N. Siriwardhana, N. S. Kalupahana, N. Moustaid-moussa, and K. Se-kwon, Eicosapentaenoic acid and docosahexaenoic acid, vol.65, pp.211-233, 2012.

M. Plourde and S. C. Cunnane, Extremely limited synthesis of long chain polyunsaturates in adults: implications for their dietary essentiality and use as supplements, Appl Physiol Nutr Metab, vol.32, issue.4, pp.619-653, 2007.

E. J. Baker, E. A. Miles, G. C. Burdge, P. Yaqoob, and P. C. Calder, Metabolism and functional effects of plant-derived omega-3 fatty acids in humans, Prog Lipid Res, vol.64, pp.30-56, 2016.

A. P. Simopoulos, Omega-3 fatty acids in inflammation and autoimmune diseases, J Am Coll Nutr, vol.21, issue.6, pp.495-505, 2002.

R. J. Deckelbaum and C. Torrejon, The omega-3 fatty acid nutritional landscape: health benefits and sources, J Nutr, vol.142, issue.3, pp.587-91, 2012.

P. C. Calder, Functional roles of fatty acids and their effects on human health, J Parenter Enter Nutr, vol.39, issue.1, pp.18-32, 2015.

J. E. Santos, T. R. Bilby, W. W. Thatcher, C. R. Staples, and F. T. Silvestre, Long chain fatty acids of diet as factors influencing reproduction in cattle, Reprod Domest Anim, vol.43, issue.2, pp.23-30, 2008.

C. E. Gulliver, M. A. Friend, B. J. King, and E. H. Clayton, The role of omega-3 polyunsaturated fatty acids in reproduction of sheep and cattle, Anim Reprod Sci, vol.131, issue.1-2, pp.9-22, 2012.

D. J. Ambrose, J. P. Kastelic, R. Corbett, P. A. Pitney, H. V. Petit et al., Lower pregnancy losses in lactating dairy cows fed a diet enriched in alpha-linolenic acid, J Dairy Sci, vol.89, issue.8, pp.3066-74, 2006.

E. Dirandeh, A. Towhidi, S. Zeinoaldini, M. Ganjkhanlou, A. Pirsaraei et al., Effects of different polyunsaturated fatty acid supplementations during the postpartum periods of early lactating dairy cows on milk yield, metabolic responses, and reproductive performances, J Anim Sci, vol.91, issue.2, pp.713-734, 2013.

S. Elis, S. Freret, A. Desmarchais, V. Maillard, J. Cognié et al., Effect of a long chain n-3 PUFA-enriched diet on production and reproduction variables in Holstein dairy cows, Anim Reprod Sci, vol.164, pp.121-153, 2016.
URL : https://hal.archives-ouvertes.fr/hal-01512139

R. Mattos, C. R. Staples, A. Arteche, M. C. Wiltbank, F. J. Diaz et al., The effects of feeding fish oil on uterine secretion of PGF2alpha, milk composition, and metabolic status of periparturient Holstein cows, J Dairy Sci, vol.87, issue.4, pp.921-953, 2004.

C. Caldari-torres, C. Rodriguez-sallaberry, E. S. Greene, and L. Badinga, Differential effects of n-3 and n-6 fatty acids on prostaglandin F2[alpha] production by bovine endometrial cells, J Dairy Sci, vol.89, issue.3, pp.971-978, 2006.

R. Mattos, C. R. Staples, J. Williams, A. Amorocho, and M. A. Mcguire, Uterine, ovarian, and production responses of lactating dairy cows to increasing dietary concentrations of menhaden fish meal, J Dairy Sci, vol.85, issue.4, pp.755-64, 2002.

E. Dirandeh, A. Towhidi, Z. A. Pirsaraei, F. A. Hashemi, M. Ganjkhanlou et al., Plasma concentrations of PGFM and uterine and ovarian responses in early lactation dairy cows fed omega-3 and omega-6 fatty acids, Theriogenology, vol.80, issue.2, pp.131-138, 2013.

U. Moallem, A. Shafran, M. Zachut, I. Dekel, Y. Portnick et al., Dietary alphalinolenic acid from flaxseed oil improved folliculogenesis and IVF performance in dairy cows, similar to eicosapentaenoic and docosahexaenoic acids from fish oil, Reproduction, vol.146, issue.6, pp.603-617, 2013.

