I. and «. Maladie, Alzheimer et démences apparentées : taux d'ALD, de patients hospitalisés et de mortalité en France métropolitaine, 2007.

J. Dyerberg and H. O. Bang, Stofferson, « Eicosapentaenoic acid and prevention of thrombosis and atherosclerosis ? », The Lance, pp.117-119, 1978.

A. Lapillonne, «. Dha, and ». Développement-du-cerveau-de-l-'enfant, DHA et d??veloppement du cerveau de l???enfant, Ol??agineux, Corps gras, Lipides, vol.14, issue.1, pp.25-27, 2007.
DOI : 10.1051/ocl.2007.0104

URL : https://doi.org/10.1051/ocl.2007.0104

M. Lavialle and S. Layé, « Acides gras poly-insaturés (oméga 3, oméga 6) et fonctionnement du système nerveux central, Innov. Agron, vol.10, pp.25-42, 2010.

J. Huertas, E. P. Alessandri, and . Guesnet, « N-3 fatty acids, neuronal activity and energy metabolism in the brain, Ol. Corps Gras Lipides, vol.19, issue.4, pp.238-244, 2012.

S. C. Cunnane, M. Plourde, F. Pifferi, M. Bégin, C. Féart et al., Fish, docosahexaenoic acid and Alzheimer???s disease, docosahexaenoic acid ans Alzheimer's disease, pp.239-256, 2009.
DOI : 10.1016/j.plipres.2009.04.001

E. Fahy, D. Cotter, M. Sud, and E. S. Subramaniam, Lipid classification, structures and tools, Lipid classification, structures and tools, pp.637-647, 2011.
DOI : 10.1016/j.bbalip.2011.06.009

URL : http://europepmc.org/articles/pmc3995129?pdf=render

R. J. Hamilton, Fatty Acids: Structure, Occurrence, Nomenclature, Biosynthesis and Properties
DOI : 10.1002/9780470697658.ch1

C. Colette and L. Monnier, Acides gras??: classification, fonction et ??quilibre entre les diff??rentes familles, Médecine Mal. Métaboliques, pp.237-245, 2011.
DOI : 10.1016/S1957-2557(11)70234-6

P. Legrand, Les Acides Gras : Structures, Fonctions, Apports Nutritionnels Conseill??s, Cahiers de Nutrition et de Di??t??tique, vol.42, pp.7-12, 2007.
DOI : 10.1016/S0007-9960(07)91234-1

C. Chenzhong and G. Shuo, Estimating enthalpies of formation of monoalkenes by the bonding orbital-connecting matrix of molecular graphics and the steric effect of the cis/trans configuration, J. Mol. Struct. THEOCHEM, vol.718, pp.153-163, 2005.

S. S. Vannieuwenhze, J. L. White, and E. A. Witztum, Dennis, « A comprehensive classification system for lipids, J. Lipid Res, vol.46, pp.839-861, 2005.

S. Eymard, Mise en évidence de l'oxydation des lipides au cours de la conservation et de la transformation du chinchard (Trachurus Trachurus) : choix de sprocédés, 2003.

M. Awada and «. L. , oxydation modifie les effets métaboliques d'acides gras polyinsaturés de la série n-3 incorporés par différents vecteurs dans des régimes hyperlipidiques : contribution de l'absorption intestinale et de la réactivité cellulaire du 4-hydroxy- hexénal, Institut National des Sciences Appliquées, 2012.

G. Barrera, S. Pizzimenti, R. Muraca, G. Barbiero, G. Bonelli et al., Effect of 4-hydroxynonenal on cell cycle progression and expression of differenciation-associated antigens in HL-60 cells », Free Radic, Biol

E. Ros, Intestinal absorption of triglyceride and cholesterol. Dietary and pharmacological inhibition to reduce cardiovascular risk, Atherosclerosis, vol.151, issue.2, pp.357-379, 2000.
DOI : 10.1016/S0021-9150(00)00456-1

P. D. Medina, M. R. Paillasse, G. Ségala, F. Khallouki, S. Brillouet et al., Importance of cholesterol and oxysterols metabolism in the pharmacology of tamoxifen and other AEBS ligands, Chemistry and Physics of Lipids, vol.164, issue.6, pp.432-437, 2011.
DOI : 10.1016/j.chemphyslip.2011.05.005

URL : https://hal.archives-ouvertes.fr/inserm-00743454

C. Magniont, G. Escadeillas, C. Oms-multon, and E. P. Cano, The benefits of incorporating glycerol carbonate into an innovative pozzolanic matrix, Cement and Concrete Research, vol.40, issue.7, pp.1072-1080, 2010.
DOI : 10.1016/j.cemconres.2010.03.009

C. Leray, « Triacylglycérols et nutrition -Absorption et digestion des lipides, Les lipides, p.69, 2013.

A. K. Agarwal and A. Garg, Congenital generalized lipodystrophy: significance of triglyceride biosynthetic pathways, Trends in Endocrinology & Metabolism, vol.14, issue.5, pp.214-221, 2003.
DOI : 10.1016/S1043-2760(03)00078-X

L. Burri, N. Hoem, and S. Banni, Marine Omega-3 Phospholipids: Metabolism and Biological Activities, International Journal of Molecular Sciences, vol.18, issue.12, pp.15401-15419, 2012.
DOI : 10.1007/s00520-009-0640-4

URL : http://www.mdpi.com/1422-0067/13/11/15401/pdf

A. Chester and «. Iupac-iub, Joint Commission on Biochemical Nomenclature - Nomenclature of glycolipids (Recommendations 1997) », Eur J Biochim, vol.257, pp.293-298, 1998.

S. Lecleire, Digestion et absorption des nutriments, Digestion et absorption des nutriments, pp.45-50, 2008.
DOI : 10.1016/S0007-9960(08)70279-7

V. Marcil, N. Peretti, E. Delvin, and E. E. Levy, Les processus digestifs et absorptifs des lipides alimentaires, Les processus digestifs et absorptifs des lipides alimentaires, pp.1257-1266, 2004.
DOI : 10.1016/S0399-8320(04)95219-0

V. Petit, I. Niot, H. Poirier, and E. P. Besnard, Absorption intestinale des??acides gras: faits et??incertitudes, Nutrition Clinique et M??tabolisme, vol.21, issue.1, pp.38-45, 2007.
DOI : 10.1016/j.nupar.2007.01.007

I. M. Atadashi, M. K. Aroua, and A. A. Aziz, Biodiesel separation and purification: A review, Renewable Energy, vol.36, issue.2, pp.437-443, 2011.
DOI : 10.1016/j.renene.2010.07.019

L. Amate, A. Gil, and E. M. Ramirez, Feeding Infant Piglets Formula with Long-Chain Polyunsaturated Fatty Acids as Triacylglycerols or Phospholipids Influences the Distribution of These Fatty Acids in Plasma Lipoprotein Fractions, The Journal of Nutrition, vol.35, issue.4, pp.1250-1255, 2001.
DOI : 10.1007/BF02536300

J. Lallès, Phosphatase alcaline intestinale??: une veille enzyme avec de nouvelles fonctions dans l???hom??ostasie intestinale et l???absorption des lipides, Cahiers de Nutrition et de Di??t??tique, vol.45, issue.6
DOI : 10.1016/j.cnd.2010.05.001

M. Lagarde, M??tabolisme des lipides bio-actifs, Métabolisme des lipides bio-actifs, pp.241-243, 2003.
DOI : 10.1016/S0369-8114(03)00076-2

N. Abumrad and C. Coburn, Ibrahimi, « Membrane proteins implicated in long-chain fatty acid uptake by mammalian cells : CD36, FATP and FABPm

C. Leray, « Triacylglycérols et nutrition -Transports sanguins, pp.70-72, 2013.

J. Dallongeville, Le m??tabolisme des lipoprot??ines, Le métabolisme des lipoprotéines, pp.55-60, 2006.
DOI : 10.1016/S0007-9960(06)70608-3

M. Beylot, Utilisation des lipides : oxydation ou stockage ?, Cahiers de Nutrition et de Di??t??tique, vol.43, issue.3, pp.131-137, 2008.
DOI : 10.1016/S0007-9960(08)73713-1

