P. N. Ravindran, K. Babu-nirmal, and K. Sivaraman, Turmeric: The Genus Curcuma, E. Review on Some Indian Traditional Medicine with Antioxidant Activity. Journal of Ethnopharmacology, vol.71, issue.212, pp.23-43, 2000.

M. Blois, Antioxidant Determinations by the Use of a Stable Free Radical, Nature, vol.55, issue.4617, pp.1199-1200, 1958.
DOI : 10.1016/0006-3002(55)90038-X

W. Ching, Y. Bin-yusoff, and W. B. Wan-amarina, Extraction of Essential Oil from Curcuma Longa, Journal of Food Chemistry and Nutrition, vol.2014, issue.01, pp.2-3

. Phytosun-aroms-huile-essentielle-curcuma-pharmacie-du-dr, Marche https, 2015.

S. Honda, F. Aoki, H. Tanaka, H. Kishida, S. Okada et al., Effects of Ingested Turmeric Oleoresin on Glucose and Lipid Metabolisms in Obese Diabetic Mice:?? A DNA Microarray Study, Journal of Agricultural and Food Chemistry, vol.54, issue.24, pp.9055-9062, 2006.
DOI : 10.1021/jf061788t

G. Xia, L. Zhou, J. Ma, Y. Wang, L. Ding et al., Sesquiterpenes from the essential oil of Curcuma wenyujin and their inhibitory effects on nitric oxide production, Fitoterapia, vol.103, issue.10, pp.143-148, 2015.
DOI : 10.1016/j.fitote.2015.03.021

V. B. Liju, K. Jeena, and R. Kuttan, Chemopreventive Activity of Turmeric Essential Oil and Possible Mechanisms of Action, Asian Pacific Journal of Cancer Prevention, vol.15, issue.16, pp.6575-6580, 2014.
DOI : 10.7314/APJCP.2014.15.16.6575

Y. Zhou, H. Liu, N. Jiang, X. Feng, X. Feng et al., Elemene, the essential oil of Curcuma wenyujin, inhibits osteogenic differentiation in ankylosing spondylitis, Joint Bone Spine, vol.82, issue.2, pp.100-103
DOI : 10.1016/j.jbspin.2014.05.004

N. G. Das, S. Dhiman, P. K. Talukdar, B. Rabha, D. Goswami et al., Synergistic mosquito-repellent activity of Curcuma longa, Pogostemon heyneanus and Zanthoxylum limonella essential oils, Journal of Infection and Public Health, vol.8, issue.4, pp.323-328
DOI : 10.1016/j.jiph.2015.02.005

O. D. Dhingra, G. N. Jham, R. C. Barcelos, and F. Mendonça, Isolation and Identification of the Principal Fungitoxic Component of Turmeric Essential Oil, Journal of Essential Oil Research, vol.9, issue.4, pp.387-391, 2007.
DOI : 10.1080/10412905.1997.9700783

T. Esatbeyoglu, P. Huebbe, I. M. Ernst, D. Chin, A. E. Wagner et al., Curcuminfrom Molecule to Biological Function, Angewandte Chemie -International Edition, vol.2012, issue.1522, pp.51-5308

V. B. Liju, K. Jeena, and R. Kuttan, Acute and Subchronic Toxicity as Well as Mutagenic Evaluation of Essential Oil from Turmeric (Curcuma Longa L) Food and Chemical Toxicology, pp.52-61, 2013.

D. Suresh, H. Manjunatha, and K. Srinivasan, Effect of heat processing of spices on the concentrations of their bioactive principles: Turmeric (Curcuma longa), red pepper (Capsicum annuum) and black pepper (Piper nigrum), Journal of Food Composition and Analysis, vol.20, issue.3-4, pp.346-351, 2007.
DOI : 10.1016/j.jfca.2006.10.002

J. H. Lee and M. G. Choung, Determination of Curcuminoid Colouring Principles in Commercial Foods by HPLC. Food Chemistry, pp.1217-1222, 2011.

M. J. Scotter, Synthesis and Chemical Characterisation of Curcuminoid Colouring Principles for Their Potential Use as HPLC Standards for the Determination of Curcumin Colour in Foods. LWT -Food Science and Technology, pp.42-1345, 2009.

L. Péret-almeida, .. P. Cherubino, R. J. Alves, L. Dufossé, and M. B. Glória, Separation and determination of the physico-chemical characteristics of curcumin, demethoxycurcumin and bisdemethoxycurcumin, Food Research International, vol.38, issue.8-9, pp.8-9, 2005.
DOI : 10.1016/j.foodres.2005.02.021

S. Yodkeeree, W. Chaiwangyen, S. Garbisa, P. Limtrakul, and . Curcumin, Curcumin, demethoxycurcumin and bisdemethoxycurcumin differentially inhibit cancer cell invasion through the down-regulation of MMPs and uPA, The Journal of Nutritional Biochemistry, vol.20, issue.2, pp.87-95, 2009.
DOI : 10.1016/j.jnutbio.2007.12.003

O. N. Gordon, P. B. Luis, R. E. Ashley, N. Osheroff, and C. Schneider, Oxidative Transformation of Demethoxy- and Bisdemethoxycurcumin: Products, Mechanism of Formation, and Poisoning of Human Topoisomerase II??, Chemical Research in Toxicology, vol.28, issue.5, pp.989-996
DOI : 10.1021/acs.chemrestox.5b00009

Y. Teng, Y. Hsieh, C. Hung, and H. Lin, Demethoxycurcumin Modulates Human P-Glycoprotein Function via Uncompetitive Inhibition of ATPase Hydrolysis Activity, Journal of Agricultural and Food Chemistry, vol.63, issue.3, pp.847-855
DOI : 10.1021/jf5042307

R. Aruna, P. Sathiyarajeswaran, K. Gopakumar, and R. S. Ramaswamy, Cardioprotective Effects of Kitchen Culinaries Mentioned in Siddha Literature, Journal of Pharmacognosy and Phytochemistry, vol.2014, issue.33, pp.71-79

K. Uma-pradeep and P. Geervani, Influence of Spices on Protein Utilisation of Winged Bean (Psophocarpus Tetragonolobus) and Horsegram (Dolichos Biflorus) Plant Foods for Human Nutrition, pp.187-193, 1994.