M. Zachut, I. Dekel, H. Lehrer, A. Arieli, A. Arav et al., Effects of dietary fats differing in n-6:n-3 ratio fed to high-yielding dairy cows on fatty acid composition of ovarian compartments, follicular status, and oocyte quality, J Dairy Sci, vol.93, issue.2, pp.529-574, 2010.

W. F. Marei, D. C. Wathes, and A. A. Fouladi-nashta, The effect of linolenic acid on bovine oocyte maturation and development, Biol Reprod, vol.81, issue.6, pp.1064-72, 2009.

M. Oseikria, S. Elis, V. Maillard, E. Corbin, and S. Uzbekova, N-3 polyunsaturated fatty acid DHA during IVM affected oocyte developmental competence in cattle, Theriogenology, vol.85, pp.1625-1659, 2016.
URL : https://hal.archives-ouvertes.fr/hal-01409334

H. V. Petit and H. Twagiramungu, Conception rate and reproductive function of dairy cows fed different fat sources, Theriogenology, vol.66, issue.5, pp.1316-1340, 2006.

S. Childs, F. Carter, C. O. Lynch, J. M. Sreenan, P. Lonergan et al., Embryo yield and quality following dietary supplementation of beef heifers with n-3 polyunsaturated fatty acids (PUFA), Theriogenology, vol.70, issue.6, pp.992-1003, 2008.

L. Sinedino, P. M. Honda, L. Souza, A. L. Lock, M. P. Boland et al., Effects of supplementation with docosahexaenoic acid on reproduction of dairy cows, Reproduction, vol.153, issue.5, pp.707-730, 2017.

A. A. Ponter, C. Guyader-joly, F. Nuttinck, B. Grimard, and P. Humblot, Oocyte and embryo production and quality after OPU-IVF in dairy heifers given diets varying in their n-6/n-3 fatty acid ratio, Theriogenology, vol.78, issue.3, pp.632-677, 2012.
URL : https://hal.archives-ouvertes.fr/hal-01003269

I. A. Hutchinson, A. A. Hennessy, S. M. Waters, R. J. Dewhurst, A. Evans et al., Effect of supplementation with different fat sources on the mechanisms involved in reproductive performance in lactating dairy cattle, Theriogenology, vol.78, issue.1, pp.12-27, 2012.

N. D. Riediger, R. A. Othman, M. Suh, and M. H. Moghadasian, A systemic review of the roles of n-3 fatty acids in health and disease, J Am Diet Assoc, vol.109, issue.4, pp.668-79, 2009.

P. C. Calder, Mechanisms of action of (n-3) fatty acids, J Nutr, vol.142, issue.3, pp.592-601, 2012.

P. C. Calder, Fatty acids and inflammation: the cutting edge between food and pharma, Eur J Pharmacol, vol.668, issue.1, pp.50-58, 2011.

P. C. Calder, Marine omega-3 fatty acids and inflammatory processes: effects, mechanisms and clinical relevance, Biochim et Biophys Acta (BBA) -Molecular and Cell Biology of Lipids, vol.1851, issue.4, pp.469-84, 2015.

D. Bagga, L. Wang, R. Farias-eisner, J. A. Glaspy, and S. T. Reddy, Differential effects of prostaglandin derived from ?-6 and ?-3 polyunsaturated fatty acids on COX-2 expression and IL-6 secretion, Proc Natl Acad Sci, vol.100, issue.4, pp.1751-1757, 2003.

S. R. Shaikh, Biophysical and biochemical mechanisms by which dietary N-3 polyunsaturated fatty acids from fish oil disrupt membrane lipid rafts, J Nutr Biochem, vol.23, issue.2, pp.101-106, 2012.

J. Miyamoto, S. Hasegawa, M. Kasubuchi, A. Ichimura, A. Nakajima et al., Nutritional signaling via free fatty acid receptors, Int J Mol Sci, vol.17, issue.4, p.450, 2016.

R. Prihandoko, E. Alvarez-curto, B. D. Hudson, A. J. Butcher, T. Ulven et al., Distinct phosphorylation clusters determine the signaling outcome of free fatty acid receptor 4/G protein-coupled receptor 120, Mol Pharmacol, vol.89, issue.5, pp.505-525, 2016.