URL : http://www.ocl-journal.org/10.1051/ocl.2008.0160/pdf

H. D. Le, J. A. Meisel, V. E. De-meijer, K. M. Gura, and E. M. Puder, The essentiality of arachidonic acid and docosahexaenoic acid, The essentiality of arachidonic acid and docosahexaenoic acid, pp.165-170, 2009.
DOI : 10.1016/j.plefa.2009.05.020

M. Mimoun, « Statut en acides gras polyinsaturés et bénéfices d'une nutrithérapie à base de glycérophospholipides enrichis en acide docosahéxaénoïque chez un modèle murin de mucoviscidose

J. T. Brenna, N. Salem, A. J. Sinclair, and S. C. , ??-Linolenic acid supplementation and conversion to n-3 long-chain polyunsaturated fatty acids in humans, Prostaglandins, Leukotrienes and Essential Fatty Acids, vol.80, issue.2-3, pp.85-91, 2009.
DOI : 10.1016/j.plefa.2009.01.004

P. Guesnet, J. Alessandri, S. Vancassel, I. Denis, and E. M. Lavialle, Acides gras om??ga 3 et fonctions c??r??brales, Acides gras omega-3 et fonctions cérébrales, pp.131-134, 2005.
DOI : 10.1016/j.nupar.2005.06.004

. Ijlst, Disorders of mitochondria fatty acyl-CoA ?-oxidation, J. Inherit. Metab. Dis, vol.22, pp.442-487, 1999.

J. Vockley and D. A. Whiteman, Defects of mitochondrial ??-oxidation: a growing group of disorders, Neuromuscular Disorders, vol.12, issue.3, pp.235-246, 2002.
DOI : 10.1016/S0960-8966(01)00308-X

O. Fischer, C. Ouerfelli, K. K. Nathan, and . Rhee, Activity-Based Metabolomic Profiling of Enzymatic Function: Identification of Rv1248c as a Mycobacterial 2-Hydroxy-3- oxoadipate Synthase, Chem. Biol, vol.17, pp.323-332, 2010.

H. Mu and T. Porsgaard, The metabolism of structured triacylglycerols, Progress in Lipid Research, vol.44, issue.6, pp.430-448, 2005.
DOI : 10.1016/j.plipres.2005.09.002

C. Leray, « Triacylglycérols et nutrition -Importance de la structure des triacylglycérols, Les lipides, pp.117-119, 2013.

R. W. Mitchell, N. H. On, M. R. Del-bigio, D. W. Miller, and G. M. Hatch, Fatty acid transport protein expression in human brain and potential role in fatty acid transport across human brain microvessel endothelial cells, Journal of Neurochemistry, vol.130, issue.5-6, pp.735-746, 2011.
DOI : 10.1016/j.molbrainres.2004.06.042

R. W. Mitchell and G. M. Hatch, Fatty acid transport into the brain: Of fatty acid fables and lipid tails, Prostaglandins, Leukotrienes and Essential Fatty Acids (PLEFA), vol.85, issue.5, pp.293-302, 2011.
DOI : 10.1016/j.plefa.2011.04.007

J. A. Hamilton, « Fatty acid transport : difficult or easy, J. Lipid Res, vol.39, pp.467-481, 1998.

E. F. Bazinet and . Calon, « Diffusion of docosahexaenoic and eicosapentaenoic acid through the blood-brain barrier : An in situ cerebral perfusion study, Neurochem. Int, vol.55, pp.476-482, 2009.

R. W. Schwenk, G. P. Holloway, J. J. Luiken, A. Bonen, and J. F. Glatz, Fatty acid transport across the cell membrane: Regulation by fatty acid transporters, Prostaglandins, Leukotrienes and Essential Fatty Acids (PLEFA), vol.82, issue.4-6, pp.149-154, 2010.
DOI : 10.1016/j.plefa.2010.02.029

F. Kamp and J. A. Hamilton, How fatty acids of different chain length enter and leave cells by free diffusion, Prostaglandins, Leukotrienes and Essential Fatty Acids, vol.75, issue.3, pp.149-159, 2006.
DOI : 10.1016/j.plefa.2006.05.003

M. Linder, N. Belhaj, P. Sautot, and E. A. , From Krill to??Whale: an??overview of??marine fatty acids and??lipid compositions, Ol??agineux, Corps gras, Lipides, vol.103, issue.4, pp.194-204, 2010.
DOI : 10.1016/j.foodchem.2006.09.013

G. Barnathan, Sources connues et potentielles de DHA pour les besoins de l'homme », Ol. Corps Gras Lipides, pp.35-47, 2007.
DOI : 10.1051/ocl.2007.0096

URL : https://doi.org/10.1051/ocl.2007.0096

M. A. Hossain, « Fish as source of n-3 polyunsaturated fatty acids (PUFAs), which one is better -farmed or wild ? », Adv, J. Food Sci. Technol, vol.3, issue.6, pp.455-466, 2011.

S. Zlatanos and K. Laskaridis, Seasonal variation in the fatty acid composition of three Mediterranean fish ??? sardine (Sardina pilchardus), anchovy (Engraulis encrasicholus) and picarel (Spicara smaris), Food Chemistry, vol.103, issue.3, pp.725-728, 2007.
DOI : 10.1016/j.foodchem.2006.09.013

N. Combe and C. , Boué-Vaysse, « Face aux besoins et à la réalité des consommations, quelles sont les spécificités des différentes sources d'acides gras oméga 3
DOI : 10.1051/ocl.2004.0103

URL : https://www.ocl-journal.org/articles/ocl/pdf/2004/02/ocl2004112p103.pdf

N. Combe and C. Boué, « Apports alimentaires en acides linoléique at alpha-linolénique d'une population d'Aquitaine », Ol. Corps Gras Lipides, pp.118-121, 2001.
DOI : 10.1051/ocl.2001.0118

URL : http://www.ocl-journal.org/articles/ocl/pdf/2001/02/ocl200182p118.pdf

B. J. Meyer, N. J. Mann, J. L. Lewis, G. C. Milligan, A. J. Sinclair et al., Dietary intakes and food sources of omega-6 and omega-3 polyunsaturated fatty acids, Lipids, vol.35, issue.Suppl. 1, pp.391-398, 2003.
DOI : 10.1007/978-1-4615-2071-9

S. Derbré and . Médicaments, M??dicaments, compl??ments alimentaires, alicaments ou nutraceutiques, comment y voir clair ?, Actualit??s Pharmaceutiques, vol.49, issue.496, pp.14-19, 2010.
DOI : 10.1016/S0515-3700(10)70693-5

A. and «. Avis, Agence Française de Sécurité Sanitaire des Aliments sur l'estimation des apports en acides gras trans de la population française, 2009.

«. Afssa, Acides gras de la fammile oméga 3 et système cardiovasculaire : intérêts nutritionnels et allégations, 2001.

P. Astorg, Apports en acides gras polyinsatur??s, notamment en DHA, dans la population fran??aise adulte : donn??es issues de l?????tude SU.VI.MAX et comparaisons avec d???autres ??tudes, Ol??agineux, Corps gras, Lipides, vol.14, issue.1, pp.28-34, 2007.
DOI : 10.1051/ocl.2007.0095

URL : https://doi.org/10.1051/ocl.2007.0095

«. Efsa, Scientific opinion on dietary reference values for fats, including saturated fatty acids, polyunsaturated fatty acids, monounsaturated fatty acids, trans fatty acids and chlesterol »

A. P. Simopoulos, « Omega-6/omega-3 essantial fatty acid ratio and chronic diseases », Food Rev, Int, vol.20, issue.1, pp.77-90, 2004.

A. P. Simopoulos, The omega-6/omega-3 fatty acid ratio: health implications, Ol??agineux, Corps gras, Lipides, vol.17, issue.5, pp.267-275, 2010.
DOI : 10.1158/1055-9965.EPI-08-0439

J. Dallongeville and N. Marecaux, Acides gras om??ga-3 et??risque cardiovasculaire, Ol??agineux, Corps gras, Lipides, vol.369, issue.4, pp.232-235, 2010.
DOI : 10.1016/S0140-6736(07)60527-3

URL : http://www.ocl-journal.org/10.1051/ocl.2010.0317/pdf

D. Kromhout and E. B. Bosschieter, De Lezenne Coulander, « The inverse relation between fish consumption and 20-year mortality from coronary heart disease

I. C. Torres, L. Mira, and C. P. Ornelas, Mélim, « Study of the effects of dietary fish intake on serum lipids an dlipoproteins in two populations with different dietary habits, Br. J. Nutr, vol.83, pp.371-379, 2000.

