M. G. Vander-heiden, L. C. Cantley, C. B. Thompson, D. Understanding-the-warburg-effect-hanahan, R. A. Weinberg et al., The Metabolic Requirements of Cell Proliferation Hallmarks of cancer: the next generation Hypersensitization of Tumor Cells to Glycolytic Inhibitors ? Hypoxia increases tumor cell sensitivity to glycolytic inhibitors: a strategy for solid tumor therapy (Model C) Glycolysis inhibition for anticancer treatment Inhibition of glycolysis in cancer cells: a novel strategy to overcome drug resistance associated with mitochondrial respiratory defect and hypoxia, Science Cell Biochemistry (Mosc. Biochem. Pharmacol. Oncogene Cancer Res C, vol.324, issue.2534627, pp.1029-1033, 2001.

E. White, R. S. Dipaola, D. Singh, A. K. Banerji, B. S. Dwarakanath et al., Targeting tumor metabolism with 2-deoxyglucose in patients with castrate-resistant prostate cancer and advanced malignancies. The Prostate Optimizing Cancer Radiotherapy with 2-Deoxy-D-Glucose: Dose Escalation Studies in Patients with Glioblastoma Multiforme, Strahlenther. Onkol. Int. J. Radiat. Oncol. Biol. Phys, vol.70, issue.351, pp.1388-1394, 1996.

L. E. Raez, K. Papadopoulos, A. D. Ricart, E. G. Chiorean, R. S. Dipaola et al., A phase I dose-escalation trial of 2-deoxy-d-glucose alone or combined with docetaxel in patients with advanced solid tumors, Cancer Chemotherapy and Pharmacology, vol.83, issue.2, pp.71-523
DOI : 10.1111/j.1600-0773.1998.tb01448.x

E. A. Struys, C. Joppich, S. Krobitsch, C. Jakobs, and H. Lehrach, A catabolic block does not sufficiently explain how 2-deoxy-D-glucose inhibits cell growth, Proc. Natl. Acad. Sci, pp.105-17807, 2008.

). Donnell, A. F. Mccartney, R. R. Chandrashekarappa, D. G. Zhang, B. B. Thorner et al., C. 2-Deoxyglucose Impairs Saccharomyces cerevisiae Growth by Stimulating Snf1-Regulated and ?-Arrestin-Mediated Trafficking of Hexose Transporters 1 and 3, Mol. Cell. Biol, vol.2015, issue.126, pp.35-939
URL : https://hal.archives-ouvertes.fr/in2p3-00353281

H. T. Kang and E. S. Hwang, 2-Deoxyglucose: An anticancer and antiviral therapeutic, but not any more a low glucose mimetic, Life Sciences, vol.78, issue.12, pp.78-1392, 2006.
DOI : 10.1016/j.lfs.2005.07.001

R. R. Mccartney, D. G. Chandrashekarappa, B. B. Zhang, and M. C. Schmidt, Genetic Analysis of Resistance and Sensitivity to 2-Deoxyglucose in Saccharomyces cerevisiae, Genetics, vol.198, issue.2, pp.635-646
DOI : 10.1534/genetics.114.169060

H. T. Jaspers and J. Van-steveninck, Transport-associated phosphorylation of 2-deoxy-d-glucose in Saccharomyces fragilis, Biochimica et Biophysica Acta (BBA) - Biomembranes, vol.406, issue.3, pp.406-370, 1975.
DOI : 10.1016/0005-2736(75)90017-6

Z. Loba and P. K. Maitra, Resistance to 2-deoxyglucose in yeast: A direct selection of mutants lacking glucose-phosphorylating enzymes, MGG Molecular & General Genetics, vol.13, issue.3, pp.297-300, 1977.
DOI : 10.1007/BF00268666

V. Farka?, A. Svoboda, and ?. Bauer, Secretion of cell-wall glycoproteins by yeast protoplasts. Effect of 2-deoxy-d-glucose and cycloheximide, Biochemical Journal, vol.118, issue.5, pp.755-758, 1970.
DOI : 10.1042/bj1180755

Z. Kratky, P. Biely, and ?. Bauer, Mechanism of 2-Deoxy-d-glucose Inhibition of Cell-Wall Polysaccharide and Glycoprotein Biosyntheses in Saccharomyces cerevisiae, European Journal of Biochemistry, vol.65, issue.2, pp.459-467, 1975.
DOI : 10.1099/00221287-65-3-291

