Skip to main content
Top
Published in: Reproductive Biology and Endocrinology 1/2018

Open Access 01-12-2018 | Research

Docosahexaenoic acid (DHA) effects on proliferation and steroidogenesis of bovine granulosa cells

Authors: Virginie Maillard, Alice Desmarchais, Maeva Durcin, Svetlana Uzbekova, Sebastien Elis

Published in: Reproductive Biology and Endocrinology | Issue 1/2018

Login to get access

Abstract

Background

Docosahexaenoic acid (DHA) is a n-3 polyunsaturated fatty acid (PUFA) belonging to a family of biologically active fatty acids (FA), which are known to have numerous health benefits. N-3 PUFAs affect reproduction in cattle, and notably directly affect follicular cells. In terms of reproduction in cattle, n-3 PUFA-enriched diets lead to increased follicle size or numbers.

Methods

The objective of the present study was to analyze the effects of DHA (1, 10, 20 and 50 μM) on proliferation and steroidogenesis (parametric and/or non parametric (permutational) ANOVA) of bovine granulosa cells in vitro and mechanisms of action through protein expression (Kruskal-Wallis) and signaling pathways (non parametric ANOVA) and to investigate whether DHA could exert part of its action through the free fatty acid receptor 4 (FFAR4).

Results

DHA (10 and 50 μM) increased granulosa cell proliferation and DHA 10 μM led to a corresponding increase in proliferating cell nuclear antigen (PCNA) expression level. DHA also increased progesterone secretion at 1, 20 and 50 μM, and estradiol secretion at 1, 10 and 20 μM. Consistent increases in protein levels were also reported for the steroidogenic enzymes, cytochrome P450 family 11 subfamily A member 1 (CYP11A1) and hydroxy-delta-5-steroid dehydrogenase, 3 beta- and steroid delta-isomerase 1 (HSD3B1), and of the cholesterol transporter steroidogenic acute regulatory protein (StAR), which are necessary for production of progesterone or androstenedione. FFAR4 was expressed in all cellular types of bovine ovarian follicles, and in granulosa cells it was localized close to the cellular membrane. TUG-891 treatment (1 and 50 μM), a FFAR4 agonist, increased granulosa cell proliferation and MAPK14 phosphorylation in a similar way to that observed with DHA treatment. However, TUG-891 treatment (1, 10 and 50 μM) showed no effect on progesterone or estradiol secretion.

