J. B. German and C. J. Dillard, Composition, structure and absorption of milk lipids: A source of energy, fatsoluble nutrients and bioactive molecules, Crit. Rev. Food Sci. Nutr, vol.46, pp.57-92, 2006.

F. Giuffrida, C. Cruz-hernandez, B. Flück, I. Tavazzi, S. K. Thakkar et al., Quantification of phospholipids classes in human milk, Lipids, vol.48, pp.1051-1058, 2013.

A. M. Kamelska, R. Pietrzak-fie?ko, and K. Bryl, Variation of the cholesterol content in breast milk during 10 days collection at early stages of lactation, Acta Biochim. Pol, vol.59, pp.243-247, 2012.

B. Delplanque, R. Gibson, B. Koletzko, A. Lapillonne, and B. Strandvik, Lipid Quality in Infant Nutrition: Current Knowledge and Future Opportunities, J. Pediatr. Gastroenterol. Nutr, vol.61, pp.8-17, 2015.
DOI : 10.1097/mpg.0000000000000818

URL : http://europepmc.org/articles/pmc4927316?pdf=render

R. V. Gow and J. R. Hibbeln, Omega-3 fatty acid and nutrient deficits in adverse neurodevelopment and childhood behaviors, Child Adolesc. Psychiatr. Clin. N. Am, vol.23, pp.555-590, 2014.

A. Zarrouk, M. Debbabi, M. Bezine, E. M. Karym, A. Badreddine et al., Lipid Biomarkers in Alzheimer's Disease. Curr. Alzheimer. Res, 2017.

R. J. Wanders and H. R. Waterham, Biochemistry of mammalian peroxisomes revisited, Annu. Rev. Biochem, vol.75, pp.295-332, 2006.
DOI : 10.1146/annurev.biochem.74.082803.133329

O. S. Ademowo, H. K. Dias, D. G. Burton, and H. R. Griffiths, Lipid (per) oxidation in mitochondria: An emerging target in the ageing process?, Biogerontology, 2017.

A. Zarrouk, I. Cheraif, S. Hadj-ahmed, W. Chaabane, S. Hammami et al., Lipid Peroxidation Markers and Cognition in Dementia, Correlations between Peripheral Trans Fatty Acids, 2004.

G. Abate, M. Marziano, W. Rungratanawanich, M. Memo, and D. Uberti, Nutrition and AGE-ing: Focusing on Alzheimer's Disease, Oxid. Med. Cell. Longev, p.7039816, 2017.
DOI : 10.1155/2017/7039816

URL : http://downloads.hindawi.com/journals/omcl/2017/7039816.pdf

G. P. Eckert, U. Lipka, and W. E. Muller, Omega-3 fatty acids in neurodegenerative diseases: Focus on mitochondria, Prostaglandins Leukot. Essent. Fatty Acids, vol.88, pp.105-114, 2013.
DOI : 10.1016/j.plefa.2012.05.006

M. C. Morris and C. C. Tangney, Dietary fat composition and dementia risk, Neurobiol. Aging, vol.2, pp.59-64, 2014.
DOI : 10.1016/j.neurobiolaging.2014.03.038

URL : https://doi.org/10.1016/j.neurobiolaging.2014.03.038

I. Björkhem, Crossing the barrier: Oxysterols as cholesterol transporters and metabolic modulators in the brain, J. Intern. Med, vol.260, pp.493-508, 2006.

A. Zarrouk, A. Vejux, J. Mackrill, Y. O'callaghan, M. Hammami et al., Involvement of oxysterols in age-related diseases and ageing processes, Ageing Res. Rev, vol.18, pp.148-162, 2014.

M. Aoun, V. Tiranti, and . Mitochondria, A crossroads for lipid metabolism defect in neurodegeneration with brain iron accumulation diseases, Int. J. Biochem. Cell Biol, vol.63, pp.25-31, 2015.

S. Yehuda and S. Rabinovitz, The role of essential fatty acids in anorexia nervosa and obesity, Crit. Rev. Food Sci. Nutr, vol.56, pp.2021-2035, 2016.

R. Palacios-pelaez, W. J. Lukiw, and N. G. Bazan, Omega-3 essential fatty acids modulate initiation and progression of neurodegenerative disease, Mol. Neurobiol, vol.41, pp.367-374, 2010.
DOI : 10.1007/s12035-010-8139-z

J. E. Karr, J. E. Alexander, and R. G. Winningham, Omega-3 polyunsaturated fatty acids and cognition throughout the lifespan: A review, Nutr. Neurosci, vol.14, pp.216-225, 2011.

N. D. Barnard, A. I. Bush, A. Ceccarelli, J. Cooper, C. A. De-jager et al., Dietary and lifestyle guidelines for the prevention of Alzheimer's disease, Neurobiol. Aging, vol.35, pp.74-78, 2014.

