L. Nicolas, Le dictionaire ou traité universel des drogues simples. 1716;Aux dépens de la Compagnie

C. W. Poerner, Instruction sur l'art de la teinture et particulièrement sur la teinture des laines, 1791.

. Andry, Matière médicale extraite des meilleurs auteurs, et principalement du Traité des médicamens de M. de Tournefort , et des leçons de M. Ferrein. 1770

, Food and Agriculture Organization of the United Nations. Spices Processing Toolkit of Curcuma, 2007.

L. Ali, A. Haque, and K. Saleem, Separation and identification of curcuminoids in turmeric powder by HPLC using phenyl column, Anal Methods, 2014.

K. J. Lee, Y. S. Kim, P. M. Jung, and J. Y. Ma, Optimization of the conditions for the analysis of curcumin and a related compound in Curcuma longa with mobile-phase composition and column temperature via RP-HPLC, In: Asian Journal fo Chemistry, 2013.

D. Harman, Aging : A theory based on free radical and radiation chemistry, 1956.

B. Halliwell and J. Gutteridge, Free Radicals in Biology and Medicine, vol.63, 1989.

J. J. Poderoso, M. C. Carreras, C. Lisdero, N. Riobo, F. Chopfer et al., Nitric oxide inhibits electron transfer and increases superoxide radical production in rat heart mitochondria and submitochondrial particles, Arch Biochem Biophys, 1996.

B. Halliwell and J. Gutteridge, Antioxidant defenses, Free Radicals in Biology and Medicine, 1999.

J. S. Wright, Predicting the antioxidant activity of curcumin and curcuminoids, Journal of Molecular Structure, pp.207-224, 2002.

S. V. Jovanovic, S. Steenken, C. W. Boone, and M. G. Simic, H-atom transfer is a preferred antioxidant mechanism of curcumin, Journal of American Chemical Society, 1999.

L. Barclay, M. R. Vinqvist, K. Mukai, H. Goto, Y. Hashimoto et al., The antioxidant mechanism of curcumin: Classical methods are needed to determine antioxidant mechanism and activity, Org Lett, vol.2, pp.2841-2843, 2000.

K. I. Priyadarsini, D. K. Maity, G. H. Naik, M. S. Kumar, M. K. Unnikrishnan et al., Role of phenolic O:H and methylene hydrogen on the free radical reaction and antioxidant activity of curcumin. Free Radical, 2003.

Y. M. Sun, H. Y. Zhang, D. Z. Chen, and C. B. Liu, Theoretical elucidation on the antioxidant mechanism of curcumin: A DFT study, Organic Letters, 2002.

L. Shen, H. Y. Zhang, and H. F. Ji, Successful application of TD-DFT in transient absorption spectra assignment, Org Lett, pp.243-246, 2005.

M. T. Huang, T. Lysz, T. Ferraro, T. F. Abidi, J. D. Laskin et al., Inhibitory effects of curcumin on in vitro lipoxygenase and cyclooxygenase activities in mouse epidermis, Cancer Res, vol.51, pp.813-819, 1991.

C. D. Funk, L. B. Funk, K. Me, A. S. Pong, and G. A. Fitzgerald, Human platelet/erythroleukemia cell prostaglandin G/H synthase:cDNA cloning, expression and gene chromosomal assignment, FASEB Journal, 1991.

K. Subbaramaiah, N. Telang, J. T. Ramonetti, R. Araki, B. Devito et al., Transcription of cyclooxygenase-2 is enhanced in transformed mammary epithelial cells, Cancer Res, issue.56, pp.4424-4433, 1996.

L. Qiao, V. Kozoni, G. J. Tsioulias, M. I. Koutsos, R. Hanif et al., Selected eicosaniods increase the proliferation rate of human colon carcinoma cell lines and mouse colonocytes in vivo, Biochim Biophys Acta, pp.215-238, 1258.

H. Y. Fang, T. S. Lin, J. P. Lin, Y. C. Wu, K. C. Chow et al., Cyclooxygenase-2 in human non-small cell lung cancer, Eur J Surg Oncol, issue.29, pp.171-178, 2003.

J. L. Masferrer, B. S. Zweifel, P. T. Manning, S. D. Hauser, K. M. Leahy et al., Selective inhibition of cyclooxygenase-2 in vivo is antiinflammatory and nonulcerogenic, Proc Natl Acad Sci, issue.91, pp.3228-3232, 1994.