J. A. Duke, M. J. Bogenschutz-godwin, J. Ducellier, and P. K. Duke, CRC Handbook of Medicinal Spices, 2002.
DOI : 10.1201/9781420040487

M. A. Azuine and S. Bhide, Protective single/combined treatment with betel leaf and turmeric against methyl (acetoxymethyl) nitrosamine-induced hamster oral carcinogenesis, International Journal of Cancer, vol.2, issue.3, pp.412-415, 1992.
DOI : 10.1001/archotol.1989.01860280082021

R. Abou-elkhair, A. H. Ahmed, and S. Selim, Effects of Black Pepper ( Piper Nigrum ) Turmeric Powder ( Curcuma Longa ) and Coriander Seeds ( Coriandrum Sativum ) and Their Combinations as Feed Additives on Growth Performance , Carcass Traits , Some Blood Parameters and Humoral Immune Response of B, pp.27-847, 2014.

K. Srinivasan, Spices as influencers of body metabolism: an overview of three decades of research, Food Research International, vol.38, issue.1, pp.77-86, 2005.
DOI : 10.1016/j.foodres.2004.09.001

D. Shetty, Y. Kim, H. Shim, and J. Snyder, Eliminating the Heart from the Curcumin Molecule: Monocarbonyl Curcumin Mimics (MACs), Molecules, vol.65, issue.1, pp.249-292
DOI : 10.1016/j.bmcl.2012.11.004

E. Schraufstatter and H. Bernt, Antibacterial Action of Curcumin and Related Compounds, Nature, vol.164, issue.4167, pp.456-457, 1949.
DOI : 10.1038/164456a0

S. Revathy, S. Elumalai, M. Benny, and B. Antony, Isolation , Purification and Identification of Curcuminoids from Turmeric ( Curcuma Longa L .) by Column Chromatography, Journal of Experimental Sciences, issue.27, pp.21-25, 2011.

A. P. Gupta, M. M. Gupta, and S. Kumar, SAMPLES USING HIGH PERFORMANCE THIN LAYER CHROMATOGRAPHY, Journal of Liquid Chromatography & Related Technologies, vol.1, issue.10, pp.1561-1569, 1999.
DOI : 10.1080/01483918808067200

Y. J. Wang, M. H. Pan, A. L. Cheng, L. I. Lin, Y. S. Ho et al., Stability of curcumin in buffer solutions and characterization of its degradation products, Journal of Pharmaceutical and Biomedical Analysis, vol.15, issue.12, pp.15-1867, 1997.
DOI : 10.1016/S0731-7085(96)02024-9

P. J. Roughley and D. A. Whiting, Experiments in the biosynthesis of curcumin, Journal of the Chemical Society, Perkin Transactions 1, vol.1, pp.2379-2388, 1973.
DOI : 10.1039/p19730002379

J. Schröder, A family of plant-specific polyketide synthases: facts and predictions, Trends in Plant Science, vol.2, issue.10, pp.373-378, 1997.
DOI : 10.1016/S1360-1385(97)87121-X

Y. Katsuyama, T. Kita, N. Funa, and S. Horinouchi, Curcuminoid Biosynthesis by Two Type III Polyketide Synthases in the Herb Curcuma Longa, Journal of Biological Chemistry, issue.17, pp.284-11160, 2009.

V. Lampe and . Synthese-von-curcumin, Berichte der deutschen chemischen Gesellschaft, pp.1347-1355, 1918.

H. J. Pabon, A Synthesis of Curcumin and Related Compounds. Recueil des Travaux Chimiques des Pays-Bas, pp.379-386, 1964.

A. Moore and C. Ran, Near-Infrared Fluorescence Molecular Imaging of Amyloid Beta Species and Monitoring Therapy in Animal Models of Alzheimer's Disease, Proceedings of the National Academy of Sciences, vol.2015, issue.31, pp.112-9734

M. Heger, R. F. Van-golen, M. Broekgaarden, and M. C. Michel, The Molecular Basis for the Pharmacokinetics and Pharmacodynamics of Curcumin and Its Metabolites in Relation to Cancer, Pharmacological Reviews, vol.66, issue.1, pp.222-307
DOI : 10.1124/pr.110.004044

T. M. Kolev, E. A. Velcheva, and B. Stamboliyska, DFT and experimental studies of the structure and vibrational spectra of curcumin, International Journal of Quantum Chemistry, vol.72, issue.6, pp.1069-1079, 2005.
DOI : 10.1016/j.saa.2003.10.018

K. Balasubramanian, Molecular Orbital Basis for Yellow Curry Spice Curcumin's Prevention of Alzheimer's Disease, Journal of Agricultural and Food Chemistry, vol.54, issue.10, pp.3512-3520, 2006.
DOI : 10.1021/jf0603533

J. Li, C. Dai, H. Sun, L. Jin, C. Guo et al., Protective effects and mechanisms of curcumin on podophyllotoxin toxicity in vitro and in vivo, Toxicology and Applied Pharmacology, vol.265, issue.2, pp.190-199
DOI : 10.1016/j.taap.2012.10.010

H. S. Alkhatib, Inhibition of Glycogen Synthase Kinase by Curcumin: Investigation by Simulated Molecular Docking and Subsequent in Vitro/in Vivo Evaluation, Journal of Enzyme Inhibition and Medicinal Chemistry, vol.24, issue.3, pp.771-778, 2009.

S. T. Ngo and M. S. Li, Curcumin Binds to A? 1?40 Peptides and Fibrils Stronger Than Ibuprofen and Naproxen, Journal of medicinal chemistry, vol.116, pp.10165-10175, 2012.

F. Ortica and M. Rodgers, A Laser Flash Photolysis Study of Curcumin in Dioxane-Water Mixtures, Photochemistry and Photobiology, vol.74, issue.6, pp.745-751, 2001.