A. Badreddine, Préparation et Caractérisation d'extraits d'Argania Spinosa et d'huile d'argan et Evaluation de Leurs Effets Neuroprotecteurs In Vivo et In Vitro

A. El-abbassi, N. Khalid, H. Zbakh, and A. Ahmad, Physicochemical characteristics, nutritional properties, and health benefits of argan oil: A review, Crit. Rev. Food Sci. Nutr, vol.54, pp.1401-1414, 2014.

H. El-monfalouti, D. Guillaume, C. Denhez, and Z. Charrouf, Therapeutic potential of argan oil-A review, J. Pharm. Pharmacol, vol.62, pp.1669-1675, 2010.

A. Drissi, J. Girona, M. Cherki, G. Godàs, A. Derouiche et al., Evidence of hypolipemiant and antioxidant properties of argan oil derived from the argan tree (Argania spinosa), Clin. Nutr, vol.23, pp.1159-1166, 2004.

M. Cherki, A. Derouiche, A. Drissi, M. El-messal, Y. Bamou et al., Consumption of argan oil may have an antiatherogenic effect by improving paraoxonase activities and antioxidant status: Intervention study in healthy men, Nutr. Metab. Cardiovasc. Dis, vol.15, pp.352-360, 2005.

A. Derouiche, M. Cherki, A. Drissi, Y. Bamou, M. El-messal et al., Nutritional intervention study with argan oil in man: Effects on lipids and apolipoproteins, Ann. Nutr. Metab, vol.49, pp.196-201, 2005.

C. Cabrera-vique, R. Marfil, R. Giménez, and O. Martínez-augustin, Bioactive compounds and nutritional significance of virgin argan oil--an edible oil with potential as a functional food, Nutr. Rev, vol.70, pp.266-279, 2012.

K. Q. Boucetta, Z. Charrouf, A. Derouiche, Y. Rahali, and Y. Bensouda, Skin hydration in postmenopausal women: Argan oil benefit with oral and/or topical use, Prz. Menopauzalny, vol.13, pp.280-288, 2014.

K. Q. Boucetta, Z. Charrouf, H. Aguenaou, A. Derouiche, and Y. Bensouda, The effect of dietary and/or cosmetic argan oil on postmenopausal skin elasticity, Clin. Interv. Aging, vol.10, pp.339-349, 2015.

Y. Bahbiti, H. Ammouri, I. Berkiks, A. El-hessni, A. Ouichou et al., Anticonvulsant effect of argan oil on pilocarpine model induced status epilepticus in wistar rats, Nutr. Neurosci, vol.2016, pp.1-7

B. Ayd?n, Effects of argan oil on the mitochondrial function, antioxidant system and the activity of NADPH-generating enzymes in acrylamide treated rat brain, Biomed. Pharmacother, vol.87, pp.476-481, 2017.

Z. A. ?ekero?lu, B. Ayd?n, and V. ?ekero?lu, Argan oil reduces oxidative stress, genetic damage and emperipolesis in rats treated with acrylamide, Biomed. Pharmacother, vol.94, pp.873-879, 2017.

F. Khallouki, C. Younos, R. Soulimani, T. Oster, Z. Charrouf et al., Consumption of argan oil (Morocco) with its unique profile of fatty acids, tocopherols, squalene, sterols and phenolic compounds should confer valuable cancer chemopreventive effects, Eur. J. Cancer Prev, vol.12, pp.67-75, 2003.

L. C. López, C. Cabrera-vique, C. Venegas, L. García-corzo, M. Luna-sánchez et al., Argan oil-contained antioxidants for human mitochondria, Nat. Prod. Commun, vol.8, pp.47-50, 2013.

D. Trompier, A. Vejux, A. Zarrouk, C. Gondcaille, F. Geillon et al., Biochimie, vol.98, pp.102-110, 2014.

M. Debbabi, T. Nury, A. Zarrouk, N. Mekahli, M. Bezine et al., Protective effects of ?-tocopherol, ?-tocopherol and oleic acid, three compounds of olive oils, and no effect of trolox, on 7-ketocholesterol-induced mitochondrial and peroxisomal dysfunction in microglial BV-2 cells, Int. J. Mol. Sci, vol.17, p.1973, 2016.
URL : https://hal.archives-ouvertes.fr/hal-01463786

G. Testa, E. Staurenghi, C. Zerbinati, S. Gargiulo, L. Iuliano et al., Changes in brain oxysterols at different stages of Alzheimer's disease: Their involvement in neuroinflammation, Redox Biol, vol.10, pp.24-33, 2016.

V. Leoni, D. Lütjohann, and T. Masterman, Levels of 7-oxocholesterol in cerebrospinal fluid are more than one thousand times lower than reported in multiple sclerosis, J. Lipid Res, vol.46, pp.191-195, 2005.