T. Kawamori, C. V. Rao, K. Seibert, and B. S. Reddy, Chemopreventive activity of celecoxib, a specific cyclooxygenase-2 inhibitor, against colon carcinogenesis, Cancer Res, issue.58, pp.409-421, 1998.

J. Hong, M. Bose, J. Ju, J. H. Ryu, X. Chen et al., Modulation of arachidonic acid metabolism by curcumin and related beta-diketone derivatives: Effects on cytosolic phospholipase A(2), cyclooxygenases and 5-lipoxygenase, Carcinogenesis, vol.25, issue.9, pp.1671-1680, 2004.

F. Zhang, N. K. Altorki, J. R. Mestre, K. Subbaramaiah, and A. J. Dannenberg, Curcumin inhibits cyclooxygenase-2 transcription in bile acid-and phorbol ester-treated human gastrointestinal epithelial cells, Carcinogenesis, issue.20, pp.445-51, 1991.

A. Goel, C. R. Boland, and D. P. Chauhan, Specific inhibition of cyclooxygenase-2 (COX-2) expression by dietary curcumin in HT-29 human colon cancer cells, Cancer Lett, issue.172, pp.111-119, 2001.

S. R. Goldstein, G. Y. Yang, X. Chen, S. K. Curtis, and C. S. Yang, Studies of iron deposits, inducible nitric oxide synthase and nitrotyrosine in a rat model for esophageal adenocarcinoma, Carcinogenesis, issue.19, pp.1445-1449, 1998.

L. L. Thomsen, D. W. Miles, L. Happerfield, L. G. Bobrow, R. G. Knowles et al., Nitric oxide synthase activity in human breast cancer, British Journal of Cancer, 1995.

M. Takahashi, K. Fukuda, T. Ohata, and K. Wakabayashi, Increased expression of inducible and endothelial nitric oxide synthases in rat colon tumors induced by azoxymethane, Cancer Res, vol.57, pp.1233-1237, 1997.

K. Chin, Y. Kurashima, T. Ogura, H. Tajiri, S. Yoshida et al., Induction of vascular endothelial growth factor by nitric oxide in human glioblastoma and hepatocellular carcinoma cells, Oncogene, 1997.

N. V. Blough and O. C. Zafiriou, Reaction of superoxide with nitric oxide to form peroxynitrite in alkaline aqueous solution, Inorganic Chemistry, 1995.

U. K. Messmer, M. Ankarcrona, P. Nicotera, and B. Brune, p53 expression in nitric oxide-induced apoptosis, FEBS Lett, issue.355, pp.23-29, 1994.

C. Szabo and H. Ohshima, DNA damage induced by peroxynitrite: subsequent biological effects, Nitric Oxide, issue.1, pp.373-385, 1997.

S. Kilaru, S. G. Frangos, A. H. Chen, D. Gottler, A. Dhadwal et al., Nicotine : A review of its role in atherosclerosis, Journal of the American College of Surgeons, 2001.

R. Olszanecki, J. Jawien, M. Gajda, L. Mateuszuk, A. Gebska et al., Effect of curcumin on atherosclerosis in apoE/LDLR-double knockout mice, J Physiol Pharmacol, vol.56, issue.4, pp.627-635, 2005.

R. Srivastava, M. Dikshit, R. C. Srimal, and B. N. Dhawan, Anti-thrombotic effect of curcumin, Thromb Res, vol.40, issue.3, pp.413-417, 1985.

N. Toda, T. Okamura, I. Shimizu, and Y. Tatsuno, Postmortem functional changes in coronary and cerebral arteries from humans and monkeys, Cardiovasc Res, vol.11, pp.707-720, 1985.

R. B. Arora, S. K. Gupta, R. C. Sharma, and H. H. Siddiqui, Isolation and characterization of a sodium retaining substance from pig heart muscle and its role in myocardial infarction, Indian J Med Res, vol.59, issue.3, pp.483-93, 1971.

S. Chandra, S. K. Mukherjee, and N. Sethi, Effect of argemone oil feeding on blood biochemistry and tissue changes in albino rats, Indian J Med Sci, vol.26, issue.5, pp.308-320, 1972.

R. L. Saul, P. Gee, and B. N. Ames, Free radicals, DNA damage and aging, Modern Biological Theories of Aging, 1987.

D. Harman, Free radical theory of aging: Effect of free radical inhibitors on the mortality rate of male LAP mice, J Gerontol, issue.23, pp.476-482, 1968.