Conclusions

These data show that DHA stimulated proliferation and steroidogenesis of bovine granulosa cells and led to MAPK14 phosphorylation. FFAR4 involvement in DHA effects requires further investigation, even if our data might suggest FFAR4 role in DHA effects on granulosa cell proliferation. Other mechanisms of DHA action should be investigated as the steroidogenic effects seemed to be independent of FFAR4 activation.
Appendix
Available only for authorised users
Literature
1.
go back to reference Siriwardhana N, Kalupahana NS, Moustaid-Moussa N, Se-Kwon K. Chapter 13 - health benefits of n-3 polyunsaturated fatty acids: Eicosapentaenoic acid and docosahexaenoic acid. In: Adv Food Nutr Res. Vol. volume 65: academic press; 2012. p. 211–22. Siriwardhana N, Kalupahana NS, Moustaid-Moussa N, Se-Kwon K. Chapter 13 - health benefits of n-3 polyunsaturated fatty acids: Eicosapentaenoic acid and docosahexaenoic acid. In: Adv Food Nutr Res. Vol. volume 65: academic press; 2012. p. 211–22.
2.
go back to reference Plourde M, Cunnane SC. Extremely limited synthesis of long chain polyunsaturates in adults: implications for their dietary essentiality and use as supplements. Appl Physiol Nutr Metab. 2007;32(4):619–34.CrossRefPubMed Plourde M, Cunnane SC. Extremely limited synthesis of long chain polyunsaturates in adults: implications for their dietary essentiality and use as supplements. Appl Physiol Nutr Metab. 2007;32(4):619–34.CrossRefPubMed
3.
go back to reference Baker EJ, Miles EA, Burdge GC, Yaqoob P, Calder PC. Metabolism and functional effects of plant-derived omega-3 fatty acids in humans. Prog Lipid Res. 2016;64:30–56.CrossRefPubMed Baker EJ, Miles EA, Burdge GC, Yaqoob P, Calder PC. Metabolism and functional effects of plant-derived omega-3 fatty acids in humans. Prog Lipid Res. 2016;64:30–56.CrossRefPubMed
4.
go back to reference Simopoulos AP. Omega-3 fatty acids in inflammation and autoimmune diseases. J Am Coll Nutr. 2002;21(6):495–505.CrossRefPubMed Simopoulos AP. Omega-3 fatty acids in inflammation and autoimmune diseases. J Am Coll Nutr. 2002;21(6):495–505.CrossRefPubMed
6.
go back to reference Calder PC. Functional roles of fatty acids and their effects on human health. J Parenter Enter Nutr. 2015;39(1 suppl):18S–32S.CrossRef Calder PC. Functional roles of fatty acids and their effects on human health. J Parenter Enter Nutr. 2015;39(1 suppl):18S–32S.CrossRef
7.
go back to reference Santos JE, Bilby TR, Thatcher WW, Staples CR, Silvestre FT. Long chain fatty acids of diet as factors influencing reproduction in cattle. Reprod Domest Anim. 2008;43(Suppl 2):23–30.CrossRefPubMed Santos JE, Bilby TR, Thatcher WW, Staples CR, Silvestre FT. Long chain fatty acids of diet as factors influencing reproduction in cattle. Reprod Domest Anim. 2008;43(Suppl 2):23–30.CrossRefPubMed
8.
go back to reference Gulliver CE, Friend MA, King BJ, Clayton EH. The role of omega-3 polyunsaturated fatty acids in reproduction of sheep and cattle. Anim Reprod Sci. 2012;131(1–2):9–22.CrossRefPubMed Gulliver CE, Friend MA, King BJ, Clayton EH. The role of omega-3 polyunsaturated fatty acids in reproduction of sheep and cattle. Anim Reprod Sci. 2012;131(1–2):9–22.CrossRefPubMed
9.
go back to reference Ambrose DJ, Kastelic JP, Corbett R, Pitney PA, Petit HV, Small JA, Zalkovic P. Lower pregnancy losses in lactating dairy cows fed a diet enriched in alpha-linolenic acid. J Dairy Sci. 2006;89(8):3066–74.CrossRefPubMed Ambrose DJ, Kastelic JP, Corbett R, Pitney PA, Petit HV, Small JA, Zalkovic P. Lower pregnancy losses in lactating dairy cows fed a diet enriched in alpha-linolenic acid. J Dairy Sci. 2006;89(8):3066–74.CrossRefPubMed
10.
go back to reference Dirandeh E, Towhidi A, Zeinoaldini S, Ganjkhanlou M, Ansari Pirsaraei Z, Fouladi-Nashta A. 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. 2013;91(2):713–21.CrossRefPubMed Dirandeh E, Towhidi A, Zeinoaldini S, Ganjkhanlou M, Ansari Pirsaraei Z, Fouladi-Nashta A. 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. 2013;91(2):713–21.CrossRefPubMed
11.
go back to reference Elis S, Freret S, Desmarchais A, Maillard V, Cognié J, Briant E, Touzé J-L, Dupont M, Faverdin P, et al. Effect of a long chain n-3 PUFA-enriched diet on production and reproduction variables in Holstein dairy cows. Anim Reprod Sci. 2016;164:121–32.CrossRefPubMed Elis S, Freret S, Desmarchais A, Maillard V, Cognié J, Briant E, Touzé J-L, Dupont M, Faverdin P, et al. Effect of a long chain n-3 PUFA-enriched diet on production and reproduction variables in Holstein dairy cows. Anim Reprod Sci. 2016;164:121–32.CrossRefPubMed
12.
