A. M. Pickrell and C. T. Moraes, What role does mitochondrial stress play in neurodegenerative diseases? [PubMed] 2. Singh, I.; Pujol, A. Pathomechanisms underlying X-adrenoleukodystrophy: A three-hit hypothesis, Lizard, G. Brain peroxisomes, pp.63-78, 2010.
DOI : 10.1007/978-1-60761-756-3_4

G. H. Kim, J. E. Kim, S. J. Rhie, and S. Yoon, The Role of Oxidative Stress in Neurodegenerative Diseases, Experimental Neurobiology, vol.24, issue.4, pp.325-340, 2015.
DOI : 10.5607/en.2015.24.4.325

S. Rizvi, S. T. Raza, F. K. Mahdi, G. Smant, J. E. Kammenga et al., Telomere length variations in aging and age-related diseases Caenorhabditis elegans DAF-16/FOXO transcription factor and its mammalian homologs associate with age-related disease Is Modulation of Oxidative Stress an Answer? The State of the Art of Redox Therapeutic Actions in Neurodegenerative Diseases, CrossRef] [PubMed] 6. Hesp, pp.161-167, 2015.

N. Cartier, C. A. Lewis, R. Zhang, and F. M. Rossi, The role of microglia in human disease: Therapeutic tool or target? Acta Neuropathol, pp.363-380, 2014.

V. Bernhardi, R. Eugenín-von-bernhardi, L. Eugenín, and J. , Microglial cell dysregulation in brain aging and neurodegeneration. Front. Aging Neurosci, 2015.

R. M. Ransohoff and J. Khoury, Microglia in Health and Disease, Cold Spring Harbor Perspectives in Biology, vol.8, issue.1, 2015.
DOI : 10.1101/cshperspect.a020560

Z. Cai, M. D. Hussain, and L. J. Yan, Microglia, neuroinflammation, and beta-amyloid protein in Alzheimer's disease, International Journal of Neuroscience, vol.148, issue.3, pp.307-321, 2014.
DOI : 10.1002/jnr.20992

J. Singh, M. Khan, and I. Singh, Silencing of Abcd1 and Abcd2 genes sensitizes astrocytes for inflammation: implication for X-adrenoleukodystrophy, Journal of Lipid Research, vol.39, issue.1, pp.135-147, 2009.
DOI : 10.1038/nbt0702-682

J. Singh, M. Khan, and I. Singh, Caffeic acid phenethyl ester induces adrenoleukodystrophy (Abcd2) gene in human X-ALD fibroblasts and inhibits the proinflammatory response in Abcd1/2 silenced mouse primary astrocytes, Biochimica et Biophysica Acta (BBA) - Molecular and Cell Biology of Lipids, vol.1831, issue.4, pp.747-758, 2013.
DOI : 10.1016/j.bbalip.2013.01.004

J. López-erauskin, S. Fourcade, J. Galino, M. Ruiz, A. Schlüter et al., Antioxidants halt axonal degeneration in a mouse model of X-adrenoleukodystrophy, Annals of Neurology, vol.256, issue.suppl 1, pp.84-92, 2011.
DOI : 10.1007/s00415-009-5130-6

F. S. Eichler, J. Q. Ren, M. Cossoy, A. M. Rietsch, S. Nagpal et al., Is microglial apoptosis an early pathogenic change in cerebral X-linked adrenoleukodystrophy?, Annals of Neurology, vol.147, issue.6, pp.729-742, 2008.
DOI : 10.4049/jimmunol.178.10.6540

A. S. Paintlia, A. G. Gilg, M. Khan, A. K. Singh, E. Barbosa et al., Correlation of very long chain fatty acid accumulation and inflammatory disease progression in childhood X-ALD: implications for potential therapies, Neurobiology of Disease, vol.14, pp.425-439, 2003.
DOI : 10.1016/j.nbd.2003.08.012

A. Kumar, H. T. Chugani, P. Chakraborty, and A. H. Huq, Evaluation of Neuroinflammation in X-Linked Adrenoleukodystrophy, Pediatric Neurology, vol.44, issue.2, pp.143-146, 2011.
DOI : 10.1016/j.pediatrneurol.2010.10.006