S. Boenzi, F. Deodato, R. Taurisano, B. M. Goffredo, C. Rizzo et al., Evaluation of plasma cholestane-3?,5?,6?-triol and 7-ketocholesterol in inherited disorders related to cholesterol metabolism, J. Lipid Res, vol.57, pp.361-367, 2016.

T. Nury, A. Zarrouk, K. Ragot, M. Debbabi, J. M. Riedinger et al., 7-Ketocholesterol is increased in the plasma of X-ALD patients and induces peroxisomal modifications in microglial cells: Potential roles of 7-ketocholesterol in the pathophysiology of X-ALD, J. Steroid. Biochem. Mol. Biol, vol.169, pp.123-136, 2017.

T. Nury, Caractérisation de la Neurotoxicité des 4-et 7-hydroxycholestérols: Impact sur la prolifération, le stress oxydant, l'apoptose et l'autophagie (notion d'oxyapoptophagie). Diplôme de l'Ecole Pratique des Hautes Etudes (EPHE), 2014.

B. Wang, P. P. Van-veldhoven, C. Brees, N. Rubio, M. Nordgren et al., Mitochondria are targets for peroxisome-derived oxidative stress in cultured mammalian cells. Free Radic, Biol. Med, vol.65, pp.882-894, 2013.
DOI : 10.1016/j.freeradbiomed.2013.08.173

C. Lismont, M. Nordgren, P. P. Van-veldhoven, and M. Fransen, Redox interplay between mitochondria and peroxisomes. Front, Cell Dev. Biol, vol.3, p.35, 2015.
DOI : 10.3389/fcell.2015.00035

URL : https://www.frontiersin.org/articles/10.3389/fcell.2015.00035/pdf

R. Sudo, F. Sato, T. Azechi, and H. Wachi, 7-Ketocholesterol-induced lysosomal dysfunction exacerbates vascular smooth muscle cell calcification via oxidative stress, Genes Cells, vol.20, pp.982-991, 2015.
DOI : 10.1111/gtc.12301

A. Chakhchar, M. Haworth, C. El-modafar, M. Lauteri, C. Mattioni et al., An assessment of genetic diversity and drought tolerance in argan tree (Argania spinosa) populations: Potential for the development of improved drought tolerance, Front Plant Sci, vol.8, p.276, 2017.

A. Adlouni, L'huile d'argan, de la nutrition à la santé, Phytothérapie, vol.8, pp.89-97, 2010.

E. J. Behrman and . Gopalan, J. Chem. Educ, vol.82, pp.1791-1793, 2005.

P. D. Sonawane, J. Pollier, S. Panda, J. Szymanski, H. Massalha et al., Plant cholesterol biosynthetic pathway overlaps with phytosterol metabolism, Nat. Plants, vol.3, p.16205, 2016.
DOI : 10.1038/nplants.2016.205

A. Vejux, L. Malvitte, and G. Lizard, Side effects of oxysterols: Cytotoxicity, oxidation, inflammation, and phospholipidosis. Braz, J. Med. Biol. Res, vol.41, pp.545-556, 2008.

G. Poli, F. Biasi, and G. Leonarduzzi, Oxysterols in the pathogenesis of major chronic diseases, Redox Biol, vol.1, pp.125-130, 2013.

M. T. Rodriguez-estrada, G. Garcia-llatas, and M. J. Lagarda, 7-Ketocholesterol as marker of cholesterol oxidation in model and food systems: When and how, Biochem. Biophys. Res. Commun, vol.446, pp.792-797, 2014.

M. G. Traber and J. F. Stevens, Vitamins C and E: Beneficial effects from a mechanistic perspective. Free Radic, Biol. Med, vol.51, pp.1000-1013, 2011.

A. Basli, S. Soulet, N. Chaher, J. M. Mérillon, M. Chibane et al., Wine polyphenols: Potential agents in neuroprotection, Oxid. Med. Cell. Longev, p.805762, 2012.

A. Cilla, A. Alegría, A. Attanzio, G. Garcia-llatas, L. Tesoriere et al., Dietary phytochemicals in the protection against oxysterol-induced damage, Chem. Phys. Lipids, pp.192-205, 2017.

M. Debbabi, A. Zarrouk, M. Bezine, W. Meddeb, T. Nury et al., Comparison of the effects of major fatty acids present in the Mediterranean diet (oleic acid, docosahexaenoic acid) and in hydrogenated oils (elaidic acid) on 7-ketocholesterol-induced oxiapoptophagy in microglial BV-2 cells, Chem. Phys. Lipids, pp.151-170, 2017.
URL : https://hal.archives-ouvertes.fr/hal-01912331

K. Ragot, D. Delmas, A. Athias, T. Nury, M. Baarine et al., Tocopherol impairs 7-ketocholesterolinduced caspase-3-dependent apoptosis involving GSK-3 activation and Mcl-1 degradation in 158N murine oligodendrocytes, Chem. Phys. Lipids, vol.164, pp.469-478, 2011.
DOI : 10.1016/j.chemphyslip.2011.04.014

G. Lizard, V. Deckert, L. Dubrez, M. Moisant, P. Gambert et al., Induction of apoptosis in endothelial cells treated with cholesterol oxides, Am. J. Pathol, vol.148, pp.1625-1638, 1996.