K. He, Y. Song, M. L. Daviglus, K. Liu, L. Van-horn et al., Accumulated Evidence on Fish Consumption and Coronary Heart Disease Mortality: A Meta-Analysis of Cohort Studies, Circulation, vol.109, issue.22, pp.2705-2711, 2004.
DOI : 10.1161/01.CIR.0000132503.19410.6B

J. L. Breslow, n???3 Fatty acids and cardiovascular disease, The American Journal of Clinical Nutrition, vol.71, issue.6, pp.1477-1482, 2006.
DOI : 10.1093/ajcn/71.1.179S

W. S. Harris, n-3 fatty acids and serum lipoproteins: animal studies, The American Journal of Clinical Nutrition, vol.65, issue.5, pp.1611-1616, 1997.
DOI : 10.1093/ajcn/65.5.1611S

URL : https://academic.oup.com/ajcn/article-pdf/65/5/1611S/23159502/1611s.pdf

U. Gogus and C. Smith, n-3 Omega fatty acids: a review of current knowledge, International Journal of Food Science & Technology, vol.102, issue.Suppl. 1
DOI : 10.1016/0005-2760(91)90246-E

L. Monnier and C. Colette, Acides gras om??ga??3 et pathologie cardiovasculaire??: la part du vrai, Médecine Mal. Métaboliques, pp.269-277, 2011.
DOI : 10.1016/S1957-2557(11)70239-5

B. H. Courtman, E. P. Strauss, and . Dorian, « Long chain n-3 polyunsaturated fatty acids reduce atrial vulnerability in a novel canine pacing model, Cardioascular Res, vol.77, pp.89-97, 2008.

J. H. Wu, R. N. Lemaitre, I. B. King, X. Song, F. Sacks et al., Association of Plasma Phospholipid Long-Chain Omega-3 Fatty Acids With Incident Atrial Fibrillation in Older Adults: The Cardiovascular Health Study, Circulation, vol.125, issue.9, pp.1084-1093, 2012.
DOI : 10.1161/CIRCULATIONAHA.111.062653

L. Frost and P. Vestergaard, n???3 Fatty acids consumed from fish and risk of atrial fibrillation or flutter: the Danish Diet, Cancer, and Health Study, The American Journal of Clinical Nutrition, vol.5, issue.1, pp.50-54, 2005.
DOI : 10.1079/PHN2002404

K. S. Broughton, C. S. Johnson, B. K. Pace, M. Liebman, and K. M. Kleppinger, Reduced asthma symptoms with n-3 fatty acid ingestion are related to 5-series leukotriene production, The American Journal of Clinical Nutrition, vol.65, issue.4, pp.1011-1017, 1997.
DOI : 10.1093/ajcn/65.4.1011

P. C. Calder, n???3 Polyunsaturated fatty acids, inflammation, and inflammatory diseases, The American Journal of Clinical Nutrition, vol.24, issue.6, pp.1505-1519, 2006.
DOI : 10.1016/j.it.2003.10.002

A. P. Simopoulos, Evolutionary aspects of??diet, the??omega-6/omega-3??ratio and??genetic variation: nutritional implications for??chronic diseases, Biomedicine & Pharmacotherapy, vol.60, issue.9, pp.502-507, 2006.
DOI : 10.1016/j.biopha.2006.07.080

L. Kremmyda, M. Vlachava, P. S. Naakes, N. D. Diaper, E. A. Miles et al., Atopy Risk in Infants and Children in Relation to Early Exposure to Fish, Oily Fish, or Long-Chain Omega-3 Fatty Acids: A Systematic Review, Clinical Reviews in Allergy & Immunology, vol.16, issue.1, pp.36-66, 2011.
DOI : 10.1183/09031936.00.16586100

J. Delarue and N. Guriec, Acides gras, insulinor??sistance, syndrome m??tabolique et diab??te de type??2, Médecine Mal. Métaboliques, pp.253-256, 2011.
DOI : 10.1016/S1957-2557(11)70236-X

T. A. Mori, D. Q. Bao, V. Burke, I. B. Puddey, G. F. Watts et al., Dietary fish as a major component of a weight-loss diet: effect on serum lipids, glucose, and insulin metabolism in overweight hypertensive subjects, The American Journal of Clinical Nutrition, vol.17, issue.5, pp.817-825, 1999.
DOI : 10.1161/01.ATV.17.2.279

G. Derosa, A. F. Cicero, E. Fogari, A. D. Angelo, A. Bonaventura et al., Effects of n-3 PUFA on insulin resistance after an oral fat load, European Journal of Lipid Science and Technology, vol.135, issue.8, pp.950-960, 2011.
DOI : 10.1016/S0002-9343(01)01114-7

«. Fowler, The effect of Metformin and intensive lifestyle intervention on the metabolic syndrome : the diabetes preention program randomized trial, Ann. Intern. Med, vol.142, issue.8, pp.611-619, 2005.

D. Faeh, K. Minehira, J. Schwarz, R. Periasami, and P. Seongsu, Toppy, « Effects of fructose overfeeding and fish oil administration on hepatic de novo lipogenesis and insulin sensitivity in healthy men, Diabetes, vol.54, 2005.

P. Bougnoux, N. Hajjaji, M. N. Ferrasson, B. Giraudeau, and C. Couet, Le Floch, « Improving outcome of chemotherapy of metastatic breast cancer by docosahexaenoic acid : a phase II trial, Br. J. Cancer, vol.101, 2009.

N. Danbara, T. Yuri, M. Tsujita-kyutoku, M. Sato, H. Senzaki et al., Tsubura, « Conjugated docosahexaenoic acid is a potent inducer af cell cycle arrest and apoptosis and inhibits growth of Colo 201 human colon cancer cells, pp.71-79, 2004.

S. Serini, E. Piccioni, and N. Merendino, Dietary polyunsaturated fatty acids as inducers of apoptosis: implications for cancer, Apoptosis, vol.13, issue.2, pp.135-152, 2009.
DOI : 10.1007/s10571-006-9064-6

H. Gleissman, J. I. Johnsen, and E. P. Kogner, Omega-3 fatty acids in cancer, the protectors of good and the killers of evil?, Experimental Cell Research, vol.316, issue.8, pp.1365-1373, 2010.
DOI : 10.1016/j.yexcr.2010.02.039

H. Gleissman, R. Yang, K. Martinod, M. Lindskog, C. N. Serhan et al., Docosahexaenoic acid metabolome in neural tumors: identification of cytotoxic intermediates, Docosahexaenoic acid in neural tumors : identification of cytotoxic intermediates, pp.906-915, 2010.
DOI : 10.1007/978-1-4020-8831-5_6

M. Lindskog, H. Gleissman, F. Ponthan, P. Kogner, and J. I. Johnsen, Neuroblastoma cell death in response to docosahexaenoic acid: Sensitization to chemotherapy and arsenic-induced oxidative stress, International Journal of Cancer, vol.139, issue.10, pp.2584-2593, 2006.
DOI : 10.1038/bjc.1997.363

P. D. Biondo, D. N. Brindley, M. B. Sawyer, and C. J. Field, The potential for treatment with dietary long-chain polyunsaturated n-3 fatty acids during chemotherapy, The Journal of Nutritional Biochemistry, vol.19, issue.12, pp.787-796, 2008.
DOI : 10.1016/j.jnutbio.2008.02.003

L. C. Chiu, K. F. Tong, and V. E. Ooi, Cytostatic and cytotoxic effects of cyclooxygenase inhibitors and their synergy with docosahexaenoic acid on the growth of human skin melanoma A-375 cells, Biomedicine & Pharmacotherapy, vol.59, pp.293-297, 2005.
DOI : 10.1016/S0753-3322(05)80049-6

X. Liu, T. Shibata, S. Hisaka, Y. Kawai, and E. T. Osawa, DHA Hydroperoxides as a Potential Inducer of Neuronal Cell Death: a Mitochondrial Dysfunction-Mediated Pathway, Journal of Clinical Biochemistry and Nutrition, vol.43, issue.1, pp.26-33, 2008.
DOI : 10.3164/jcbn.2008040