R. Datema and R. T. Schwarz, Formation of 2-Deoxyglucose-Containing Lipid-Linked Oligosaccharides. Interference with Glycosylation of Glycoproteins, European Journal of Biochemistry, vol.33, issue.3, pp.505-516, 1978.
DOI : 10.1271/bbb1961.39.2089

H. Xi, M. Kurtoglu, H. Liu, M. Wangpaichitr, M. You et al., 2-Deoxy-d-glucose activates autophagy via endoplasmic reticulum stress rather than ATP depletion, Cancer Chemotherapy and Pharmacology, vol.26, issue.4, pp.899-910, 2011.
DOI : 10.1016/S0968-0004(01)01908-9

URL : http://europepmc.org/articles/pmc3093301?pdf=render

H. Xi, J. C. Barredo, J. R. Merchan, and T. J. Lampidis, Endoplasmic reticulum stress induced by 2-deoxyglucose but not glucose starvation activates AMPK through CaMKK?? leading to autophagy, Biochemical Pharmacology, vol.85, issue.10, pp.85-1463
DOI : 10.1016/j.bcp.2013.02.037

S. Ramirez-peinado, F. Alcazar-limones, L. Lagares-tena, N. Mjiyad, A. Caro-maldonado et al., Munoz-Pinedo, C. 2-Deoxyglucose Induces Noxa-Dependent Apoptosis in Alveolar Rhabdomyosarcoma, Cancer Res, issue.21, pp.71-6796, 2011.

S. Mahamoud, Y. Aite, M. Martin, C. Zhadobov, M. Sauleau et al., Additive Effects of Millimeter Waves and 2-Deoxyglucose Co-Exposure on the Human Keratinocyte Transcriptome, PLOS ONE, vol.2016, issue.8, pp.11-0160810
URL : https://hal.archives-ouvertes.fr/hal-01367149

M. ?uperlovi?-culf, D. A. Barnett, and A. S. Culf, Chute, I. Cell culture metabolomics: applications and future directions, Drug Discov. Today, issue.15, pp.15-610, 2010.

U. Ceglarek, C. Shackleton, F. Z. Stancyk, and J. Adamski, Steroid profiling and analytics: Going towards sterome, The Journal of Steroid Biochemistry and Molecular Biology, vol.121, issue.3-5, pp.479-480, 2010.
DOI : 10.1016/j.jsbmb.2010.07.002

R. Wang-sattler, Y. Yu, K. Mittelstrass, E. Lattka, E. Altmaier et al., Metabolic Profiling Reveals Distinct Variations Linked to Nicotine Consumption in Humans ??? First Results from the KORA Study, Metabolic Profiling Reveals Distinct Variations Linked to Nicotine Consumption in Humans ? First Results from the KORA Study, p.3863, 2008.
DOI : 10.1371/journal.pone.0003863.s007

URL : https://doi.org/10.1371/journal.pone.0003863

E. Altmaier, G. Kastenmüller, W. Römisch-margl, B. Thorand, K. M. Weinberger et al., Variation in the human lipidome associated with coffee consumption as revealed by quantitative targeted metabolomics, Molecular Nutrition & Food Research, vol.163, issue.11, pp.53-1357, 2009.
DOI : 10.1161/01.ATV.20.12.2614

D. B. Kell and R. Goodacre, Metabolomics and systems pharmacology: why and how to model the human metabolic network for drug discovery, Drug Discovery Today, vol.19, issue.2, pp.171-182
DOI : 10.1016/j.drudis.2013.07.014

URL : https://doi.org/10.1016/j.drudis.2013.07.014

A. Sreekumar, L. M. Poisson, T. M. Rajendiran, A. P. Khan, Q. Cao et al., Metabolomic profiles delineate potential role for sarcosine in prostate cancer progression, Nature, vol.12, issue.7231, pp.457-910, 2009.
DOI : 10.1038/nature07762

URL : http://europepmc.org/articles/pmc2724746?pdf=render

D. E. Misek and E. H. Kim, Protein Biomarkers for the Early Detection of Breast Cancer, International Journal of Proteomics, vol.51, issue.2, pp.1-9, 2011.
DOI : 10.1021/ac100131e

R. Wang-sattler, Z. Yu, C. Herder, A. C. Messias, A. Floegel et al., Novel biomarkers for pre-diabetes identified by metabolomics, Molecular Systems Biology, vol.41, p.615, 2012.
DOI : 10.1007/s11306-010-0203-1

URL : http://msb.embopress.org/content/msb/8/1/615.full.pdf

T. Hyötyläinen, Novel methodologies in metabolic profiling with a focus on molecular diagnostic applications, Expert Review of Molecular Diagnostics, vol.883, issue.5, pp.527-538
DOI : 10.1093/nar/gkl923

W. B. Dunn and D. I. Ellis, Metabolomics: Current analytical platforms and methodologies, TrAC Trends Anal. Chem, vol.24, issue.4, pp.285-294, 2005.