J. M. Zahm, S. Baconnais, S. Monier, N. Bonnet, G. Bessède et al., Chronology of cellular alterations during 7-ketocholesterol-induced cell death on A7R5 rat smooth muscle cells: Analysis by time lapse-video microscopy and conventional fluorescence microscopy, Cytometry A, vol.52, pp.57-69, 2003.
URL : https://hal.archives-ouvertes.fr/inserm-00148786

A. Zarrouk, T. Nury, E. M. Karym, A. Vejux, R. Sghaier et al., Attenuation of 7-ketocholesterol-induced overproduction of reactive oxygen species, apoptosis, and autophagy by dimethyl fumarate in 158N murine oligodendrocytes, J. Steroid. Biochem. Mol. Biol, vol.169, pp.29-38, 2017.

S. Monier, M. Samadi, C. Prunet, M. Denance, A. Laubriet et al., Impairment of the cytotoxic and oxidative activities of 7beta-hydroxycholesterol and 7-ketocholesterol by esterification with oleate, Biochem. Biophys. Res. Commun, vol.303, pp.814-824, 2003.

T. Nury, A. Zarrouk, J. J. Mackrill, M. Samadi, P. Durand et al., Induction of oxiapoptophagy in 158N murine oligodendrocytes treated by 7-ketocholesterol-, 7?-hydroxycholesterol-, or 24(S)-hydroxycholesterol: Protective effects of ?-tocopherol and docosahexaenoic acid (DHA, Steroids 2015, 99 Pt B, vol.22, pp.194-203

M. Baes and P. Aubourg, Peroxisomes, myelination, and axonal integrity in the CNS, Neuroscientist, vol.15, pp.367-379, 2009.

E. Gray, C. Rice, K. Hares, J. Redondo, K. Kemp et al., Reductions in neuronal peroxisomes in multiple sclerosis grey matter, Mult. Scler, vol.20, pp.651-659, 2014.

A. Ghosh, M. Jana, K. Modi, F. J. Gonzalez, K. B. Sims et al., Activation of peroxisome proliferator-activated receptor ? induces lysosomal biogenesis in brain cells: Implications for lysosomal storage disorders, J. Biol. Chem, vol.290, pp.10309-10324, 2015.

M. A. Esmaeili, S. Yadav, R. K. Gupta, G. R. Waggoner, A. Deloach et al., Preferential PPAR-? activation reduces neuroinflammation, and blocks neurodegeneration in vivo, Hum. Mol. Genet, vol.25, pp.317-327, 2016.

S. Agarwal, A. Yadav, and R. K. Chaturvedi, Peroxisome proliferator-activated receptors (PPARs) as therapeutic target in neurodegenerative disorders, Biochem. Biophys. Res. Commun, vol.483, pp.1166-1177, 2017.

K. Ragot, J. J. Mackrill, A. Zarrouk, T. Nury, V. Aires et al., Absence of correlation between oxysterol accumulation in lipid raft microdomains, calcium increase, and apoptosis induction in 158N murine oligodendrocytes, Biochem. Pharmacol, vol.86, pp.67-79, 2013.

T. Nury, A. Zarrouk, A. Vejux, M. Doria, J. M. Riedinger et al., Induction of oxiapoptophagy, a mixed mode of cell death associated with oxidative stress, apoptosis and autophagy, on 7-ketocholesterol-treated 158N murine oligodendrocytes: Impairment by ?-tocopherol, Biochem. Biophys. Res. Commun, vol.446, pp.714-719, 2014.

T. M. Goldfinger, Beyond the French paradox: The impact of moderate beverage alcohol and wine consumption in the prevention of cardiovascular disease, Cardiol. Clin, vol.21, pp.449-457, 2003.

J. Ferrières, The French paradox: Lessons for other countries, Heart, vol.90, pp.107-111, 2004.

S. Lecour and K. T. Lamont, Natural polyphenols and cardioprotection. Mini Rev. Med. Chem, vol.11, pp.1191-1199, 2011.

S. M. Nadtochiy and E. K. Redman, Mediterranean diet and cardioprotection: The role of nitrite, vol.27, pp.733-744, 2011.

M. A. Martínez-gonzález, J. Salas-salvadó, R. Estruch, D. Corella, M. Fitó et al., Ppredimed investigators. Benefits of the mediterranean diet: Insights from the predimed study, Prog. Cardiovasc. Dis, vol.58, pp.50-60, 2015.