M. B. Schaefer, A. Wenzel, T. Fischer, R. C. Braun-dullaeus, F. Renner et al., Fatty acids differentially influence phosphatidylinositol 3-kinase signal transduction in endothelial cells: Impact on adhesion and apoptosis, Atherosclerosis, vol.197, issue.2, pp.630-637, 2008.
DOI : 10.1016/j.atherosclerosis.2007.09.004

L. Spencer, C. Mann, M. Metcalfe, M. Webb, C. Pollard et al., The effect of omega-3 FAs on tumour angiogenesis and their therapeutic potential, European Journal of Cancer, vol.45, issue.12, pp.2077-2086, 2009.
DOI : 10.1016/j.ejca.2009.04.026

S. Antoun, M. Merad, G. Nitenberg, P. Ruffié, and E. B. Raynard, Acide gras N-3 et cancer d??clar????: int??r??t r??el ou effet de mode???, Nutrition Clinique et M??tabolisme, vol.19, issue.3, pp.160-165, 2005.
DOI : 10.1016/j.nupar.2005.06.002

P. B. Adams, S. Lawson, A. Sanigorski, and A. J. Sinclair, Arachidonic acid to eicosapentaenoic acid ratio in blood correlates positively with clinical symptoms of depression, Lipids, vol.3, issue.1, pp.157-161, 1996.
DOI : 10.1016/0925-4439(92)90063-S

C. Féart, E. Peuchant, L. Letenneur, C. Samieri, D. Montagnier et al., Plasma eicosapentaenoic acid is inversely associated with severity of depressive symptomatology in the elderly: data from the Bordeaux sample of the Three-City Study, The American Journal of Clinical Nutrition, vol.69, issue.5, pp.1156-1162, 2008.
DOI : 10.1016/j.plefa.2003.08.020

A. L. Stoll, E. Severus, M. P. Freeman, S. Rueter, H. A. Zboyan et al., Omega 3 Fatty Acids in Bipolar Disorder, Archives of General Psychiatry, vol.56, issue.5, pp.407-412, 1999.
DOI : 10.1001/archpsyc.56.5.407

B. Nemets, Z. Staht, and R. H. Belmaker, Addition of omega-3 fatty acid to maintenant medication treatment for recurrent unipolar depressive disorder, Am. Psychiatr. Publ, vol.159, issue.3, pp.447-479, 2002.

E. Sublette, S. P. Ellis, A. L. Geant, J. J. Mann, and . Meta, Meta-Analysis of the Effects of Eicosapentaenoic Acid (EPA) in Clinical Trials in Depression, The Journal of Clinical Psychiatry, vol.72, issue.12, pp.12-1577, 2011.
DOI : 10.4088/JCP.10m06634

J. Sontrop and M. K. Campbell, ??-3 polyunsaturated fatty acids and depression: A review of the evidence and a methodological critique, Preventive Medicine, vol.42, issue.1, pp.4-13, 2006.
DOI : 10.1016/j.ypmed.2005.11.005

C. M. Da-rocha and G. Kac, High dietary ratio of omega-6 to omega-3 polyunsaturated acids during pregnancy and prevalence of post-partum depression, Maternal & Child Nutrition, vol.159, issue.1, pp.36-48, 2012.
DOI : 10.1093/aje/kwh090

C. Leray, « Santé mentale : troubles de l'humeur, Les lipides, pp.236-249, 2013.

M. Maes, R. Smith, and S. Scharpe, The monocyte-T-lymphocyte hypothesis of major depression, Psychoneuroendocrinology, vol.20, issue.2, pp.111-116, 1995.
DOI : 10.1016/0306-4530(94)00066-J

R. K. Mcnamara and S. E. Carlson, Role of omega-3 fatty acids in brain development and function: Potential implications for the pathogenesis and prevention of psychopathology, Prostaglandins, Leukotrienes and Essential Fatty Acids, vol.75, issue.4-5, pp.329-349, 2006.
DOI : 10.1016/j.plefa.2006.07.010

C. Lee and A. K. Hajra, Molecular Species of Diacylglycerols and Phosphoglycerides and the Postmortem Changes in the Molecular Species of Diacylglycerols in Rat Brains, Journal of Neurochemistry, vol.44, issue.2, pp.370-379, 1991.
DOI : 10.1016/S0021-9673(01)80033-9

I. Carrié, M. Clément, D. De-javel, H. Francès, and J. Bourre, « Specific phospholipid fatty acid composition of brain regions in mice : effects of n-3 polyunsaturated fatty acid deficiency and phospholipid supplementation, J. Lipid Res, vol.41, pp.465-472, 2000.

M. Lavialle, G. Champeil-potokar, I. Denis, P. Guesnet, F. Pifferi et al., Le DHA dans la neurotransmission, Ol??agineux, Corps gras, Lipides, vol.14, issue.1, pp.11-15, 2007.
DOI : 10.1051/ocl.2007.0099

URL : https://doi.org/10.1051/ocl.2007.0099

A. J. Borgdorff and D. Choquet, Regulation of AMPA receptor lateral movements, Nature, vol.17, issue.6889, pp.649-653, 2002.
DOI : 10.1016/S0959-4388(00)00099-4

R. Duelli and W. Kuschinsky, Brain Glucose Transporters: Relationship to Local Energy Demand, Physiology, vol.16, issue.2, pp.71-76, 2001.
DOI : 10.1016/0165-3806(95)00177-8

J. Bourre, M. Francois, A. Youyou, O. Dumont, M. Piciotti et al., The Effects of Dietary ??-Linolenic Acid on the Composition of Nerve Membranes, Enzymatic Activity, Amplitude of Electrophysiological Parameters, Resistance to Poisons and Performance of Learning Tasks in Rats, The Journal of Nutrition, vol.119, issue.12, pp.1880-1892, 1989.
DOI : 10.1093/jn/119.12.1880

F. Pifferi, F. Roux, B. Langelier, J. Alessandri, S. Vancassel et al., (n-3) Polyunsaturated Fatty Acid Deficiency Reduces the Expression of Both Isoforms of the Brain Glucose Transporter GLUT1 in Rats, The Journal of Nutrition, vol.135, issue.9, pp.2241-2246, 2005.
DOI : 10.1093/jn/135.9.2241

D. , K. Cullen, M. E. Gilroy, H. R. Irons, and M. C. Laplaca, « Synapse-to-neuron ratio is inversely related to neuronal density in mature neuronal cultures, Brain Res, vol.1359, pp.44-55, 2010.

S. C. Heinrichs, Dietary ??-3 fatty acid supplementation for optimizing neuronal structure and function, Molecular Nutrition & Food Research, vol.155, issue.4, pp.447-456, 2010.
DOI : 10.1177/026010600601800306

C. Derouesné and . La-maladie-d, Alzheimer : regards sur le présent à la lumière du passé

G. E. Berrios, Dementia during the seventeenth and eighteenth centuries: a conceptual history, Psychological Medicine, vol.9, issue.04, pp.829-837, 1987.
DOI : 10.1002/1520-6696(196510)1:43.0.CO;2-C

D. R. George, P. J. Whitehouse, and E. J. Ballenger, The Evolving Classification of Dementia: Placing the DSM-V in a Meaningful Historical and Cultural Context and Pondering the Future of ???Alzheimer???s???, Culture, Medicine, and Psychiatry, vol.24, issue.1, pp.417-435, 2011.
DOI : 10.1016/S1474-4422(05)70075-2

G. E. Berrios, Alzheimer's disease: A conceptual history, International Journal of Geriatric Psychiatry, vol.17, issue.6, pp.355-365, 1990.
DOI : 10.1001/archneurpsyc.1935.02250160162015

C. Gallez, Rapport sur la maladie d'Alzheimer et les maladies apparentées -Office parlementaire d'évaluation des politiques de santé, 2005.

«. Berg, Aphasia in senile dementia of the Alzheimer type, Ann. Neurobiol, vol.23, issue.4, pp.365-370, 1988.

