Stocktake Sale on now: wide range of books at up to 70% off!
Register      Login
Reproduction, Fertility and Development Reproduction, Fertility and Development Society
Vertebrate reproductive science and technology
REVIEW

Metabolic exchanges between the oocyte and its environment: focus on lipids

Svetlana Uzbekova A B * , Priscila Silvana Bertevello A , Rozenn Dalbies-Tran A , Sebastien Elis A , Valerie Labas A C , Philippe Monget A and Ana-Paula Teixeira-Gomes A C
+ Author Affiliations
- Author Affiliations

A CNRS, IFCE, INRAE, Université de Tours, PRC, F-37380 Nouzilly, France.

B LK Ernst Federal Science Centre for Animal Husbandry, Podolsk, Russia.

C INRAE, Université de Tours, CHRU Tours, Plate-Forme PIXANIM, F-37380 Nouzilly, France.

* Correspondence to: svetlana.uzbekova@inrae.fr

Reproduction, Fertility and Development 34(2) 1-26 https://doi.org/10.1071/RD21249
Published online: 11 October 2021

© 2022 The Author(s) (or their employer(s)). Published by CSIRO Publishing on behalf of the IETS

Abstract

Finely regulated fatty acid (FA) metabolism within ovarian follicles is crucial to follicular development and influences the quality of the enclosed oocyte, which relies on the surrounding intra-follicular environment for its growth and maturation. A growing number of studies have examined the association between the lipid composition of follicular compartments and oocyte quality. In this review, we focus on lipids, their possible exchanges between compartments within the ovarian follicle and their involvement in different pathways during oocyte final growth and maturation. Lipidomics provides a detailed snapshot of the global lipid profiles and identified lipids, clearly discriminating the cells or fluid from follicles at distinct physiological stages. Follicular fluid appears as a main mediator of lipid exchanges between follicular somatic cells and the oocyte, through vesicle-mediated and non-vesicular transport of esterified and free FA. A variety of expression data allowed the identification of common and cell-type-specific actors of lipid metabolism in theca cells, granulosa cells, cumulus cells and oocytes, including key regulators of FA uptake, FA transport, lipid transformation, lipoprotein synthesis and protein palmitoylation. They act in harmony to accompany follicular development, and maintain intra-follicular homeostasis to allow the oocyte to accumulate energy and membrane lipids for subsequent meiotic divisions and first embryo cleavages.

Keywords: extracellular vesicles, fatty acid metabolism, follicular cells, follicular fluid, lipids, oocyte, ovary, protein palmitoylation.


References

Aardema, H, Vos, PLAM, Lolicato, F, Roelen, BAJ, Knijn, HM, Vaandrager, AB, Helms, JB, and Gadella, BM (2011). Oleic acid prevents detrimental effects of saturated fatty acids on bovine oocyte developmental competence. Biology of Reproduction 85, 62–69.
Oleic acid prevents detrimental effects of saturated fatty acids on bovine oocyte developmental competence.Crossref | GoogleScholarGoogle Scholar | 21311036PubMed |

Aardema, H, Lolicato, F, van de Lest, CHA, Brouwers, JF, Vaandrager, AB, van Tol, HTA, Roelen, BAJ, Vos, PLAM, Helms, JB, and Gadella, BM (2013). Bovine cumulus cells protect maturing oocytes from increased fatty acid levels by massive intracellular lipid storage. Biology of Reproduction 88, 1–15.
Bovine cumulus cells protect maturing oocytes from increased fatty acid levels by massive intracellular lipid storage.Crossref | GoogleScholarGoogle Scholar |

Aardema, H, Gadella, BM, van de Lest, CH, Brouwers, JFHM, Stout, TAE, Roelen, BAJ, and Vos, PLAM (2015). Free fatty acid levels in fluid of dominant follicles at the preferred insemination time in dairy cows are not affected by early postpartum fatty acid stress. Journal of Dairy Science 98, 2322–2336.
Free fatty acid levels in fluid of dominant follicles at the preferred insemination time in dairy cows are not affected by early postpartum fatty acid stress.Crossref | GoogleScholarGoogle Scholar | 25648816PubMed |

Aardema, H, van Tol, HTA, Wubbolts, RW, Brouwers, JFHM, Gadella, BM, and Roelen, BAJ (2017). Stearoyl-CoA desaturase activity in bovine cumulus cells protects the oocyte against saturated fatty acid stress. Biology of Reproduction 96, 982–992.
Stearoyl-CoA desaturase activity in bovine cumulus cells protects the oocyte against saturated fatty acid stress.Crossref | GoogleScholarGoogle Scholar | 28486699PubMed |

Aardema, H, Vos, PLAM, and Gadella, BM (2018). Cumulus cells protect the oocyte against saturated free fatty acids. Animal Reproduction 15, 737–750.
Cumulus cells protect the oocyte against saturated free fatty acids.Crossref | GoogleScholarGoogle Scholar |

Annes, K, Muller, DB, Vilela, JAP, Valente, RS, Caetano, DP, Cibin, FWS, Milazzotto, MP, Mesquita, FS, Belaz, KRA, Eberlin, MN, and Sudano, MJ (2019). Influence of follicle size on bovine oocyte lipid composition, follicular metabolic and stress markers, embryo development and blastocyst lipid content. Reproduction, Fertility and Development 31, 462–472.
Influence of follicle size on bovine oocyte lipid composition, follicular metabolic and stress markers, embryo development and blastocyst lipid content.Crossref | GoogleScholarGoogle Scholar |