go back to reference Mattos R, Staples CR, Arteche A, Wiltbank MC, Diaz FJ, Jenkins TC, Thatcher WW. The effects of feeding fish oil on uterine secretion of PGF2alpha, milk composition, and metabolic status of periparturient Holstein cows. J Dairy Sci. 2004;87(4):921–32.CrossRefPubMed Mattos R, Staples CR, Arteche A, Wiltbank MC, Diaz FJ, Jenkins TC, Thatcher WW. The effects of feeding fish oil on uterine secretion of PGF2alpha, milk composition, and metabolic status of periparturient Holstein cows. J Dairy Sci. 2004;87(4):921–32.CrossRefPubMed
13.
go back to reference Caldari-Torres C, Rodriguez-Sallaberry C, Greene ES, Badinga L. Differential effects of n-3 and n-6 fatty acids on prostaglandin F2[alpha] production by bovine endometrial cells. J Dairy Sci. 2006;89(3):971–7.CrossRefPubMed Caldari-Torres C, Rodriguez-Sallaberry C, Greene ES, Badinga L. Differential effects of n-3 and n-6 fatty acids on prostaglandin F2[alpha] production by bovine endometrial cells. J Dairy Sci. 2006;89(3):971–7.CrossRefPubMed
14.
go back to reference Mattos R, Staples CR, Williams J, Amorocho A, McGuire MA. Uterine, ovarian, and production responses of lactating dairy cows to increasing dietary concentrations of menhaden fish meal. J Dairy Sci. 2002;85(4):755–64.CrossRefPubMed Mattos R, Staples CR, Williams J, Amorocho A, McGuire MA. Uterine, ovarian, and production responses of lactating dairy cows to increasing dietary concentrations of menhaden fish meal. J Dairy Sci. 2002;85(4):755–64.CrossRefPubMed
15.
go back to reference Dirandeh E, Towhidi A, Pirsaraei ZA, Hashemi FA, Ganjkhanlou M, Zeinoaldini S, Roodbari AR, Saberifar T, Petit HV. Plasma concentrations of PGFM and uterine and ovarian responses in early lactation dairy cows fed omega-3 and omega-6 fatty acids. Theriogenology. 2013;80(2):131–7.CrossRefPubMed Dirandeh E, Towhidi A, Pirsaraei ZA, Hashemi FA, Ganjkhanlou M, Zeinoaldini S, Roodbari AR, Saberifar T, Petit HV. Plasma concentrations of PGFM and uterine and ovarian responses in early lactation dairy cows fed omega-3 and omega-6 fatty acids. Theriogenology. 2013;80(2):131–7.CrossRefPubMed
16.
go back to reference Moallem U, Shafran A, Zachut M, Dekel I, Portnick Y, Arieli A. Dietary alpha-linolenic acid from flaxseed oil improved folliculogenesis and IVF performance in dairy cows, similar to eicosapentaenoic and docosahexaenoic acids from fish oil. Reproduction. 2013;146(6):603–14.CrossRefPubMed Moallem U, Shafran A, Zachut M, Dekel I, Portnick Y, Arieli A. Dietary alpha-linolenic acid from flaxseed oil improved folliculogenesis and IVF performance in dairy cows, similar to eicosapentaenoic and docosahexaenoic acids from fish oil. Reproduction. 2013;146(6):603–14.CrossRefPubMed
17.
go back to reference Zachut M, Dekel I, Lehrer H, Arieli A, Arav A, Livshitz L, Yakoby S, Moallem U. 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. 2010;93(2):529–45.CrossRefPubMed Zachut M, Dekel I, Lehrer H, Arieli A, Arav A, Livshitz L, Yakoby S, Moallem U. 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. 2010;93(2):529–45.CrossRefPubMed
18.
go back to reference Marei WF, Wathes DC, Fouladi-Nashta AA. The effect of linolenic acid on bovine oocyte maturation and development. Biol Reprod. 2009;81(6):1064–72.CrossRefPubMed Marei WF, Wathes DC, Fouladi-Nashta AA. The effect of linolenic acid on bovine oocyte maturation and development. Biol Reprod. 2009;81(6):1064–72.CrossRefPubMed
19.
go back to reference Oseikria M, Elis S, Maillard V, Corbin E, Uzbekova S. N-3 polyunsaturated fatty acid DHA during IVM affected oocyte developmental competence in cattle. Theriogenology. 2016;85:1625–34.CrossRefPubMed Oseikria M, Elis S, Maillard V, Corbin E, Uzbekova S. N-3 polyunsaturated fatty acid DHA during IVM affected oocyte developmental competence in cattle. Theriogenology. 2016;85:1625–34.CrossRefPubMed
20.
go back to reference Petit HV, Twagiramungu H. Conception rate and reproductive function of dairy cows fed different fat sources. Theriogenology. 2006;66(5):1316–24.CrossRefPubMed Petit HV, Twagiramungu H. Conception rate and reproductive function of dairy cows fed different fat sources. Theriogenology. 2006;66(5):1316–24.CrossRefPubMed
21.
go back to reference Childs S, Carter F, Lynch CO, Sreenan JM, Lonergan P, Hennessy AA, Kenny DA. Embryo yield and quality following dietary supplementation of beef heifers with n-3 polyunsaturated fatty acids (PUFA). Theriogenology. 2008;70(6):992–1003.CrossRefPubMed Childs S, Carter F, Lynch CO, Sreenan JM, Lonergan P, Hennessy AA, Kenny DA. Embryo yield and quality following dietary supplementation of beef heifers with n-3 polyunsaturated fatty acids (PUFA). Theriogenology. 2008;70(6):992–1003.CrossRefPubMed
22.