B. Gao, Q. Huang, Q. Jie, W. G. Lu, L. Wang et al., GPR120: a bi-potential mediator to modulate the osteogenic and adipogenic differentiation of, BMMSCs. Sci Rep, vol.5, p.14080, 2015.

B. I. Gómez, C. A. Gifford, D. M. Hallford, H. Gifford, and J. A. , Protein kinase B is required for follicle-stimulating hormone mediated beta-catenin accumulation and estradiol production in granulosa cells of cattle, Anim Reprod Sci, vol.163, pp.97-104, 2015.

J. M. Silva, M. Hamel, M. Sahmi, and C. A. Price, Control of oestradiol secretion and of cytochrome P450 aromatase messenger ribonucleic acid accumulation by FSH involves different intracellular pathways in oestrogenic bovine granulosa cells in vitro, Reproduction, vol.132, issue.6, pp.909-926, 2006.

K. E. Ryan, C. Glister, P. Lonergan, F. Martin, P. G. Knight et al., Functional significance of the signal transduction pathways Akt and Erk in ovarian follicles: in vitro and in vivo studies in cattle and sheep, J Ovarian Res, vol.1, issue.1, p.2, 2008.

X. H. Du, X. L. Zhou, R. Cao, P. Xiao, Y. Teng et al., FSH-induced p38-MAPK-mediated dephosphorylation at serine 727 of the signal transducer and activator of transcription 1 decreases Cyp1b1 expression in mouse granulosa cells, Cell Signal, vol.27, issue.1, pp.6-14, 2015.

K. Inagaki, F. Otsuka, T. Miyoshi, M. Yamashita, M. Takahashi et al., p38-mitogen-activated protein kinase stimulated steroidogenesis in granulosa cell-oocyte cocultures: role of bone morphogenetic proteins 2 and 4, Endocrinology, vol.150, issue.4, pp.1921-1951, 2009.

A. A. Abughazaleh, R. B. Potu, and S. Ibrahim, Short communication: the effect of substituting fish oil in dairy cow diets with docosahexaenoic acidmicro algae on milk composition and fatty acids profile, J Dairy Sci, vol.92, issue.12, pp.6156-6165, 2009.

C. Boeckaert, B. Vlaeminck, J. Dijkstra, A. Issa-zacharia, T. Van-nespen et al., Effect of dietary starch or micro algae supplementation on rumen fermentation and milk fatty acid composition of dairy cows, J Dairy Sci, vol.91, issue.12, pp.4714-4741, 2008.

B. D. Hudson, B. Shimpukade, A. E. Mackenzie, A. J. Butcher, J. D. Pediani et al., The pharmacology of TUG-891, a potent and selective agonist of the free fatty acid receptor 4 (FFA4/GPR120), demonstrates both potential opportunity and possible challenges to therapeutic agonism, Mol Pharmacol, vol.84, issue.5, pp.710-735, 2013.

A. Hirasawa, K. Tsumaya, T. Awaji, S. Katsuma, T. Adachi et al., Free fatty acids regulate gut incretin glucagonlike peptide-1 secretion through GPR120, Nat Med, vol.11, issue.1, pp.90-94, 2005.

L. Tosca, C. Chabrolle, S. Uzbekova, and J. Dupont, Effects of metformin on bovine granulosa cells steroidogenesis: possible involvement of adenosine 5? monophosphate-activated protein kinase (AMPK), Biol Reprod, vol.76, issue.3, pp.368-78, 2007.

S. Canepa, A. B. Laine, A. Fagu, C. Flon, C. Monniaux et al., Validation d'une methode immunoenzymatique pour le dosage de la progesterone dans le plasma des ovins et des bovins, Les Cahiers Techniques de L'INRA, vol.64, pp.19-30, 2008.

J. Lefils, A. Géloën, H. Vidal, M. Lagarde, and N. Bernoud-hubac, Dietary DHA: time course of tissue uptake and effects on cytokine secretion in mice, Br J Nutr, vol.104, issue.9, pp.1304-1316, 2010.

T. Hothorn, F. Bretz, and P. Westfall, Simultaneous inference in general parametric models, Biom J, vol.50, issue.3, pp.346-63, 2008.