A. Vejux, M. Samadi, and G. Lizard, Contribution of Cholesterol and Oxysterols in the Physiopathology of Cataract: Implication for the Development of Pharmacological Treatments, Journal of Ophthalmology, vol.13, issue.4, p.471947, 2011.
DOI : 10.1016/j.jnutbio.2009.01.001

L. Iuliano, Pathways of cholesterol oxidation via non-enzymatic mechanisms, Chemistry and Physics of Lipids, vol.164, issue.6, pp.457-468, 2011.
DOI : 10.1016/j.chemphyslip.2011.06.006

V. Mutemberezi, O. Guillemot-legris, and G. G. Muccioli, Oxysterols: From cholesterol metabolites to key mediators, Progress in Lipid Research, vol.64
DOI : 10.1016/j.plipres.2016.09.002

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, Journal of Lipid Research, vol.37, issue.2, pp.191-195, 2005.
DOI : 10.1533/9781845698409.5.309

T. Nury, A. Zarrouk, K. Ragot, M. Debbabi, J. M. Riedinger et al., Lizard, G. 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, 2016.

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, Journal of Lipid Research, vol.57, issue.3, pp.361-367, 2016.
DOI : 10.1186/1743-7075-8-23

S. Mukhopadhyay, K. Fellows, R. W. Browne, P. Khare, S. Krishnan-radhakrishnan et al., Interdependence of oxysterols with cholesterol profiles in multiple sclerosis, Multiple Sclerosis Journal, vol.43, issue.3
DOI : 10.1073/pnas.1413561111

H. T. Phan, T. Hata, M. Morita, T. Yoda, T. Hamada et al., The effect of oxysterols on the interaction of Alzheimer's amyloid beta with model membranes, Biochimica et Biophysica Acta (BBA) - Biomembranes, vol.1828, issue.11, pp.2487-2495, 2013.
DOI : 10.1016/j.bbamem.2013.06.021

C. Prunet, T. Montange, A. Véjux, A. Laubriet, J. F. Rohmer et al., Multiplexed flow cytometric analyses of pro- and anti-inflammatory cytokines in the culture media of oxysterol-treated human monocytic cells and in the sera of atherosclerotic patients, Cytometry Part A, vol.61, issue.5, pp.359-373, 2006.
DOI : 10.5551/jat1994.1.Supplemment1_S10

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, Molecular Aspects of Medicine, vol.30, issue.3, pp.153-170, 2009.
DOI : 10.1016/j.mam.2009.02.006

A. Zarrouk, A. Vejux, J. Mackrill, Y. O-'callaghan, M. Hammami et al., Involvement of oxysterols in age-related diseases and ageing processes, Ageing Research Reviews, vol.18, pp.148-162, 2014.
DOI : 10.1016/j.arr.2014.09.006

F. Sedel, D. Bernard, D. M. Mock, and A. Tourbah, Targeting demyelination and virtual hypoxia with high-dose biotin as a treatment for progressive multiple sclerosis, Neuropharmacology, vol.110
DOI : 10.1016/j.neuropharm.2015.08.028

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, The Journal of Steroid Biochemistry and Molecular Biology, vol.169, 2016.
DOI : 10.1016/j.jsbmb.2016.03.029

M. Nordgren and M. Fransen, Peroxisomal metabolism and oxidative stress, Biochimie, vol.98, pp.56-62, 2014.
DOI : 10.1016/j.biochi.2013.07.026

URL : https://lirias.kuleuven.be/bitstream/123456789/412512/2/Nordgren+and+Fransen_2014_Peroxisomal+metabolism+and+oxidative+stress.pdf

C. Lismont, M. Nordgren, P. P. Van-veldhoven, and M. Fransen, Redox interplay between mitochondria and peroxisomes, Frontiers in Cell and Developmental Biology, vol.94, issue.83, p.35
DOI : 10.1152/physrev.00026.2013

URL : http://www.ncbi.nlm.nih.gov/pmc/articles/PMC4444963

S. Manivannan, C. Q. Scheckhuber, M. Veenhuis, and I. J. Van-der-klei, The Impact of Peroxisomes on Cellular Aging and Death, Frontiers in Oncology, vol.2, issue.2, p.50, 2012.
DOI : 10.3389/fonc.2012.00050

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 on 158N murine oligodendrocytes, Biochemical Pharmacology, vol.86, issue.1, pp.67-79, 2013.
DOI : 10.1016/j.bcp.2013.02.028