S. Canhada, K. Castro, I. S. Perry, and V. C. Luft, Omega-3 fatty acids' supplementation in Alzheimer's disease: A systematic review, Nutr. Neurosci, vol.2017, pp.1-10

F. Casamenti and M. Stefani, Olive polyphenols: New promising agents to combat aging-associated neurodegeneration, Expert Rev. Neurother, vol.7, pp.345-358, 2017.

V. K. Burg, H. S. Grimm, T. L. Rothhaar, S. Grösgen, B. Hundsdörfer et al., Plant sterols the better cholesterol in Alzheimer's disease? A mechanistical study, J. Neurosci, vol.33, pp.16072-16087, 2013.

R. Shuang, X. Rui, L. Wenfang, and D. Phytosterols, Plant Foods Hum. Nutr, vol.71, pp.347-354, 2016.

M. O. Grimm, J. Mett, and T. Hartmann, The Impact of Vitamin E and Other Fat-Soluble Vitamins on Alzheimer's Disease, Int. J. Mol. Sci, vol.17, p.1785, 2016.

G. Lizard, Y. Filali-zegzouti, and A. E. Midaoui, Benefits of Argan Oil on Human Health, Errachidia, Morocco. Int. J. Mol. Sci, vol.18, p.1383, 2017.
URL : https://hal.archives-ouvertes.fr/inserm-01552936

H. J. Tritschler, . Packer, and R. Medori, Oxidative stress and mitochondrial dysfunction in neurodegeneration, Biochem. Mol. Biol. Int, vol.34, pp.169-181, 1994.

V. I. Titorenko and S. R. Terlecky, Peroxisome metabolism and cellular aging, Traffic, vol.12, pp.252-259, 2011.
DOI : 10.1111/j.1600-0854.2010.01144.x

URL : https://onlinelibrary.wiley.com/doi/pdf/10.1111/j.1600-0854.2010.01144.x

M. Fransen, M. Nordgren, B. Wang, O. Apanasets, and P. P. Van-veldhoven, Aging, age-related diseases and peroxisomes, Subcell. Biochem, vol.69, pp.45-65, 2013.
DOI : 10.1007/978-94-007-6889-5_3

E. Galea, N. Launay, M. Portero-otin, M. Ruiz, R. Pamplona et al., Oxidative stress underlying axonal degeneration in adrenoleukodystrophy: A paradigm for multifactorial neurodegenerative diseases?, Biochim. Biophys. Acta, vol.1822, pp.1475-1488, 2012.

M. Depreter, M. Espeel, and F. Roels, Human peroxisomal disorders, Microsc. Res. Tech, vol.61, pp.203-223, 2003.
DOI : 10.1002/jemt.10330

V. K. Senanayake, W. Jin, A. Mochizuki, B. Chitou, and D. B. Goodenowe, Metabolic dysfunctions in multiple sclerosis: Implications as to causation, early detection, and treatment, a case control study, BMC Neurol, p.154, 2015.

J. Kou, G. G. Kovacs, R. Höftberger, W. Kulik, A. Brodde et al., Peroxisomal alterations in Alzheimer's disease, Acta Neuropathol, vol.122, pp.271-283, 2011.

G. Lizard, O. Rouaud, J. Demarquoy, M. Cherkaoui-malki, and L. Iuliano, Potential roles of peroxisomes in Alzheimer's disease and in dementia of the Alzheimer's type, J. Alzheimers Dis, vol.29, pp.241-254, 2012.

A. Zarrouk, J. M. Riedinger, S. H. Ahmed, S. Hammami, W. Chaabane et al., Fatty acid profiles in demented patients: Identification of hexacosanoic acid (C26:0) as a blood lipid biomarker of dementia, J. Alzheimers Dis, vol.44, pp.1349-1359, 2015.

B. Loos, D. J. Klionsky, and E. Wong, Augmenting brain metabolism to increase macro-and chaperonemediated autophagy for decreasing neuronal proteotoxicity and aging, Prog. Neurobiol, vol.156, pp.90-106, 2017.

G. Rimbach, A. M. Minihane, J. Majewicz, A. Fischer, J. Pallauf et al., Regulation of cell signalling by vitamin E, Proc. Nutr. Soc, vol.61, pp.415-425, 2002.

P. A. Grimsrud, H. Xie, T. J. Griffin, and D. A. Bernlohr, Oxidative stress and covalent modification of protein with bioactive aldehydes, J. Biol. Chem, vol.283, pp.21837-21841, 2008.

E. Guyot, X. Coumoul, J. F. Chassé, F. Khallouki, J. F. Savouret et al., Identification of a new stilbene-derived inducer of paraoxonase 1 and ligand of the Aryl hydrocarbon Receptor, Biochem. Pharmacol, vol.83, pp.627-632, 2012.
URL : https://hal.archives-ouvertes.fr/inserm-00743456

F. Khallouki, A. Breuer, E. Merieme, C. M. Ulrich, and R. W. Owen, Characterization and quantitation of the polyphenolic compounds detected in methanol extracts of Pistacia atlantica Desf. Fruits from the Guelmim region of Morocco, J. Pharm. Biomed. Anal, vol.134, pp.310-318, 2017.