D. Harman, The aging process, Proc Natl Acad Sci, issue.78, pp.124-128, 1981.

G. B. Sijitlal, P. Chitra, and G. Chandrakasan, Effect of curcumin on the advanced glycation and crosslinking of collagen in diabetic rats, Biochem Pharmacol J, vol.56, issue.12, pp.1067-1074, 1998.

C. A. Janeway, The immune system evolved to discriminate infectious nonself from noninfectious self, Immunology Today, p.11, 1992.

H. C. Huang, T. R. Jan, and S. F. Yeh, Inhibitory effect of curcumin, an antiinflammatory agent, on vascular smooth muscle cell proliferation, Eur J Pharmacol, issue.221, pp.381-385, 1992.

M. Kitagawa, H. Mitsui, H. Nakamura, S. Yoshino, S. Miyakawa et al., Differential regulation of rheumatoid synovial cell interleukin-12 production by tumor necrosis factor alpha and CD 40 signals, Arthritis Rheumatol, issue.42, pp.1917-1926, 1999.

B. Bosman, Testing of lipoxygenase inhibitors, cycloxigenase inhibitors, drugs with immunomodulating properties and some reference antipsoriatic drugs in the modified mouse tail test, an animal model of psoriasis, Skin Pharmacol, vol.7, pp.324-334, 1994.

M. C. Heng, M. K. Song, J. Harker, and M. K. Heng, Drug-induced suppression of phosphorylase kinase activity correlates with resolution of psoriasis as assessed by clinical, histological and immunohistochemical parameters, Br J Dermatol, issue.143, pp.937-949, 2000.

R. Rukkumani, K. Aruna, P. S. Varma, and V. P. Menon, Curcumin influences hepatic expression patterns of matrix metalloproteinases in liver toxicity, Ital J Biochem, issue.53, pp.61-67, 2004.

T. Mahesh, S. Balasubashini, M. Menon, and V. P. , Photo irradiated curcumin supplementation in streptozotocin-induced diabetic rats : Effect on lipid peroxidation, Therapie, vol.59, issue.6, pp.639-644, 2004.

A. Bierhaus, M. A. Hofmann, R. Ziegler, and P. Nawroth, AGE and other interaction with AGE-receptors in vascular disease and diabetes-I. The AGE concept, Cardiovasc Res, issue.37, pp.586-590, 1998.

G. L. King, H. Ishii, and D. Koya, Diabetic vascular dysfunction:Amodel of excessive activation of protein kinase C, Kidney Int, vol.60, pp.77-85, 1997.

J. R. Williamson, K. Chang, M. Frangos, K. S. Hasan, Y. Ido et al., Hyperglycemic pseudohypoxia and diabetic complications, Diabetes, issue.42, pp.801-814, 1993.

B. Goossens, J. Grooten, D. Vos, K. Fiers, and W. , Direct evidence for tumor necrosis factor-induced mitochondrial reactive oxygen intermediates and their involvement in cytotoxicity, Proc Natl Acad Sci, vol.92, issue.18, pp.8115-8119, 1995.

G. Boden, Role of fatty acids in the pathogenesis of insulin resistance and NIDDM, Diabetes, vol.46, issue.1, pp.3-10, 1997.

C. R. Kahn, Causes of insulin resistance, Nature, vol.373, issue.6513, pp.384-385, 1995.

R. Ross, Atherosclerosis -An inflammatory disease, N Engl J Med, issue.340, pp.115-126, 1999.

A. Tsoupras, R. Lordan, and I. Zabetakis, Inflammation, not Cholesterol, Is a Cause of Chronic Disease, Nutrients, issue.10, p.604, 2018.

X. Ma and Y. Feng, Hypercholesterolemia Tunes Hematopoietic Stem/Progenitor Cells for Inflammation and Atherosclerosis, Int J Mol Sci, issue.17, p.1162, 2016.

A. L. Catapano, A. Pirillo, and G. D. Norata, Vascular inflammation and low-density lipoproteins: Is cholesterol the link? A lesson from the clinical trials, Br J Pharmacol, issue.174, pp.3973-3985, 2017.

S. P. Weisberg, D. Mccann, M. Desai, M. Rosenbaum, R. L. Leibel et al., Obesity is associated with macrophage accumulation in adipose tissue, J Clin Invest, issue.112, pp.1796-1808, 2003.

H. Xu, G. T. Barnes, Q. Yang, G. Tan, D. Yang et al., Chronic inflammation in fat plays a crucial role in the development of obesity-related insulin resistance, J Clin Invest, issue.112, pp.1821-1830, 2003.

