatcher, K. Olsen, T. Garcia-bates, C. Baglole et al., Peroxisome proliferator-activated receptor-?? ligands induce heme oxygenase-1 in lung fibroblasts by a PPAR??-independent, glutathione-dependent mechanism, American Journal of Physiology-Lung Cellular and Molecular Physiology, vol.59, issue.5, pp.912-919, 2009.
DOI : 10.1016/S0002-9440(10)62229-8

C. Li, M. Kim, L. Godoy, A. Thiantanawat, L. Trudel et al., Nitric oxide activation of Keap1/Nrf2 signaling in human colon carcinoma cells, Proceedings of the National Academy of Sciences, vol.2001, issue.86, pp.14547-14551, 2009.
DOI : 10.1126/scisignal.862001pl1

A. Chia, C. Goldring, N. Kitteringham, S. Wong, P. Morgan et al., Differential effect of covalent protein modification and glutathione depletion on the transcriptional response of Nrf2 and NF-??B, Biochemical Pharmacology, vol.80, issue.3, pp.410-421, 2010.
DOI : 10.1016/j.bcp.2010.04.004

G. Kronke, A. Kadl, E. Ikonomu, S. Bluml, A. Furnkranz et al., Expression of Heme Oxygenase-1 in Human Vascular Cells Is Regulated by Peroxisome Proliferator-Activated Receptors, Arteriosclerosis, Thrombosis, and Vascular Biology, vol.27, issue.6, pp.1276-1282, 2007.
DOI : 10.1161/ATVBAHA.107.142638

F. 2. Metz-technopôle, Université de Lorraine -UMR CNRS 7565 -Structure et Réactivité des

G. Delestraint, ?. , M. Cedex, *. France, and . Corresponding-author, Fax: + 33 3 87 31 58 01. SMP3 apparatus and are uncorrected. The 1 H and 13 C NMR spectra were measured on an AC Bruker 250 MHz spectrometer in CDCl 3 or DMSO-d 6 ; chemical shifts are reported in parts per million (ppm) All coupling constants (J) are given in Hz. MS spectra were recorded on an Agilent Technologies GC-MS instrument equipped with a 7683 injector, 6890N gas chromatograph and a 5973 mass selective detector. The mass spectrometer was operated in EI mode at 70 eV, and MS spectra were recorded from m/z 50 to 650, Elemental analyses (C, H, N, S) were used to confirm the purity of all compounds (>95%) and were performed on a CHN ThermoScientific Flash, pp.33-36, 2000.

C. Nmr, 17 (s, 1 H, NH)6; HRMS (ESI) : m/z calcd for(4-Chlorophenyl)-3-thienyl]imino}-5-(4-methylbenzylidene)-1,3-thiazolidin- 4-one (4c) Yellow solid ; yield : 69% ; mp 284°C, pp.39-46

H. Esi, (4-Chlorophenyl)-3-thienyl]imino}-5-(4-bromobenzylidene)-1, pp.3-4

H. Nmr, 16 (s, 1 H, CH)39 (s, 1 H, CH), 7.45-7.77 (m, 9 H, 9 × CH), 12.33 (s, 1 H, NH); HRMS (ESI) : m/z calcd for, Green, vol.11, issue.7, pp.3-4

H. Nmr and C. Nmr, 33 (s, 1 H, NH)3; HRMS (ESI) : m/z calcd for 2-{[5-(4-Chlorophenyl)-3-thienyl]imino}-5-(2-naphtylidene)-1,3-thiazolidin-4-one (4h) Yellow solid ; yield : 89% ; mp 288°C ; 1 H NMR (DMSO-d 6 ) ? H 7.21 (s, 1 H, CH), 7.41-8.17 (m, 13 H, 13 × CH)29 (s, 1 H, NH), 13 C NMR (DMSO-d 6 ) ? C 112.2, 112 HRMS (ESI) : m/z cald forChlorophenyl)-3-thienyl]imino}-5-heptylidene-1,3-thiazolidin-4-one (4i) Brown solid ; yield : 60% ; mp 191°C, Green, vol.17081, issue.707, pp.84-90

H. Nmr and C. Nmr, 19 (s, 1 H, NH)6; HRMS (ESI) : m/z cald forChlorophenyl)-3-thienyl]imino}-5-(3,4-dimethoxybenzylidene)-1,3- thiazolidin-4-one (4k) Yellow solid ; yield : 72%, (m, 2 H, 2 × CH) HRMS (ESI) : m/z calcd for, pp.31-38