Antonucci, F, Turola, E, Riganti, L, Caleo, M, Gabrielli, M, Perrotta, C, Novellino, L, Clementi, E, Giussani, P, Viani, P, Matteoli, M, and Verderio, C (2012). Microvesicles released from microglia stimulate synaptic activity via enhanced sphingolipid metabolism. EMBO Journal 31, 1231–1240.
Microvesicles released from microglia stimulate synaptic activity via enhanced sphingolipid metabolism.Crossref | GoogleScholarGoogle Scholar |

Argov, N, and Sklan, D (2004). Expression of mRNA of lipoprotein receptor related protein 8, low density lipoprotein receptor, and very low density lipoprotein receptor in bovine ovarian cells during follicular development and corpus luteum formation and regression. Molecular Reproduction and Development 68, 169–175.
Expression of mRNA of lipoprotein receptor related protein 8, low density lipoprotein receptor, and very low density lipoprotein receptor in bovine ovarian cells during follicular development and corpus luteum formation and regression.Crossref | GoogleScholarGoogle Scholar | 15095337PubMed |

Argov, N, Moallem, U, and Sklan, D (2004). Lipid transport in the developing bovine follicle: messenger RNA expression increases for selective uptake receptors and decreases for endocytosis receptors. Biology of Reproduction 71, 479–485.
Lipid transport in the developing bovine follicle: messenger RNA expression increases for selective uptake receptors and decreases for endocytosis receptors.Crossref | GoogleScholarGoogle Scholar | 15056566PubMed |

Assey, RJ, Hyttel, P, Greve, T, and Purwantara, B (1994). Oocyte morphology in dominant and subordinate follicles. Molecular Reproduction and Development 37, 335–344.
Oocyte morphology in dominant and subordinate follicles.Crossref | GoogleScholarGoogle Scholar | 8185939PubMed |

Auclair, S, Uzbekov, R, Elis, S, Sanchez, L, Kireev, I, Lardic, L, Dalbies-Tran, R, and Uzbekova, S (2013). Absence of cumulus cells during in vitro maturation affects lipid metabolism in bovine oocytes. American Journal of Physiology-Endocrinology and Metabolism 304, E599–E613.
Absence of cumulus cells during in vitro maturation affects lipid metabolism in bovine oocytes.Crossref | GoogleScholarGoogle Scholar | 23321473PubMed |

Azhar, S, Leers-Sucheta, S, and Reaven, E (2003). Cholesterol uptake in adrenal and gonadal tissues: the SR-BI and ‘selective’ pathway connection. Frontiers in Bioscience 8, s998–s1029.
Cholesterol uptake in adrenal and gonadal tissues: the SR-BI and ‘selective’ pathway connection.Crossref | GoogleScholarGoogle Scholar | 12957864PubMed |

Bausenwein, J, Serke, H, Eberle, K, Hirrlinger, J, Jogschies, P, Hmeidan, FA, Blumenauer, V, and Spanel-Borowski, K (2010). Elevated levels of oxidized low-density lipoprotein and of catalase activity in follicular fluid of obese women. Molecular Human Reproduction 16, 117–124.
Elevated levels of oxidized low-density lipoprotein and of catalase activity in follicular fluid of obese women.Crossref | GoogleScholarGoogle Scholar | 19729414PubMed |

Bender, K, Walsh, S, Evans, ACO, Fair, T, and Brennan, L (2010). Metabolite concentrations in follicular fluid may explain differences in fertility between heifers and lactating cows. Reproduction 139, 1047–1055.
Metabolite concentrations in follicular fluid may explain differences in fertility between heifers and lactating cows.Crossref | GoogleScholarGoogle Scholar | 20385782PubMed |

Bertevello, P, Ghazouani, O, Banliat, C, Elis, S, Teixeira-Gomes, AP, Maillard, V, Labas, V, and Uzbekova, S (2016). MALDI-TOF mass spectrometry analysis of lipids in single bovine oocytes during IVM. Scientific Meeting of the AETE 13, 712.

Bertevello, PS, Teixeira-Gomes, AP, Seyer, A, Vitorino Carvalho, A, Labas, V, Blache, MC, Banliat, C, Cordeiro, LAV, Duranthon, V, Papillier, P, Maillard, V, Elis, S, and Uzbekova, S (2018). Lipid identification and transcriptional analysis of controlling enzymes in bovine ovarian follicle. International Journal of Molecular Sciences 19, 3261.
Lipid identification and transcriptional analysis of controlling enzymes in bovine ovarian follicle.Crossref | GoogleScholarGoogle Scholar |

Bertevello, PS, Teixeira-Gomes, AP, Labas, V, Cordeiro, L, Blache, MC, Papillier, P, Singina, G, Uzbekov, R, Maillard, V, and Uzbekova, S (2020). MALDI-TOF mass spectrometry revealed significant lipid variations in follicular fluid and somatic follicular cells but not in enclosed oocytes between the large dominant and small subordinate follicles in bovine ovary. International Journal of Molecular Sciences 21, .
MALDI-TOF mass spectrometry revealed significant lipid variations in follicular fluid and somatic follicular cells but not in enclosed oocytes between the large dominant and small subordinate follicles in bovine ovary.Crossref | GoogleScholarGoogle Scholar | 32932995PubMed |

Bijlmakers, MJ, and Marsh, M (2003). The on–off story of protein palmitoylation. Trends in Cell Biology 13, 32–42.
The on–off story of protein palmitoylation.Crossref | GoogleScholarGoogle Scholar | 12480338PubMed |