T. Yamauchi and T. Kadowaki, Adiponectin receptor as a key player in healthy longevity and obesityrelated diseases, Cell Metab, issue.17, pp.185-196, 2013.

S. T. Hasan, J. M. Zingg, P. Kwan, T. Noble, D. Smith et al., Curcumin modulation of high fat dietinduced atherosclerosis and steatohepatosis in LDL receptor deficient mice, Atherosclerosis, issue.232, pp.40-51, 2014.

J. M. Zingg, S. T. Hasan, and M. Meydani, Molecular mechanisms of hypolipidemic effects of curcumin, Biofactors, issue.39, pp.101-121, 2013.

A. Pongchaidecha, N. Lailerd, W. Boonprasert, and N. Chattipakorn, Effects of curcuminoid supplement on cardiac autonomic status in high-fat-induced obese rats, Nutrition, vol.25, pp.870-878, 2009.

X. Peng, C. Dai, Q. Liu, L. J. Qiu, and J. , Curcumin Attenuates on Carbon Tetrachloride-Induced Acute Liver Injury in Mice via Modulation of the Nrf2/HO-1 and TGF-1/Smad3 Pathway, Molecules, issue.23, p.215, 2018.

X. Cui, H. Song, and J. Su, Curcumin attenuates hypoxic-ischemic brain injury in neonatal rats through induction of nuclear factor erythroid-2-related factor 2 and heme oxygenase-1, Exp Ther Med, issue.14, pp.1512-1518, 2017.

T. Morita, T. Imai, T. Sugiyama, S. Katayama, and G. Yoshino, Heme oxygenase-1 in vascular smooth muscle cells counteracts cardiovascular damage induced by angiotensin II, Curr Neurovasc Res, issue.2, pp.113-120, 2005.

S. P. Weisberg, R. Leibel, and D. V. Tortoriello, Dietary curcumin significantly improves obesity-associated inflammation and diabetes in mouse models of diabesity, Endocrinology, issue.149, pp.3549-3558, 2008.

M. D. Maines, Heme oxygenase: Function, multiplicity, regulatory mechanisms, and clinical applications, FASEB J, issue.2, pp.2557-2568, 1988.

W. Luo, Y. Wang, H. Yang, C. Dai, H. H. Li et al., Heme oxygenase-1 ameliorates oxidative stress-induced endothelial senescence via regulating endothelial nitric oxide synthase activation and coupling, Aging, issue.10, pp.1722-1744, 2018.

S. K. Shin, T. Y. Ha, R. A. Mcgrego, and M. S. Choi, Long-term curcumin administration protects against atherosclerosis via hepatic regulation of lipoprotein cholesterol metabolism, Mol Nutr Food Res, issue.55, pp.1829-1840, 2011.

J. M. Zingg, S. T. Hasan, K. Nakagawa, E. Canepa, R. Ricciarelli et al., Modulation of cAMP levels by high-fat diet and curcumin and regulatory effects on CD36/FAT scavenger receptor/fatty acids transporter gene expression, Biofactors, issue.43, pp.42-53, 2017.

Q. Zhang, Z. Wang, and H. Chen, Endothelium-specific overexpression of class III deacetylase SIRT1 decreases atherosclerosis in apolipoprotein E-deficient mice, Cardiovasc Res, issue.80, pp.191-199, 2008.

M. Notarbartolo, P. Poma, D. Perri, L. Dusonchet, M. Cervello et al., Antitumor effects of curcumin, alone or in combination with cisplatin or doxorubicin, on human hepatic cancer cells. Analysis of their possible relationship to changes in NF-kB activation levels and in IAP gene expression, Cancer Lett, issue.53, p.224, 2005.

B. Hill, Drug resistance, and overview of the current state of the art, Int J Oncol, issue.9, p.197, 1996.

P. S. Lacombe, J. Vicente, J. G. Pages, and P. L. Morselli, Causes and problems of nonresponse or poor response to drugs, Drugs, issue.51, p.552, 1996.

M. Dietel, What's new in cytostatic drug resistance and pathology, Pathol Res Pract J, issue.187, p.892, 1991.

W. T. Beck, Mechanisms of multidrug resistance in human tumor cells. The roles of Pglycoprotein,DNAtopoisomerase II, and other factors, Cancer Treat Rev, issue.17, p.11, 1990.

C. S. Morrow and K. H. Cowan, Glutathione S-transferases and drug resistance, Cancer Cells, vol.15, issue.2, 1990.

J. R. Hammond, R. M. Johnstone, and P. Gros, Enhanced efflux of [3H]vinblastine from Chinese hamster ovary cells transfected with a full-length complementaryDNA clone for the mdr1 gene, Cancer Res, issue.49, p.3867, 1989.