J. Folch, M. Lees, and G. H. Sloane-stanley, A simple method for the isolation and purification of total lipides from animal tissues, J. Biol. Chem, vol.226, pp.497-509, 1957.

E. G. Bligh and W. J. Dyer, A rapid method of total lipid extraction and purification, Can. J. Biochem. Physiol, issue.8, pp.37-911, 1959.
DOI : 10.1139/y59-099

A. D. Watson, Lipidomics: a global approach to lipid analysis in biological systems, Journal of Lipid Research, vol.25, issue.69, pp.47-2101, 2006.
DOI : 10.1161/01.CIR.0000130844.01174.55

K. Dettmer, N. Nürnberger, H. Kaspar, M. A. Gruber, M. F. Almstetter et al., Metabolite extraction from adherently growing mammalian cells for metabolomics studies: optimization of harvesting and extraction protocols, Analytical and Bioanalytical Chemistry, vol.97, issue.410, pp.399-1127, 2011.
DOI : 10.1093/jxb/eri069

V. Matyash, G. Liebisch, T. V. Kurzchalia, A. Shevchenko, and D. Schwudke, -butyl ether for high-throughput lipidomics, Journal of Lipid Research, vol.33, issue.5, pp.49-1137, 2008.
DOI : 10.1016/j.jasms.2004.07.009

URL : http://www.jlr.org/content/49/5/1137.full.pdf

J. F. Xiao, B. Zhou, and H. W. Ressom, Metabolite identification and quantitation in LC-MS/MS-based metabolomics, TrAC Trends in Analytical Chemistry, vol.32, pp.1-14, 2012.
DOI : 10.1016/j.trac.2011.08.009

URL : http://europepmc.org/articles/pmc3278153?pdf=render

J. Trygg, E. Holmes, and T. Lundstedt, Chemometrics in Metabonomics, Journal of Proteome Research, vol.6, issue.2, pp.469-479, 2007.
DOI : 10.1021/pr060594q

URL : http://www.metabolomics.se/Courses/Systems Biology/References/Trygg et al_2007 Chemometrics in metabonomics_review.pdf

L. Quément, C. Nicolaz, C. N. Habauzit, D. Zhadobov, M. Sauleau et al., Impact of 60- GHz millimeter waves and corresponding heat effect on endoplasmic reticulum stress sensor gene expression: ER Stress Under MMW Exposure, Bioelectromagnetics, vol.2014, issue.6, pp.35-444

A. Halama, Metabolomics in cell culture???A strategy to study crucial metabolic pathways in cancer development and the response to treatment, Archives of Biochemistry and Biophysics, vol.564, pp.100-109, 2014.
DOI : 10.1016/j.abb.2014.09.002

G. Kerdivel, A. Boudot, D. Habauzit, F. Percevault, F. Demay et al., Activation of the MKL1/actin signaling pathway induces hormonal escape in estrogen-responsive breast cancer cell lines, Molecular and Cellular Endocrinology, vol.390, issue.1-2, pp.34-44
DOI : 10.1016/j.mce.2014.03.009

URL : https://hal.archives-ouvertes.fr/hal-01061363

Z. León, J. C. García-cañaveras, M. T. Donato, and A. Lahoz, Mammalian cell metabolomics: Experimental design and sample preparation, ELECTROPHORESIS, vol.708, pp.2762-2775, 2013.
DOI : 10.1007/978-1-61737-985-7_15

U. Batista, M. Garvas, M. Nemec, M. Schara, P. Verani? et al., Effects of different detachment procedures on viability, nitroxide reduction kinetics and plasma membrane heterogeneity of V-79 cells, Cell Biology International, vol.103, issue.6, pp.34-663, 2010.
DOI : 10.1016/j.abb.2006.09.030