B. Wheeler, lmPerm: Permutation tests for linear models, 2010.

F. Konietschke, M. Placzek, F. Schaarschmidt, and L. A. Hothorn, Nparcomp: an R software package for nonparametric multiple comparisons and simultaneous confidence intervals, J Stat Softw, vol.64, issue.9, pp.1-17, 2015.

. R_core_team, R: a language and environment for statistical computing. Vienna: R-project, 2015.

K. N. Choe and G. Moldovan, Forging ahead through darkness: PCNA, still the principal conductor at the replication fork, Mol Cell, vol.65, issue.3, pp.380-92, 2017.

K. E. Wonnacott, W. Y. Kwong, J. Hughes, A. M. Salter, R. G. Lea et al., Dietary omega-3 and -6 polyunsaturated fatty acids affect the composition and development of sheep granulosa cells, oocytes and embryos, Reproduction, vol.139, issue.1, pp.57-69, 2010.

A. Moussavi, R. O. Gilbert, T. R. Overton, D. E. Bauman, and W. R. Butler, Effects of feeding fish meal and n-3 fatty acids on ovarian and uterine responses in early lactating dairy cows, J Dairy Sci, vol.90, issue.1, pp.145-54, 2007.

R. S. Robinson, P. G. Pushpakumara, Z. Cheng, A. R. Peters, D. R. Abayasekara et al., Effects of dietary polyunsaturated fatty acids on ovarian and uterine function in lactating dairy cows, Reproduction, vol.124, issue.1, pp.119-150, 2002.

D. M. Stocco, A. H. Zhao, L. N. Tu, K. Morohaku, and V. Selvaraj, A brief history of the search for the protein(s) involved in the acute regulation of steroidogenesis, Mol Cell Endocrinol, vol.441, pp.7-16, 2017.

R. J. Rodgers, Steroidogenic cytochrome P450 enzymes and ovarian steroidogenesis, Reprod Fertil Dev, vol.2, issue.2, pp.153-63, 1990.

B. Bao and H. A. Garverick, Expression of steroidogenic enzyme and gonadotropin receptor genes in bovine follicles during ovarian follicular waves: a review, J Anim Sci, vol.76, issue.7, pp.1903-1924, 1998.

A. R. Benkert, M. Young, D. Robinson, C. Hendrickson, P. A. Lee et al., Severe Salt-Losing 3?-Hydroxysteroid Dehydrogenase Deficiency: Treatment and Outcomes of HSD3B2 c.35G>A Homozygotes, J Clinical Endocrinol Metabol, vol.100, issue.8, pp.1105-1120, 2015.

J. Hughes, W. Y. Kwong, D. Li, A. M. Salter, R. G. Lea et al., Effects of omega-3 and -6 polyunsaturated fatty acids on ovine follicular cell steroidogenesis, embryo development and molecular markers of fatty acid metabolism, Reproduction, vol.141, issue.1, pp.105-123, 2011.

S. M. Waters, G. S. Coyne, D. A. Kenny, D. E. Machugh, and D. G. Morris, Dietary n-3 polyunsaturated fatty acid supplementation alters the expression of genes involved in the control of fertility in the bovine uterine endometrium, Physiol Genomics, vol.44, issue.18, pp.878-88, 2012.

X. Wang, M. T. Dyson, Y. Jo, and D. M. Stocco, Inhibition of Cyclooxygenase-2 activity enhances steroidogenesis and steroidogenic acute regulatory gene expression in MA-10 mouse Leydig cells, Endocrinology, vol.144, issue.8, pp.3368-75, 2003.

T. Ringbom, U. Huss, Å. Stenholm, S. Flock, L. Skattebøl et al., COX-2 inhibitory effects of naturally occurring and modified fatty acids, J Nat Prod, vol.64, issue.6, pp.745-754, 2001.

M. E. Pavone and S. E. Bulun, Aromatase inhibitors for the treatment of endometriosis: a review, Fertil Steril, vol.98, issue.6, pp.1370-1379, 2012.