M. B. Patterson, J. L. Mack, D. S. Geldmacher, and P. J. Whotehouse, Executive functions and Alzheimer's disease: problems and prospects, European Journal of Neurology, vol.5, issue.suppl., pp.5-15, 1996.
DOI : 10.1176/jnp.5.3.241

M. Pancrazi, P. Metais, and . Maladie, Alzheimer, diagnostic des troubles psychologiques et comportementaux, Presse Médicale, pp.661-666, 2005.
DOI : 10.1016/s0755-4982(05)84010-2

H. C. Chui, S. A. Lyness, E. Sobel, and L. S. Schneider, Extrapyramidal Signs and Psychiatric Symptoms Predict Faster Cognitive Decline in Alzheimer's Disease, Archives of Neurology, vol.51, issue.7, pp.676-681, 1994.
DOI : 10.1001/archneur.1994.00540190056015

J. Touchon, F. Portet-tarodo, and E. K. Ritchie, « Troubles psychocomportementaux de la maladie d'Alzheimer, Rev. Prat, vol.48, pp.17-1898, 1998.

C. Duyckaerts, M. Panchal, B. Delatour, and M. Potier, Neuropathologie morphologique et mol??culaire de la maladie d???Alzheimer, Neuropathologie morphologique et moléculaire de la maladie d'Alzheimer », pp.127-135, 2009.
DOI : 10.1016/j.pharma.2009.01.001

F. Checler and L. Buée, Donn??es fondamentales sur les pathologies amylo??de et Tau dans la maladie d???Alzheimer??: quelles perspectives th??rapeutiques???, Annales Pharmaceutiques Fran??aises, vol.67, issue.2
DOI : 10.1016/j.pharma.2009.01.002

J. A. Carson and A. J. Turner, ??-Amyloid catabolism: roles for neprilysin (NEP) and other metallopeptidases?, Journal of Neurochemistry, vol.81, issue.Suppl.1, pp.1-8, 2002.
DOI : 10.1016/0143-4179(92)90059-6

P. Delaère, C. Duyckaerts, Y. He, F. Piette, and J. J. Hauw, Subtypes and differential laminar distributions of ?A4 deposits in Alzheimer's disease: relationship with the intellectual status of 26 cases, Acta Neuropathologica, vol.85, issue.3, pp.328-335, 1991.
DOI : 10.1007/BF00305876

S. W. Suh, K. B. Jensen, M. S. Jensen, D. S. Silva, P. J. Kesslak et al., Histochemically-reactive zinc in amyloid plaques, angiopathy, and degenerating neurons of Alzheimer's diseased brains, Brain Research, vol.852, issue.2, pp.274-278, 2000.
DOI : 10.1016/S0006-8993(99)02096-X

T. Wisniewski, J. Ghiso, and E. B. Frangione, Biology of A?? Amyloid in Alzheimer's Disease, Neurobiology of Disease, vol.4, issue.5, pp.313-328, 1997.
DOI : 10.1006/nbdi.1997.0147

M. Kidd, Paired Helical Filaments in Electron Microscopy of Alzheimer's Disease, Nature, vol.86, issue.4863, pp.192-193, 1963.
DOI : 10.1113/jphysiol.1961.sp006744

L. Buée, T. Bussière, V. Buée-scherrer, A. Delacourte, and P. R. Hof, Tau protein isoforms, phosphorylation and role in neurodegenerative disorders11These authors contributed equally to this work., Brain Research Reviews, vol.33, issue.1, pp.95-130, 2000.
DOI : 10.1016/S0165-0173(00)00019-9

E. B. Parisi and . Hyman, « Neuronal loss corelates with but exceeds neurofibrillary tangles in Alzheimer's disease, Ann. Neurol, vol.41, issue.1, pp.17-24, 1997.

Y. Grignon, C. Duyckaerts, M. Bennecib, and J. J. Hauw, Cytoarchitectonic alterations in the supramarginal gyrus of late onset Alzheimer's disease, Acta Neuropathologica, vol.95, issue.4, pp.395-406, 1998.
DOI : 10.1007/s004010050816

R. T. Bartus, R. L. Dean, B. Beer, and A. S. Lippa, The cholinergic hypothesis of geriatric memory dysfunction, Science, vol.217, issue.4558, pp.408-417, 1982.
DOI : 10.1126/science.7046051

T. D. Bird, Genetic aspects of Alzheimer disease, Genetic aspects of Alzheimer disease, pp.231-239, 2008.
DOI : 10.1212/WNL.54.11.2061

M. Gatz, C. A. Reynold, L. Fratiglioni, B. Johansson, J. A. Mortimer et al., Role of Genes and Environments for Explaining Alzheimer Disease, Archives of General Psychiatry, vol.63, issue.2, pp.168-174, 2006.
DOI : 10.1001/archpsyc.63.2.168

N. Kadmiri, K. Hamzi, B. Moutawakil, I. Slassi, and E. S. Nadifi, Les aspects g??n??tiques de la maladie d???Alzheimer (Revue), Pathologie Biologie, vol.61, issue.6, pp.228-238, 2013.
DOI : 10.1016/j.patbio.2013.04.001

H. and «. Maladie, Alzheimer et maladies apparentées : diagnostic et prise en charge, Recommandations, 2011.

M. Habert, J. Horn, M. Sarazin, J. Lotterie, M. Puel et al., Brain perfusion SPECT with an automated quantitative tool can identify prodromal Alzheimer's disease among patients with mild cognitive impairment, Neurobiology of Aging, vol.32, issue.1, pp.15-23, 2011.
DOI : 10.1016/j.neurobiolaging.2009.01.013

I. Alvarez-illan, J. M. Gorriz, J. Ramirez, D. Salas-gonzalez, M. Lopez et al., Projecting independent components of SPECT images for computer aided diagnosis of Alzheimer???s disease, Pattern Recognition Letters, vol.31, issue.11, pp.1342-1347, 2010.
DOI : 10.1016/j.patrec.2010.03.004

A. Isanski, R. L. Hamilton, and S. T. Dekosky, « Post-mortem correlates of in vivo PIB- PET amyloid imaging in a typical case of Alzheimer's disease, pp.1630-1645, 2008.

D. A. Loewenstein, A. Acevedo, S. J. Czaja, and E. R. Duara, Cognitive Rehabilitation of Mildly Impaired Alzheimer Disease Patients on Cholinesterase Inhibitors, The American Journal of Geriatric Psychiatry, vol.12, issue.4
DOI : 10.1097/00019442-200407000-00007

I. Burnside and B. K. Haight, Reminiscence and life review: analysing each concept, Journal of Advanced Nursing, vol.15, issue.4, pp.855-862, 1992.
DOI : 10.1111/j.2044-8260.1987.tb00736.x

E. P. Games and . Seubert, « Immunization with amyloid-? attenuates Alzheimer-disease-like pathology in the PDAPP mouse, Nature, vol.400, pp.173-177, 1999.

C. Wischik and R. Staff, « Challenges in the conduction of disease-modifying trials in AD : pratical experience from a phase 2 trial of tau-aggregation inhibitor therapy, J. Nutr, vol.13, issue.4, pp.367-369, 2009.

S. C. Cunnane, R. Chouinard-watkins, C. A. Castellano, P. Barberger, and . Gateau, Docosahexaenoic acid homeostasis, brain aging and Alzheimer's disease: Can we reconcile the evidence?, Prostaglandins, Leukotrienes and Essential Fatty Acids (PLEFA), vol.88, issue.1, pp.61-70, 2013.
DOI : 10.1016/j.plefa.2012.04.006

T. Fraser, H. Tayler, and E. S. Love, Fatty Acid Composition of Frontal, Temporal and Parietal Neocortex in the Normal Human Brain and in Alzheimer???s Disease, Neurochemical Research, vol.26, issue.Pt 3, pp.503-513, 2010.
DOI : 10.1016/j.bbalip.2005.11.011

E. Pifferi and S. C. Freemantle, Cunnane, « Higher plasma n-3 fatty acid status in the moderately healthy elderly in southern Québec : higher fish intake or aging-related change in n-3 fatty acid metabolism ?, Prostaglandins Leukot. Essent. Fatty Acids, vol.82, pp.277-280, 2010.

M. Plourde, R. Chouinard-watkin, M. Vandal, Y. Zhang, P. Lawrence et al., Cunnane, « Plasma incorporation, apparent retroconversion and ?-oxidation of 13C-docosahexaenoic acid in the elderly, Nutr. Metab, vol.8, issue.5, 2011.

