A. R. Weseler, E. J. Ruijters, M. J. Drittij-reijnders, K. D. Reesink, and G. R. Haenen, Pleiotropic benefit of monomeric and oligomeric flavanols on vascular health-a randomized controlled clinical pilot study, PLoS One, vol.6, p.28460, 2011.

C. Heiss, C. L. Keen, and M. Kelm, Flavanols and cardiovascular disease prevention, Eur Heart J, vol.31, pp.2583-2592, 2010.

H. Schroeter, C. Heiss, J. P. Spencer, C. L. Keen, and J. R. Lupton, Recommending flavanols and procyanidins for cardiovascular health: current knowledge and future needs, Mol Aspects Med, vol.31, pp.546-557, 2010.

D. Milenkovic, B. Jude, and C. Morand, ) miRNA as molecular target of polyphenols underlying their biological effects, Free Radic Biol Med, vol.64, pp.40-51, 2013.

V. C. De-boer, E. M. Van-schothorst, A. A. Dihal, H. Van-der-woude, and I. C. Arts, Chronic quercetin exposure affects fatty acid catabolism in rat lung, Cell Mol Life Sci, vol.63, pp.2847-2858, 2006.

A. Chanet, D. Milenkovic, C. Deval, M. Potier, and J. Constans, Naringin, the major grapefruit flavonoid, specifically affects atherosclerosis development in diet-induced hypercholesterolemia in mice, J Nutr Biochem, vol.23, pp.469-477, 2012.
URL : https://hal.archives-ouvertes.fr/hal-02648869

D. Coban, D. Milenkovic, A. Chanet, J. Khallou-laschet, and L. Sabbe, Dietary curcumin inhibits atherosclerosis by affecting the expression of genes involved in leukocyte adhesion and transendothelial migration, Mol Nutr Food Res, vol.56, pp.1270-1281, 2012.
URL : https://hal.archives-ouvertes.fr/hal-02647896

A. Chanet, D. Milenkovic, C. S. Maier, J. A. , K. Khan et al., Flavanone metabolites decrease monocyte adhesion to TNF-alpha-activated endothelial cells by modulating expression of atherosclerosis-related genes, Br J Nutr, vol.110, pp.587-598, 2013.
URL : https://hal.archives-ouvertes.fr/hal-02642629

D. Kaul, K. Sikand, and A. R. Shukla, Effect of green tea polyphenols on the genes with atherosclerotic potential, Phytother Res, vol.18, pp.177-179, 2004.

J. Leiro, J. A. Arranz, N. Fraiz, M. L. Sanmartin, and E. Quezada, Effect of cis-resveratrol on genes involved in nuclear factor kappa B signaling, Int Immunopharmacol, vol.5, pp.393-406, 2005.

A. Overman, A. Bumrungpert, A. Kennedy, K. Martinez, and C. C. Chuang, Polyphenol-rich grape powder extract (GPE) attenuates inflammation in human macrophages and in human adipocytes exposed to macrophageconditioned media, Int J Obes, vol.34, pp.800-808, 2011.

I. Boomgaarden, S. Egert, G. Rimbach, S. Wolffram, and M. J. Muller, Quercetin supplementation and its effect on human monocyte gene expression profiles in vivo, Br J Nutr, vol.104, pp.336-345, 2010.

D. Milenkovic, C. Deval, C. Dubray, A. Mazur, and C. Morand, Hesperidin displays relevant role in the nutrigenomic effect of orange juice on blood leukocytes in human volunteers: a randomized controlled cross-over study, PLoS One, vol.6, p.26669, 2011.
URL : https://hal.archives-ouvertes.fr/hal-02645333

V. Van-der-velpen, A. Geelen, E. G. Schouten, P. C. Hollman, and L. A. Afman, Estrogen receptor-mediated effects of isoflavone supplementation were not observed in whole-genome gene expression profiles of peripheral blood mononuclear cells in postmenopausal, equol-producing women, J Nutr, vol.143, pp.774-780, 2013.

W. Vanden-berghe, Epigenetic impact of dietary polyphenols in cancer chemoprevention: lifelong remodeling of our epigenomes, Pharmacol Res, vol.65, pp.565-576, 2012.