M. Borsari, E. Ferrari, R. Grandi, and M. Saladini, Curcuminoids as potential new iron-chelating agents: spectroscopic, polarographic and potentiometric study on their Fe(III) complexing ability, Inorganica Chimica Acta, vol.328, issue.1, pp.61-68, 2002.
DOI : 10.1016/S0020-1693(01)00687-9

A. J. Thomson and H. B. Gray, Bio-inorganic chemistry, Current Opinion in Chemical Biology, vol.2, issue.2, pp.155-158, 1998.
DOI : 10.1016/S1367-5931(98)80056-2

A. T. Dinkova-kostova, M. A. Massiah, R. E. Bozak, R. J. Hicks, and P. Talalay, Potency of Michael reaction acceptors as inducers of enzymes that protect against carcinogenesis depends on their reactivity with sulfhydryl groups, Proceedings of the National Academy of Sciences of the United States of America, pp.98-3404, 2000.
DOI : 10.1021/jm00294a001

J. Fang, J. Lu, and A. Holmgren, Thioredoxin Reductase Is Irreversibly Modified by Curcumin, Journal of Biological Chemistry, vol.11, issue.26, pp.25284-25290, 2005.
DOI : 10.1016/j.drup.2004.01.004

URL : http://www.jbc.org/content/280/26/25284.full.pdf

J. Jankun, A. M. Aleem, S. Malgorzewicz, M. Szkudlarek, M. I. Zavodszky et al., Synthetic curcuminoids modulate the arachidonic acid metabolism of human platelet 12-lipoxygenase and reduce sprout formation of human endothelial cells, Molecular Cancer Therapeutics, vol.5, issue.5, pp.1371-1383, 2006.
DOI : 10.1158/1535-7163.MCT-06-0021

T. Loftsson and H. H. Tønnesen, Symmetric and Asymmetric Curcuminoids: Stability, Activity and Complexation with Cyclodextrin, International Journal of Pharmaceutics, vol.338, issue.12, pp.27-34, 2007.

M. Patil and B. Shivapraskash, Pharmacology and Clinical Use of Dimethyl Sulfoxide (DMSO): A Review, International Journal of Molecular Veterinary Research, vol.2013, issue.36, pp.23-33
DOI : 10.5376/ijmvr.2013.03.0006

Y. Liu, Z. Yang, J. Du, X. Yao, and R. Lei, Interaction of curcumin with intravenous immunoglobulin: A fluorescence quenching and Fourier transformation infrared spectroscopy study, Immunobiology, vol.213, issue.8, pp.651-661, 2008.
DOI : 10.1016/j.imbio.2008.02.003

A. Karewicz, D. Bielska, B. Gzyl-malcher, M. Kepczynski, R. Lach et al., Interaction of curcumin with lipid monolayers and liposomal bilayers, Colloids and Surfaces B: Biointerfaces, vol.88, issue.1, pp.231-239, 2011.
DOI : 10.1016/j.colsurfb.2011.06.037

W. Hung, F. Chen, C. Lee, Y. Sun, M. Lee et al., Membrane-Thinning Effect of Curcumin, Biophysical Journal, vol.94, issue.11, pp.94-4331, 2008.
DOI : 10.1529/biophysj.107.126888

Y. Sun, C. Lee, W. Hung, F. Chen, M. Lee et al., The Bound States of Amphipathic Drugs in Lipid Bilayers: Study of Curcumin, Biophysical Journal, vol.95, issue.5, pp.95-2318, 2008.
DOI : 10.1529/biophysj.108.133736

J. Baell and M. Walters, Chemistry: Chemical con artists foil drug discovery, Nature, vol.513, issue.7519, pp.481-483, 2014.
DOI : 10.1038/513481a

M. Subramanian, . Sreejayan, M. N. Rao, T. P. Devasagayam, and B. B. Singh, Diminution of Singlet Oxygen-Induced DNA Damage by Curcumin and Related Antioxidants. Fundamental and Molecular Mechanisms of Mutagenesis, pp.249-255, 1994.

O. N. Gordon, P. B. Luis, H. O. Sintim, and C. Schneider, Unraveling Curcumin Degradation, Journal of Biological Chemistry, vol.242, issue.8, pp.290-4817
DOI : 10.1158/0008-5472.CAN-04-4539

URL : http://www.jbc.org/content/290/8/4817.full.pdf

A. Khurana and C. Ho, High Performance Liquid Chromatographic Analysis of Curcuminoids and Their Photo-oxidative Decomposition Compounds in Curcuma Longa L, Journal of Liquid Chromatography, vol.12, issue.11, pp.11-2295, 1988.
DOI : 10.1007/BF01041928

H. H. Tonnesen, J. Karlsen, and G. Beijersbergen-van-henegouwen, Photochemical Stability of Curcumin, In Studies on curcumin and curcuminoids, pp.116-122, 1986.

K. Priyadarsini, The Chemistry of Curcumin: From Extraction to Therapeutic Agent, Molecules, vol.7, issue.12, pp.20091-20112
DOI : 10.1158/1535-7163.MCT-12-1227

C. F. Chignell, P. Bilski, K. J. Reszka, A. G. Motten, R. H. Sik et al., Spectral and Photochemical Propoerties of Curcumin, Photochemistry and Photobiology, issue.3, pp.59-295, 1994.

A. J. Gescher and W. P. Steward, Prolonged Biologically Active Colonic Tissue Levels of Curcumin Achieved After Oral Administration ? A Clinical Pilot Study Including Assessment of Patient Acceptability, Cancer Prevention Research, vol.2013, issue.62, pp.119-129

D. Hanahan and R. A. Weinberg, Hallmarks of Cancer: The Next Generation, Cell, vol.144, issue.5, pp.646-674, 2011.
DOI : 10.1016/j.cell.2011.02.013

D. Hanahan, R. A. Weinberg, and S. Francisco, The Hallmarks of Cancer, Cell, vol.100, issue.1, pp.57-70, 2000.
DOI : 10.1016/S0092-8674(00)81683-9

G. J. Hannon and D. Beach, P15INK4B Is a Potentiel Effector of TGF-Beta-Induced Cell Cycle Arrest, Nature, vol.371, pp.267-261, 1994.

S. E. Artandi and R. A. Depinho, Telomeres and telomerase in cancer, Carcinogenesis, vol.31, issue.1, pp.9-18
DOI : 10.1093/carcin/bgp268

URL : https://academic.oup.com/carcin/article-pdf/31/1/9/7094383/bgp268.pdf

T. G. Cotter, Apoptosis and cancer: the genesis of a research field, Nature Reviews Cancer, vol.112, issue.7, pp.501-507, 2009.
DOI : 10.1172/JCI20039

C. Murdoch, M. Muthana, S. B. Coffet, and C. E. Lewis, The role of myeloid cells in the promotion of tumour angiogenesis, Nature Reviews Cancer, vol.13, issue.8, pp.618-631, 2008.
DOI : 10.1593/neo.07871

A. Nürnberg, T. Kitzing, and R. Grosse, Nucleating actin for invasion, Nature Reviews Cancer, vol.60, issue.3, pp.177-187, 2011.
DOI : 10.1136/thx.2004.037622

M. Olayioye, R. M. Neve, H. Lane, and N. Hynes, The ErbB Signaling Network: Receptor Heterodimerization in Development and Cancer. The EMBO journal, pp.3159-3167, 2000.