D. J. Klionsky, K. Abdelmohsen, A. Abe, M. J. Abedin, H. Abeliovich et al., Guidelines for the use and interpretation of assays for monitoring autophagy, vol.12, pp.1-222, 2016.
URL : https://hal.archives-ouvertes.fr/hal-01396896

T. Guina, M. Deiana, S. Calfapietra, B. Cabboi, M. Maina et al., The role of p38 MAPK in the induction of intestinal inflammation by dietary oxysterols: Modulation by wine phenolics, Food Funct, vol.6, p.1218, 2015.

Y. Steffen, I. Wiswedel, D. Peter, T. Schewe, and H. Sies, Cytotoxicity of myeloperoxidase/nitrite-oxidized low-density lipoprotein toward endothelial cells is due to a high 7beta-hydroxycholesterol to 7-ketocholesterol ratio. Free Radic, Biol. Med, vol.41, pp.1139-1150, 2006.

B. Dugas, S. Charbonnier, M. Baarine, K. Ragot, D. Delmas et al., Effects of oxysterols on cell viability, inflammatory cytokines, VEGF, and reactive oxygen species production on human retinal cells: Cytoprotective effects and prevention of VEGF secretion by resveratrol, Eur. J. Nutr, vol.49, pp.435-446, 2010.
URL : https://hal.archives-ouvertes.fr/hal-00514897

L. Tesoriere, A. Attanzio, M. Allegra, C. Gentile, and M. A. Livrea, Phytochemical indicaxanthin suppresses 7-ketocholesterol-induced THP-1 cell apoptosis by preventing cytosolic Ca 2+ increase and oxidative stress, Br. J. Nutr, vol.110, pp.230-240, 2013.
DOI : 10.1017/s000711451200493x

URL : https://www.cambridge.org/core/services/aop-cambridge-core/content/view/6B1799393BA2B5DF659DA713497C6D2B/S000711451200493Xa.pdf/div-class-title-phytochemical-indicaxanthin-suppresses-7-ketocholesterol-induced-thp-1-cell-apoptosis-by-preventing-cytosolic-ca-span-class-sup-2-span-increase-and-oxidative-stress-div.pdf

A. Vejux, S. Guyot, T. Montange, J. M. Riedinger, E. Kahn et al., Phospholipidosis and downregulation of the PI3-K/PDK-1/Akt signalling pathway are vitamin E inhibitable events associated with 7-ketocholesterol-induced apoptosis, J. Nutr. Biochem, pp.45-61, 1920.

T. Said, J. Tremblay-mercier, H. Berrougui, P. Rat, and A. Khalil, Effects of vegetable oils on biochemical and biophysical properties of membrane retinal pigment epithelium cells. Can, J. Physiol. Pharmacol, vol.91, pp.812-817, 2013.

A. Altmeyer, A. C. Jung, M. Ignat, S. Benzina, J. M. Denis et al., Pharmacological enhancement of autophagy induced in a hepatocellular carcinoma cell line by high-LET radiation, Anticancer Res, vol.30, pp.303-310, 2010.

Y. El-kharrassi, M. Samadi, T. Lopez, T. Nury, R. El-kebbaj et al., Biological activities of Schottenol and Spinasterol, two natural phytosterols present in argan oil and in cactus pear seed oil, on murine miroglial BV2 cells, Biochem. Biophys. Res. Commun, vol.446, pp.798-804, 2014.
URL : https://hal.archives-ouvertes.fr/hal-01518331

R. El-kebbaj, S. El-kamouni, H. I. El-hajj, P. Andreoletti, J. Gresti et al., Modulation of peroxisomes abundance by argan oil and lipopolysaccharides in acyl-CoA oxidase 1-deficient fibroblasts, vol.5, pp.62-69, 2013.

R. El-kebbaj, P. Andreoletti, H. I. El-hajj, Y. El-kharrassi, J. Vamecq et al., Argan oil prevents down-regulation induced by endotoxin on liver fatty acid oxidation and gluconeogenesis and on peroxisome proliferator-activated receptor gamma coactivator-1?, (PGC-1?), peroxisome proliferator-activated receptor ? (PPAR?) and estrogen related receptor ? (ERR?), Biochim. Open, vol.1, pp.51-59, 2015.

L. Malvitte, T. Montange, A. Vejux, C. Joffre, A. Bron et al., Activation of a caspase-3-independent mode of cell death associated with lysosomal destabilization in cultured human retinal pigment epithelial cells (ARPE-19) exposed to 7beta-hydroxycholesterol, Curr. Eye Res, vol.33, pp.769-781, 2008.