Y. A. Hannun, Apoptosis and the dilemma of cancer chemotherapy, Blood, p.1845, 1997.

Y. Y. Liu, T. Y. Han, A. E. Giuliano, and M. C. Cabot, Ceramide glycosylation potentiates cellular multidrug resistance, FASEB J, issue.15, p.719, 2001.

Z. E. Sauna, M. M. Smith, M. Muller, K. M. Kerr, and S. V. Ambudkar, The mechanism of action of multidrugresistance-linked P-glycoprotein, J Bioenerg Biomembr, issue.33, p.481, 2001.

T. Litman, T. E. Druley, W. D. Stein, and S. E. Bates, From MDR to MXR : New understanding of multidrug resistance systems, their properties and clinical significance, Cell Mol Life Sci, issue.58, p.931, 2001.

H. A. Bardelmeijer, J. H. Beijnen, K. R. Brouwer, H. Rosing, W. J. Nooijen et al., Increased oral bioavailability of paclitaxel by GF120918 in mice through selective modulation of Pglycoprotein, Clin Cancer Res, issue.6, p.4416, 2000.

L. J. Green, P. Marder, and C. A. Slapak, Modulation by LY335979 of P-glycoprotein function in multidrugresistant cell lines and human natural killer cells, Biochem Pharmacol J, p.1393, 2001.

N. Chainani-wu, Safety and anti-inflammatory activity of curcumin: A component of tumeric (Curcuma longa), J Altern Complement Med, issue.9, p.161, 2003.

A. L. Cheng, C. H. Hsu, J. K. Lin, M. M. Hsu, Y. F. Ho et al., Phase I clinical trial of curcumin, a chemopreventive agent, in patients with high-risk or pre-malignant lesions, Anticancer Res, issue.21, p.2895, 2001.

R. L. Juliano and V. Ling, A surface glycoprotein modulating drug permeability in Chinese hamster ovary cell mutants, Biochim Biophys Acta, issue.455, p.152, 1976.

D. Nielson, C. Maare, and T. Skovsgaard, Influx of daunorubicin in multidrug resistance Erlich ascites tumor cells, correlation to expression of P-glycoprotein and efflux. Influence of verapamil, Biochem Pharmacol J, issue.50, p.443, 1995.

A. R. Safa, Photoaffinity labeling of p-glycoprotein in multidrug resistance cells, Cancer Invest, issue.10, p.295, 1992.

R. G. Deeley and S. P. Cole, Function, evolution and structure of multidrug resistance protein (MRP), Semin Cancer Biol, issue.8, p.193, 1997.

P. Gros and C. Shustik, Multidrug resistance: A novel class of membrane-associated transport proteins is identified, Cancer Invest, issue.9, p.563, 1991.

M. M. Cornwell, I. Pastan, and M. M. Gottesman, Certain calcium channel blockers bind specifically to multidrug-resistant humanKBcarcinoma membrane vesicles and inhibit drug binding to Pglycoprotein, J Biol Chem, issue.262, p.2166, 1987.

T. W. Loo and D. M. Clarke, Functional consequences of glycine mutations in the predicted cytoplasmic loops of P-glycoprotein, J Biol Chem, issue.269, p.7243, 1994.

F. J. Sharom, X. Yu, and C. A. Doige, Functional reconstitution of drug transportand ATPase activity in proteoliposomes containing partially purified P-glycoprotein, J Biol Chem, issue.268, p.197, 1993.

K. Ueda, A. Yoshida, and T. Amachi, Recent progress in P-glycoprotein research, Anticancer Drug Des, issue.14, p.115, 1999.

A. F. Castro, J. K. Horton, C. G. Vanoye, and G. A. Altenberg, Mechanism of inhibition of P-glycoproteinmediated drug transport by protein kinase C blockers, Biochem Pharmacol, issue.58, p.1723, 1999.

G. Conseil, J. M. Perez-victoria, J. M. Jault, F. Gamarro, A. Goffeau et al., Protein kinase C effectors bind to multidrug ABC transporters and inhibit their activity, Biochemistry, issue.40, p.2564, 2001.
URL : https://hal.archives-ouvertes.fr/hal-00313619

A. C. Lockhart, R. G. Tirona, and R. B. Kim, Pharmacogenetics of ATP-binding cassette transporters in cancer and chemotherapy, Mol Cancer Ther, issue.2, p.685, 2003.