H. Nmr, (m, 5 H, 5 × CH)21 (s, 1 H, NH); 13 C NMR (DMSO-d 6 ) ? C 116, Orange, vol.1, issue.1, pp.3-4, 1920.

×. Ch, 93 (s, 1 H, CH)51 (s, 1 H, NH) HRMS (ESI) : m/z calcd for(4-Chlorophenyl)-1,3-thiazol-2-yl]imino}-5-(4-methylbenzylidene)-1,3- thiazolidin-4-one (21) Yellow solid ; yield : 68% ; mp> 300°C66 (s, 1 H, CH), 7.93 (s, 1 H, CH)68 (s, 1 H, NH); HRMS (ESI) : m/z calcd forChlorophenyl)-1,3-thiazol-2-yl]imino}-5-(4-methoxybenzylidene)-1,3- thiazolidin-4-one (22) Yellow solid ; yield : 79% ; mp> 300°C ; 1 H NMR (DMSO-d 6 ) ? H 3 Yellow solid ; yield : 85% ; mp> 300°C, 86% ; mp> 300°C ; 1 H NMR (DMSO-d 6 ) ? H 3.05 (s, 6 H, 2 × CH 3 H NMR (DMSO-d 6 ) ? H 2.38 (s, 3 H, CH 3 ), 7.40 (d, 2 H, 2 × CH, J= 8.25Hz)d, 2 H, 2 × CH, J=8.5Hz) ,12.74 (s, 1 H, NH);HRMS (ESI) : m/z calcd for.05 (m, 5 H, 5 × CH) CH) HRMS (ESI) : m/z calcd for, pp.8951-8958

×. Ch, 79 (s, 1 H, NH) HRMS (ESI) : m/z calcd for General procedure for the synthesis of N-substituted thiazolidinones, pp.12-17

. Mmol, The mixture was then heated at 40°C overnight After cooling to room temperature, the mixture was poured into cold water (25 mL) and extracted with ethyl acetate. The organic layer was dried with magnesium sulfate, filtered and evaporated under reduced pressure. The expected product was purified using silica gel chromatography

H. , C. , J. H. , C. , and J. C. Nmr, 50 (m, 4 H, 4 × CH)0; HRMS (ESI) : m/z calcd for(4-chlorophenyl)-3-thienyl]imino}-1,3-thiazolidin-4-one (13) Pink solid ; yield : 83% ; mp 152°C, H NMR (CDCl 3 ) ? H 4.05 (s (m, 4 H, 4 × CH), pp.29-36

×. Nmr, 3; HRMS (ESI) : m/z calcd for [C 19 H 13 ClN 2 OS 2 + H] + : 385.0231 ; found : 385.0234. 3.1.22. 3-(4-Methylphenyl)-2-{[5-(4-chlorophenyl)-3-thienyl]imino}-1,3-thiazolidin-4-one (14) Brown solid ; yield : 90% ; mp 107°C, 6.91 (d, 1 H, CH, J=1.5Hz), 7.16 (d, 2 H, 2 × CH, J=9Hz), p.97

H. , C. , J. H. , C. , J. et al., J=8Hz) ; 13 C NMR (CDCl 3 ) ? C 325; HRMS (ESI) : m/z calcd for(1-Naphtyl)-2-{[5-(4-chlorophenyl)-3-thienyl]imino}-1,3-thiazolidin-4-one (17) Yellow solid ; yield : 92% ; mp 98°C ; 1 H NMR (CDCl 3 ) ? H 4, Pale brown solid ; yield : 59% ; mp 181°C ; 1 H NMR (CDCl 3 ) ? H 3.905Hz), 7.32 (d, 2 H, 2 × CH, J=8Hz), 7.47 (d, 2 H, 2 × CH H, CH, J=2Hz), 7.22 (d, 2 H, 2 × CH, J=8.5Hz), 7.36 (d, 2 H, 2 × CH, J=8.5Hz), pp.91-95

H. and ×. Ch, 89 (s, 1 H, CH), NH, vol.7, issue.796, pp.12-14

C. Nmr, 1; HRMS (ESI) : m/z cald for, pp.309-9876

A. Mut, K. , and H. Apc-wt, K-RAS mut ) cell lines have been established from colon adenocarcinoma tissue and represented colon cancer cell model behaving chromosomic and microsatellite instability, respectively. SW620 cells (APC wt , P53 mut , K-RAS mut ) were originated from a lymphoid nodule and have been characterised as metastatic cells. MCF7 cells derived from mammary adenocarcinoma tissue. MDA-MB-231 cells were isolated from pleural effusion and characterised as metastatic cells. Colon cancer cells were cultivated in Dulbecco minimum essential medium DMEM (Eurobio, Courtaboeuf, France) supplemented with 10% (v/v) heat-inactivated (30 min at 56°C) fetal calf serum (Eurobio), Biological methods 3.2.1. Cell culture HT29) and 2 mM L-Glutamine (Eurobio). Mammary cancer cell were cultivated in RPMI medium containing 10% heatinactivated fetal calf serum, p.50

E. Pasquier, S. Honore, and D. Braguer, Microtubule-targeting agents in angiogenesis: Where do we stand?, Drug Resistance Updates, vol.9, issue.1-2, pp.74-86, 2006.
DOI : 10.1016/j.drup.2006.04.003