Blanc, M, David, FPA, Abrami, L, Migliozzi, D, Armand, F, and Burgi, J Blanc, M, David, FPA, Abrami, L, Migliozzi, D, Armand, F, and Burgi, J (2015). SwissPalm: protein palmitoylation database. F1000Research 4, 261.
SwissPalm: protein palmitoylation database.Crossref | GoogleScholarGoogle Scholar | 26339475PubMed |

Blondin, P, and Sirard, MA (1995). Oocyte and follicular morphology as determining characteristics for developmental competence in bovine oocytes. Molecular Reproduction and Development 41, 54–62.
Oocyte and follicular morphology as determining characteristics for developmental competence in bovine oocytes.Crossref | GoogleScholarGoogle Scholar | 7619506PubMed |

Bonnet, A, Servin, B, Mulsant, P, and Mandon-Pepin, B (2015). Spatio-temporal gene expression profiling during in vivo early ovarian folliculogenesis: integrated transcriptomic study and molecular signature of early follicular growth. PLoS ONE 10, e0141482.
Spatio-temporal gene expression profiling during in vivo early ovarian folliculogenesis: integrated transcriptomic study and molecular signature of early follicular growth.Crossref | GoogleScholarGoogle Scholar | 26540452PubMed |

Bowman, AP, Bogie, JFJ, Hendriks, JJA, Haidar, M, Belov, M, Heeren, RMA, and Ellis, SR (2020). Evaluation of lipid coverage and high spatial resolution MALDI-imaging capabilities of oversampling combined with laser post-ionisation. Analytical and Bioanalytical Chemistry 412, 2277–2289.
Evaluation of lipid coverage and high spatial resolution MALDI-imaging capabilities of oversampling combined with laser post-ionisation.Crossref | GoogleScholarGoogle Scholar | 31879798PubMed |

Brantmeier, SA, Grummer, RR, and Ax, RL (1987). Concentrations of high density lipoproteins vary among follicular sizes in the bovine. Journal of Dairy Science 70, 2145–2149.
Concentrations of high density lipoproteins vary among follicular sizes in the bovine.Crossref | GoogleScholarGoogle Scholar | 3680733PubMed |

Brisard, D, Chesnel, F, Elis, S, Desmarchais, A, Sanchez-Lazo, L, Chasles, M, Maillard, V, and Uzbekova, S (2014). Tribbles expression in cumulus cells is related to oocyte maturation and fatty acid metabolism. Journal of Ovarian Research 7, 44.
Tribbles expression in cumulus cells is related to oocyte maturation and fatty acid metabolism.Crossref | GoogleScholarGoogle Scholar | 24834131PubMed |

Bunel, A, Jorssen, EP, Merckx, E, Leroy, JL, Bols, PE, and Sirard, MA (2015). Individual bovine in vitro embryo production and cumulus cell transcriptomic analysis to distinguish cumulus-oocyte complexes with high or low developmental potential. Theriogenology 83, 228–237.
Individual bovine in vitro embryo production and cumulus cell transcriptomic analysis to distinguish cumulus-oocyte complexes with high or low developmental potential.Crossref | GoogleScholarGoogle Scholar | 25442391PubMed |

Buschiazzo, J, Ríos, GL, Canizo, JR, Antollini, SS, and Alberio, RH (2017). Free cholesterol and cholesterol esters in bovine oocytes: Implications in survival and membrane raft organization after cryopreservation. PLoS ONE 12, e0180451.
Free cholesterol and cholesterol esters in bovine oocytes: Implications in survival and membrane raft organization after cryopreservation.Crossref | GoogleScholarGoogle Scholar | 28686720PubMed |

Butler, WR, and Smith, RD (1989). Interrelationships between energy balance and postpartum reproductive function in dairy cattle. Journal of Dairy Science 72, 767–783.
Interrelationships between energy balance and postpartum reproductive function in dairy cattle.Crossref | GoogleScholarGoogle Scholar | 2654227PubMed |

Campbell, DI, Ferreira, CR, Eberlin, LS, and Cooks, RG (2012). Improved spatial resolution in the imaging of biological tissue using desorption electrospray ionization. Analytical and Bioanalytical Chemistry 404, 389–398.
Improved spatial resolution in the imaging of biological tissue using desorption electrospray ionization.Crossref | GoogleScholarGoogle Scholar | 22706326PubMed |

Charlier, C, Montfort, J, Chabrol, O, Brisard, D, Nguyen, T, Le Cam, A, Richard-Parpaillon, L, Moreews, F, Pontarotti, P, Uzbekova, S, Chesnel, F, and Bobe, J (2012). Oocyte-somatic cells interactions, lessons from evolution. BMC Genomics 13, 560.
Oocyte-somatic cells interactions, lessons from evolution.Crossref | GoogleScholarGoogle Scholar | 23083410PubMed |

Cheong, SH, Filho, OGS, Absalón-Medina, VA, Pelton, SH, Butler, WR, and Gilbert, RO (2016). Metabolic and endocrine differences between dairy cows that do or do not ovulate first postpartum dominant follicles. Biology of Reproduction 94, 18.
Metabolic and endocrine differences between dairy cows that do or do not ovulate first postpartum dominant follicles.Crossref | GoogleScholarGoogle Scholar | 26632612PubMed |

Chmurzyńska, A (2006). The multigene family of fatty acid-binding proteins (FABPs): function, structure and polymorphism. Journal of Applied Genetics 47, 39–48.
The multigene family of fatty acid-binding proteins (FABPs): function, structure and polymorphism.Crossref | GoogleScholarGoogle Scholar | 16424607PubMed |