Y. Jung, W. Xu, H. Kim, N. Ha, and L. Neckers, Curcumin-induced degradation of ErbB2: A role for the E3 ubiquitin ligase CHIP and the Michael reaction acceptor activity of curcumin, Biochimica et Biophysica Acta (BBA) - Molecular Cell Research, vol.1773, issue.3, pp.383-390, 2007.
DOI : 10.1016/j.bbamcr.2006.11.004

B. Bharat, *. Aggarwal, S. Banerjee, U. Bhardwaj, B. Sung et al., Curcumin Induces the Degradation of Cyclin E Expression through Ubiquitin-Dependent Pathway and up-Regulates Cyclin-Dependent Kinase Inhibitors p21 and p27 Multiple Human Tumor Cell Lines, Biochemical Pharmacology, vol.73, pp.1024-1032, 2007.

J. H. Lee and I. K. Chung, Curcumin inhibits nuclear localization of telomerase by dissociating the Hsp90 co-chaperone p23 from hTERT, Cancer Letters, vol.290, issue.1, pp.76-86, 2010.
DOI : 10.1016/j.canlet.2009.08.026

B. D. Manning, L. C. Cantley, A. Review, and . Signaling, AKT/PKB Signaling: Navigating Downstream, Cell, vol.129, issue.7, pp.1261-1274, 2007.
DOI : 10.1016/j.cell.2007.06.009

URL : http://doi.org/10.1016/j.cell.2007.06.009

S. Nagai, M. Kurimoto, K. Washiyama, Y. Hirashima, and T. Kumanishi, Inhibition of Cellular Proliferation and Induction of Apoptosis by Curcumin in Human Malignant Astrocytoma Cell Lines, Journal of Neuro-Oncology, vol.21, issue.2, pp.105-111, 2005.
DOI : 10.1128/MCB.12.10.4412

E. M. Grinner and M. G. Kazanietz, Protein kinase C and other diacylglycerol effectors in cancer, Nature Reviews Cancer, vol.10, issue.4, pp.281-294, 2007.
DOI : 10.1038/bjc.1998.13

A. Majhi, G. M. Rahman, S. Panchal, and J. Das, Binding of curcumin and its long chain derivatives to the activator binding domain of novel protein kinase C, Bioorganic & Medicinal Chemistry, vol.18, issue.4, pp.1591-1598, 2011.
DOI : 10.1016/j.bmc.2009.12.075

D. W. Scott and G. Loo, Curcumin-induced GADD153 gene up-regulation in human colon cancer cells, Carcinogenesis, vol.25, issue.11, pp.25-2155, 2004.
DOI : 10.1093/carcin/bgh239

S. I. Grivennikov and M. Karin, Inflammation and oncogenesis: a vicious connection, Current Opinion in Genetics & Development, vol.20, issue.1, pp.65-71, 2010.
DOI : 10.1016/j.gde.2009.11.004

URL : http://www.ncbi.nlm.nih.gov/pmc/articles/PMC2821983

W. Maldonado-rojas and J. Olivero-verbel, Potential interaction of natural dietary bioactive compounds with COX-2, Journal of Molecular Graphics and Modelling, vol.30, pp.157-166, 2011.
DOI : 10.1016/j.jmgm.2011.07.002

D. J. Kerr, J. A. Dunn, M. J. Langman, J. L. Smith, R. S. Midgley et al., Rofecoxib and Cardiovascular Adverse Events in Adjuvant Treatment of Colorectal Cancer, New England Journal of Medicine, vol.357, issue.4, pp.360-369, 2007.
DOI : 10.1056/NEJMoa071841

J. Hong, M. Bose, J. Ju, J. Ryu, X. Chen et al., Modulation of arachidonic acid metabolism by curcumin and related ??-diketone derivatives: effects on cytosolic phospholipase A2, cyclooxygenases and 5-lipoxygenase, Carcinogenesis, vol.25, issue.9, pp.1671-1679, 2004.
DOI : 10.1093/carcin/bgh165

B. G. Hernández, O. V. Volpert, M. A. Íñiguez, E. Lorenzo, S. Martínez-martínez et al., Selective Inhibition of Vascular Endothelial Growth Factor???Mediated Angiogenesis by Cyclosporin a, The Journal of Experimental Medicine, vol.26, issue.5, pp.607-620, 2001.
DOI : 10.1056/NEJM199412013312203

L. Bartik, G. K. Whitfield, M. Kaczmarska, C. L. Lowmiller, W. Moffet et al., Curcumin: a novel nutritionally derived ligand of the vitamin D receptor with implications for colon cancer chemoprevention, The Journal of Nutritional Biochemistry, vol.21, issue.12, pp.1153-1161, 2011.
DOI : 10.1016/j.jnutbio.2009.09.012

M. V. Koonammackal, U. V. Nellipparambil, and C. Sudarsanakumar, Molecular dynamics simulations and binding free energy analysis of DNA minor groove complexes of curcumin, Journal of Molecular Modeling, vol.294, issue.11, pp.17-2805, 2011.
DOI : 10.1006/jmbi.1999.3280

R. A. Stelzma, H. N. Schnitzlein, and F. Murllagh, An english translation of alzheimer's 1907 paper, ????ber eine eigenartige erkankung der hirnrinde?, Clinical Anatomy, vol.26, issue.6, pp.429-431, 1995.
DOI : 10.1001/archneur.1972.00490080015001

M. Muller, S. Sigurdsson, O. Kjartansson, P. V. Jonsson, M. Garcia et al., Birth Size and Brain Function 75 Years Later, PEDIATRICS, vol.134, issue.4, pp.761-770
DOI : 10.1542/peds.2014-1108

URL : http://www.ncbi.nlm.nih.gov/pmc/articles/PMC4179101

M. F. Folstein, S. E. Folstein, and P. Mchugh, ???Mini-mental state???, Journal of Psychiatric Research, vol.12, issue.3, pp.189-198, 1975.
DOI : 10.1016/0022-3956(75)90026-6

C. Derouesné, J. Poitreneau, L. Hugonot, M. Kalafat, B. Dubois et al., Le Mini- Mental State Examination ( MMSE ) : Un Outil Pratique Pour L'évaluation de L'état Cognitif Des Patients Par Le Clinicien, pp.28-1141, 1999.