S. Elis, A. Desmarchais, S. Freret, V. Maillard, V. Labas et al., Effect of a long-chain n-3 polyunsaturated fatty acidenriched diet on adipose tissue lipid profiles and gene expression in Holstein dairy cows, J Dairy Sci, vol.99, issue.12, pp.10109-10136, 2016.

A. Agrawal, A. Alharthi, M. Vailati-riboni, Z. Zhou, and J. J. Loor, Expression of fatty acid sensing G-protein coupled receptors in peripartal Holstein cows, J Anim Sci Biotechnol, vol.8, issue.1, p.20, 2017.

T. Song, J. Peng, J. Ren, H. , W. Peng et al., Cloning and characterization of spliced variants of the porcine G protein coupled receptor 120, Biomed Res Int, p.813816, 2015.

C. Gotoh, Y. H. Hong, T. Iga, D. Hishikawa, Y. Suzuki et al., The regulation of adipogenesis through GPR120, Biochem Biophys Res Commun, vol.354, issue.2, pp.591-598, 2007.

S. Miyauchi, A. Hirasawa, T. Iga, N. Liu, C. Itsubo et al., Distribution and regulation of protein expression of the free fatty acid receptor GPR120, Naunyn Schmiedeberg's Arch Pharmacol, vol.379, issue.4, pp.427-461, 2009.

L. M. Cornall, M. L. Mathai, D. H. Hryciw, and A. J. Mcainch, Diet-induced obesity upregulates the abundance of GPR43 and GPR120 in a tissue specific manner, Cell Physiol Biochem, vol.28, issue.5, pp.949-58, 2011.

S. Lager, V. I. Ramirez, F. Gaccioli, T. Jansson, and T. L. Powell, Expression and localization of the omega-3 fatty acid receptor GPR120 in human term placenta, Placenta, vol.35, issue.7, pp.523-528, 2014.

B. Shimpukade, B. D. Hudson, C. K. Hovgaard, G. Milligan, and T. Ulven, Discovery of a potent and selective GPR120 agonist, J Med Chem, vol.55, issue.9, pp.4511-4516, 2012.

T. Song, Y. Zhou, J. Peng, Y. Tao, Y. Yang et al., GPR120 promotes adipogenesis through intracellular calcium and extracellular signal-regulated kinase 1/2 signal pathway, Mol Cell Endocrinol, vol.434, pp.1-13, 2016.

A. N. Anbazhagan, S. Priyamvada, T. Gujral, S. Bhattacharyya, W. A. Alrefai et al., A novel anti-inflammatory role of GPR120 in intestinal epithelial cells, Am J Physiol Cell Physiol, vol.310, issue.7, pp.612-633, 2016.

X. Li, L. L. Ballantyne, C. X. Mewburn, J. D. Kang, J. X. Barkley et al., Endogenously generated Omega-3 fatty acids attenuate vascular inflammation and Neointimal hyperplasia by interaction with free fatty acid receptor 4 in mice, J Am Heart Assoc: Cardiovasc Cerebrovasc Dis, vol.4, issue.4, p.1856, 2015.

E. J. Calabrese and L. A. Baldwin, U-shaped dose-responses in biology, toxicology, and public health, Annu Rev Public Health, vol.22, pp.15-33, 2001.

D. Y. Oh, S. Talukdar, E. J. Bae, T. Imamura, H. Morinaga et al., GPR120 is an omega-3 fatty acid receptor mediating potent anti-inflammatory and insulin-sensitizing effects, Cell, vol.142, issue.5, pp.687-98, 2010.

P. Yang and S. K. Roy, Transforming growth factor B1 stimulated DNA synthesis in the granulosa cells of preantral follicles: negative interaction with epidermal growth factor, Biol Reprod, vol.75, issue.1, pp.140-148, 2006.

H. Vaidya and S. K. Cheema, Arachidonic acid has a dominant effect to regulate lipogenic genes in 3T3-L1 adipocytes compared to omega-3 fatty acids, Food Nutr Res, vol.59, 2015.

V. Casañas-sánchez, J. A. Pérez, N. Fabelo, D. Quinto-alemany, and M. L. Díaz, Docosahexaenoic (DHA) modulates phospholipid-hydroperoxide glutathione peroxidase (Gpx4) gene expression to ensure self-protection from oxidative damage in hippocampal cells, Front Physiol, vol.6, p.203, 2015.