G. Paglia, S. Hrafnsdóttir, M. Magnúsdóttir, R. M. Fleming, S. Thorlacius et al., Thiele, I. Monitoring metabolites consumption and secretion in cultured cells using ultraperformance liquid chromatography quadrupole?time of flight mass spectrometry (UPLC?Q?ToF- MS), Anal. Bioanal. Chem, vol.2012, issue.3, pp.402-1183

R. Wehrens, J. A. Hageman, F. Van-eeuwijk, R. Kooke, P. J. Flood et al., Improved batch correction in untargeted MS-based metabolomics Large-scale untargeted LC-MS metabolomics data correction using between-batch feature alignment and cluster-based within-batch signal intensity drift correction, Metabolomics Metabolomics, vol.2016, issue.201611, pp.88-136

A. J. Chetwynd, A. Sada, S. G. Holt, and E. M. Hill, Use of a pre-analysis osmolality normalisation method to correct for variable urine concentrations and for improved metabolomic analyses, Journal of Chromatography A, vol.1431, pp.1431-103, 2016.
DOI : 10.1016/j.chroma.2015.12.056

C. A. Smith, E. J. Want, G. O-'maille, R. Abagyan, G. Siuzdak et al., Processing Mass Spectrometry Data for Metabolite Profiling Using Nonlinear Peak Alignment, Matching, and Identification, pp.78-779, 2006.

C. A. Smith, G. O-'maille, E. J. Want, C. Qin, S. A. Trauger et al., METLIN, Therapeutic Drug Monitoring, vol.27, issue.6, pp.27-747, 2005.
DOI : 10.1097/01.ftd.0000179845.53213.39

R. Tautenhahn, G. J. Patti, D. Rinehart, G. Siuzdak, and . Online, A Web-Based Platform to Process Untargeted Metabolomic Data, Anal. Chem, vol.2012, issue.8411, pp.5035-5039

D. S. Wishart, D. Tzur, C. Knox, R. Eisner, A. C. Guo et al., HMDB: the Human Metabolome Database, Database), pp.35-521, 2007.
DOI : 10.1093/nar/gkl923

URL : https://hal.archives-ouvertes.fr/hal-01712873

D. S. Wishart, T. Jewison, A. C. Guo, M. Wilson, C. Knox et al., 0--The Human Metabolome Database in 2013, Nucleic Acids Res, issue.D1, pp.41-801, 2013.

E. Fahy, S. Subramaniam, R. C. Murphy, M. Nishijima, C. R. Raetz et al., Update of the LIPID MAPS comprehensive classification system for lipids, Journal of Lipid Research, vol.37, issue.Supplement, pp.50-59, 2008.
DOI : 10.1016/0031-9422(88)83013-9

F. Desmots, R. Sauleau, D. Thouroude, D. Michel, and Y. Le-drean, Absence of direct effect of low-power millimeter-wave radiation at 60.4 GHz on endoplasmic reticulum stress, Cell Biol. Toxicol, issue.565, pp.25-471, 2009.
URL : https://hal.archives-ouvertes.fr/hal-00406594

J. Ikeda, S. Kaneda, K. Kuwabara, S. Ogawa, T. Kobayashi et al., Cloning and Expression of cDNA Encoding the Human 150 kDa Oxygen-Regulated Protein, ORP150, Biochemical and Biophysical Research Communications, vol.230, issue.1, pp.94-99, 1997.
DOI : 10.1006/bbrc.1996.5890

H. Zhang, M. Lu, H. B. Qi, and J. H. Zhang, Up-regulation of glucose regulated protein 78 induced by 2-deoxy-glucose plays a protective role for fetal rat cerebral neuron following intrauterine distress, Zhonghua Fu Chan Ke Za Zhi, vol.43, pp.356-360, 2008.

K. Nagano, H. Kano, H. Arito, S. Yamamoto, and T. Matsushima, Enhancement of Renal Carcinogenicity by Combined Inhalation and Oral Exposures to Chloroform in Male Rats, Journal of Toxicology and Environmental Health, Part A, vol.13, issue.20, pp.69-1827, 2006.
DOI : 10.1539/joh.44.283

P. Schmid, E. Hunter, and J. Calvert, Extraction and purification of lipids. III. Serious limitations of chloroform and chloroform-methanol in lipid investigations, Physiol. Chem. Phys. Med. NMR, vol.5, pp.151-155, 1973.