H. M. Pinedo, G. Giaccone, and . P-glycoprotein, A marker of cancer-cell behavior, N Engl J Med, issue.333, p.1417, 1995.

J. Renes, E. G. De-vries, E. F. Nienhuis, P. L. Jansen, and M. Muller, ATP and glutathione-dependent transport of chemotherapeutic drugs by the multidrug resistance protein MRP1, Br J Pharmacol, issue.126, p.681, 1999.

K. Barnouin, I. Leier, G. Jedlitschky, A. Pourtier-manzanedo, J. Konig et al., Multidrug resistance protein-mediated transport of chlorambucil and melphalan conjugated to glutathione, Br J Cancer, issue.77, p.201, 1998.

W. Chearwae, C. P. Wu, H. Y. Chu, T. R. Lee, S. V. Ambudkar et al., Curcuminoids purified from turmeric powder modulate the function of human multidrug resistance protein 1 (ABCC1)

, Cancer Chemother Pharmacol, issue.57, p.376, 2006.

S. E. Chuang, P. Y. Yeh, Y. S. Lu, G. M. Lai, C. M. Liao et al., Basal levels and patterns of anticancer drug-induced activation of nuclear factor-?B (NF-?B), and its attenuation by tamoxifen, dexamethasone, and curcumin in carcinoma cells, Biochem Pharmacol, issue.63, p.1709, 2002.

S. Aggarwal, H. Ichikawa, Y. Takada, S. K. Sandur, S. Shishodia et al., Curcumin (diferuloylmethane) down-regulates expression of cell proliferation and antiapoptotic and metastatic gene products through suppression of I?B? kinase and Akt activation, Mol Pharmacol, p.195, 2006.

S. V. Bava, V. T. Puliappadamba, A. Deepti, A. Nair, D. Karunagaran et al., Sensitization of taxolinduced apoptosis by curcumin involves down-regulation of nuclear factor-kappaB and the serine/threonine kinase Akt and is independent of tubulin polymerization, J Biol Chem, issue.280, p.6301, 2005.

V. Sundram, S. Chauhan, and M. Jaggi, Emerging Roles of Protein Kinase D1 in Cancer, Mol Cancer Res, issue.9, pp.985-996, 2011.

M. Jaggi, C. Du, W. Zhang, and K. Balaji, Protein kinase D1 : a protein of emerging translational interest, Front Biosci, issue.12, pp.3757-3767, 2007.

E. Rozengurt, O. Rey, and R. Waldron, Protein kinase D signaling, J Biol Chem, issue.280, pp.13205-13208, 2005.

J. Van-lint, A. Rykx, Y. Maeda, T. Vantus, and S. Sturany, Protein kinase D : an intracellular traffic regulator on the move, Trends Cell Biol, issue.12, pp.193-200, 2002.

A. Rykx, L. De-kimpe, S. Mikhalap, T. Vantus, and T. Seufferlein, Protein kinase D : a family affair, FEBS Lett, issue.546, pp.81-86, 2003.

S. Guha, S. Tanasanvimon, J. Sinnett-smith, and E. Rozengurt, Role of protein kinase D signaling in pancreatic cancer, Biochem Pharmacol J, pp.1946-1954, 2010.

L. Valle, C. George, K. Sharlow, E. Lazo, J. Wipf et al., Protein kinase D as a potential new target for cancer therapy, Biochim Biophys Acta, pp.183-192, 1806.

A. Valverde, J. Sinnett-smith, J. Van-lint, and E. Rozengurt, Molecular cloning and characterization of protein kinase D: a target for diacylglycerol and phorbol esters with a distinctive catalytic domain, Proc Natl Acad Sci, issue.91, pp.8572-8576, 1994.

A. Su, T. Wiltshire, S. Batalov, H. Lapp, and K. Ching, A gene atlas of the mouse and human proteinencoding transcriptomes, Proc Natl Acad Sci, issue.101, pp.6062-6067, 2004.

M. Jaggi, S. Chauhan, C. Du, and K. Balaji, Bryostatin 1 modulates betacatenin subcellular localization and transcription activity through protein kinase D1 activation, Mol Cancer Ther, issue.7, pp.2703-2712, 2008.

M. Biswas, C. Du, C. Zhang, J. Straubhaar, and L. Languino, Protein kinase D1 inhibits cell proliferation through matrix metalloproteinase-2 and matrix metalloproteinase-9 secretion in prostate cancer, Cancer Res, issue.70, pp.2095-2104, 2010.