N. Boque, R. De-la-iglesia, A. L. De-la-garza, F. I. Milagro, and M. Olivares, Prevention of diet-induced obesity by apple polyphenols in Wistar rats through regulation of adipocyte gene expression and DNA methylation patterns, Mol Nutr Food Res, vol.57, pp.1473-1478, 2013.

J. M. Ordovas and C. E. Smith, Epigenetics and cardiovascular disease, Nat Rev Cardiol, vol.7, pp.510-519, 2010.

M. E. Symonds, S. P. Sebert, M. A. Hyatt, and H. Budge, Nutritional programming of the metabolic syndrome, Nat Rev Endocrinol, vol.5, pp.604-610, 2009.

P. D. Gluckman, M. A. Hanson, T. Buklijas, F. M. Low, and A. S. Beedle, Epigenetic mechanisms that underpin metabolic and cardiovascular diseases, Nat Rev Endocrinol, vol.5, pp.401-408, 2009.

S. Tonna, A. El-osta, M. E. Cooper, and C. Tikellis, Metabolic memory and diabetic nephropathy: potential role for epigenetic mechanisms, Nat Rev Nephrol, vol.6, pp.332-341, 2010.

M. K. Shanmugam and G. Sethi, Role of epigenetics in inflammation-associated diseases, Subcell Biochem, vol.61, pp.627-657, 2012.

C. Scoccianti, F. Ricceri, P. Ferrari, C. Cuenin, and C. Sacerdote, Methylation patterns in sentinel genes in peripheral blood cells of heavy smokers: Influence of cruciferous vegetables in an intervention study, Epigenetics, vol.6, pp.1114-1119, 2011.

S. Friso, S. Udali, P. Guarini, C. Pellegrini, and P. Pattini, Global DNA hypomethylation in peripheral blood mononuclear cells as a biomarker of cancer risk, Cancer Epidemiol Biomarkers Prev, vol.22, pp.348-355, 2013.

Y. Gao, K. Killian, H. Zhang, K. Yu, and Q. Z. Li, Leukocyte DNA methylation and colorectal cancer among male smokers, World J Gastrointest Oncol, vol.4, pp.193-201, 2012.

A. Crescenti, R. Sola, R. M. Valls, A. Caimari, D. Bas et al., Cocoa Consumption Alters the Global DNA Methylation of Peripheral Leukocytes in Humans with Cardiovascular Disease Risk Factors: A Randomized Controlled Trial, PLoS One, vol.8, p.65744, 2013.

A. Bird, DNA methylation patterns and epigenetic memory, Genes Dev, vol.16, pp.6-21, 2002.

A. M. Deaton and A. Bird, CpG islands and the regulation of transcription, Genes Dev, vol.25, pp.1010-1022, 2011.

J. R. Gibbs, M. P. Van-der-brug, D. G. Hernandez, B. J. Traynor, and M. A. Nalls, Abundant quantitative trait loci exist for DNA methylation and gene expression in human brain, PLoS Genet, vol.6, p.1000952, 2010.

J. T. Bell, A. A. Pai, J. K. Pickrell, D. J. Gaffney, and R. Pique-regi, DNA methylation patterns associate with genetic and gene expression variation in HapMap cell lines, Genome Biol, vol.12, issue.6, p.405, 2011.

D. Zhang, L. Cheng, J. A. Badner, C. Chen, and Q. Chen, Genetic control of individual differences in gene-specific methylation in human brain, Am J Hum Genet, vol.86, pp.411-419, 2010.

I. Paur, L. M. Austenaa, and R. Blomhoff, Extracts of dietary plants are efficient modulators of nuclear factor kappa B, Food Chem, vol.46, pp.1288-1297, 2008.

W. Vanden-berghe, S. Plaisance, E. Boone, D. Bosscher, K. Schmitz et al., and extracellular signal-regulated kinase mitogen-activated protein kinase pathways are required for nuclear factor-kappaB p65 transactivation mediated by tumor necrosis factor, J Biol Chem, vol.273, pp.3285-3290, 1998.

M. E. Price, A. M. Cotton, L. L. Lam, P. Farre, and E. Emberly, Additional annotation enhances potential for biologically-relevant analysis of the Illumina Infinium HumanMethylation450 BeadChip array, Epigenetics Chromatin, vol.6, p.4, 2013.