T. Nury, A. Zarrouk, J. J. Mackrill, M. Samadi, P. Durand et al., Induction of oxiapoptophagy on 158N murine oligodendrocytes treated by 7-ketocholesterol-, 7??-hydroxycholesterol-, or 24(S)-hydroxycholesterol: Protective effects of ??-tocopherol and docosahexaenoic acid (DHA; C22:6 n-3), Steroids, vol.99, pp.194-203, 2015.
DOI : 10.1016/j.steroids.2015.02.003

URL : https://hal.archives-ouvertes.fr/hal-01518110

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; C22:6 n-3), Steroids, vol.99, pp.238-247, 2015.
DOI : 10.1016/j.steroids.2015.01.018

URL : https://hal.archives-ouvertes.fr/hal-01518107

M. Forkink, J. A. Smeitink, R. Brock, P. H. Willems, and W. J. Koopman, Detection and manipulation of mitochondrial reactive oxygen species in mammalian cells, Biochimica et Biophysica Acta (BBA) - Bioenergetics, vol.1797, issue.6-7, pp.1034-1044, 2010.
DOI : 10.1016/j.bbabio.2010.01.022

G. Rimbach, A. M. Minihane, J. Majewicz, A. Fischer, J. Pallauf et al., Regulation of cell signalling by vitamin E, Proceedings of the Nutrition Society, vol.131, issue.04, pp.61-415, 2002.
DOI : 10.1016/S0300-483X(00)00209-2

G. Rimbach, J. Moehring, P. Huebbe, and J. K. Lodge, Gene-Regulatory Activity of ??-Tocopherol, Molecules, vol.15, issue.3, pp.1746-1761, 2010.
DOI : 10.3390/molecules15031746

G. Lizard, C. Miguet, G. Besséde, S. Monier, S. Gueldry et al., Impairment with various antioxidants of the loss of mitochondrial transmembrane potential and of the cytosolic release of cytochrome c occuring during 7-ketocholesterol-induced apoptosis, Free Radical Biology and Medicine, vol.28, issue.5, pp.743-753, 2000.
DOI : 10.1016/S0891-5849(00)00163-5

M. C. Royer, S. Lemaire-ewing, C. Desrumaux, S. Monier, J. P. Pais-de-barros et al., 7-Ketocholesterol Incorporation into Sphingolipid/Cholesterol-enriched (Lipid Raft) Domains Is Impaired by Vitamin E, Journal of Biological Chemistry, vol.8, issue.23, pp.15826-15834, 2009.
DOI : 10.1016/j.freeradbiomed.2007.03.024

URL : http://www.jbc.org/content/284/23/15826.full.pdf

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, Biochemical and Biophysical Research Communications, vol.446, issue.3, pp.2014-714
DOI : 10.1016/j.bbrc.2013.11.081

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 on 158N murine oligodendrocytes, J. Steroid Biochem. Mol. Biol, 2016.

T. Khalfaoui and A. Bron, 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

C. Mascia, M. Maina, E. Chiarpotto, G. Leonarduzzi, G. Poli et al., Proinflammatory effect of cholesterol and its oxidation products on CaCo-2 human enterocyte-like cells: effective protection by epigallocatechin-3-gallate, Free Radical Biology and Medicine, vol.49, issue.12, pp.2049-2057, 2010.
DOI : 10.1016/j.freeradbiomed.2010.09.033

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.234, issue.4, pp.1218-1228, 2015.
DOI : 10.3181/0901-RM-1

M. Allegra, F. Carletti, G. Gambino, M. Tutone, A. Attanzio et al., Crosses the Blood???Brain Barrier and Modulates Neuronal Bioelectric Activity in Rat Hippocampus at Dietary-Consistent Amounts, Journal of Agricultural and Food Chemistry, vol.63, issue.33, pp.7353-7360, 2015.
DOI : 10.1021/acs.jafc.5b02612

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 Ca2+ increase and oxidative stress, British Journal of Nutrition, vol.80, issue.02, pp.230-240, 2013.
DOI : 10.1016/S0891-5849(00)00163-5

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

L. Tesoriere, A. Attanzio, M. Allegra, and M. A. Livrea, Dietary indicaxanthin from cactus pear (Opuntia ficus-indica L. Mill) fruit prevents eryptosis induced by oxysterols in a hypercholesterolaemia-relevant proportion and adhesion of human erythrocytes to endothelial cell layers, British Journal of Nutrition, vol.10, issue.03, pp.368-375, 2015.
DOI : 10.1016/j.freeradbiomed.2007.02.023

P. Gamba, G. Testa, S. Gargiulo, E. Staurenghi, G. Poli et al., Oxidized cholesterol as the driving force behind the development of Alzheimer's disease. Front, Aging Neurosci, vol.7, p.119, 2015.