L. Li, X. Lu, J. Zhao, J. Zhang, Y. Zhao et al., Lipidome and metabolome analysis of fresh tobacco leaves in different geographical regions using liquid chromatography???mass spectrometry, Analytical and Bioanalytical Chemistry, vol.88, issue.7, pp.407-5009
DOI : 10.2307/3558330

L. Li, J. Zhao, Y. Zhao, X. Lu, Z. Zhou et al., Comprehensive investigation of tobacco leaves during natural early senescence via multi-platform metabolomics analyses, 37976. (63) Cajka, T.; Fiehn, O. Toward Merging Untargeted and Targeted Methods in Mass Spectrometry- Based Metabolomics and Lipidomics, pp.524-545
DOI : 10.1021/ac051437y

K. Nzoughet, J. Bocca, C. Simard, G. Prunier-mirebeau, D. Chao-de-la-barca et al., A Nontargeted UHPLC-HRMS Metabolomics Pipeline for Metabolite Identification: Application to Cardiac Remote Ischemic Preconditioning, Anal. Chem, vol.2017, issue.3, pp.89-2138

E. J. Want, I. D. Wilson, H. Gika, G. Theodoridis, R. S. Plumb et al., Global metabolic profiling procedures for urine using UPLC???MS, Nature Protocols, vol.406, issue.6, pp.1005-1018, 2010.
DOI : 10.1016/S0378-4347(00)00360-1

N. Kim, S. M. Ryu, D. Lee, J. W. Lee, E. Seo et al., A metabolomic approach to determine the geographical origins of Anemarrhena asphodeloides by using UPLC???QTOF MS, Journal of Pharmaceutical and Biomedical Analysis, vol.92, pp.47-52, 2014.
DOI : 10.1016/j.jpba.2013.12.040

S. Grösch, S. Schiffmann, and G. Geisslinger, Chain length-specific properties of ceramides, Progress in Lipid Research, vol.51, issue.1, pp.50-62
DOI : 10.1016/j.plipres.2011.11.001

B. J. Pettus, C. E. Chalfant, and Y. A. Hannun, Ceramide in apoptosis: an overview and current perspectives, Biochimica et Biophysica Acta (BBA) - Molecular and Cell Biology of Lipids, vol.1585, issue.2-3, pp.1585-114, 2002.
DOI : 10.1016/S1388-1981(02)00331-1

L. J. Siskind, R. N. Kolesnick, and M. Colombini, Ceramide Channels Increase the Permeability of the Mitochondrial Outer Membrane to Small Proteins, Journal of Biological Chemistry, vol.59, issue.30, pp.277-26796, 2002.
DOI : 10.1016/S0014-5793(98)00744-3

URL : http://www.jbc.org/content/277/30/26796.full.pdf

Y. Uchida, Ceramide signaling in mammalian epidermis, Biochimica et Biophysica Acta (BBA) - Molecular and Cell Biology of Lipids, vol.1841, issue.3, pp.1841-453
DOI : 10.1016/j.bbalip.2013.09.003

URL : http://europepmc.org/articles/pmc3943494?pdf=render

R. J. Bleicher and M. C. Cabot, Glucosylceramide synthase and apoptosis, Biochimica et Biophysica Acta (BBA) - Molecular and Cell Biology of Lipids, vol.1585, issue.2-3, pp.1585-172, 2002.
DOI : 10.1016/S1388-1981(02)00338-4

S. C. Datta and N. S. Radin, Stimulation of liver growth and DNA synthesis by glucosylceramide, Lipids, vol.38, issue.5, pp.508-510, 1988.
DOI : 10.1016/0304-4165(79)90134-X

J. A. Shayman, G. D. Deshmukh, S. Mahdiyoun, T. P. Thomas, D. Wu et al., Modulation of renal epithelial cell growth by glucosylceramide. Association with protein kinase C, sphingosine, and diacylglycerol, J. Biol. Chem, vol.266, pp.22968-22974, 1991.

N. L. Marchell, Y. Uchida, B. E. Brown, P. M. Elias, and W. M. Holleran, Glucosylceramides Stimulate Mitogenesisin Aged Murine Epidermis, Journal of Investigative Dermatology, vol.110, issue.4, pp.383-387, 1998.
DOI : 10.1046/j.1523-1747.1998.00145.x

URL : https://doi.org/10.1046/j.1523-1747.1998.00145.x

Y. Lavie, H. Cao, S. L. Bursten, A. E. Giuliano, and M. C. Cabot, Accumulation of Glucosylceramides in Multidrug-resistant Cancer Cells, Journal of Biological Chemistry, vol.267, issue.32, pp.271-19530, 1996.
DOI : 10.1016/0092-8674(94)90446-4