go back to reference Sinedino LDP, Honda PM, Souza LRL, Lock AL, Boland MP, Staples CR, Thatcher WW, Santos JEP. Effects of supplementation with docosahexaenoic acid on reproduction of dairy cows. Reproduction. 2017;153(5):707–23.CrossRefPubMed Sinedino LDP, Honda PM, Souza LRL, Lock AL, Boland MP, Staples CR, Thatcher WW, Santos JEP. Effects of supplementation with docosahexaenoic acid on reproduction of dairy cows. Reproduction. 2017;153(5):707–23.CrossRefPubMed
23.
go back to reference Ponter AA, Guyader-Joly C, Nuttinck F, Grimard B, Humblot P. 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. 2012;78(3):632–45.CrossRefPubMed Ponter AA, Guyader-Joly C, Nuttinck F, Grimard B, Humblot P. 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. 2012;78(3):632–45.CrossRefPubMed
24.
go back to reference Hutchinson IA, Hennessy AA, Waters SM, Dewhurst RJ, Evans ACO, Lonergan P, Butler ST. Effect of supplementation with different fat sources on the mechanisms involved in reproductive performance in lactating dairy cattle. Theriogenology. 2012;78(1):12–27.CrossRefPubMed Hutchinson IA, Hennessy AA, Waters SM, Dewhurst RJ, Evans ACO, Lonergan P, Butler ST. Effect of supplementation with different fat sources on the mechanisms involved in reproductive performance in lactating dairy cattle. Theriogenology. 2012;78(1):12–27.CrossRefPubMed
25.
go back to reference Riediger ND, Othman RA, Suh M, Moghadasian MH. A systemic review of the roles of n-3 fatty acids in health and disease. J Am Diet Assoc. 2009;109(4):668–79.CrossRefPubMed Riediger ND, Othman RA, Suh M, Moghadasian MH. A systemic review of the roles of n-3 fatty acids in health and disease. J Am Diet Assoc. 2009;109(4):668–79.CrossRefPubMed
27.
go back to reference Calder PC. Fatty acids and inflammation: the cutting edge between food and pharma. Eur J Pharmacol. 2011;668(Suppl 1):S50–8.CrossRefPubMed Calder PC. Fatty acids and inflammation: the cutting edge between food and pharma. Eur J Pharmacol. 2011;668(Suppl 1):S50–8.CrossRefPubMed
28.
go back to reference Calder PC. Marine omega-3 fatty acids and inflammatory processes: effects, mechanisms and clinical relevance. Biochim et Biophys Acta (BBA) - Molecular and Cell Biology of Lipids. 2015;1851(4):469–84.CrossRef Calder PC. Marine omega-3 fatty acids and inflammatory processes: effects, mechanisms and clinical relevance. Biochim et Biophys Acta (BBA) - Molecular and Cell Biology of Lipids. 2015;1851(4):469–84.CrossRef
29.
go back to reference Bagga D, Wang L, Farias-Eisner R, Glaspy JA, Reddy ST. Differential effects of prostaglandin derived from ω-6 and ω-3 polyunsaturated fatty acids on COX-2 expression and IL-6 secretion. Proc Natl Acad Sci U S A. 2003;100(4):1751–6.CrossRefPubMedPubMedCentral Bagga D, Wang L, Farias-Eisner R, Glaspy JA, Reddy ST. Differential effects of prostaglandin derived from ω-6 and ω-3 polyunsaturated fatty acids on COX-2 expression and IL-6 secretion. Proc Natl Acad Sci U S A. 2003;100(4):1751–6.CrossRefPubMedPubMedCentral
30.
go back to reference Shaikh SR. Biophysical and biochemical mechanisms by which dietary N-3 polyunsaturated fatty acids from fish oil disrupt membrane lipid rafts. J Nutr Biochem. 2012;23(2):101–5.CrossRefPubMed Shaikh SR. Biophysical and biochemical mechanisms by which dietary N-3 polyunsaturated fatty acids from fish oil disrupt membrane lipid rafts. J Nutr Biochem. 2012;23(2):101–5.CrossRefPubMed
31.
go back to reference Miyamoto J, Hasegawa S, Kasubuchi M, Ichimura A, Nakajima A, Kimura I. Nutritional signaling via free fatty acid receptors. Int J Mol Sci. 2016;17(4):450.CrossRefPubMedPubMedCentral Miyamoto J, Hasegawa S, Kasubuchi M, Ichimura A, Nakajima A, Kimura I. Nutritional signaling via free fatty acid receptors. Int J Mol Sci. 2016;17(4):450.CrossRefPubMedPubMedCentral
32.
go back to reference Prihandoko R, Alvarez-Curto E, Hudson BD, Butcher AJ, Ulven T, Miller AM, Tobin AB, Milligan G. Distinct phosphorylation clusters determine the signaling outcome of free fatty acid receptor 4/G protein-coupled receptor 120. Mol Pharmacol. 2016;89(5):505–20.CrossRefPubMed Prihandoko R, Alvarez-Curto E, Hudson BD, Butcher AJ, Ulven T, Miller AM, Tobin AB, Milligan G. Distinct phosphorylation clusters determine the signaling outcome of free fatty acid receptor 4/G protein-coupled receptor 120. Mol Pharmacol. 2016;89(5):505–20.CrossRefPubMed
33.
go back to reference Gao B, Huang Q, Jie Q, Lu WG, Wang L, Li XJ, Sun Z, Hu YQ, Chen L, et al. GPR120: a bi-potential mediator to modulate the osteogenic and adipogenic differentiation of BMMSCs. Sci Rep. 2015;5:14080.CrossRefPubMedPubMedCentral Gao B, Huang Q, Jie Q, Lu WG, Wang L, Li XJ, Sun Z, Hu YQ, Chen L, et al. GPR120: a bi-potential mediator to modulate the osteogenic and adipogenic differentiation of BMMSCs. Sci Rep. 2015;5:14080.CrossRefPubMedPubMedCentral