T. Eiseler, H. Doppler, I. Yan, S. Goodison, and P. Storz, Protein kinase D1 regulates matrix metalloproteinase expression and inhibits breast cancer cell invasion, Breast Cancer Res, issue.11, 2009.

T. Dorai, Y. Cao, B. Dorai, R. Buttyan, and A. Katz, Therapeutic potential of curcumin in human prostate cancer. III. Curcumin inhibits proliferation, induces apoptosis, and inhibits angiogenesis of LNCaP prostate cancer cells in vivo, Prostate, issue.47, pp.293-303, 2001.

T. Dorai, N. Gehani, and A. Katz, Therapeutic potential of curcumin in human prostate cancer-I. curcumin induces apoptosis in both androgen-dependent and androgen-independent prostate cancer cells, Prostate Cancer Prostatic Dis, issue.3, pp.84-93, 2000.

T. Dorai, N. Gehani, and A. Katz, Therapeutic potential of curcumin in human prostate cancer. II. Curcumin inhibits tyrosine kinase activity of epidermal growth factor receptor and depletes the protein, Mol Urol, issue.4, pp.1-6, 2000.

J. Hardy, Amyloid, the presenilins and Alzheimer's disease, Trends Neurosci, vol.20, pp.154-159, 1997.

D. J. Selkoe, Alzheimer's disease: Genotypes, phenotypes, and treatments, Science, vol.275, pp.630-631, 1997.

D. Games, D. Adams, R. Alessandrini, R. Barbour, P. Berthelette et al., Alzheimertype neuropathology in transgenic mice overexpressing V717F b-amyloid precursor protein, Nature, issue.373, pp.523-527, 1995.

K. Hsiao, P. Chapman, S. Nilsen, C. Eckman, Y. Harigaya et al., Correlative memory deficits, Ab elevation and amyloid plaques in transgenic mice, Science, issue.274, pp.99-102, 1996.

G. P. Lim, T. Chu, F. Yang, W. Beech, S. A. Frautschy et al., The curry spice curcumin reduces oxidative damage and amyloid pathology in an Alzheimer transgenic mouse, J Neurosci, issue.21, pp.8370-8377, 2001.

S. A. Frautschy, W. Hu, S. A. Miller, P. Kim, M. E. Harris-white et al., Phenolic anti-inflammatory antioxidant reversal of A?-induced cognitive deficits and neuropathology, Neurobiol Aging, issue.22, pp.991-1003, 2001.

G. M. Cole, F. Yang, G. P. Lim, J. L. Cummings, D. L. Masterman et al., A rationale for curcuminoids for the prevention or treatment of Alzheimer's disease, Immunol Endocr Metab Agents, issue.3, pp.15-25, 2003.

F. Yang, G. P. Lim, A. N. Begum, O. J. Ubeda, M. R. Simmons et al., Curcumin inhibits formation of amyloid beta oligomers and fibrils, binds plaques, and reduces amyloid in vivo, J Biol Chem, issue.280, pp.5892-5901, 2005.

X. Huang, C. S. Atwood, R. D. Moir, M. A. Hartshorn, R. E. Tanzi et al., Trace metal contamination initiates the apparent auto-aggregation, amyloidosis, and oligomerization of Alzheimer's Abeta peptides, J Biol Inorg Chem, issue.9, pp.954-960, 2004.

K. Fassbender, M. Simons, C. Bergmann, M. Stroick, D. Lutjohann et al., Simvastatin strongly reduces levels of Alzheimer's disease beta -amyloid peptides Abeta 42 and Abeta 40 in vitro and in vivo, Proc Natl Acad Sci, issue.98, pp.5856-5861, 2001.

L. M. Refolo, M. A. Pappola, J. Lafrancois, B. Malester, S. D. Schmidt et al., A cholesterollowering drug reduces ?-amyloid pathology in a transgenic mouse model of Alzheimer's disease, Neurobiol Dis, issue.8, pp.890-899, 2001.

M. Sastre, I. Dewachter, G. E. Landreth, T. M. Willson, T. Klockgether et al., Nonsteroidal anti-inflammatory drugs and peroxisome proliferatoractivated-receptor-gamma agonists modulate immunostimulated processing of amyloid precursor protein through regulation of beta-secretase, J Neurosci, issue.23, pp.9796-9804, 2003.