S. Dedeurwaerder, M. Defrance, M. Bizet, E. Calonne, and G. Bontempi, A comprehensive overview of Infinium HumanMethylation450 data processing, 2013.

D. Rio, D. Rodriguez-mateos, A. Spencer, J. P. Tognolini, M. Borges et al., Dietary (poly)phenolics in human health: structures, bioavailability, and evidence of protective effects against chronic diseases, Antioxid Redox Signal, vol.18, pp.1818-1892, 2013.

H. H. Feringa, D. A. Laskey, J. E. Dickson, and C. I. Coleman, The effect of grape seed extract on cardiovascular risk markers: a meta-analysis of randomized controlled trials, J Am Diet Assoc, vol.111, pp.1173-1181, 2011.

J. Tome-carneiro, M. Larrosa, M. J. Yanez-gascon, A. Davalos, and J. Gil-zamorano, One-year supplementation with a grape extract containing resveratrol modulates inflammatory-related microRNAs and cytokines expression in peripheral blood mononuclear cells of type 2 diabetes and hypertensive patients with coronary artery disease, Pharmacol Res, vol.72, pp.69-82, 2013.

C. Weber, A. Schober, and A. Zernecke, Chemokines: key regulators of mononuclear cell recruitment in atherosclerotic vascular disease, Arterioscler Thromb Vasc Biol, vol.24, pp.1997-2008, 2004.

S. Man, B. Tucky, A. Cotleur, J. Drazba, and Y. Takeshita, CXCL12-induced monocyte-endothelial interactions promote lymphocyte transmigration across an in vitro blood-brain barrier, Sci Transl Med, vol.4, pp.119-114, 2012.

P. Ray, S. A. Lewin, L. A. Mihalko, S. C. Lesher-perez, and S. Takayama, Secreted CXCL12 (SDF-1) forms dimers under physiological conditions, Biochem J, vol.442, pp.433-442, 2012.

S. Nola, M. Sebbagh, S. Marchetto, N. Osmani, and C. Nourry, Scrib regulates PAK activity during the cell migration process, Hum Mol Genet, vol.17, pp.3552-3565, 2008.

M. Moser, M. Bauer, S. Schmid, R. Ruppert, and S. Schmidt, Kindlin-3 is required for beta2 integrin-mediated leukocyte adhesion to endothelial cells, Nat Med, vol.15, pp.300-305, 2009.

P. Von-hundelshausen, R. R. Koenen, and C. Weber, Platelet-mediated enhancement of leukocyte adhesion, Microcirculation, vol.16, pp.84-96, 2009.

C. L. Chao, N. C. Chang, C. S. Weng, K. R. Lee, and S. T. Kao, Grape seed extract ameliorates tumor necrosis factor-alpha-induced inflammatory status of human umbilical vein endothelial cells, Eur J Nutr, vol.50, pp.401-409, 2011.

S. Auclair, D. Milenkovic, C. Besson, S. Chauvet, and E. Gueux, Catechin reduces atherosclerotic lesion development in apo E-deficient mice: a transcriptomic study, Atherosclerosis, vol.204, pp.21-27, 2009.

L. M. Coussens and Z. Werb, Inflammation and cancer, Nature, vol.420, pp.860-867, 2002.

P. Libby, Inflammation and cardiovascular disease mechanisms, Am J Clin Nutr, vol.83, pp.456-460, 2006.

G. Mundy, R. Van-den-berg, G. R. Haenen, H. Van-den-berg, and A. Bast, Nuclear factor-kappaB activation is higher in peripheral blood mononuclear cells of male smokers, Environ Toxicol Pharmacol, vol.65, pp.147-151, 2001.

E. S. Wan, W. Qiu, A. Baccarelli, V. J. Carey, and H. Bacherman, Cigarette smoking behaviors and time since quitting are associated with differential DNA methylation across the human genome, Hum Mol Genet, vol.21, pp.3073-3082, 2012.

S. Zeilinger, B. Kuhnel, N. Klopp, H. Baurecht, and A. Kleinschmidt, Tobacco smoking leads to extensive genome-wide changes in DNA methylation, PLoS One, vol.8, p.63812, 2013.