M. C. Burke, M. S. Carlson, and . Carlson, « Benefits of fatty fish on dementia risk are stronger for those without APOE ?4, Neurology, vol.65, pp.1409-1414, 2005.

M. Plourde, M. Vohl, M. Vandal, P. Couture, S. Lemieux et al., Plasma n-3 fatty acid response to an n-3 fatty acid supplement is modulated by apoE 4 but not by the common PPAR-?? L162V polymorphism in men, British Journal of Nutrition, vol.31, issue.08, pp.1121-1124, 2009.
DOI : 10.1159/000137555

U. Mccloy, M. A. Ryan, P. B. Pencharz, R. J. Ross, and S. C. , Cunnane, « A comparison of the metabolism of eighteen-carbon 13C-unsaturated fatty acids in healthy women, J

S. Corsi, M. P. Bandinelli, E. L. Mattson, and . Ferrucci, « Low plasma n-3 fatty acids and dementia in older persons : the InCHIANTI study, J. Gerontol, vol.62, issue.10, pp.1120-1126, 2007.

M. A. Beydoun, J. S. Kaufman, J. A. Satia, W. Rosamond, and A. R. Folsom, Plasma n???3 fatty acids and the risk of cognitive decline in older adults: the Atherosclerosis Risk in Communities Study, The American Journal of Clinical Nutrition, vol.26, issue.suppl, pp.1103-1111, 2007.
DOI : 10.1016/j.neurobiolaging.2004.10.002

T. A. Sanders, A. Hinds, and C. C. Pereira, Influence of n-3 fatty acids on blood lipids in normal subjects, Journal of Internal Medicine, vol.57, issue.S731, pp.99-104, 1989.
DOI : 10.1079/BJN19870046

«. Lavialle, Glucose transport and utilization are altered in the brain of rats deficient in n- 3 polyunsaturated fatty acids, J. Neurochem, vol.81, pp.1328-1337, 2002.

P. Foley, Lipids in Alzheimer's disease: A century-old story, Biochimica et Biophysica Acta (BBA) - Molecular and Cell Biology of Lipids, vol.1801, issue.8, pp.750-753, 2010.
DOI : 10.1016/j.bbalip.2010.05.004

A. A. Farooqui, L. A. Horrocks, and E. T. Farooqui, Glycerophospholipids in brain: their metabolism, incorporation into membranes, functions, and involvement in neurological disorders, Chemistry and Physics of Lipids, vol.106, issue.1, pp.1-29, 2000.
DOI : 10.1016/S0009-3084(00)00128-6

J. W. Phillis, L. A. Horrocks, A. A. Farooqui, and . Cyclooxygenases, Cyclooxygenases, lipoxygenases, and epoxygenases in CNS: Their role and involvement in neurological disorders, CNS : their role and involvement in neurological disorders, pp.201-243, 2006.
DOI : 10.1016/j.brainresrev.2006.02.002

J. C. Demar-jr, H. Lee, K. Ma, L. Chang, J. M. Bell et al., Brain elongation of linoleic acid is a negligible source of the arachidonate in brain phospholipids of adult rats, Biochimica et Biophysica Acta (BBA) - Molecular and Cell Biology of Lipids, vol.1761, issue.9, pp.1050-1059, 2006.
DOI : 10.1016/j.bbalip.2006.06.006

A. A. Farooqui, L. A. Horrocks, and E. T. Farooqui, Modulation of inflammation in brain: a matter of fat, Journal of Neurochemistry, vol.338, issue.Suppl 1, pp.577-599, 2007.
DOI : 10.1007/978-1-4899-0179-8_47

R. P. Bazinet, Is the brain arachidonic acid cascade a common target of drugs used to manage bipolar disorder?, Biochemical Society Transactions, vol.37, issue.5, pp.1104-1109, 2009.
DOI : 10.1042/BST0371104

G. Goracci, M. Ferrini, and E. V. Nardicchi, Low Molecular Weight Phospholipases A2 in Mammalian Brain and Neural Cells: Roles in Functions and Dysfunctions, Molecular Neurobiology, vol.277, issue.R2, pp.274-289, 2010.
DOI : 10.1042/bj3190385

«. Goracci and . Group, IIa secretory phospholipase A2 (GIIA) mediates apoptotic death during NMDA receptor activation in rat primary cortical neurons, J. Neurochem, vol.112, pp.1574-1583, 2010.

K. Kishimoto, R. Li, J. Zhang, J. A. Klaus, K. K. Kibler et al., Sapirstein, « Cytosolic phospholipase A2 alpha amplifies early cyclooxygenase-2 expression, oxidative stress and MAP kinase phosphorylation after cerebral ischemia in mice, J. Neuroinflammation, vol.7, pp.42-43, 2010.

W. Dai, O. L. Lopez, O. T. Carmichael, J. T. Becker, L. H. Kuller et al., Mild Cognitive Impairment and Alzheimer Disease: Patterns of Altered Cerebral Blood Flow at MR Imaging, Radiology, vol.250, issue.3, pp.856-866, 2009.
DOI : 10.1148/radiol.2503080751

T. Murphy, A. Parikh, R. Schnaar, and E. J. Coyle, Arachidonic Acid Metabolism in Glutamate Neurotoxicity, Annals of the New York Academy of Sciences, vol.559, issue.1 Arachidonie A, pp.474-477, 1989.
DOI : 10.1016/0006-8993(88)90941-9

K. Arai, Y. Ikegaya, Y. Nakatani, I. Kudo, N. Nishiyama et al., mediates ischemic injury in the hippocampus: a regional difference of neuronal vulnerability, European Journal of Neuroscience, vol.270, issue.12, pp.2319-2323, 2001.
DOI : 10.1074/jbc.270.12.6433

M. T. Heneka and M. K. O-'banion, Inflammatory processes in Alzheimer's disease, Journal of Neuroimmunology, vol.184, issue.1-2, pp.69-91, 2007.
DOI : 10.1016/j.jneuroim.2006.11.017

B. P. Imbimbo, V. Solfrizzi, and E. F. Panza, Are NSAIDs useful to treat Alzheimer's disease or mild cognitive impairment ? », Front, Aging Neurosci, vol.2, pp.1-14, 2010.
DOI : 10.3389/fnagi.2010.00019

URL : http://journal.frontiersin.org/article/10.3389/fnagi.2010.00019/pdf

P. J. Cimino, C. D. Keene, R. M. Breyer, K. S. Montine, and T. J. Montine, Therapeutic Targets in Prostaglandin E2 Signaling for Neurologic Disease, Therapeutic targets in prostaglandin E2 signaling for neurologic disease, pp.1863-1869, 2009.
DOI : 10.2174/092986708785132915

O. Pepicelli, E. Fedele, M. Berardi, M. Raiteri, G. Levi et al., Cyclo-oxygenase-1 and -2 differently contribute to prostaglandin???E2 synthesis and lipid peroxidation after in vivo activation of N-methyl-d-aspartate receptors in rat hippocampus, Journal of Neurochemistry, vol.58, issue.6, pp.1561-1567, 2005.
DOI : 10.1161/01.RES.88.6.600

P. T. Jantzen, K. E. Connor, G. Dicarlo, G. L. Wenk, J. L. Wallace et al., « Microglial activation and ?-amyloid reduction caused by a nitric oxide-releasing nonsteroidal anti-inflammatory drug in amyloid precursor protein plus presenilin-1 transgenic mice, J. Neurosci, vol.22, issue.6, pp.2246-2254, 2002.