G. T. Vatassery, H. T. Quach, W. E. Smith, and M. Kuskowski, Alpha and Gamma Tocopherols in Cerebrospinal Fluid and Serum from Older, Male, Human Subjects, Journal of the American College of Nutrition, vol.77, issue.3, pp.233-238, 2004.
DOI : 10.1016/S0304-4165(96)00090-6

R. W. Mitchell, C. L. Edmundson, D. W. Miller, and G. M. Hatch, On the mechanism of oleate transport across human brain microvessel endothelial cells, Journal of Neurochemistry, vol.53, issue.3, pp.1049-1057, 2009.
DOI : 10.1152/ajpendo.00462.2001

E. Stefanoudaki, M. Williams, K. Chartzoulakis, and J. Harwood, Effect of Irrigation on Quality Attributes of Olive Oil, Journal of Agricultural and Food Chemistry, vol.57, issue.15, pp.7048-7055, 2009.
DOI : 10.1021/jf900862w

E. Pouliarekou, A. Badeka, M. Tasioula-margari, S. Kontakos, F. Longobardi et al., Characterization and classification of Western Greek olive oils according to cultivar and geographical origin based on volatile compounds, Journal of Chromatography A, vol.1218, issue.42, pp.7534-7542, 1218.
DOI : 10.1016/j.chroma.2011.07.081

B. Brahim, S. Gargouri, B. Marrakchi, F. Bouaziz, and M. , The Effects of Different Irrigation Treatments on Olive Oil Quality and Composition: A Comparative Study between Treated and Olive Mill Wastewater, Journal of Agricultural and Food Chemistry, vol.64, issue.6
DOI : 10.1021/acs.jafc.5b05030

N. Merchak, E. Bacha, R. Bou-khouzam, T. Rizk, S. Akoka et al., Geoclimatic, morphological, and temporal effects on Lebanese olive oils composition and classification: A 1 H NMR metabolomic study, Food Chemistry, vol.217, issue.217, pp.379-388
DOI : 10.1016/j.foodchem.2016.08.110

A. Incani, G. Serra, A. Atzeri, M. P. Melis, G. Serreli et al., Extra virgin olive oil phenolic extracts counteract the pro-oxidant effect of dietary oxidized lipids in human intestinal cells, Food and Chemical Toxicology, vol.90, pp.171-180, 2016.
DOI : 10.1016/j.fct.2016.02.015

G. Rigane, M. Boukhris, R. B. Salem, S. Sayadi, and M. Bouaziz, Analytical evaluation of two monovarietal virgin olive oils cultivated in the south of Tunisia: Jemri-Bouchouka and Chemlali-Tataouin cultivars, Journal of the Science of Food and Agriculture, vol.93, issue.5, pp.1242-1248, 2013.
DOI : 10.1002/jsfa.6101

T. H. Borges, J. A. Pereira, C. Cabrera-vique, L. Lara, and A. F. Oliveira, Characterization of Arbequina virgin olive oils produced in different regions of Brazil and Spain: Physicochemical properties, oxidative stability and fatty acid profile, Food Chemistry, vol.215, pp.454-462, 2017.
DOI : 10.1016/j.foodchem.2016.07.162

A. Nakbi, W. Tayeb, A. Grissa, M. Issaoui, S. Dabbou et al., Effects of olive oil and its fractions on oxidative stress and the liver's fatty acid composition in 2,4-Dichlorophenoxyacetic acid-treated rats, Nutrition & Metabolism, vol.7, issue.1, p.80, 2010.
DOI : 10.1186/1743-7075-7-80

A. Nakbi, W. Tayeb, S. Dabbou, M. Issaoui, A. K. Grissa et al., Dietary olive oil effect on antioxidant status and fatty acid profile in the erythrocyte of 2,4-D- exposed rats, Lipids in Health and Disease, vol.9, issue.1, p.89, 2010.
DOI : 10.1186/1476-511X-9-89