Chu, T, Dufort, I, and Sirard, MA (2012). Effect of ovarian stimulation on oocyte gene expression in cattle. Theriogenology 77, 1928–1938.
Effect of ovarian stimulation on oocyte gene expression in cattle.Crossref | GoogleScholarGoogle Scholar | 22444561PubMed |

Collado-Fernandez, E, Picton, HM, and Dumollard, R (2012). Metabolism throughout follicle and oocyte development in mammals. The International Journal of Developmental Biology 56, 799–808.
Metabolism throughout follicle and oocyte development in mammals.Crossref | GoogleScholarGoogle Scholar | 23417402PubMed |

Conti, M, Hsieh, M, Zamah, AM, and Oh, JS (2012). Novel signaling mechanisms in the ovary during oocyte maturation and ovulation. Molecular and Cellular Endocrinology 356, 65–73.
Novel signaling mechanisms in the ovary during oocyte maturation and ovulation.Crossref | GoogleScholarGoogle Scholar | 22101318PubMed |

Cordeiro, FB, Jarmusch, AK, León, M, Ferreira, CR, Pirro, V, Eberlin, LS, Hallett, J, Miglino, MA, and Cooks, RG (2020). Mammalian ovarian lipid distributions by desorption electrospray ionization-mass spectrometry (DESI-MS) imaging. Analytical and Bioanalytical Chemistry 412, 1251–1262.
Mammalian ovarian lipid distributions by desorption electrospray ionization-mass spectrometry (DESI-MS) imaging.Crossref | GoogleScholarGoogle Scholar | 31953714PubMed |

D’Occhio, MJ, Baruselli, PS, and Campanile, G (2019). Influence of nutrition, body condition, and metabolic status on reproduction in female beef cattle: a review. Theriogenology 125, 277–284.
Influence of nutrition, body condition, and metabolic status on reproduction in female beef cattle: a review.Crossref | GoogleScholarGoogle Scholar | 30497026PubMed |

D’Souza, K, Paramel, GV, and Kienesberger, PC (2018). Lysophosphatidic acid signaling in obesity and insulin resistance. Nutrients 10, 399.
Lysophosphatidic acid signaling in obesity and insulin resistance.Crossref | GoogleScholarGoogle Scholar |

da Silveira, JC, de Avila, ACFCM, Garrett, HL, Bruemmer, JE, Winger, QA, and Bouma, GJ (2018). Cell-secreted vesicles containing microRNAs as regulators of gamete maturation. Journal of Endocrinology 236, R15–R27.
Cell-secreted vesicles containing microRNAs as regulators of gamete maturation.Crossref | GoogleScholarGoogle Scholar |

Dadarwal, D, Adams, GP, Hyttel, P, Brogliatti, GM, Caldwell, S, and Singh, J (2015). Organelle reorganization in bovine oocytes during dominant follicle growth and regression. Reproductive Biology and Endocrinology 13, 124.
Organelle reorganization in bovine oocytes during dominant follicle growth and regression.Crossref | GoogleScholarGoogle Scholar | 26577904PubMed |

Dalbies-Tran, R, Cadoret, V, Desmarchais, A, Elis, S, Maillard, V, Monget, P, Monniaux, D, Reynaud, K, Saint-Dizier, M, and Uzbekova, S (2020). A comparative analysis of oocyte development in mammals. Cells 9, 1002.
A comparative analysis of oocyte development in mammals.Crossref | GoogleScholarGoogle Scholar |

de Almeida Monteiro Melo Ferraz, M, Fujihara, M, Nagashima, JB, Noonan, MJ, Inoue-Murayama, M, and Songsasen, N (2020). Follicular extracellular vesicles enhance meiotic resumption of domestic cat vitrified oocytes. Scientific Reports 10, 8619.
Follicular extracellular vesicles enhance meiotic resumption of domestic cat vitrified oocytes.Crossref | GoogleScholarGoogle Scholar | 32451384PubMed |

de Andrade Melo-Sterza, F, and Poehland, R (2021). Lipid metabolism in bovine oocytes and early embryos under in vivoin vitro, and stress conditions. International Journal of Molecular Sciences 22, 3421.
Lipid metabolism in bovine oocytes and early embryos under in vivoin vitro, and stress conditions.Crossref | GoogleScholarGoogle Scholar | 33810351PubMed |

del Collado, M, da Silveira, JC, Sangalli, JR, Andrade, GM, Sousa, LRDS, Silva, LA, Meirelles, FV, and Perecin, F (2017). Fatty acid binding protein 3 and transzonal projections are involved in lipid accumulation during in vitro maturation of bovine oocytes. Scientific Reports 7, 2645.
Fatty acid binding protein 3 and transzonal projections are involved in lipid accumulation during in vitro maturation of bovine oocytes.Crossref | GoogleScholarGoogle Scholar | 28572619PubMed |

Demarquoy, J (2015). Crosstalk between mitochondria and peroxisomes. World Journal of Biological Chemistry 6, 301.
Crosstalk between mitochondria and peroxisomes.Crossref | GoogleScholarGoogle Scholar | 26629313PubMed |

Di Pietro, C (2016). Exosome-mediated communication in the ovarian follicle. Journal of Assisted Reproduction and Genetics 33, 303–311.
Exosome-mediated communication in the ovarian follicle.Crossref | GoogleScholarGoogle Scholar | 26814471PubMed |

Douville, G, and Sirard, MA (2014). Changes in granulosa cells gene expression associated with growth, plateau and atretic phases in medium bovine follicles. Journal of Ovarian Research 7, 50.
Changes in granulosa cells gene expression associated with growth, plateau and atretic phases in medium bovine follicles.Crossref | GoogleScholarGoogle Scholar | 24955130PubMed |