34.
go back to reference Gómez BI, Gifford CA, Hallford DM, Hernandez Gifford JA. Protein kinase B is required for follicle-stimulating hormone mediated beta-catenin accumulation and estradiol production in granulosa cells of cattle. Anim Reprod Sci. 2015;163:97–104.CrossRefPubMed Gómez BI, Gifford CA, Hallford DM, Hernandez Gifford JA. Protein kinase B is required for follicle-stimulating hormone mediated beta-catenin accumulation and estradiol production in granulosa cells of cattle. Anim Reprod Sci. 2015;163:97–104.CrossRefPubMed
35.
go back to reference Silva JM, Hamel M, Sahmi M, Price CA. 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. 2006;132(6):909–17.CrossRefPubMed Silva JM, Hamel M, Sahmi M, Price CA. 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. 2006;132(6):909–17.CrossRefPubMed
36.
go back to reference Ryan KE, Glister C, Lonergan P, Martin F, Knight PG, Evans AC. 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. 2008;1(1):2.CrossRefPubMedPubMedCentral Ryan KE, Glister C, Lonergan P, Martin F, Knight PG, Evans AC. 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. 2008;1(1):2.CrossRefPubMedPubMedCentral
37.
go back to reference Du XH, Zhou XL, Cao R, Xiao P, Teng Y, Ning CB, Liu HL. 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. 2015;27(1):6–14.CrossRefPubMed Du XH, Zhou XL, Cao R, Xiao P, Teng Y, Ning CB, Liu HL. 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. 2015;27(1):6–14.CrossRefPubMed
38.
go back to reference Inagaki K, Otsuka F, Miyoshi T, Yamashita M, Takahashi M, Goto J, Suzuki J, Makino H. p38-mitogen-activated protein kinase stimulated steroidogenesis in granulosa cell-oocyte cocultures: role of bone morphogenetic proteins 2 and 4. Endocrinology. 2009;150(4):1921–30.CrossRefPubMed Inagaki K, Otsuka F, Miyoshi T, Yamashita M, Takahashi M, Goto J, Suzuki J, Makino H. p38-mitogen-activated protein kinase stimulated steroidogenesis in granulosa cell-oocyte cocultures: role of bone morphogenetic proteins 2 and 4. Endocrinology. 2009;150(4):1921–30.CrossRefPubMed
39.
go back to reference Abughazaleh AA, Potu RB, Ibrahim S. Short communication: the effect of substituting fish oil in dairy cow diets with docosahexaenoic acid-micro algae on milk composition and fatty acids profile. J Dairy Sci. 2009;92(12):6156–9.CrossRefPubMed Abughazaleh AA, Potu RB, Ibrahim S. Short communication: the effect of substituting fish oil in dairy cow diets with docosahexaenoic acid-micro algae on milk composition and fatty acids profile. J Dairy Sci. 2009;92(12):6156–9.CrossRefPubMed
40.
go back to reference Boeckaert C, Vlaeminck B, Dijkstra J, Issa-Zacharia A, Van Nespen T, Van Straalen W, Fievez V. Effect of dietary starch or micro algae supplementation on rumen fermentation and milk fatty acid composition of dairy cows. J Dairy Sci. 2008;91(12):4714–27.CrossRefPubMed Boeckaert C, Vlaeminck B, Dijkstra J, Issa-Zacharia A, Van Nespen T, Van Straalen W, Fievez V. Effect of dietary starch or micro algae supplementation on rumen fermentation and milk fatty acid composition of dairy cows. J Dairy Sci. 2008;91(12):4714–27.CrossRefPubMed
41.
go back to reference Hudson BD, Shimpukade B, Mackenzie AE, Butcher AJ, Pediani JD, Christiansen E, Heathcote H, Tobin AB, Ulven T, 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. 2013;84(5):710–25.CrossRefPubMedPubMedCentral Hudson BD, Shimpukade B, Mackenzie AE, Butcher AJ, Pediani JD, Christiansen E, Heathcote H, Tobin AB, Ulven T, 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. 2013;84(5):710–25.CrossRefPubMedPubMedCentral
42.
go back to reference Hirasawa A, Tsumaya K, Awaji T, Katsuma S, Adachi T, Yamada M, Sugimoto Y, Miyazaki S, Tsujimoto G. Free fatty acids regulate gut incretin glucagon-like peptide-1 secretion through GPR120. Nat Med. 2005;11(1):90–4.CrossRefPubMed Hirasawa A, Tsumaya K, Awaji T, Katsuma S, Adachi T, Yamada M, Sugimoto Y, Miyazaki S, Tsujimoto G. Free fatty acids regulate gut incretin glucagon-like peptide-1 secretion through GPR120. Nat Med. 2005;11(1):90–4.CrossRefPubMed
43.
go back to reference Tosca L, Chabrolle C, Uzbekova S, Dupont J. Effects of metformin on bovine granulosa cells steroidogenesis: possible involvement of adenosine 5′ monophosphate-activated protein kinase (AMPK). Biol Reprod. 2007;76(3):368–78.CrossRefPubMed Tosca L, Chabrolle C, Uzbekova S, Dupont J. Effects of metformin on bovine granulosa cells steroidogenesis: possible involvement of adenosine 5′ monophosphate-activated protein kinase (AMPK). Biol Reprod. 2007;76(3):368–78.CrossRefPubMed
44.