N. Amel, T. Wafa, D. Samia, B. Yousra, C. Issam et al., Extra virgin olive oil modulates brain docosahexaenoic acid level and oxidative damage caused by 2,4-Dichlorophenoxyacetic acid in rats, Journal of Food Science and Technology, vol.15, issue.6, pp.1454-1464, 2016.
DOI : 10.1002/med.1028

URL : http://www.ncbi.nlm.nih.gov/pmc/articles/PMC4984713

E. Gray, C. Rice, K. Hares, J. Redondo, K. Kemp et al., Reductions in neuronal peroxisomes in multiple sclerosis grey matter, Multiple Sclerosis Journal, vol.51, issue.6, pp.651-659, 2014.
DOI : 10.1177/1352458508091370

R. J. Wanders and H. Waterham, Biochemistry of Mammalian Peroxisomes Revisited, Annual Review of Biochemistry, vol.75, issue.1, pp.295-332, 2006.
DOI : 10.1146/annurev.biochem.74.082803.133329

M. Baes and P. Aubourg, Peroxisomes, Myelination, and Axonal Integrity in the CNS, The Neuroscientist, vol.28, issue.4, pp.367-379, 2009.
DOI : 10.1523/JNEUROSCI.0458-08.2008

C. M. Kassmann, C. Lappe-siefke, M. Baes, B. Brügger, A. Mildner et al., Axonal loss and neuroinflammation caused by peroxisome-deficient oligodendrocytes, Nature Genetics, vol.7, issue.8, pp.969-976, 2007.
DOI : 10.1139/o59-099

URL : http://pubman.mpdl.mpg.de/pubman/item/escidoc:595611/component/escidoc:595610/319657.pdf

A. Bottelbergs, S. Verheijden, L. Hulshagen, D. H. Gutmann, S. Goebbels et al., Axonal integrity in the absence of functional peroxisomes from projection neurons and astrocytes, Glia, vol.12, pp.1532-1543, 2010.
DOI : 10.1016/S1388-1981(00)00077-9

B. Wang, P. P. Van-veldhoven, C. Brees, N. Rubio, M. Nordgren et al., Agostinis, P.; Fransen, M. Mitochondria are targets for peroxisome-derived oxidative stress in cultured mammalian cells. Free Radic, Biol. Med, vol.65, pp.882-894, 2013.

E. Baumgart, I. Vanhorebeek, M. Grabenbauer, M. Borgers, P. E. Declercq et al., Mitochondrial Alterations Caused by Defective Peroxisomal Biogenesis in a Mouse Model for Zellweger Syndrome (PEX5 Knockout Mouse), The American Journal of Pathology, vol.159, issue.4, pp.1477-1494, 2001.
DOI : 10.1016/S0002-9440(10)62534-5

R. Dirkx, I. Vanhorebeek, K. Martens, A. Schad, M. Grabenbauer et al., Absence of peroxisomes in mouse hepatocytes causes mitochondrial and ER abnormalities, Hepatology, vol.274, issue.4, pp.868-878, 2005.
DOI : 10.1042/bj3300889

I. R. Rodriguez, M. E. Clark, J. W. Lee, and C. Curcio, 7-ketocholesterol accumulates in ocular tissues as a consequence of aging and is present in high levels in drusen, Experimental Eye Research, vol.128, pp.151-155, 2014.
DOI : 10.1016/j.exer.2014.09.009

M. Indaram, W. Ma, L. Zhao, R. N. Fariss, I. R. Rodriguez et al., 7-Ketocholesterol Increases Retinal Microglial Migration, Activation and Angiogenicity: A Potential Pathogenic Mechanism Underlying Age-related Macular Degeneration, Scientific Reports, vol.3, issue.9, p.9144, 2015.
DOI : 10.1038/nprot.2008.172

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?, Biochimica et Biophysica Acta (BBA) - Molecular Basis of Disease, vol.1822, issue.9, pp.1475-1488, 2012.
DOI : 10.1016/j.bbadis.2012.02.005

L. Morató, E. Bertini, D. Verrigni, A. Ardissone, M. Ruiz et al., Mitochondrial dysfunction in central nervous system white matter disorders, Glia, vol.58, issue.Suppl 1, pp.1878-1894, 2014.
DOI : 10.1002/glia.21052