Drzazga, A, Sowinska, A, and Koziolkiewicz, M (2014). Lysophosphatidylcholine and lysophosphatidylinosiol--novel promissing signaling molecules and their possible therapeutic activity. Acta Poloniae Pharmaceutica 71, 887–899.
| 25745761PubMed |

Duchez, AC, Boudreau, LH, Naika, GS, Bollinger, J, Belleannee, C, Cloutier, N, Laffont, B, Mendoza-Villarroel, RE, Levesque, T, Rollet-Labelle, E, Rousseau, M, Allaeys, I, Tremblay, JJ, Poubelle, PE, Lambeau, G, Pouliot, M, Provost, P, Soulet, D, Gelb, MH, and Boilard, E (2015). Platelet microparticles are internalized in neutrophils via the concerted activity of 12-lipoxygenase and secreted phospholipase A2-IIA. Proceedings of the National academy of Sciences of the United States of America 112, E3564–E3573.
Platelet microparticles are internalized in neutrophils via the concerted activity of 12-lipoxygenase and secreted phospholipase A2-IIA.Crossref | GoogleScholarGoogle Scholar | 26106157PubMed |

Dunning, KR, Anastasi, MR, Zhang, VJ, Russell, DL, and Robker, RL (2014a). Regulation of fatty acid oxidation in mouse cumulus-oocyte complexes during maturation and modulation by PPAR agonists. PLoS ONE 9, e87327.
Regulation of fatty acid oxidation in mouse cumulus-oocyte complexes during maturation and modulation by PPAR agonists.Crossref | GoogleScholarGoogle Scholar | 24505284PubMed |

Dunning, KR, Russell, DL, and Robker, RL (2014b). Lipids and oocyte developmental competence: the role of fatty acids and β-oxidation. Reproduction 148, R15–R27.
Lipids and oocyte developmental competence: the role of fatty acids and β-oxidation.Crossref | GoogleScholarGoogle Scholar | 24760880PubMed |

Dupont, J, Reverchon, M, Cloix, L, Froment, P, and Rame, C (2012). Involvement of adipokines, AMPK, PI3K and the PPAR signaling pathways in ovarian follicle development and cancer. The International Journal of Developmental Biology 56, 959–967.
Involvement of adipokines, AMPK, PI3K and the PPAR signaling pathways in ovarian follicle development and cancer.Crossref | GoogleScholarGoogle Scholar | 23417417PubMed |

Eberlé, D, Hegarty, B, Bossard, P, Ferré, P, and Foufelle, F (2004). SREBP transcription factors: master regulators of lipid homeostasis. Biochimie 86, 839–848.
SREBP transcription factors: master regulators of lipid homeostasis.Crossref | GoogleScholarGoogle Scholar | 15589694PubMed |

Edidin, M (2003). Lipids on the frontier: a century of cell-membrane bilayers. Nature Reviews Molecular Cell Biology 4, 414–418.
Lipids on the frontier: a century of cell-membrane bilayers.Crossref | GoogleScholarGoogle Scholar | 12728275PubMed |

Elis, S, Freret, S, Desmarchais, A, Maillard, V, Cognie, J, Briant, E, Touzé, JL, Dupont, M, Faverdin, P, Chajès, V, Uzbekova, S, Monget, P, and Dupont, J (2016). Effect of a long chain n−3 PUFA-enriched diet on production and reproduction variables in Holstein dairy cows. Animal Reproduction Science 164, 121–132.
Effect of a long chain n−3 PUFA-enriched diet on production and reproduction variables in Holstein dairy cows.Crossref | GoogleScholarGoogle Scholar | 26651949PubMed |

Elis, S, Oseikria, M, Vitorino Carvalho, A, Bertevello, PS, Corbin, E, Teixeira-Gomes, AP, Lecardonnel, J, Archilla, C, Duranthon, V, Labas, V, and Uzbekova, S (2017). Docosahexaenoic acid mechanisms of action on the bovine oocyte-cumulus complex. Journal of Ovarian Research 10, 74.
Docosahexaenoic acid mechanisms of action on the bovine oocyte-cumulus complex.Crossref | GoogleScholarGoogle Scholar | 29122003PubMed |

Erion, DM, and Shulman, GI (2010). Diacylglycerol-mediated insulin resistance. Nature Medicine 16, 400–402.
Diacylglycerol-mediated insulin resistance.Crossref | GoogleScholarGoogle Scholar | 20376053PubMed |

Esposito, G, Irons, PC, Webb, EC, and Chapwanya, A (2014). Interactions between negative energy balance, metabolic diseases, uterine health and immune response in transition dairy cows. Animal Reproduction Science 144, 60–71.
Interactions between negative energy balance, metabolic diseases, uterine health and immune response in transition dairy cows.Crossref | GoogleScholarGoogle Scholar | 24378117PubMed |

Estienne, A, Brossaud, A, Reverchon, M, Ramé, C, Froment, P, and Dupont, J (2020). Adipokines expression and effects in oocyte maturation, fertilization and early embryo development: lessons from mammals and birds. International Journal of Molecular Sciences 21, 3581.
Adipokines expression and effects in oocyte maturation, fertilization and early embryo development: lessons from mammals and birds.Crossref | GoogleScholarGoogle Scholar |

Fahy, E, Cotter, D, Sud, M, and Subramaniam, S (2011). Lipid classification, structures and tools. Biochimica et Biophysica Acta (BBA) – Molecular and Cell Biology of Lipids 1811, 637–647.
Lipid classification, structures and tools.Crossref | GoogleScholarGoogle Scholar |