go back to reference Canepa S, Laine AB, A., Fagu C, Flon C, Monniaux D: 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 2008; 64:19–30. Canepa S, Laine AB, A., Fagu C, Flon C, Monniaux D: 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 2008; 64:19–30.
45.
go back to reference Lefils J, Géloën A, Vidal H, Lagarde M, Bernoud-Hubac N. Dietary DHA: time course of tissue uptake and effects on cytokine secretion in mice. Br J Nutr. 2010;104(9):1304–12.CrossRefPubMed Lefils J, Géloën A, Vidal H, Lagarde M, Bernoud-Hubac N. Dietary DHA: time course of tissue uptake and effects on cytokine secretion in mice. Br J Nutr. 2010;104(9):1304–12.CrossRefPubMed
46.
go back to reference Hothorn T, Bretz F, Westfall P. Simultaneous inference in general parametric models. Biom J. 2008;50(3):346–63.CrossRefPubMed Hothorn T, Bretz F, Westfall P. Simultaneous inference in general parametric models. Biom J. 2008;50(3):346–63.CrossRefPubMed
47.
go back to reference Wheeler B: lmPerm: Permutation tests for linear models. 2010. Wheeler B: lmPerm: Permutation tests for linear models. 2010.
48.
go back to reference Konietschke F, Placzek M, Schaarschmidt F, Hothorn LA. Nparcomp: an R software package for nonparametric multiple comparisons and simultaneous confidence intervals. J Stat Softw. 2015;64(9):1–17.CrossRef Konietschke F, Placzek M, Schaarschmidt F, Hothorn LA. Nparcomp: an R software package for nonparametric multiple comparisons and simultaneous confidence intervals. J Stat Softw. 2015;64(9):1–17.CrossRef
49.
go back to reference R_Core_Team. R: a language and environment for statistical computing. Vienna: R-project; 2015. R_Core_Team. R: a language and environment for statistical computing. Vienna: R-project; 2015.
50.
51.
go back to reference Wonnacott KE, Kwong WY, Hughes J, Salter AM, Lea RG, Garnsworthy PC, Sinclair KD. Dietary omega-3 and -6 polyunsaturated fatty acids affect the composition and development of sheep granulosa cells, oocytes and embryos. Reproduction. 2010;139(1):57–69.CrossRefPubMed Wonnacott KE, Kwong WY, Hughes J, Salter AM, Lea RG, Garnsworthy PC, Sinclair KD. Dietary omega-3 and -6 polyunsaturated fatty acids affect the composition and development of sheep granulosa cells, oocytes and embryos. Reproduction. 2010;139(1):57–69.CrossRefPubMed
52.
go back to reference Moussavi ARH, Gilbert RO, Overton TR, Bauman DE, Butler WR. Effects of feeding fish meal and n-3 fatty acids on ovarian and uterine responses in early lactating dairy cows. J Dairy Sci. 2007;90(1):145–54.CrossRefPubMed Moussavi ARH, Gilbert RO, Overton TR, Bauman DE, Butler WR. Effects of feeding fish meal and n-3 fatty acids on ovarian and uterine responses in early lactating dairy cows. J Dairy Sci. 2007;90(1):145–54.CrossRefPubMed
53.
go back to reference Robinson RS, Pushpakumara PG, Cheng Z, Peters AR, Abayasekara DR, Wathes DC. Effects of dietary polyunsaturated fatty acids on ovarian and uterine function in lactating dairy cows. Reproduction. 2002;124(1):119–31.CrossRefPubMed Robinson RS, Pushpakumara PG, Cheng Z, Peters AR, Abayasekara DR, Wathes DC. Effects of dietary polyunsaturated fatty acids on ovarian and uterine function in lactating dairy cows. Reproduction. 2002;124(1):119–31.CrossRefPubMed
54.
go back to reference Stocco DM, Zhao AH, Tu LN, Morohaku K, Selvaraj V. A brief history of the search for the protein(s) involved in the acute regulation of steroidogenesis. Mol Cell Endocrinol. 2017;441:7–16.CrossRefPubMed Stocco DM, Zhao AH, Tu LN, Morohaku K, Selvaraj V. A brief history of the search for the protein(s) involved in the acute regulation of steroidogenesis. Mol Cell Endocrinol. 2017;441:7–16.CrossRefPubMed
55.
go back to reference Rodgers RJ. Steroidogenic cytochrome P450 enzymes and ovarian steroidogenesis. Reprod Fertil Dev. 1990;2(2):153–63.CrossRefPubMed Rodgers RJ. Steroidogenic cytochrome P450 enzymes and ovarian steroidogenesis. Reprod Fertil Dev. 1990;2(2):153–63.CrossRefPubMed
56.
go back to reference Bao B, Garverick HA. Expression of steroidogenic enzyme and gonadotropin receptor genes in bovine follicles during ovarian follicular waves: a review. J Anim Sci. 1998;76(7):1903–21.CrossRefPubMed Bao B, Garverick HA. Expression of steroidogenic enzyme and gonadotropin receptor genes in bovine follicles during ovarian follicular waves: a review. J Anim Sci. 1998;76(7):1903–21.CrossRefPubMed
57.
go back to reference Benkert AR, Young M, Robinson D, Hendrickson C, Lee PA, Strauss KA. Severe Salt-Losing 3β-Hydroxysteroid Dehydrogenase Deficiency: Treatment and Outcomes of HSD3B2 c.35G>A Homozygotes. J Clinical Endocrinol Metabol. 2015;100(8):E1105–15.CrossRef Benkert AR, Young M, Robinson D, Hendrickson C, Lee PA, Strauss KA. Severe Salt-Losing 3β-Hydroxysteroid Dehydrogenase Deficiency: Treatment and Outcomes of HSD3B2 c.35G>A Homozygotes. J Clinical Endocrinol Metabol. 2015;100(8):E1105–15.CrossRef