S. Fourcade, I. Ferrer, and A. Pujol, Oxidative stress, mitochondrial and proteostasis malfunction in adrenoleukodystrophy: A paradigm for axonal degeneration, Free Radical Biology and Medicine, vol.88, pp.18-29, 2015.
DOI : 10.1016/j.freeradbiomed.2015.05.041

G. R. Campbell, J. T. Worrall, and D. J. Mahad, The central role of mitochondria in axonal degeneration in multiple sclerosis, Multiple Sclerosis Journal, vol.485, issue.14, pp.1806-1813
DOI : 10.1093/brain/awh118

J. M. Zingg, Vitamin E: A Role in Signal Transduction, Annual Review of Nutrition, vol.35, issue.1, pp.135-173
DOI : 10.1146/annurev-nutr-071714-034347

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.
DOI : 10.1016/j.freeradbiomed.2011.05.017

URL : http://www.ncbi.nlm.nih.gov/pmc/articles/PMC3156342

K. Ragot, D. Delmas, A. Athias, T. Nury, and M. Baarine, ??-Tocopherol impairs 7-ketocholesterol-induced caspase-3-dependent apoptosis involving GSK-3 activation and Mcl-1 degradation on 158N murine oligodendrocytes, Chemistry and Physics of Lipids, vol.164, issue.6, pp.469-478, 2011.
DOI : 10.1016/j.chemphyslip.2011.04.014

C. J. Yeh, B. L. His, and W. P. Faulk, Propidium iodide as a nuclear marker in immunofluorescence. II. Use with cellular identification and viability studies, Journal of Immunological Methods, vol.43, issue.3, pp.269-275, 1981.
DOI : 10.1016/0022-1759(81)90174-5

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.43, issue.3, pp.275-283, 1995.
DOI : 10.1177/25.7.70450

L. Ryan, Y. C. O-'callaghan, and N. M. O-'brien, Comparison of the apoptotic processes induced by the oxysterols 7??-hydroxycholesterol and cholesterol-5??,6??-epoxide, Cell Biology and Toxicology, vol.456, issue.5, pp.313-323, 2004.
DOI : 10.1016/0165-1161(91)90247-6

D. A. Mickle, R. K. Li, R. D. Weisel, L. C. Tumiati, and T. W. Wu, Water-soluble antioxidant specificity against free radical injury using cultured human ventricular myocytes and fibroblasts and saphenous vein endothelial cells, Journal of Molecular and Cellular Cardiology, vol.22, issue.11, pp.1297-1304, 1990.
DOI : 10.1016/0022-2828(90)90065-A

R. Albertini and P. M. Abuja, Prooxidant and antioxidant properties of Trolox C, analogue of vitamin E, in oxidation of low-density lipoprotein, Free Radical Research, vol.3, issue.3, pp.181-188, 1999.
DOI : 10.1021/tx00014a001

R. Orihuela, C. A. Mcpherson, and G. J. Harry, Microglial M1/M2 polarization and metabolic states, British Journal of Pharmacology, vol.116, issue.Suppl. 1, pp.649-665, 2016.
DOI : 10.1182/blood-2010-02-258558

URL : http://onlinelibrary.wiley.com/doi/10.1111/bph.13139/pdf

A. Vejux, S. Guyot, T. Montange, J. M. Riedinger, E. Kahn et al., Phospholipidosis and down-regulation of the PI3-K/PDK-1/Akt signalling pathway are vitamin E inhibitable events associated with 7-ketocholesterol-induced apoptosis, The Journal of Nutritional Biochemistry, vol.20, issue.1, pp.45-61, 2009.
DOI : 10.1016/j.jnutbio.2007.12.001

R. González, J. A. Collado, S. Nell, J. Briceño, M. J. Tamayo et al., Cytoprotective properties of ??-tocopherol are related to gene regulation in cultured d-galactosamine-treated human hepatocytes, Free Radical Biology and Medicine, vol.43, issue.10, pp.1439-1452, 2007.
DOI : 10.1016/j.freeradbiomed.2007.07.023

Y. Y. Tyurina, M. A. Tungekar, M. Y. Jung, V. A. Tyurin, J. S. Greenberger et al., Mitochondria targeting of non-peroxidizable triphenylphosphonium conjugated oleic acid protects mouse embryonic cells against apoptosis: Role of cardiolipin remodeling, FEBS Letters, vol.45, issue.3, pp.235-241, 2012.
DOI : 10.1021/bi0525573