Fair, T, Hulshof, SCJ, Hyttel, P, Greve, T, and Boland, M (1997). Oocyte ultrastructure in bovine primordial to early tertiary follicles. Anatomy and Embryology 195, 327–336.
Oocyte ultrastructure in bovine primordial to early tertiary follicles.Crossref | GoogleScholarGoogle Scholar | 9108198PubMed |

Falkenburger, BH, Jensen, JB, Dickson, EJ, Suh, BC, and Hille, B (2010). Symposium review: phosphoinositides: lipid regulators of membrane proteins. Journal of Physiology 588, 3179–3185.
Symposium review: phosphoinositides: lipid regulators of membrane proteins.Crossref | GoogleScholarGoogle Scholar |

Fang, J, Wang, H, Miao, L, Kuang, X, Ma, W, Wang, C, Zhang, J, and Xia, G (2016). Involvement of protein acyltransferase ZDHHC3 in maintaining oocyte meiotic arrest in Xenopus laevis1. Biology of Reproduction 95, 1–9.
Involvement of protein acyltransferase ZDHHC3 in maintaining oocyte meiotic arrest in Xenopus laevis1.Crossref | GoogleScholarGoogle Scholar |

Ferreira, CR, Saraiva, SA, Catharino, RR, Garcia, JS, Gozzo, FC, Sanvido, GB, Santos, LFA, Lo Turco, EG, Pontes, JHF, Basso, AC, Bertolla, RP, Sartori, R, Guardieiro, MM, Perecin, F, Meirelles, FV, Sangalli, JR, and Eberlin, MN (2010). Single embryo and oocyte lipid fingerprinting by mass spectrometry. Journal of Lipid Research 51, 1218–1227.
Single embryo and oocyte lipid fingerprinting by mass spectrometry.Crossref | GoogleScholarGoogle Scholar | 19965589PubMed |

Fiore, E, Arfuso, F, Gianesella, M, Vecchio, D, Morgante, M, Mazzotta, E, Badon, T, Rossi, P, Bedin, S, and Piccione, G (2018). Metabolic and hormonal adaptation in Bubalus bubalis around calving and early lactation. PLoS ONE 13, e0193803.
Metabolic and hormonal adaptation in Bubalus bubalis around calving and early lactation.Crossref | GoogleScholarGoogle Scholar | 29617370PubMed |

Flis, VV, and Daum, G (2013). Lipid transport between the endoplasmic reticulum and mitochondria. Cold Spring Harbor Perspectives in Biology 5, a013235.
Lipid transport between the endoplasmic reticulum and mitochondria.Crossref | GoogleScholarGoogle Scholar | 23732475PubMed |

Freret, S, Oseikria, M, Le Bourhis, D, Desmarchais, A, Briant, E, Desnoes, O, Dupont, M, Le Berre, L, Ghazouani, O, Bertevello, PS, Teixeira-Gomes, AP, Labas, V, Uzbekova, S, Salvetti, P, Maillard, V, and Elis, S (2019). Effects of a n−3 PUFA enriched diet on embryo production in dairy cows. Reproduction 158, 71–83.
Effects of a n−3 PUFA enriched diet on embryo production in dairy cows.Crossref | GoogleScholarGoogle Scholar | 31013477PubMed |

Fukata, Y, Murakami, T, Yokoi, N, and Fukata, M (2016). Local palmitoylation cycles and specialized membrane domain organization. Current Topics in Membranes 77, 97–141.
Local palmitoylation cycles and specialized membrane domain organization.Crossref | GoogleScholarGoogle Scholar | 26781831PubMed |

Garnsworthy, PC, Lock, A, Mann, GE, Sinclair, KD, and Webb, R (2008). Nutrition, metabolism, and fertility in dairy cows: 2. Dietary fatty acids and ovarian function. Journal of Dairy Science 91, 3824–3833.
Nutrition, metabolism, and fertility in dairy cows: 2. Dietary fatty acids and ovarian function.Crossref | GoogleScholarGoogle Scholar | 18832204PubMed |

Gautier, T, Becker, S, Drouineaud, V, Ménétrier, F, Sagot, P, Nofer, J-R, von Otte, S, Lagrost, L, Masson, D, and Tietge, UJF (2010). Human luteinized granulosa cells secrete apoB100-containing lipoproteins. Journal of Lipid Research 51, 2245–2252.
Human luteinized granulosa cells secrete apoB100-containing lipoproteins.Crossref | GoogleScholarGoogle Scholar | 20407020PubMed |

Gegenfurtner, K, Flenkenthaler, F, Frohlich, T, Wolf, E, and Arnold, GJ (2020). The impact of transcription inhibition during in vitro maturation on the proteome of bovine oocytes. Biology of Reproduction 103, 1000–1011.
The impact of transcription inhibition during in vitro maturation on the proteome of bovine oocytes.Crossref | GoogleScholarGoogle Scholar | 32856698PubMed |

Gilbert, I, Robert, C, Vigneault, C, Blondin, P, and Sirard, MA (2012). Impact of the LH surge on granulosa cell transcript levels as markers of oocyte developmental competence in cattle. Reproduction 143, 735–747.
Impact of the LH surge on granulosa cell transcript levels as markers of oocyte developmental competence in cattle.Crossref | GoogleScholarGoogle Scholar | 22457433PubMed |

Gilchrist, RB, Lane, M, and Thompson, JG (2008). Oocyte-secreted factors: regulators of cumulus cell function and oocyte quality. Human Reproduction Update 14, 159–177.
Oocyte-secreted factors: regulators of cumulus cell function and oocyte quality.Crossref | GoogleScholarGoogle Scholar | 18175787PubMed |