C. He, H. Zhu, W. Zhang, I. Okon, Q. Wang et al., 7-Ketocholesterol induces autophagy in vascular smooth muscle cells through Nox4 and Atg4B, Am. J. Pathol, vol.183, pp.626-637, 2013.
DOI : 10.1016/j.ajpath.2013.04.028

URL : https://doi.org/10.1016/j.ajpath.2013.04.028

B. Levine and G. Kroemer, Autophagy in the pathogenesis of disease, Cell, vol.132, pp.27-42, 2009.

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

URL : http://www.nature.com:80/polopoly_fs/1.15991!/menu/main/topColumns/topLeftColumn/pdf/513481a.pdf

D. Santos, J. L. Chin, and C. M. , Pan-Assay interference compounds (PAINS): Warning signs in biochemicalpharmacological evaluations, Biochem. Pharmacol, 2015.

S. Jasial, Y. Hu, and J. Bajorath, How Frequently Are Pan-Assay Interference Compounds Active? Large-Scale Analysis of Screening Data Reveals Diverse Activity Profiles, Low Global Hit Frequency, and Many Consistently Inactive Compounds, J. Med. Chem, vol.60, pp.3879-3886, 2017.

A. Vejux and G. Lizard, Cytotoxic effects of oxysterols associated with human diseases: Induction of cell death (apoptosis and/or oncosis), oxidative and inflammatory activities, and phospholipidosis, Mol. Asp. Med, vol.30, pp.153-170, 2009.

S. J. Capuzzi, E. N. Muratov, and A. Tropsha, Phantom PAINS: Problems with the utility of alerts for Pan-Assay INterference CompoundS, J. Chem. Inf. Model, vol.57, pp.417-427, 2017.

E. Vandenhaute, L. Dehouck, M. C. Boucau, E. Sevin, R. Uzbekov et al., Modelling the neurovascular unit and the blood-brain barrier with the unique function of pericytes, Curr. Neurovasc. Res, vol.8, pp.258-269, 2011.

F. Gosselet, J. Saint-pol, and L. Fenart, Effects of oxysterols on the blood-brain barrier: Implications for Alzheimer's disease, Biochem. Biophys. Res. Commun, vol.446, pp.687-691, 2014.

M. Baarine, K. Ragot, E. C. Genin, H. El-hajj, D. Trompier et al., Peroxisomal and mitochondrial status of two murine oligodendrocytic cell lines (158N, 158JP) potential models for the study of peroxisomal disorders associated with dysmyelination processes, J. Neurochem, vol.111, pp.119-131, 2009.
URL : https://hal.archives-ouvertes.fr/hal-00514890

V. Leoni, T. Nury, A. Vejux, A. Zarrouk, C. Caccia et al., Mitochondrial dysfunctions in 7-ketocholesterol-treated 158N oligodendrocytes without or with ?-tocopherol: Impacts on the cellular profil of tricarboxylic cycle-associated organic acids, long chain saturated and unsaturated fatty acids, oxysterols, cholesterol and cholesterol precursors, J. Steroid. Biochem. Mol. Biol, vol.169, pp.96-110, 2017.

A. Zarrouk, T. Nury, M. Samadi, Y. O'callaghan, M. Hammami et al., Effects of cholesterol oxides on cell death induction and calcium increase in human neuronal cells (SK-N-BE) and evaluation of the protective effects of docosahexaenoic acid (DHA, Steroids 2015, 99 Pt B, vol.22, pp.238-247
URL : https://hal.archives-ouvertes.fr/hal-01518107

C. Miguet, S. Monier, A. Bettaieb, A. Athias, G. Besséde et al., Ceramide generation occurring during 7beta-hydroxycholesterol-and 7-ketocholesterolinduced apoptosis is caspase independent and is not required to trigger cell death, Cell Death Differ, vol.8, pp.83-99, 2001.

E. Kahn, F. Ménétrier, A. Vejux, T. Montange, D. Dumas et al., Flow cytometry and spectral imaging multiphoton microscopy analysis of CD36 expression with quantumdots 605 of untreated and 7-ketocholesterol-treated human monocytic cells, Anal. Quant. Cytol. Histol, vol.28, pp.316-330, 2006.

B. Vurusaner, P. Gamba, G. Testa, S. Gargiulo, F. Biasi et al., Survival signalingelicited by 27-hydroxycholesterol through the combined modula-tion of cellular redox state and ERK/Akt phosphorylation. Free Radic, Biol. Med, vol.77, pp.376-385, 2014.

Z. Charrouf, Valorisation des produits de l'arganier pour une gestion durable des zones arides du sud-ouest marocain, Proceedings of the Actes du 4ème Colloque Produits naturels d'origine végétale, pp.26-29, 1998.

G. Collin, F. X. Et-garneau, . Ed, and . Université-du-québec, , pp.195-209, 1999.

T. Moilanen and T. Nikkari, The effect of storage on the fatty acid composition of human serum, Clin. Chim. Acta, vol.114, pp.111-116, 1981.