S. Weggen, J. L. Eriksen, S. A. Sagi, C. U. Pietrzik, V. Ozols et al., Evidence That Nonsteroidal Anti-inflammatory Drugs Decrease Amyloid ??42 Production by Direct Modulation of ??-Secretase Activity, Journal of Biological Chemistry, vol.4, issue.34, pp.31831-31837, 2003.
DOI : 10.1074/jbc.M304824200

B. S. Berlett and E. R. Stadtman, Protein Oxidation in Aging, Disease, and Oxidative Stress, Journal of Biological Chemistry, vol.60, issue.33, pp.20313-20316, 1997.
DOI : 10.1016/B978-0-12-152828-7.50010-X

M. D. Ikonomovic, E. E. Abrahamson, T. Uz, H. Manev, and S. T. Dekosky, Increased 5-Lipoxygenase Immunoreactivity in the Hippocampus of Patients With Alzheimer's Disease, Journal of Histochemistry & Cytochemistry, vol.104, issue.12, pp.12-1065, 2008.
DOI : 10.1016/j.lfs.2006.05.022

K. J. Kwon, Y. Jung, S. H. Lee, C. Moon, and E. J. Baik, Arachidonic acid induces neuronal death through lipoxygenase and cytochrome P450 rather than cyclooxygenase, Arachidonic acid induces neuronal death through lipoxygenase and cytochrome P450 rather than cyclooxygenase, pp.73-84, 2005.
DOI : 10.1038/jcbfm.1987.43

URL : http://repository.ajou.ac.kr/bitstream/201003/2924/3/73-84.pdf

C. N. Serhan, K. Gotlinger, and S. Hong, Resolvins, docosatrienes, and neuroprotectins, novel omega-3-derived mediators, and their aspirin-triggered endogenous epimers: an overview of their protective roles in catabasis, Prostaglandins & Other Lipid Mediators, vol.73, issue.3-4, pp.155-172, 2004.
DOI : 10.1016/j.prostaglandins.2004.03.005

C. N. Serhan, N. Chiang, and T. E. Van-dyke, Resolving inflammation: dual anti-inflammatory and pro-resolution lipid mediators, Nature Reviews Immunology, vol.13, issue.5, pp.349-361, 2008.
DOI : 10.4049/jimmunol.164.5.2660

URL : http://europepmc.org/articles/pmc2744593?pdf=render

N. G. Bazan, Cellular and molecular events mediated by docosahexaenoic acid-derived neuroprotectin D1 signaling in photoreceptor cell survival and brain protection, Prostaglandins, Leukotrienes and Essential Fatty Acids, vol.81, issue.2-3, pp.205-211, 2009.
DOI : 10.1016/j.plefa.2009.05.024

C. N. Serhan, R. Yang, K. Martinod, K. Kasuga, P. S. Pillai et al., Maresins: novel macrophage mediators with potent antiinflammatory and proresolving actions, The Journal of Experimental Medicine, vol.192, issue.1, pp.15-23, 2008.
DOI : 10.1016/S0076-6879(07)32012-0

URL : http://jem.rupress.org/content/jem/206/1/15.full.pdf

V. Martin, N. Fabelo, G. Santpere, B. Puig, R. Marin et al., Lipid Alterations in Lipid Rafts from Alzheimer's Disease Human Brain Cortex, Journal of Alzheimer's Disease, vol.19, issue.2, pp.489-502, 2010.
DOI : 10.3233/JAD-2010-1242

J. Yeo, W. Ong, S. Ling, and A. A. Farooqui, Intracerebroventricular injection of phospholipases A2 inhibitors modulates allodynia after facial carrageenan injection in mice, Pain, vol.112, issue.1, pp.148-155, 2004.
DOI : 10.1016/j.pain.2004.08.009

T. Schilling, F. Lehmann, B. Rückert, and E. C. Eder, Physiological mechanisms of lysophosphatidylcholine-induced de-ramification of murine microglia, The Journal of Physiology, vol.286, issue.1, pp.105-120, 2004.
DOI : 10.1161/hc0802.104457

E. P. Scheltens and . Scheffer, « Decreased lysophosphatidylcholine/phosphatidylcholine ratio in cerebrospinal fluid in Alzheimer's disease, J. Oh Neural Transm, vol.110, pp.949-955, 2003.

S. D. Ryan, C. S. Harris, C. L. Carswell, J. E. Baenziger, and S. A. Bennet, -1 carbon chain of platelet-activating factor glycerophospholipids determines pro- or anti-apoptotic signaling in primary neurons, Journal of Lipid Research, vol.148, issue.10, pp.2250-2258, 2008.
DOI : 10.1002/1097-4547(20010101)63:1<10::AID-JNR2>3.0.CO;2-R

I. Izquierdo, C. Fin, P. K. Schmitz, R. C. Da-silva, D. Jerusalinsky et al., Memory enhancement by intrahippocampal, intraamygdala, or intraentorhinal infusion of platelet-activating factor measured in an inhibitory avoidance task., Proc. Natl. Acad. Sci. USA, pp.5047-5051, 1995.
DOI : 10.1073/pnas.92.11.5047

S. Ishii and T. Shimizu, Platelet-activating factor (PAF) receptor and genetically engineered PAF receptor mutant mice, Progress in Lipid Research, vol.39, issue.1, pp.41-82, 2000.
DOI : 10.1016/S0163-7827(99)00016-8

K. Nogami, Y. Hirashima, and S. Endo, Involvement of platelet-activating factor (PAF) in glutamate neurotoxicity in rat neuronal cultures, Brain Research, vol.754, issue.1-2, pp.72-78, 1997.
DOI : 10.1016/S0006-8993(97)00047-4

A. D. Butterfield and M. L. Bader-lange, Involvements of the lipid peroxidation product, HNE, in the pathogenesis and progression of Alzheimer's disease???, Biochimica et Biophysica Acta (BBA) - Molecular and Cell Biology of Lipids, vol.1801, issue.8, pp.924-929, 2010.
DOI : 10.1016/j.bbalip.2010.02.005

H. Raza, A. John, E. M. Brown, and S. Benedict, Kambal, « Alterations in mitochondrial respiratory functions, redox metabolism and apoptosis by oxidant 4- hydroxynonenal and antioxidants curcumin and melatonin in PC12 cells

M. Guglielmotto, L. Giliberto, and E. Tamagno, Tabaton, « Oxidative stress mediates the pathogenic effects of different Alzheimer's disease risk factors », Front, Aging Neurosci, vol.2, pp.1-8, 2010.

M. A. Bradley, S. Xiong-fister, W. R. Markesbery, and M. A. Lovell, Elevated 4-hydroxyhexenal in Alzheimer's disease (AD) progression, Neurobiology of Aging, vol.33, issue.6, pp.1034-1044, 2012.
DOI : 10.1016/j.neurobiolaging.2010.08.016

D. Pratico, The neurobiology of isoprostanes and Alzheimer's disease, Biochimica et Biophysica Acta (BBA) - Molecular and Cell Biology of Lipids, vol.1801, issue.8, pp.930-933, 2010.
DOI : 10.1016/j.bbalip.2010.01.009

«. Montine, Brain regional quantification of F-ring and D-/E-ring isoprostanes and neuroprostanes in Alzheimer's disease », Am. J. Pathol, vol.158, issue.1, pp.293-297, 2001.

C. B. Sullivan, E. Matafonova, L. J. Roberts, I. , V. Amarnath et al., Isoketals form cytotoxic phosphatidylethanolamine adducts in cells, Isoketals form cytotoxic phosphatidylethanolamine adducts in cells, pp.999-1009, 2010.
DOI : 10.1074/jbc.M604660200

N. Bernoud-hubac, S. S. Davies, O. Boutaud, T. J. Montine, and L. J. , Roberts II, « Formation of highly reactive ?-ketoaldehydes (neuroketals) as product of the neuroprostane pathway, J. Biol. Chem, vol.276, pp.33-30964, 2001.

J. D. Fessel, C. Hulette, S. Powell, L. J. Roberts, I. et al., Isofurans, but not F2-isoprostanes, are increased in the substantia nigra of patients with Parkinson's disease and with dementia with Lewy body disease, Journal of Neurochemistry, vol.154, issue.3, pp.645-650, 2003.
DOI : 10.1016/S0002-9440(10)65396-5

«. Fitzgerald and . Neurofurans, novel indices of oxidant stress derived from docosahexaenoic acid, J. Biol. Chem, vol.283, pp.6-16, 2008.

H. Aybek, F. Ercan, D. Aslan, and E. T. Sahiner, Determination of malondialdehyde, reduced glutathione levels and APOE4 allele frequency in late-onset Alzheimer's disease in Denizli, Turkey, Clinical Biochemistry, vol.40, issue.3-4, pp.172-176, 2007.
DOI : 10.1016/j.clinbiochem.2006.09.005

P. H. Reddy and M. F. Beal, Are mitochondria critical in the pathogenesis of Alzheimer's disease?, Brain Research Reviews, vol.49, issue.3, pp.618-632, 2005.
DOI : 10.1016/j.brainresrev.2005.03.004

I. Sponne, A. Fifre, B. Kriem, V. Koziel, B. Bihain et al., « Membrane cholesterol interfers with neuronal apoptosis induced by soluble oligomers but not fibrils of the amyloid-? peptide », FASEB J, 2004.