S. H. Barage and K. D. Sonawane, Amyloid cascade hypothesis: Pathogenesis and therapeutic strategies in Alzheimer's disease, Neuropeptides, vol.52, issue.110, 2015.
DOI : 10.1016/j.npep.2015.06.008

M. E. Jönhagen, A. Nordberg, K. Amberla, L. Bäckman, T. Ebendal et al., Intracerebroventricular Infusion of Nerve Growth Factor in Three Patients with Alzheimer's Disease. Dementia and Geriatric Cognitive Disorders, pp.246-257, 1998.

C. L. Masters, G. Simms, N. Weinman, G. Multhaup, B. L. Mcdonald et al., Amyloid plaque core protein in Alzheimer disease and Down syndrome., Proceedings of the National Academy of Sciences, vol.82, issue.12, pp.82-4245, 1985.
DOI : 10.1073/pnas.82.12.4245

J. Walter and D. R. Thal, Biochemical Stages of Amyloid-? Peptide Aggregation and Accumulation in the Human Brain and Their Association with Symptomatic and Pathologically Preclinical Alzheimer's Disease, Brain, vol.2014, issue.1373, pp.887-903

K. Garai and C. Frieden, Quantitative analysis of the time course of A?? oligomerization and subsequent growth steps using tetramethylrhodamine-labeled A??, Proceedings of the National Academy of Sciences of the United States of America 2013, pp.3321-3326
DOI : 10.1016/j.ab.2008.12.024

L. Hendriks, The betaA4 Amyloid Precursor Protein Gene and Alzheimer's Disease, European Journal of Biochemistry, vol.245, issue.1, pp.6-15, 1996.
DOI : 10.1016/0092-8674(95)90073-X

P. R. Turner, K. O-'connor, W. P. Tate, and W. C. Abraham, Roles of amyloid precursor protein and its fragments in regulating neural activity, plasticity and memory, Progress in Neurobiology, vol.70, issue.1, 2003.
DOI : 10.1016/S0301-0082(03)00089-3

S. Gandy, The Role of Cerebral Amyloid Beta Accumulation in Common Forms of Alzheimer Disease, The Journal of Clinical Investigation, vol.115, issue.1195, pp.1121-1129, 2005.

Y. Xu, J. Shen, X. Luo, W. Zhu, K. Chen et al., Conformational Transition of Amyloid Beta-Peptide, Proceedings of the National Academy of Sciences of the United States of America, issue.12015, pp.102-5403, 2005.

M. Yang and D. B. Teplow, Amyloid ??-Protein Monomer Folding: Free-Energy Surfaces Reveal Alloform-Specific Differences, Journal of Molecular Biology, vol.384, issue.2, pp.450-464, 2008.
DOI : 10.1016/j.jmb.2008.09.039

URL : http://www.ncbi.nlm.nih.gov/pmc/articles/PMC2673916

K. A. Ball, A. H. Phillips, D. E. Wemmer, and T. Head-gordon, Differences in ??-strand Populations of Monomeric A??40 and A??42, Biophysical Journal, vol.104, issue.12, pp.2714-2724, 2013.
DOI : 10.1016/j.bpj.2013.04.056

J. Hardy and D. J. Selkoe, The Amyloid Hypothesis of Alzheimer's Disease: Progress and Problems on the Road to Therapeutics, Science, vol.297, issue.5580, pp.297-353, 2002.
DOI : 10.1126/science.1072994

N. Carulla, M. Zhou, E. Giralt, C. V. Robinson, and C. M. Dobson, Structure and Intermolecular Dynamics of Aggregates Populated during Amyloid Fibril Formation Studied by Hydrogen/Deuterium Exchange, Accounts of Chemical Research, vol.43, issue.8, pp.43-1072, 2010.
DOI : 10.1021/ar9002784

L. O. Tjernberg, J. Näslund, F. Lindqvist, J. Johansson, R. Karlström et al., Arrest of Beta-Amyloid Fibril Formation by a Pentapeptide Ligand, The Journal of Biological Chemistry, issue.12515, pp.271-8545, 1996.

P. Cheng, C. Liu, M. Zhao, D. Eisenberg, and J. S. Nowick, Amyloid ??-sheet mimics that antagonize protein aggregation and reduce amyloid toxicity, Nature Chemistry, vol.4, issue.11, pp.927-933
DOI : 10.1126/science.1213151

URL : http://www.ncbi.nlm.nih.gov/pmc/articles/PMC3481199

J. Geng, C. Zhao, J. Ren, and X. Qu, Alzheimer's disease amyloid beta converting left-handed Z-DNA back to right-handed B-form, Chemical Communications, vol.45, issue.38, pp.46-7187, 2010.
DOI : 10.1039/c0cc02049d

B. Urbanc, M. Betnel, L. Cruz, G. Bltan, and D. B. Teplow, Elucidation of Amyloid ??-Protein Oligomerization Mechanisms: Discrete Molecular Dynamics Study, Journal of the American Chemical Society, vol.132, issue.12, pp.132-4266, 2010.
DOI : 10.1021/ja9096303

P. Cizas, R. Budvytyte, R. Morkuniene, R. Moldovan, M. Broccio et al., Size-dependent neurotoxicity of ??-amyloid oligomers, Archives of Biochemistry and Biophysics, vol.496, issue.2, pp.84-92, 2010.
DOI : 10.1016/j.abb.2010.02.001

M. Rosini and E. Simoni, Milelli, a; Minarini, a; Melchiorre, C. Oxidative Stress in Alzheimer's Disease: Are We Connecting the Dots, Journal of Medicinal Chemistry, vol.2014, issue.577, pp.2821-2831

D. A. Butterfield, T. Reed, S. F. Newman, and R. Sultana, Roles of amyloid ??-peptide-associated oxidative stress and brain protein modifications in the pathogenesis of Alzheimer's disease and mild cognitive impairment, Free Radical Biology and Medicine, vol.43, issue.5, pp.43-658, 2007.
DOI : 10.1016/j.freeradbiomed.2007.05.037