M. Tabaton, Oxidative stress and beta-APP proteolytic processing, Neurobiol Aging, issue.25, 2004.

E. Tamagno, M. Parola, P. Bardini, A. Piccini, R. Borhi et al., Beta-site APP cleaving enzyme up-regulation induced by 4-hydroxynonenal is mediated by stress-activated protein kinasses pathways, J Neurochem, issue.92, pp.628-636, 2005.

S. Sung, Y. Yao, K. Uryu, H. Yang, V. M. Lee et al., Early vitamin E supplementation in young but not aged mice reduces Abeta levels and amyloid deposition in a transgenic model of Alzheimer's disease, FASEB J, issue.18, pp.323-325, 2004.

L. Baum and A. Ng, Curcumin interaction with copper and iron suggests one possible mechanism of action in Alzheimer's disease animal models, J Alzheimers Dis, issue.6, pp.367-377, 2004.

K. B. Suni and R. Kuttan, Effect of oral curcumin administration on serum peroxides and cholesterol levels in human volunteers, Indian J Physiol Pharmacol, issue.36, pp.273-275, 1992.

P. Venkatesanand, Structure-activity relationships for the inhibition of lipid peroxidation and the scavenging of free radicals by synthetic symmetrical curcumin analogues, J Pharm Pharmacol, issue.52, pp.1123-1128, 2000.

D. Peschel, R. Koerting, and N. Nass, Curcumin induces changes in expression of genes involved in cholesterol homeostasis, J Nutr Biochem, 2006.

C. J. Cummings, Y. Sun, P. Opal, B. Antalffy, R. Mestril et al., Over-expression of inducible HSP70 chaperone suppresses neuropathology and improves motor function in SCA1 mice, Hum Mol Genet, issue.10, pp.1511-1518, 2001.

K. Kato, H. Ito, K. Kamei, and I. Iwamoto, Stimulation of the stress-induced expression of stress proteins by curcumin in cultured cells and in rat tissues in vivo, Cell Stress Chaperones, issue.3, pp.152-160, 1998.

V. Chandra, R. Pandav, H. H. Dodge, J. M. Johnston, S. H. Belle et al., Incidence of Alzheimer's disease in a rural community in India, the Indo-US study, Neurology, issue.57, pp.985-989, 2001.

R. J. Mehlhornand and G. M. Cole, The free radical theory of aging : A critical review, Adv Free Radic Biol Med, issue.1, pp.165-223, 1985.

Y. Zhou, G. Gu, D. R. Goodlett, T. Zhang, C. Pan et al., Analysis of alpha -synucleinassociated proteins by quantitative proteomics, J Biol Chem, issue.279, pp.39155-39164, 2004.

J. S. Hong, Role of inflammation in the pathogenesis of Parkinson's disease: Models, mechanisms, and therapeutic interventions, Ann N Y Acad Sci, pp.151-152, 1053.

T. Takahashi, H. Yamashita, T. Nakamura, Y. Nagano, and S. Nakamura, Tyrosine 125 of alphasynuclein plays a critical role for dimerization following nitrative stress, Brain Res, issue.938, pp.73-80, 2002.

K. Onoand and M. Yamada, Antioxidant compounds have potent anti-fibrillogenic and fibrildestabilizing effects for alpha-synuclein fibrils in vitro, J Neurochem, issue.97, pp.105-115, 2006.

N. Pandey and J. E. Galvin, Curcumin prevents aggregation of alpha-synuclein, Soc Neurosci, issue.31, 2005.

P. Kuner, R. Schubenel, and C. Hertel, Beta-amyloid binds to p57NTR and activates NFkappaB in human neuroblastoma cells, J Neurosci Res, issue.54, pp.798-804, 1998.

K. Santacruz, J. Lewis, T. Spires, J. Paulson, and L. Kotilinek, Tau suppression in a neurodegenerative mouse model improves memory function, Science, issue.309, pp.476-481, 2005.

K. Bhattacharya, K. B. Rank, B. D. Evans, and S. K. Sharma, Role of cysteine-291 and cysteine-322 in the polymerization of human tau into Alzheimer-like filaments, Biochem Biophys Res Commun, issue.285, pp.20-26, 2001.

B. E. Dwyer, A. K. Raina, G. Perry, and M. A. Smith, Homocysteine and Alzheimer's disease: A modifiable risk?, Free Radic Biol Med, issue.36, pp.1471-1475, 2004.

G. Ramaswami, H. Chai, Q. Yao, P. H. Lin, A. B. Lumsden et al., Curcumin blocks homocysteineinduced endothelial dysfunction in porcine coronary arteries, J Vasc Surg, issue.40, pp.1216-1222, 2004.