N. S. Shenker, S. Polidoro, K. Van-veldhoven, C. Sacerdote, and F. Ricceri, Epigenome-wide association study in the European Prospective Investigation into Cancer and Nutrition (EPIC-Turin) identifies novel genetic loci associated with smoking, Hum Mol Genet, vol.22, pp.843-851, 2013.

M. F. Fraga, E. Ballestar, M. F. Paz, S. Ropero, and F. Setien, Epigenetic differences arise during the lifetime of monozygotic twins, Proc Natl Acad Sci U S A, vol.102, pp.10604-10609, 2005.

M. V. Dogan, B. Shields, C. Cutrona, L. Gao, and F. X. Gibbons, The effect of smoking on DNA methylation of peripheral blood mononuclear cells from African American women, BMC Genomics, vol.15, p.151, 2014.

H. R. Elliott, T. Tillin, W. L. Mcardle, K. Ho, and A. Duggirala, Differences in smoking associated DNA methylation patterns in South Asians and Europeans, Clin Epigenetics, vol.6, p.4, 2014.

B. R. Joubert, S. E. Haberg, R. M. Nilsen, X. Wang, and S. E. Vollset, 450K epigenome-wide scan identifies differential DNA methylation in newborns related to maternal smoking during pregnancy, Environ Health Perspect, vol.120, pp.1425-1431, 2012.

S. Ehrlich, E. Walton, J. L. Roffman, D. Weiss, and I. Puls, Smoking, but not malnutrition, influences promoter-specific DNA methylation of the proopiomelanocortin gene in patients with and without anorexia nervosa, Can J Psychiatry, vol.57, pp.168-176, 2012.

L. P. Breitling, R. Yang, B. Korn, B. Burwinkel, and H. Brenner, Tobaccosmoking-related differential DNA methylation: 27K discovery and replication, Am J Hum Genet, vol.88, pp.450-457, 2011.

A. Bellavia, B. Urch, M. Speck, R. D. Brook, and J. A. Scott, DNA hypomethylation, ambient particulate matter, and increased blood pressure: findings from controlled human exposure experiments, J Am Heart Assoc, vol.2, p.212, 2013.

D. C. Sulistyoningrum, R. Singh, and A. M. Devlin, Epigenetic regulation of glucocorticoid receptor expression in aorta from mice with hyperhomocysteinemia, Epigenetics, vol.7, pp.514-521, 2012.

T. D. Howard, S. M. Ho, L. Zhang, J. Chen, and W. Cui, Epigenetic changes with dietary soy in cynomolgus monkeys, PLoS One, vol.6, p.26791, 2011.

S. Udali, P. Guarini, S. Moruzzi, S. W. Choi, and S. Friso, Cardiovascular epigenetics: from DNA methylation to microRNAs, Mol Aspects Med, vol.34, pp.883-901, 2013.

L. E. Reinius, N. Acevedo, M. Joerink, G. Pershagen, and S. E. Dahlen, Differential DNA methylation in purified human blood cells: implications for cell lineage and studies on disease susceptibility, PLoS One, vol.7, p.41361, 2012.

M. Jacoby, S. Gohrbandt, V. Clausse, N. H. Brons, and C. P. Muller, Interindividual variability and co-regulation of DNA methylation differ among blood cell populations, Epigenetics, vol.7, pp.1421-1434, 2012.

B. T. Adalsteinsson, H. Gudnason, T. Aspelund, T. B. Harris, and L. J. Launer, Heterogeneity in white blood cells has potential to confound DNA methylation measurements, PLoS One, vol.7, p.46705, 2012.

K. Vanhees, S. Coort, E. J. Ruijters, R. W. Godschalk, and F. J. Van-schooten, Epigenetics: prenatal exposure to genistein leaves a permanent signature on the hematopoietic lineage, FASEB J, vol.25, pp.797-807, 2011.

C. Faulk and D. C. Dolinoy, Timing is everything: the when and how of environmentally induced changes in the epigenome of animals, Epigenetics, vol.6, pp.791-797, 2011.

M. L. Simoons and A. F. Casparie, Therapy and prevention of coronary heart diseases through lowering of the serum cholesterol levels, CBO. Ned Tijdschr Geneeskd, vol.142, pp.2096-2101, 1998.