D. A. Butterfield, V. Galvan, M. B. Lange, H. Tang, R. Sowell et al., In vivo oxidative stress in brain of Alzheimer disease transgenic mice: Requirement for methionine 35 in amyloid ??-peptide of APP, Free Radical Biology and Medicine, vol.48, issue.1, pp.136-144, 2010.
DOI : 10.1016/j.freeradbiomed.2009.10.035

E. Gaggelli, H. Kozlowski, D. Valensin, and G. Valensin, Copper Homeostasis and Neurodegenerative Disorders (Alzheimer's, Prion, and Parkinson's Diseases and Amyotrophic Lateral Sclerosis), Chemical Reviews, vol.106, issue.6, pp.1995-2044, 2006.
DOI : 10.1021/cr040410w

K. Iqbal, Alzheimer Review Series: Alzheimer neurofibrillary degeneration: significance, etiopathogenesis, therapeutics and prevention, Journal of Cellular and Molecular Medicine, vol.9, issue.1, pp.38-55, 2008.
DOI : 10.1212/WNL.34.7.939

URL : http://onlinelibrary.wiley.com/doi/10.1111/j.1582-4934.2008.00225.x/pdf

S. L. Shammas, G. A. Garcia, S. Kumar, M. Kjaergaard, M. H. Horrocks et al., A mechanistic model of tau amyloid aggregation based on direct observation of oligomers, Nature Communications, vol.97, issue.135, pp.1-10, 2015.
DOI : 10.1073/pnas.97.10.5129

H. Kadavath, R. V. Hofele, J. Biernat, S. Kumar, K. Tepper et al., Tau stabilizes microtubules by binding at the interface between tubulin heterodimers, Proceedings of the National Academy of Sciences, vol.86, issue.2, 2015.
DOI : 10.1038/nature02393

M. Kolarova, F. Garc, A. Bartos, J. Ricny, D. Ripova et al., Structure and Pathology of Tau Protein in Alzheimer Disease, International Journal of Alzheimer's Disease, vol.14, issue.4, pp.1-13
DOI : 10.3233/JAD-2008-14407

M. Bergen, . Von, P. Friedhoff, J. Biernat, J. Heberle et al., Assembly of Tau Protein into Alzheimer Paired Helical Filaments Depends on a Local Sequence Motif ( 306 VQIVYK 311 ) Forming ? Structure, Proceedings of the National Academy of Sciences of the United States of America, pp.97-5129, 2000.

P. H. Reddy and . Tau, Abnormal tau, mitochondrial dysfunction, impaired axonal transport of mitochondria, and synaptic deprivation in Alzheimer's disease, Brain Research, vol.1415, issue.139, pp.1415-136, 2011.
DOI : 10.1016/j.brainres.2011.07.052

I. Morales, L. Guzmán-martínez, C. Cerda-troncoso, G. Farías, and R. B. Maccioni, Neuroinflammation in the Pathogenesis of Alzheimer's Disease. A Rational Framework for the Search of Novel Therapeutic Approaches, Frontiers in Cellular Neuroscience, vol.2014, issue.8112, pp.1-9

T. C. Frank-cannon, L. T. Alto, F. E. Mcalpine, and M. G. Tansey, Does Neuroinflammation Fan the Flame in Neurodegenerative Diseases? Molecular Neurodegeneration, pp.1-13, 2009.

W. Lee, C. Loo, M. Bebawy, F. Luk, R. S. Mason et al., Curcumin and its Derivatives: Their Application in Neuropharmacology and Neuroscience in the 21st Century, Current Neuropharmacology, vol.11, issue.4, pp.338-378, 2013.
DOI : 10.2174/1570159X11311040002

S. Kotler, P. Walsh, J. R. Brender, and A. Ramamoorthy, Differences between amyloid-?? aggregation in solution and on the membrane: insights into elucidation of the mechanistic details of Alzheimer's disease, Chem. Soc. Rev., vol.39, issue.Suppl, pp.8-10, 2014.
DOI : 10.1007/s10858-007-9176-4

L. N. Zhao, H. Long, Y. Mu, and L. Chew, The Toxicity of Amyloid ? Oligomers, International Journal of Molecular Sciences, vol.2012, issue.1446, pp.13-7303

S. M. Butterfield and H. Lashuel, Amyloidogenic Protein-Membrane Interactions: Mechanistic Insight from Model Systems, Angewandte Chemie International Edition, vol.24, issue.33, pp.49-5628, 2010.
DOI : 10.1128/jb.175.18.5953-5961.1993

URL : https://infoscience.epfl.ch/record/142971/files/5628_ftp.pdf

H. Jang, J. Zheng, R. Lal, and R. Nussinov, New structures help the modeling of toxic amyloid?? ion channels, Trends in Biochemical Sciences, vol.33, issue.2, pp.91-100, 2008.
DOI : 10.1016/j.tibs.2007.10.007

T. L. Williams and L. C. Serpell, Membrane and surface interactions of Alzheimer???s A?? peptide - insights into the mechanism of cytotoxicity, FEBS Journal, vol.132, issue.Suppl 1, pp.278-3905, 2011.
DOI : 10.1021/ja9096303

J. Zhao, Y. Fu, C. Liu, M. Shinohara, H. M. Nielsen et al., Retinoic Acid Isomers Facilitate Apolipoprotein E Production and Lipidation in Astrocytes through the Retinoid X Receptor/Retinoic Acid Receptor Pathway, Journal of Biological Chemistry, vol.42, issue.16, pp.289-11282
DOI : 10.1186/1750-1326-8-18

P. Hobson, Lack of Support for Bexarotene as a Treatment for Alzheimer's Disease, Neuropharmacology, vol.100, pp.124-130, 2016.

D. M. Michaelson, Reversal of apoE4-Driven Brain Pathology and Behavioral Deficits by Bexarotene, Journal of Neuroscience, vol.150, issue.3421, pp.7293-7301

Q. Ma, F. Yang, E. R. Rosario, O. J. Ubeda, W. Beech et al., Beta-Amyloid Oligomers Induce Phosphorylation of Tau and Inactivation of Insulin Receptor Substrate via c- Jun N-Terminal Kinase Signaling: Suppression by Omega-3 Fatty Acids and Curcumin, The Journal of Neuroscience : the Official Journal of the Society for Neuroscience, issue.15128, pp.29-9078, 2009.