58.
go back to reference Hughes J, Kwong WY, Li D, Salter AM, Lea RG, Sinclair KD. Effects of omega-3 and -6 polyunsaturated fatty acids on ovine follicular cell steroidogenesis, embryo development and molecular markers of fatty acid metabolism. Reproduction. 2011;141(1):105–18.CrossRefPubMed Hughes J, Kwong WY, Li D, Salter AM, Lea RG, Sinclair KD. Effects of omega-3 and -6 polyunsaturated fatty acids on ovine follicular cell steroidogenesis, embryo development and molecular markers of fatty acid metabolism. Reproduction. 2011;141(1):105–18.CrossRefPubMed
59.
go back to reference Waters SM, Coyne GS, Kenny DA, MacHugh DE, Morris DG. 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. 2012;44(18):878–88.CrossRefPubMed Waters SM, Coyne GS, Kenny DA, MacHugh DE, Morris DG. 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. 2012;44(18):878–88.CrossRefPubMed
60.
go back to reference Wang X, Dyson MT, Jo Y, Stocco DM. Inhibition of Cyclooxygenase-2 activity enhances steroidogenesis and steroidogenic acute regulatory gene expression in MA-10 mouse Leydig cells. Endocrinology. 2003;144(8):3368–75.CrossRefPubMed Wang X, Dyson MT, Jo Y, Stocco DM. Inhibition of Cyclooxygenase-2 activity enhances steroidogenesis and steroidogenic acute regulatory gene expression in MA-10 mouse Leydig cells. Endocrinology. 2003;144(8):3368–75.CrossRefPubMed
61.
go back to reference Ringbom T, Huss U, Stenholm Å, Flock S, Skattebøl L, Perera P, Bohlin L. COX-2 inhibitory effects of naturally occurring and modified fatty acids. J Nat Prod. 2001;64(6):745–9.CrossRefPubMed Ringbom T, Huss U, Stenholm Å, Flock S, Skattebøl L, Perera P, Bohlin L. COX-2 inhibitory effects of naturally occurring and modified fatty acids. J Nat Prod. 2001;64(6):745–9.CrossRefPubMed
63.
go back to reference Elis S, Desmarchais A, Freret S, Maillard V, Labas V, Cognié J, Briant E, Hivelin C, Dupont J, et al. Effect of a long-chain n-3 polyunsaturated fatty acid–enriched diet on adipose tissue lipid profiles and gene expression in Holstein dairy cows. J Dairy Sci. 2016;99(12):10109–27.CrossRefPubMed Elis S, Desmarchais A, Freret S, Maillard V, Labas V, Cognié J, Briant E, Hivelin C, Dupont J, et al. Effect of a long-chain n-3 polyunsaturated fatty acid–enriched diet on adipose tissue lipid profiles and gene expression in Holstein dairy cows. J Dairy Sci. 2016;99(12):10109–27.CrossRefPubMed
64.
go back to reference Agrawal A, Alharthi A, Vailati-Riboni M, Zhou Z, Loor JJ. Expression of fatty acid sensing G-protein coupled receptors in peripartal Holstein cows. J Anim Sci Biotechnol. 2017;8(1):20.CrossRefPubMedPubMedCentral Agrawal A, Alharthi A, Vailati-Riboni M, Zhou Z, Loor JJ. Expression of fatty acid sensing G-protein coupled receptors in peripartal Holstein cows. J Anim Sci Biotechnol. 2017;8(1):20.CrossRefPubMedPubMedCentral
65.
go back to reference Song T, Peng J, Ren J, H-k W, Peng J. Cloning and characterization of spliced variants of the porcine G protein coupled receptor 120. Biomed Res Int. 2015;2015:813816.PubMedPubMedCentral Song T, Peng J, Ren J, H-k W, Peng J. Cloning and characterization of spliced variants of the porcine G protein coupled receptor 120. Biomed Res Int. 2015;2015:813816.PubMedPubMedCentral
66.
go back to reference Gotoh C, Hong YH, Iga T, Hishikawa D, Suzuki Y, Song SH, Choi KC, Adachi T, Hirasawa A, et al. The regulation of adipogenesis through GPR120. Biochem Biophys Res Commun. 2007;354(2):591–7.CrossRefPubMed Gotoh C, Hong YH, Iga T, Hishikawa D, Suzuki Y, Song SH, Choi KC, Adachi T, Hirasawa A, et al. The regulation of adipogenesis through GPR120. Biochem Biophys Res Commun. 2007;354(2):591–7.CrossRefPubMed
67.
go back to reference Miyauchi S, Hirasawa A, Iga T, Liu N, Itsubo C, Sadakane K, Hara T, Tsujimoto G. Distribution and regulation of protein expression of the free fatty acid receptor GPR120. Naunyn Schmiedeberg's Arch Pharmacol. 2009;379(4):427–34.CrossRef Miyauchi S, Hirasawa A, Iga T, Liu N, Itsubo C, Sadakane K, Hara T, Tsujimoto G. Distribution and regulation of protein expression of the free fatty acid receptor GPR120. Naunyn Schmiedeberg's Arch Pharmacol. 2009;379(4):427–34.CrossRef
68.
go back to reference Cornall LM, Mathai ML, Hryciw DH, McAinch AJ. Diet-induced obesity up-regulates the abundance of GPR43 and GPR120 in a tissue specific manner. Cell Physiol Biochem. 2011;28(5):949–58.CrossRefPubMed Cornall LM, Mathai ML, Hryciw DH, McAinch AJ. Diet-induced obesity up-regulates the abundance of GPR43 and GPR120 in a tissue specific manner. Cell Physiol Biochem. 2011;28(5):949–58.CrossRefPubMed