A. C. Tripathi, S. J. Gupta, G. N. Fatima, P. K. Sonar, A. Vermab et al., 4-Thiazolidinones: The advances continue???, European Journal of Medicinal Chemistry, vol.72, pp.52-77, 2014.
DOI : 10.1016/j.ejmech.2013.11.017

C. D. Wright, D. J. Schrier, and R. D. Dyer, Synthesis and biological evaluation of 5-[[3,5-bis(1,1- dimethylethyl)-4-hydroxyphenyl)methylene]oxazoles, -thiazoles, and ?imidazoles: novel dual 5-lipoxygenase and cyclooxygenase inhibitors with anti-inflammatory activity, J. Med. Chem, pp.322-328, 1994.

. Glamoclija, Novel 4-thiazolidinone derivatives as potential antifungal and antibacterial drugs, Bioorg. Med. Chem, vol.18, pp.426-432, 2010.

B. Yan and . Fang, Anticancer activity of 5-benzylidene-2-phenylimino-1,3-thiazolidin-4-one (BPT) analogs, Med. Chem, pp.597-605, 2006.

B. L. Swaan, B. Fang, B. Zhang, and . Yan, Design, synthesis, cytoselective toxicity, structutreactivity relationships, and pharmacophore of thiazolidinone derivatives targeting drugresistant lung cancer cells, J. Med. Chem, pp.51-1242, 2008.

E. Mini, Gong, Design, synthesis and biological evaluation of novel 4-thiazolidinones containing indolin-2-one moiety as potential antitumor agent, Bioorg. Med. Chem. Lett. Eur. J. Med. Chem, vol.15, pp.3930-3933, 2005.

I. Abdillahi, G. Revelant, Y. Datoussaid, and G. Kirsch, Synthesis of novel 2- thienylimino-1,3-thiazolidin-4-ones, Synthesis, pp.2543-2546, 2010.

G. Revelant, S. Hesse, and G. Kirsch, Synthesis of novel N-subtituted 2-(hetero)arylimino- 1,3-thiazolidin-4-ones, Synthesis, pp.3319-3324, 2010.

G. Revelant, S. Hesse, and G. Kirsch, Synthesis of novel 2-aminothieno[3,2-d]thiazoles and selenolo, Tetrahedron, vol.3, pp.9352-9357, 2011.

F. Teraishi, S. Wu, J. Sasaki, L. Zhang, W. Guo et al., -Terminal Kinase???Dependent Apoptosis in Human Colon Cancer Cells, Cancer Research, vol.65, issue.14, pp.65-6380, 2005.
DOI : 10.1158/0008-5472.CAN-05-0575

M. Dodson, V. Darley-usmar, and J. Zhang, Cellular metabolic and autophagic pathways: Traffic control by redox signaling, Free Radical Biology and Medicine, vol.63, pp.207-221, 2013.
DOI : 10.1016/j.freeradbiomed.2013.05.014

Z. Liu and M. J. Lenardo, Reactive Oxygen Species Regulate Autophagy through Redox-Sensitive Proteases, Developmental Cell, vol.12, issue.4, pp.484-485, 2007.
DOI : 10.1016/j.devcel.2007.03.016

S. K. Niture, J. W. Kaspar, J. Shen, and A. K. , Nrf2 signaling and cell survival, Nrf2 signaling and cell survival, pp.37-42, 2010.
DOI : 10.1016/j.taap.2009.06.009

T. Xie and A. K. , AP-2-mediated regulation of human NAD(P)H: Quinone oxidoreductase1 (NQO1) gene expression, Biochemical Pharmacology, vol.51, issue.6, pp.51-771, 1996.
DOI : 10.1016/0006-2952(95)02394-1

J. Wu, H. Xu, P. F. Wong, S. Xia, J. Xu et al., Icaritin attenuates cigarette smoke-mediated oxidative stress in human lung epithelial cells via activation of PI3K-AKT and Nrf2 signaling, Food and Chemical Toxicology, vol.64, pp.307-313, 2014.
DOI : 10.1016/j.fct.2013.12.006

D. Trachootham, J. Alexandre, and P. Huang, Targeting cancer cells by ROS-mediated mechanisms: a radical therapeutic approach?, Nature Reviews Drug Discovery, vol.104, issue.7, pp.579-591, 2009.
DOI : 10.4161/cbt.7.12.7067

M. M. Hippert, P. S. O-'toole, and A. Thorburn, Autophagy in cancer: good, bad, or both?

K. Lewicki, S. Marchand, S. , L. Matoub, J. Lulek et al., Development of a fluorescence-based microtiter plate method for the measurement of glutathione in yeast Protein measurement with the Folin phenol reagent, Talanta J. Biol. Chem, vol.70, issue.193, pp.265-275, 1951.