L. Texidó, M. Martín-satué, E. Alberdi, C. Solsona, and C. Matute, Amyloid ?? peptide oligomers directly activate NMDA receptors, Cell Calcium, vol.49, issue.3, pp.184-190, 2011.
DOI : 10.1016/j.ceca.2011.02.001

H. B. Nygaard and S. M. Strittmatter, Cellular Prion Protein Mediates the Toxicity of Beta- Amyloid Oligomers: Implications for Alzheimer Disease, Archives of Neurology, issue.15311, pp.66-1325, 2009.

A. I. Bush, C. L. Masters, and R. E. Tanzi, Copper, ??-amyloid, and Alzheimer's disease: Tapping a sensitive connection, Proceedings of the National Academy of Sciences, vol.274, issue.5284, pp.100-11193, 2003.
DOI : 10.1126/science.274.5284.99

URL : http://www.ncbi.nlm.nih.gov/pmc/articles/PMC208731

L. M. Sayre, G. Perry, P. L. Harris, Y. Liu, and K. Schubert, In Situ Oxidative Catalysis by Neurofibrillary Tangles and Senile Plaques in Alzheimer???s Disease, Situ, pp.270-279, 2000.
DOI : 10.1016/S0304-4165(97)00039-1

K. Ono, K. Hasegawa, H. Naiki, and M. Yamada, Curcumin has potent anti-amyloidogenic effects for Alzheimer's ?-amyloid fibrils in vitro, Journal of Neuroscience Research, vol.67, issue.6, pp.75-742, 2004.
DOI : 10.1007/BF02797132

P. P. Rao, T. Mohamed, K. Teckwani, and G. Tin, Curcumin Binding to Beta Amyloid: A Computational Study, Chemical Biology & Drug Design, vol.109, issue.4, 2015.
DOI : 10.1073/pnas.1218792109

A. Reinke and J. Gestwicki, Structure?activity Relationships of Amyloid Beta-aggregation Inhibitors Based on Curcumin: Influence of Linker Length and Flexibility, Chemical Biology & Drug Design, vol.44, issue.3, pp.206-215, 2007.
DOI : 10.1177/37.8.2666510

H. Levine, Thioflavine T Interaction with Synthetic Alzheimer's Disease Beta-Amyloid Peptides: Detection of Amyloid Aggregation in Solution Protein science : a publication of the, pp.404-410, 1993.

M. Biancalana and S. Koide, Molecular mechanism of Thioflavin-T binding to amyloid fibrils, Biochimica et Biophysica Acta (BBA) - Proteins and Proteomics, vol.1804, issue.7, pp.1804-1405, 2010.
DOI : 10.1016/j.bbapap.2010.04.001

L. S. Wolfe, M. F. Calabrese, A. Nath, D. Blaho, A. D. Miranker et al., Protein-induced photophysical changes to the amyloid indicator dye thioflavin T, Proceedings of the National Academy of Sciences, vol.18, issue.Pt 11, pp.16863-16868, 2010.
DOI : 10.1063/1.1747632

M. R. Krebs, E. H. Bromley, and . M. Donald, The binding of thioflavin-T to amyloid fibrils: localisation and implications, Journal of Structural Biology, vol.149, issue.1, pp.30-37, 2005.
DOI : 10.1016/j.jsb.2004.08.002

K. Murata, S. Matsumura, Y. Yoshioka, Y. Ueno, and H. Matsuda, Screening of ??-secretase and acetylcholinesterase inhibitors from plant resources, Journal of Natural Medicines, vol.288, issue.1, pp.123-129
DOI : 10.1074/jbc.M112.444703

T. Hamaguchi, K. Ono, and M. Yamada, REVIEW: Curcumin and Alzheimer's Disease, CNS Neuroscience & Therapeutics, vol.62, issue.524, pp.285-297, 2010.
DOI : 10.1016/S0027-5107(02)00328-7

M. Landau, M. R. Sawaya, K. F. Faull, A. Laganowsky, L. Jiang et al., Towards a Pharmacophore for Amyloid, PLoS Biology, vol.278, issue.6, pp.25-27, 2011.
DOI : 10.1371/journal.pbio.1001080.s009

W. Berhanu and A. Masunov, Atomistic mechanism of polyphenol amyloid aggregation inhibitors: molecular dynamics study of Curcumin, Exifone, and Myricetin interaction with the segment of tau peptide oligomer, Journal of Biomolecular Structure and Dynamics, vol.22, issue.7, 2014.
DOI : 10.1021/jf063299z

M. Bergen, . Von, P. Friedhoff, A. Schneider, T. Kampera et al., Rapid Assembly of Microtubule-Associated Protein Tau into Alzheimer-like Paired Helical Filaments Monitored by Fluorescence in Solution, Biochemistry, issue.16928, pp.37-10223, 1998.

S. V. Jovanovic, S. Steenken, C. W. Boone, and M. G. Simic, H-Atom Transfer Is A Preferred Antioxidant Mechanism of Curcumin, Journal of the American Chemical Society, vol.121, issue.41, pp.121-9677, 1999.
DOI : 10.1021/ja991446m

T. Ak and I. Gülçin, Antioxidant and radical scavenging properties of curcumin, Chemico-Biological Interactions, vol.174, issue.1, pp.27-37, 2008.
DOI : 10.1016/j.cbi.2008.05.003

C. Beauchamp and I. Fridovich, Superoxide dismutase: Improved assays and an assay applicable to acrylamide gels, Analytical Biochemistry, vol.44, issue.1, pp.276-287, 1971.
DOI : 10.1016/0003-2697(71)90370-8

T. C. Dinis, V. M. Madeira, and L. M. Almeida, Action of Phenolic Derivatives (Acetaminophen, Salicylate, and 5-Aminosalicylate) as Inhibitors of Membrane Lipid Peroxidation and as Peroxyl Radical Scavengers, Archives of Biochemistry and Biophysics, vol.315, issue.1, pp.315-161
DOI : 10.1006/abbi.1994.1485

R. E. Tanzi, The Zinc Dyshomeostasis Hypothesis of Alzheimer's Disease, PLoS ONE, vol.2012, issue.73, p.33552

S. M. Hancock and D. I. Finkelstein, Adlard, P. a. Glia and Zinc in Ageing and Alzheimer's Disease: A Mechanism for Cognitive Decline?, Frontiers in Aging Neuroscience, vol.2014, issue.6137, pp.1-6

G. Shoba, D. Joy, T. Joseph, M. Majeed, R. Rajendran et al., Influence of Piperine on the Pharmacokinetics of Curcumin in Animals and Human Volunteers, Influence of Piperine on the Pharmacokinetics of Curcumin in Animals and Human Volunteers, pp.353-356, 1998.
DOI : 10.1055/s-2006-957450

D. J. Greenblatt, M. Divoll, J. S. Harmatz, and R. I. Shader, Pharmacokinetic Comparison of Sublingual Lorazepam with Intravenous, Intramuscular, and Oral Lorazepam, Journal of pharmaceutical sciences, issue.1772, pp.71-248, 1982.