URL : http://www.jbc.org/content/271/32/19530.full.pdf

G. Gutiérrez-iglesias, Y. Hurtado, and I. Palma-lara, López-Marure, R. Resistance to the antiproliferative effect induced by a short-chain ceramide is associated with an increase of glucosylceramide synthase, P-glycoprotein, and multidrug-resistance gene-1 in cervical cancer cells, Cancer Chemother. Pharmacol, vol.2014, issue.744, pp.809-817

Y. Uchida, S. Murata, M. Schmuth, M. J. Behne, J. D. Lee et al., Glucosylceramide synthesis and synthase expression protect against ceramide-induced stress, J. Lipid Res, issue.8, pp.43-1293, 2002.

J. A. Bowden, A. Heckert, C. Z. Ulmer, C. M. Jones, J. P. Koelmel et al., Harmonizing Lipidomics: NIST Interlaboratory Comparison Exercise for Lipidomics using Standard Reference Material 1950 Metabolites in Frozen Human Plasma, J. Lipid Res, vol.doi, p.79012, 2017.
DOI : 10.1194/jlr.m079012

M. C. Messner and M. C. Cabot, Glucosylceramides in human In Sphingolipids as signaling and regulatory molecules Advances in experimental medicine and biology, pp.156-164, 2010.

S. Kavaliauskiene, T. Skotland, T. Sylvänne, H. Simolin, T. I. Klokk et al., Novel actions of 2-deoxy-D-glucose: protection against Shiga toxins and changes in cellular lipids, Biochemical Journal, vol.470, issue.1, pp.23-37
DOI : 10.1042/BJ20141562

G. Liebisch, J. A. Vizcaíno, H. Köfeler, M. Trötzmüller, W. J. Griffiths et al., Shorthand notation for lipid structures derived from mass spectrometry, Journal of Lipid Research, vol.1686, issue.6, pp.1523-1530, 2013.
DOI : 10.1371/journal.pone.0061951

URL : http://www.jlr.org/content/54/6/1523.full.pdf

J. Garate, R. Fernández, S. Lage, J. Bestard-escalas, D. H. Lopez et al., Amengual, I.; et al. Imaging mass spectrometry increased resolution using 2- mercaptobenzothiazole and 2,5-diaminonaphtalene matrices: application to lipid distribution in human colon, Anal. Bioanal. Chem, vol.2015, issue.16, pp.407-4697

J. S. Hamilton, R. Aguilar, R. A. Petros, and G. Verbeck, DAPNe with micro-capillary separatory chemistry-coupled to MALDI-MS for the analysis of polar and non-polar lipid metabolism in one cell, Journal of The American Society for Mass Spectrometry, vol.24, issue.5, pp.28-918
DOI : 10.1007/s13361-013-0607-z

E. K. Perttu, A. G. Kohli, and F. Szoka, Inverse-Phosphocholine Lipids: A Remix of a Common Phospholipid, Journal of the American Chemical Society, vol.134, issue.10, pp.134-4485
DOI : 10.1021/ja210989h

URL : http://europepmc.org/articles/pmc3303984?pdf=render

S. N. Jackson, H. J. Wang, and A. S. Woods, In situ structural characterization of phosphatidylcholines in brain tissue using MALDI-MS/MS, Journal of the American Society for Mass Spectrometry, vol.226, issue.12, pp.16-2052, 2005.
DOI : 10.1006/abio.1995.1228

M. Gaudin, M. Panchal, N. Auzeil, C. Duyckaerts, A. Brunelle et al., Choline-containing phospholipids in microdissected human Alzheimer???s disease brain senile plaque versus neuropil, Bioanalysis, vol.4, issue.17, pp.2153-2159
DOI : 10.1016/j.neurobiolaging.2006.05.022

E. G. Stanley, B. J. Jenkins, C. G. Walker, A. Koulman, L. Browning et al.,

S. A. Jebb and J. L. Griffin, Lipidomics Profiling of Human Adipose Tissue Identifies a Pattern of Lipids Associated with Fish Oil Supplementation, J. Proteome Res, vol.2017, issue.169, pp.3168-3179

Z. Li and D. E. Vance, Phosphatidylcholine and choline homeostasis, Journal of Lipid Research, vol.16, issue.6, pp.1187-1194, 2008.
DOI : 10.1146/annurev.bi.16.070147.001205