D. Sommerweiss, T. Gorski, S. Richter, A. Garten, and W. Kiess, Oleate rescues INS-1E ??-cells from palmitate-induced apoptosis by preventing activation of the unfolded protein response, Biochemical and Biophysical Research Communications, vol.441, issue.4, pp.770-776, 2013.
DOI : 10.1016/j.bbrc.2013.10.130

M. Servili, Nutrigenomics of extra-virgin olive oil: A review, Biofactors, 2016.

L. Fernández-del-río, E. Gutiérrez-casado, A. Varela-lópez, and J. M. Villalba, Olive Oil and the Hallmarks of Aging, Molecules, vol.110, issue.2, p.163, 2016.
DOI : 10.1021/jf404421p

J. J. Lemasters, Dying a Thousand Deaths: Redundant Pathways From Different Organelles to Apoptosis and Necrosis, Gastroenterology, vol.129, issue.1, pp.351-360, 2005.
DOI : 10.1053/j.gastro.2005.06.006

P. S. Tang, M. Mura, R. Seth, and M. Liu, Acute lung injury and cell death: how many ways can cells die?, AJP: Lung Cellular and Molecular Physiology, vol.294, issue.4, pp.632-641, 2008.
DOI : 10.1152/ajplung.00262.2007

URL : http://ajplung.physiology.org/content/ajplung/294/4/L632.full.pdf

M. Baarine, P. Andréoletti, A. Athias, T. Nury, A. Zarrouk et al., Evidence of oxidative stress in very long chain fatty acid ??? Treated oligodendrocytes and potentialization of ROS production using RNA interference-directed knockdown of ABCD1 and ACOX1 peroxisomal proteins, Neuroscience, vol.213, pp.1-18, 2012.
DOI : 10.1016/j.neuroscience.2012.03.058

J. Kou, G. G. Kovacs, R. Höftberger, W. Kulik, A. Brodde et al., Peroxisomal alterations in Alzheimer???s disease, Acta Neuropathologica, vol.35, issue.Suppl 1, pp.271-283, 2011.
DOI : 10.1503/jpn.090059

O. Rouaud, M. Frih, and G. Lizard, 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.

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 Neurology, vol.101, issue.5, pp.15-154, 2015.
DOI : 10.1111/j.1471-4159.2007.04487.x

S. Kemp, F. L. Theodoulou, and R. J. Wanders, Mammalian peroxisomal ABC transporters: from endogenous substrates to pathology and clinical significance, British Journal of Pharmacology, vol.32, issue.7, pp.1753-1766, 2011.
DOI : 10.1385/CBB:32:1-3:239

URL : http://www.ncbi.nlm.nih.gov/pmc/articles/PMC3246701

M. Schrader, S. Grille, H. D. Fahimi, and M. Islinger, Peroxisome Interactions and Cross-Talk with Other Subcellular Compartments in Animal Cells, Subcell. Biochem, vol.69, pp.1-22, 2013.
DOI : 10.1007/978-94-007-6889-5_1

D. Effelsberg, L. D. Cruz-zaragoza, W. Schliebs, and R. Erdmann, Pex9p is a novel yeast peroxisomal import receptor for PTS1-proteins, Journal of Cell Science, vol.2016, issue.129, pp.4057-4066
DOI : 10.1242/jcs.195271

F. Sedaghat, M. Jessri, M. Behrooz, M. Mirghotbi, and B. Rashidkhani, Mediterranean diet adherence and risk of multiple sclerosis: A case-control study. Asia Pac, J. Clin. Nutr, vol.25, pp.377-384, 2016.