C. D. Lao, M. T. Ruffin, D. Normolle, D. D. Heath, S. I. Murray et al., Dose escalation of a curcuminoid formulation, BMC Complementary and Alternative Medicine, vol.61, issue.3, pp.1-4, 2006.
DOI : 10.1016/0300-483X(81)90056-1

P. Anand, A. B. Kunnumakkara, R. Newman, and B. B. Aggarwal, Bioavailability of Curcumin: Problems and Promises, Molecular Pharmaceutics, vol.4, issue.6, pp.807-818, 2007.
DOI : 10.1021/mp700113r

M. H. Pan, T. M. Huang, and J. K. Lin, Biotransformation of Curcumin through Reduction and Glucuronidation in Mice, Drug Metabolism and Disposition, vol.27, issue.1804, pp.486-494, 1999.

M. Cruz-correa, D. A. Shoskes, P. Sanchez, R. Zhao, L. M. Hylind et al., Combination Treatment With Curcumin and Quercetin of Adenomas in Familial Adenomatous Polyposis, Clinical Gastroenterology and Hepatology, vol.4, issue.8, pp.1035-1038, 2006.
DOI : 10.1016/j.cgh.2006.03.020

A. Kondo, T. Takeda, B. Li, K. Tsuiji, M. Kitamura et al., Epigallocatechin-3-gallate potentiates curcumin???s ability to suppress uterine leiomyosarcoma cell growth and induce apoptosis, International Journal of Clinical Oncology, vol.256, issue.3, pp.380-388, 2013.
DOI : 10.1006/excr.2000.4834

X. Chen, L. Zou, J. Niu, W. Liu, S. Peng et al., The Stability, Sustained Release and Cellular Antioxidant Activity of Curcumin Nanoliposomes, Molecules, vol.65, issue.8, pp.20-14293
DOI : 10.1021/jf304821k

S. Sood, K. Jain, and K. Gowthamarajan, Optimization of curcumin nanoemulsion for intranasal delivery using design of experiment and its toxicity assessment, Colloids and Surfaces B: Biointerfaces, vol.113, issue.185, pp.330-337, 2014.
DOI : 10.1016/j.colsurfb.2013.09.030

A. N. Khanji, F. Michaux, J. Jasniewski, J. Petit, E. Lahimer et al., Structure and Gelation Properties of Casein Micelles Doped with Curcumin under Acidic Conditions. Food and FunctionFunction, pp.10-1039, 2015.
URL : https://hal.archives-ouvertes.fr/hal-01264776

Y. Yu, X. Zhang, and L. Qiu, The anti-tumor efficacy of curcumin when delivered by size/charge-changing multistage polymeric micelles based on amphiphilic poly(??-amino ester) derivates, Biomaterials, vol.35, issue.10, pp.35-3467
DOI : 10.1016/j.biomaterials.2013.12.096

K. Lee, C. Hc, I. Sa, P. Yh, K. Cs et al., Multicenter phase II trial of Genexol-PM, a Cremophor-free, polymeric micelle formulation of paclitaxel, in patients with metastatic breast cancer, Breast Cancer Research and Treatment, vol.97, issue.2, pp.241-250, 2008.
DOI : 10.1007/s10549-007-9591-y

K. Nagahama, Y. Sano, and T. Kumano, Anticancer drug-based multifunctional nanogels through self-assembly of dextran???curcumin conjugates toward cancer theranostics, Bioorganic & Medicinal Chemistry Letters, vol.25, issue.12
DOI : 10.1016/j.bmcl.2015.04.062

S. S. Bansal, H. Kausar, F. Aqil, J. Jeyabalan, M. V. Vadhanam et al., Curcumin implants for continuous systemic delivery: safety and biocompatibility, Drug Delivery and Translational Research, vol.9, issue.4, pp.332-341, 2011.
DOI : 10.1016/0142-9612(88)90063-4

M. Ghosh and R. O. Ryan, ApoE enhances nanodisk-mediated curcumin delivery to glioblastoma multiforme cells, Nanomedicine, vol.9, issue.6, pp.763-771
DOI : 10.2217/nnm.13.35

URL : http://www.ncbi.nlm.nih.gov/pmc/articles/PMC3904962

B. B. Aggarwal and K. B. Harikumar, Potential therapeutic effects of curcumin, the anti-inflammatory agent, against neurodegenerative, cardiovascular, pulmonary, metabolic, autoimmune and neoplastic diseases, The International Journal of Biochemistry & Cell Biology, vol.41, issue.1, pp.41-81, 2009.
DOI : 10.1016/j.biocel.2008.06.010

J. W. Betts and D. W. Wareham, In vitro activity of curcumin in combination with epigallocatechin gallate (EGCG) versus multidrug-resistant Acinetobacter baumannii, BMC Microbiology, vol.14, issue.1, p.172
DOI : 10.1016/j.dyepig.2004.05.008

O. N. Tembo, P. Dallemagne, S. Rault, and M. Robba, An Efficient Synhtesis of New Phenylpyrrolizine and Phenylpyrrolopyrazine Derivatives, Heterocycles, vol.36, issue.9, pp.2129-2137, 1993.

G. Atassi, D. Caignard, and P. Renard, Rault, S. Design, Synthesis, and Evaluation of Novel Thienopyrrolizinones as Antitubulin Agents, Journal of medicinal chemistry, issue.6, pp.47-1448, 2004.

S. Rault, M. Cugnon-de-sevicourt, A. M. Godard, and M. Robba, A Convenient Synthesis of New Aminopyrroloindoles via an Iminium Salt, Tetrahedron Letters, issue.19, pp.26-2305, 1985.