69.
go back to reference Lager S, Ramirez VI, Gaccioli F, Jansson T, Powell TL. Expression and localization of the omega-3 fatty acid receptor GPR120 in human term placenta. Placenta. 2014;35(7):523–5.CrossRefPubMedPubMedCentral Lager S, Ramirez VI, Gaccioli F, Jansson T, Powell TL. Expression and localization of the omega-3 fatty acid receptor GPR120 in human term placenta. Placenta. 2014;35(7):523–5.CrossRefPubMedPubMedCentral
70.
go back to reference Shimpukade B, Hudson BD, Hovgaard CK, Milligan G, Ulven T. Discovery of a potent and selective GPR120 agonist. J Med Chem. 2012;55(9):4511–5.CrossRefPubMed Shimpukade B, Hudson BD, Hovgaard CK, Milligan G, Ulven T. Discovery of a potent and selective GPR120 agonist. J Med Chem. 2012;55(9):4511–5.CrossRefPubMed
71.
go back to reference Song T, Zhou Y, Peng J, Tao Y-X, Yang Y, Xu T, Peng J, Ren J, Xiang Q, et al. GPR120 promotes adipogenesis through intracellular calcium and extracellular signal-regulated kinase 1/2 signal pathway. Mol Cell Endocrinol. 2016;434:1–13.CrossRefPubMed Song T, Zhou Y, Peng J, Tao Y-X, Yang Y, Xu T, Peng J, Ren J, Xiang Q, et al. GPR120 promotes adipogenesis through intracellular calcium and extracellular signal-regulated kinase 1/2 signal pathway. Mol Cell Endocrinol. 2016;434:1–13.CrossRefPubMed
72.
go back to reference Anbazhagan AN, Priyamvada S, Gujral T, Bhattacharyya S, Alrefai WA, Dudeja PK, Borthakur A. A novel anti-inflammatory role of GPR120 in intestinal epithelial cells. Am J Physiol Cell Physiol. 2016;310(7):C612–21.CrossRefPubMedPubMedCentral Anbazhagan AN, Priyamvada S, Gujral T, Bhattacharyya S, Alrefai WA, Dudeja PK, Borthakur A. A novel anti-inflammatory role of GPR120 in intestinal epithelial cells. Am J Physiol Cell Physiol. 2016;310(7):C612–21.CrossRefPubMedPubMedCentral
73.
go back to reference Li X, Ballantyne LL, Che X, Mewburn JD, Kang JX, Barkley RM, Murphy RC, Yu Y, Funk CD. 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. 2015;4(4):e001856.CrossRef Li X, Ballantyne LL, Che X, Mewburn JD, Kang JX, Barkley RM, Murphy RC, Yu Y, Funk CD. 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. 2015;4(4):e001856.CrossRef
74.
go back to reference Calabrese EJ, Baldwin LA. U-shaped dose-responses in biology, toxicology, and public health. Annu Rev Public Health. 2001;22:15–33.CrossRefPubMed Calabrese EJ, Baldwin LA. U-shaped dose-responses in biology, toxicology, and public health. Annu Rev Public Health. 2001;22:15–33.CrossRefPubMed
75.
go back to reference Oh DY, Talukdar S, Bae EJ, Imamura T, Morinaga H, Fan W, Li P, Lu WJ, Watkins SM, et al. GPR120 is an omega-3 fatty acid receptor mediating potent anti-inflammatory and insulin-sensitizing effects. Cell. 2010;142(5):687–98.CrossRefPubMedPubMedCentral Oh DY, Talukdar S, Bae EJ, Imamura T, Morinaga H, Fan W, Li P, Lu WJ, Watkins SM, et al. GPR120 is an omega-3 fatty acid receptor mediating potent anti-inflammatory and insulin-sensitizing effects. Cell. 2010;142(5):687–98.CrossRefPubMedPubMedCentral
76.
go back to reference Yang P, Roy SK. Transforming growth factor B1 stimulated DNA synthesis in the granulosa cells of preantral follicles: negative interaction with epidermal growth factor. Biol Reprod. 2006;75(1):140–8.CrossRefPubMedPubMedCentral Yang P, Roy SK. Transforming growth factor B1 stimulated DNA synthesis in the granulosa cells of preantral follicles: negative interaction with epidermal growth factor. Biol Reprod. 2006;75(1):140–8.CrossRefPubMedPubMedCentral
78.
go back to reference Casañas-Sánchez V, Pérez JA, Fabelo N, Quinto-Alemany D, Díaz ML. Docosahexaenoic (DHA) modulates phospholipid-hydroperoxide glutathione peroxidase (Gpx4) gene expression to ensure self-protection from oxidative damage in hippocampal cells. Front Physiol. 2015;6:203.CrossRefPubMedPubMedCentral Casañas-Sánchez V, Pérez JA, Fabelo N, Quinto-Alemany D, Díaz ML. Docosahexaenoic (DHA) modulates phospholipid-hydroperoxide glutathione peroxidase (Gpx4) gene expression to ensure self-protection from oxidative damage in hippocampal cells. Front Physiol. 2015;6:203.CrossRefPubMedPubMedCentral
Metadata
Title
Docosahexaenoic acid (DHA) effects on proliferation and steroidogenesis of bovine granulosa cells
Authors
Virginie Maillard
Alice Desmarchais
Maeva Durcin
Svetlana Uzbekova
Sebastien Elis
Publication date
01-12-2018
Publisher
BioMed Central
Published in
Reproductive Biology and Endocrinology / Issue 1/2018
Electronic ISSN: 1477-7827
DOI
https://doi.org/10.1186/s12958-018-0357-7

Other articles of this Issue 1/2018

Reproductive Biology and Endocrinology 1/2018 Go to the issue