A. Haghikia, S. Jörg, A. Duscha, J. Berg, A. Manzel et al., Dietary Fatty Acids Directly Impact Central Nervous System Autoimmunity via the Small Intestine, Dietary fatty acids directly impact central nervous system autoimmunity via the small intestine, pp.817-829, 2015.
DOI : 10.1016/j.immuni.2015.09.007

URL : http://doi.org/10.1016/j.immuni.2016.04.006

G. Null, L. Pennesi, and M. Feldman, Nutrition and Lifestyle Intervention on Mood and Neurological Disorders, Journal of Evidence-Based Complementary & Alternative Medicine, vol.10, issue.12, 2016.
DOI : 10.1089/acm.2004.10.891

F. Luchetti, B. Canonico, E. Cesarini, M. Betti, L. Galluzzi et al., 7-Ketocholesterol and 5,6-secosterol induce human endothelial cell dysfunction by differential mechanisms, Steroids, vol.99, pp.204-211, 2015.
DOI : 10.1016/j.steroids.2015.02.008

E. Pedruzzi, C. Guichard, V. Ollivier, F. Driss, M. Fay et al., NAD(P)H Oxidase Nox-4 Mediates 7-Ketocholesterol-Induced Endoplasmic Reticulum Stress and Apoptosis in Human Aortic Smooth Muscle Cells, Molecular and Cellular Biology, vol.24, issue.24, pp.10703-10717, 2004.
DOI : 10.1128/MCB.24.24.10703-10717.2004

C. Helmschrodt, S. Becker, J. Schröter, M. Hecht, G. Aust et al., Fast LC???MS/MS analysis of free oxysterols derived from reactive oxygen species in human plasma and carotid plaque, Clinica Chimica Acta, vol.425, pp.3-8, 2013.
DOI : 10.1016/j.cca.2013.06.022

C. Miguet, S. Monier, A. Bettaieb, A. Athias, G. Besséde et al., Ceramide generation occurring during 7??-hydroxycholesterol- and 7-ketocholesterol-induced apoptosis is caspase independent and is not required to trigger cell death, Cell Death and Differentiation, vol.8, issue.1, pp.83-99, 2001.
DOI : 10.1038/sj.cdd.4400792

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.

G. Poli, Survival signalingelicited by 27-hydroxycholesterol through the combined modula-tion of cellular redox state and ERK/Akt phosphorylation. Free Radic, Biol. Med, vol.2014, issue.77, pp.376-385

A. Teubert, J. Thome, A. Büttner, J. Richter, and G. Irmisch, Elevated oleic acid serum concentrations in patients suffering from alcohol dependence, Journal of Molecular Psychiatry, vol.1, issue.1, p.13, 2013.
DOI : 10.1186/1476-511X-10-234

URL : http://www.ncbi.nlm.nih.gov/pmc/articles/PMC4224011

T. Moilanen and T. Nikkari, The effect of storage on the fatty acid composition of human serum, Clinica Chimica Acta, vol.114, issue.1, pp.111-116, 1981.
DOI : 10.1016/0009-8981(81)90235-7

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, Journal of Chromatography B: Biomedical Sciences and Applications, vol.751, issue.2, pp.297-303, 2001.
DOI : 10.1016/S0378-4347(00)00488-6

I. F. Benzie and J. J. Strain, The Ferric Reducing Ability of Plasma (FRAP) as a Measure of ???Antioxidant Power???: The FRAP Assay, Analytical Biochemistry, vol.239, issue.1, pp.70-76, 1996.
DOI : 10.1006/abio.1996.0292

M. Oyaizu, Studies on products of browning reaction. Antioxidative activities of products of browning reaction prepared from glucosamine., The Japanese Journal of Nutrition and Dietetics, vol.44, issue.6, pp.307-316, 1986.
DOI : 10.5264/eiyogakuzashi.44.307

O. P. Sharma and T. K. Bhat, DPPH antioxidant assay revisited, Food Chemistry, vol.113, issue.4, pp.1202-1205, 2009.
DOI : 10.1016/j.foodchem.2008.08.008

S. B. Kedare and R. P. Singh, Genesis and development of DPPH method of antioxidant assay, Journal of Food Science and Technology, vol.50, issue.1, pp.412-422, 2011.
DOI : 10.1021/jf0206163

T. Mosmann, Rapid colorimetric assay for cellular growth and survival: Application to proliferation and cytotoxicity assays, Journal of Immunological Methods, vol.65, issue.1-2, pp.55-63, 1983.
DOI : 10.1016/0022-1759(83)90303-4

J. Pérez-hernández, V. J. Zaldívar-machorro, D. Villanueva-porras, E. Vega-Ávila, and A. Chavarría, A Potential Alternative against Neurodegenerative Diseases: Phytodrugs, Oxidative Medicine and Cellular Longevity, vol.131, issue.3, p.8378613, 2016.
DOI : 10.1016/0197-0186(95)00117-4