Gilchrist, RB, Luciano, AM, Richani, D, Zeng, HT, Wang, X, Vos, MD, Sugimura, S, Smitz, J, Richard, FJ, and Thompson, JG (2016). Oocyte maturation and quality: role of cyclic nucleotides. Reproduction 152, R143–R157.
Oocyte maturation and quality: role of cyclic nucleotides.Crossref | GoogleScholarGoogle Scholar | 27422885PubMed |

Girard, A, Dufort, I, Douville, G, and Sirard, MA (2015). Global gene expression in granulosa cells of growing, plateau and atretic dominant follicles in cattle. Reproductive Biology and Endocrinology 13, 17–15.
Global gene expression in granulosa cells of growing, plateau and atretic dominant follicles in cattle.Crossref | GoogleScholarGoogle Scholar | 25879740PubMed |

Goddard, AD, and Watts, A (2012). Regulation of G protein-coupled receptors by palmitoylation and cholesterol. BMC Biology 10, S27.
Regulation of G protein-coupled receptors by palmitoylation and cholesterol.Crossref | GoogleScholarGoogle Scholar |

Gonçalves, RF, Ferreira, MS, de Oliveira, DN, Canevarolo, R, Achilles, MA, D’Ercole, DL, Bols, PE, Visintin, JA, Killian, GJ, and Catharino, RR (2016). Analysis and characterisation of bovine oocyte and embryo biomarkers by matrix-assisted desorption ionisation mass spectrometry imaging. Reproduction, Fertility and Development 28, 293–301.
Analysis and characterisation of bovine oocyte and embryo biomarkers by matrix-assisted desorption ionisation mass spectrometry imaging.Crossref | GoogleScholarGoogle Scholar |

Goto-Inoue, N, Hayasaka, T, Zaima, N, and Setou, M (2011). Imaging mass spectrometry for lipidomics. Biochimica et Biophysica Acta – Molecular and Cell Biology of Lipids 1811, 961–969.
Imaging mass spectrometry for lipidomics.Crossref | GoogleScholarGoogle Scholar |

Gross, JJ, Schwarz, FJ, Eder, K, van Dorland, HA, and Bruckmaier, RM (2013). Liver fat content and lipid metabolism in dairy cows during early lactation and during a mid-lactation feed restriction. Journal of Dairy Science 96, 5008–5017.
Liver fat content and lipid metabolism in dairy cows during early lactation and during a mid-lactation feed restriction.Crossref | GoogleScholarGoogle Scholar | 23746584PubMed |

Guzel, Y, Bildik, G, and Oktem, O (2018). Sphingosine-1-phosphate protects human ovarian follicles from apoptosis in vitro. European Journal of Obstetrics & Gynecology and Reproductive Biology 222, 19–24.
Sphingosine-1-phosphate protects human ovarian follicles from apoptosis in vitro.Crossref | GoogleScholarGoogle Scholar |

Hailay, T, Hoelker, M, Poirier, M, Gebremedhn, S, Rings, F, Saeed-Zidane, M, Salilew-Wondim, D, Dauben, C, Tholen, E, Neuhoff, C, Schellander, K, and Tesfaye, D (2019). Extracellular vesicle-coupled miRNA profiles in follicular fluid of cows with divergent post-calving metabolic status. Scientific Reports 9, 12851.
Extracellular vesicle-coupled miRNA profiles in follicular fluid of cows with divergent post-calving metabolic status.Crossref | GoogleScholarGoogle Scholar | 31492906PubMed |

Hannun, YA, and Obeid, LM (2008). Principles of bioactive lipid signalling: lessons from sphingolipids. Nature Reviews Molecular Cell Biology 9, 139–150.
Principles of bioactive lipid signalling: lessons from sphingolipids.Crossref | GoogleScholarGoogle Scholar | 18216770PubMed |

Hao, Q, Zhu, Z, Xu, D, Liu, W, Lyu, L, and Li, P (2019). Proteomic characterization of bovine granulosa cells in dominant and subordinate follicles. Hereditas 156, 21.
Proteomic characterization of bovine granulosa cells in dominant and subordinate follicles.Crossref | GoogleScholarGoogle Scholar | 31293364PubMed |

Hao, JW, Wang, J, Guo, H, Zhao, YY, Sun, HH, Li, YF, Lai, XY, Zhao, N, Wang, X, Xie, C, Hong, L, Huang, X, Wang, HR, Li, CB, Liang, B, Chen, S, and Zhao, TJ (2020). CD36 facilitates fatty acid uptake by dynamic palmitoylation-regulated endocytosis. Nature Communications 11, 4765.
CD36 facilitates fatty acid uptake by dynamic palmitoylation-regulated endocytosis.Crossref | GoogleScholarGoogle Scholar | 32958780PubMed |

Haraszti, RA, Didiot, MC, Sapp, E, Leszyk, J, Shaffer, SA, Rockwell, HE, Gao, F, Narain, NR, DiFiglia, M, Kiebish, MA, Aronin, N, and Khvorova, A (2016). High-resolution proteomic and lipidomic analysis of exosomes and microvesicles from different cell sources. Journal of Extracellular Vesicles 5, 32570.
High-resolution proteomic and lipidomic analysis of exosomes and microvesicles from different cell sources.Crossref | GoogleScholarGoogle Scholar | 27863537PubMed |

Harris, SE, Leese, HJ, Gosden, RG, and Picton, HM (2009). Pyruvate and oxygen consumption throughout the growth and development of Murine Oocytes. Molecular Reproduction and Development 76, 231–238.
Pyruvate and oxygen consumption throughout the growth and development of Murine Oocytes.Crossref | GoogleScholarGoogle Scholar | 18618608PubMed |