W. R. Morrison and L. M. Smith, Preparation of fatty acid methyl esters and dimethylacetals from lipids with boron fluoride-methanol, J. Lipid Res, vol.5, pp.600-608, 1964.

B. Lyan, V. Azaïs-braesco, N. Cardinault, V. Tyssandier, P. Borel et al., Simple method for clinical determination of 13 carotenoids in human plasma using an isocratic high-performance liquid chromatographic method, J. Chromatogr. B Biomed. Sci. Appl, vol.751, pp.297-303, 2001.

S. Mahmoudi, M. Khali, and N. Mahmoudi, Etude de l'extraction des composés phénoliques de différentes parties de la fleur d'artichaut (Cynara scolymus L.), Nat. Technol. B Sci. Agron. Biol, vol.9, pp.35-40, 2013.

R. Rossi, G. Pastorelli, and C. Corino, Application of KRL test to assess total antioxidant activity in pigs: Sensitivity to dietary antioxidants, Res. Vet. Sci, vol.94, pp.372-377, 2013.

M. Oyaizu, Studies on products of browning reaction: Antioxidative activities of products of browning reaction prepared from glucosamine, Jpn. J. Nutr, p.307, 1986.

I. F. Benzie and J. J. Strain, The ferric reducing ability of plasma (FRAP) as ameasure of "antioxidant power": The FRAP assay, Anal. Biochem, vol.239, pp.70-76, 1996.

V. L. Singleton and J. A. Rossi, Colorunetry of total phenolics with phosphomolybdic-phosphotungstic acid reagents, Am. J. Enol. Viticult, vol.16, pp.144-158, 1965.

D. Colin, E. Limagne, S. Jeanningros, A. Jacquel, G. Lizard et al., Endocytosis of resveratrol via lipid rafts and activation of downstream signaling pathways in cancer cells, Cancer Prev. Res. (Phila.), vol.4, pp.1095-1106, 2011.

G. Lizard, S. Gueldry, V. Deckert, P. Gambert, and L. Lagrost, Evaluation of the cytotoxic effects of some oxysterols and of cholesterol on endothelial cell growth: Methodological aspects, Pathol. Biol, vol.45, pp.281-290, 1997.

T. Mosmann, Rapid colorimetric assay for cellular growth and survival: Application to proliferation and cytotoxicity assays, J. Immunol. Methods, vol.65, pp.55-63, 1983.

M. Olsson, I. Rundquist, and U. Brunk, Flow cytofluorometry of lysosomal acridine orange uptake by living cultured cells, Acta Path. Microbiol. Immunol. Scand, vol.95, pp.159-165, 1987.

G. M. Olsson, J. Rungby, I. Rundquist, and U. T. Brunk, Evaluation of lysosomal stability in living cultured macrophages by cytofluorometry. Effect of silver lactate and hypotonic conditions, Virchows Arch. B Cell Pathol. Incl. Mol. Pathol, vol.56, pp.263-269, 1989.

X. M. Yuan, W. Li, U. T. Brunk, H. Dalen, Y. H. Chang et al., Lysosomal destabilization during macrophage damage induced by cholesterol oxidation products. Free Radic, Biol. Med, vol.28, pp.208-218, 2000.

A. Pierzy?ska-mach, P. A. Janowski, and J. W. Dobrucki, Evaluation of acridine orange, LysoTracker Red, and quinacrine as fluorescent probes for long-term tracking of acidic vesicles, Cytometry A, vol.85, pp.729-737, 2014.

G. Rothe and G. Valet, Flow cytometric analysis of respiratory burst activity in phagocytes with hydroethidine and 2?,7?-dichlorofluorescin, J. Leukoc. Biol, vol.47, pp.440-448, 1990.

A. Zarrouk, A. Vejux, T. Nury, H. I. El-hajj, M. Haddad et al., Induction of mitochondrial changes associated with oxidative stress on very long chain fatty acids (C22:0, C24:0, or C26:0)-treated human neuronal cells (SK-NB-E), Oxid. Med. Cell. Longev, p.623257, 2012.

G. Lizard, S. Fournel, L. Genestier, N. Dhedin, C. Chaput et al., Kinetics of plasma membrane and mitochondrial alterations in cells undergoing apoptosis, Cytometry, vol.21, pp.275-283, 1995.

C. Zerbinati and L. Iuliano, Cholesterol and related sterols autoxidation. Free Radic, Biol. Med, vol.111, pp.151-155, 2017.
DOI : 10.1016/j.freeradbiomed.2017.04.013

A. Badreddine, . Karym, .. M. El, A. Zarrouk, T. Nury et al., An expeditious synthesis of spinasterol and schottenol, two phytosterols present in argan oil and in cactus pear seed oil, and evaluation of their biological activities on cells of the central nervous system, Steroids, vol.99, pp.119-124, 2015.
URL : https://hal.archives-ouvertes.fr/hal-01518108