H. Cheng, K. S. Vetrivel, P. Gong, and A. Parent, Mechanisms of Disease: new therapeutic strategies for Alzheimer's disease???targeting APP processing in lipid rafts, Nature Clinical Practice Neurology, vol.25, issue.7, pp.374-382, 2007.
DOI : 10.1038/ncpneuro0549

M. Sastre, T. Klockgether, and M. T. Heneka, Contribution of inflammatory processes to Alzheimer's disease: molecular mechanisms, International Journal of Developmental Neuroscience, vol.24, issue.2-3, pp.167-176, 2006.
DOI : 10.1016/j.ijdevneu.2005.11.014

A. Tixier-vidal, R. Picart, and C. Loudes, Effects of polyunsaturated fatty acids and hormones on synaptogenesis in serum-free medium cultures of mouse fetal hypothalamic cells, Neuroscience, vol.17, issue.1, pp.115-132, 1986.
DOI : 10.1016/0306-4522(86)90230-7

S. Kotani, E. Sakaguchi, S. Warashina, N. Matsukawa, Y. Ishikura et al., Dietary supplementation of arachidonic and docosahexaenoic acids improves cognitive dysfunction, Dietary supplementation of arachidonic and docosahexaenoic acids improves cognitive dysfunction, pp.159-164, 2006.
DOI : 10.1016/j.neures.2006.06.010

T. L. Kaduce, Y. Chen, J. W. Hell, and A. A. , Docosahexaenoic acid synthesis from n-3 fatty acid precursors in rat hippocampal neurons, Journal of Neurochemistry, vol.42, issue.2, pp.1525-1535, 2008.
DOI : 10.1016/j.plefa.2006.05.014

L. Jiang, Y. Shi, L. Wang, and E. Z. Yang, The influence of orally administered docosahexaenoic acid on cognitive ability in aged mice, The Journal of Nutritional Biochemistry, vol.20, issue.9, pp.735-741, 2009.
DOI : 10.1016/j.jnutbio.2008.07.003

G. P. Lim, F. Calon, T. Morihara, F. Yang, B. Teter et al., A Diet Enriched with the Omega-3 Fatty Acid Docosahexaenoic Acid Reduces Amyloid Burden in an Aged Alzheimer Mouse Model, Journal of Neuroscience, vol.25, issue.12, pp.12-3032, 2005.
DOI : 10.1523/JNEUROSCI.4225-04.2005

D. S. Martin, P. Spencer, D. F. Horrobin, and M. A. Lynch, Long-term potentiation in aged rats is restored when the age-related decrease in polyunsaturated fatty acid concentration is reversed, Prostaglandins, Leukotrienes and Essential Fatty Acids, vol.67, issue.2-3, pp.121-130, 2002.
DOI : 10.1054/plef.2002.0408

T. D. Niemoller and N. G. Bazan, Docosahexaenoic acid neurolipidomics, Docosahexaenoic acid neurolipidomics, pp.85-89, 2010.
DOI : 10.1016/j.prostaglandins.2009.09.005

URL : http://europepmc.org/articles/pmc2905848?pdf=render

C. He, X. Qu, L. Cui, J. Wang, and J. X. Kang, Improved spatial learning performance of fat-1 mice is associated with enhanced neurogenesis and neuritogenesis by docosahexaenoic acid, Proc. Natl. Acad. Sci. USA, pp.27-11370, 2009.
DOI : 10.1016/0165-0270(84)90007-4

T. Sakamoto, M. Cansev, and R. J. Wurtman, Oral supplementation with docosahexaenoic acid and uridine-5???-monophosphate increases dendritic spine density in adult gerbil hippocampus, Brain Research, vol.1182, pp.50-59, 2007.
DOI : 10.1016/j.brainres.2007.08.089

Q. Ma, B. Teter, O. Ubeda, T. Morihara, D. Dhoot et al., « Omega-3 fatty acid docosahexaenoic acid increses SorLA/LR11, a sorting protein with reduced expression in sporadic Alzheimer's disease (AD) : relevance to AD prevention, J. Neurosci, vol.27, pp.52-14299, 2007.

L. G. Puskas, K. Kitajka, C. Nyakas, and G. Barcelo-coblijn, Farkas, « Short-term administration of omega 3 fatty acids from fish oil results in increased tranthyretin transcription in old rat hippocampus, Proc. Natl. Acad. Sci. USA, pp.1580-1585, 2003.

T. Reijmer, L. M. Van-groen, D. Broersen, A. Lütjohann, A. J. Heerschap et al., « DHA ans cholesterol containing diets influence Alzheimer-like pathology, cognition and cerebral asculature in APPswe, Neurobiol. Dis, vol.33, pp.482-498, 2009.

S. Hossain, M. Hashimoto, M. Katakura, T. Shimada, and E. O. Shido, -induced toxicity in SH-S5Y5 cells, Journal of Neurochemistry, vol.278, issue.2, pp.568-579, 2009.
DOI : 10.1016/S0925-4439(00)00029-6

A. Johansson, A. Garlind, F. Berglind-dehlin, G. Karlsson, K. Edwards et al., Docosahexaenoic acid stabilizes soluble amyloid-?? protofibrils and sustains amyloid-??-induced neurotoxicity in???vitro, FEBS Journal, vol.174, issue.Part 1, pp.990-1000, 2007.
DOI : 10.1016/S0927-7757(00)00516-1

C. Bate, V. Marshall, L. Colombo, L. Diomede, M. Salmona et al., Docosahexaenoic and eicosapentaenoic acids increase neuronal death in response to HuPrP82???146 and A??1???42, Docosahexaenoic and eicosapentaenoic acids increase neuronal death in response tu HuPrP82-146 and A?1-42, pp.934-943, 2008.
DOI : 10.1016/j.neuropharm.2008.02.003

T. Oster and T. Pillot, Docosahexaenoic acid and synaptic protection in Alzheimer's disease mice, Biochimica et Biophysica Acta (BBA) - Molecular and Cell Biology of Lipids, vol.1801, issue.8, pp.791-798, 2010.
DOI : 10.1016/j.bbalip.2010.02.011

M. Akbar, F. Calderon, Z. Wen, and H. Kim, Docosahexaenoic acid: A positive modulator of Akt signaling in neuronal survival, Proc. Natl. Acad. Sci. USA, pp.31-10858, 2005.
DOI : 10.1073/pnas.0402531101

. Bazinet, Polunsaturated fatty acid deprivation in rats decreases frontal cortex BDNF via a p38 MAPK-dependent mechanis, Mol. Psychiatry, vol.12, pp.36-46, 2007.

K. Jung and M. Kwak, « The Nrf2 system as potetial target for the development of indirect antioxidants, pp.7266-7291, 2010.

M. C. Marin, E. M. Gonzalez-montelongo, and . Diaz, Addition of docosahexaenoic acid, but not arachidonic acid, activates glutathione and thioredoxin antioxidant system in murine hippocampal HT22 cells : potential implications in neuroprotection, J. Neurochem, vol.131, pp.470-483, 2014.

Y. Levi, I. Vedin, T. Cederholm, H. Basun, G. Irving et al., Transfer of omega-3 fatty acids across the blood-brain barrier after dietary supplementation with a docosahexaenoic acid-rich omega-3 fatty acid preparation in patients with Alzheimer's disease: the OmegAD study, Journal of Internal Medicine, vol.33, issue.4, pp.428-436, 2014.
DOI : 10.1016/j.neurobiolaging.2011.12.014

E. J. Béguin and . Heemskerk, « Vitamin K-dependent and vitamin K-independent hypocoagulant effects of dietary fish oil in rats, Thromb. Res, vol.104, pp.137-147, 2001.

K. Ritchie, I. Carrière, A. De-mendonça, F. Portet, J. F. Dartigues et al., Ancelin, « The neuroprotective effects of caffeine : a prospective population study (the Three City Study), Neurology, vol.69, 2007.

L. Guihenneuc-jouyaux, C. Cynober, and . Moinard, Allinquant, « Citrulline diet supplementation improves specific age-related raft changes in wild-type rodent hippocampus, Age, vol.35, pp.1589-1606, 2013.