Hashemi, HF, and Goodman, JM (2015). The life cycle of lipid droplets. Current Opinion in Cell Biology 33, 119–124.
The life cycle of lipid droplets.Crossref | GoogleScholarGoogle Scholar | 25703629PubMed |

Hatzirodos, N, Hummitzsch, K, Irving-Rodgers, HF, Harland, ML, Morris, SE, and Rodgers, RJ (2014a). Transcriptome profiling of granulosa cells from bovine ovarian follicles during atresia. BMC Genomics 15, 40.
Transcriptome profiling of granulosa cells from bovine ovarian follicles during atresia.Crossref | GoogleScholarGoogle Scholar | 24438529PubMed |

Hatzirodos, N, Irving-Rodgers, HF, Hummitzsch, K, Harland, ML, Morris, SE, and Rodgers, RJ (2014b). Transcriptome profiling of granulosa cells of bovine ovarian follicles during growth from small to large antral sizes. BMC Genomics 15, 24.
Transcriptome profiling of granulosa cells of bovine ovarian follicles during growth from small to large antral sizes.Crossref | GoogleScholarGoogle Scholar | 24422759PubMed |

Hatzirodos, N, Hummitzsch, K, Irving-Rodgers, HF, and Rodgers, RJ (2015). Transcriptome comparisons identify new cell markers for theca interna and granulosa cells from small and large antral ovarian follicles. PLoS ONE 10, e0119800.
Transcriptome comparisons identify new cell markers for theca interna and granulosa cells from small and large antral ovarian follicles.Crossref | GoogleScholarGoogle Scholar | 25775029PubMed |

Hernández-Araiza, I, Morales-Lázaro, SL, Canul-Sánchez, JA, Islas, LD, and Rosenbaum, T (2018). Role of lysophosphatidic acid in ion channel function and disease. Journal of Neurophysiology 120, 1198–1211.
Role of lysophosphatidic acid in ion channel function and disease.Crossref | GoogleScholarGoogle Scholar | 29947596PubMed |

Hotamisligil, GS, and Bernlohr, DA (2015). Metabolic functions of FABPs–mechanisms and therapeutic implications. Nature Reviews Endrocrinology 11, 592–605.
Metabolic functions of FABPs–mechanisms and therapeutic implications.Crossref | GoogleScholarGoogle Scholar |

Huang, J, Zhu, R, and Shi, D (2021). The role of FATP1 in lipid accumulation: a review. Molecular and Cellular Biochemistry 476, 1897–1903.
The role of FATP1 in lipid accumulation: a review.Crossref | GoogleScholarGoogle Scholar | 33486652PubMed |

Hung, WT, Hong, X, Christenson, LK, and McGinnis, LK (2015). Extracellular vesicles from bovine follicular fluid support cumulus expansion. Biology of Reproduction 93, 117.
Extracellular vesicles from bovine follicular fluid support cumulus expansion.Crossref | GoogleScholarGoogle Scholar | 26423123PubMed |

Hung, WT, Navakanitworakul, R, Khan, T, Zhang, P, Davis, JS, McGinnis, LK, and Christenson, LK (2017). Stage-specific follicular extracellular vesicle uptake and regulation of bovine granulosa cell proliferation. Biology of Reproduction 97, 644–655.
Stage-specific follicular extracellular vesicle uptake and regulation of bovine granulosa cell proliferation.Crossref | GoogleScholarGoogle Scholar | 29025042PubMed |

Hurwitz, SN, Rider, MA, Bundy, JL, Liu, X, Singh, RK, and Meckes, DG (2016). Proteomic profiling of NCI-60 extracellular vesicles uncovers common protein cargo and cancer type-specific biomarkers. Oncotarget 7, 86999–87015.
Proteomic profiling of NCI-60 extracellular vesicles uncovers common protein cargo and cancer type-specific biomarkers.Crossref | GoogleScholarGoogle Scholar | 27894104PubMed |

Hyttel, P, Fair, T, Callesen, H, and Greve, T (1997). Oocyte growth, capacitation and final maturartion in cattle. Theriogenology 47, 23–32.
Oocyte growth, capacitation and final maturartion in cattle.Crossref | GoogleScholarGoogle Scholar |

Ioannou, MS, Jackson, J, Sheu, SH, Chang, CL, Weigel, AV, Liu, H, Pasolli, HA, Xu, CS, Pang, S, Matthies, D, Hess, HF, Lippincott-Schwartz, J, and Liu, Z (2019). Neuron-astrocyte metabolic coupling protects against activity-induced fatty acid toxicity. Cell 177, 1522–1535.e14.
Neuron-astrocyte metabolic coupling protects against activity-induced fatty acid toxicity.Crossref | GoogleScholarGoogle Scholar | 31130380PubMed |

Jaffe, LA, and Egbert, JR (2017). Regulation of mammalian oocyte meiosis by intercellular communication within the ovarian follicle. Annual Review of Physiology 79, 237–260.
Regulation of mammalian oocyte meiosis by intercellular communication within the ovarian follicle.Crossref | GoogleScholarGoogle Scholar | 27860834PubMed |

Jeong, WJ, Cho, SJ, Lee, HS, Deb, GK, Lee, YS, Kwon, TH, and Kong, IK (2009). Effect of cytoplasmic lipid content on in vitro developmental efficiency of bovine IVP embryos. Theriogenology 72, 584–589.
Effect of cytoplasmic lipid content on in vitro developmental efficiency of bovine IVP embryos.Crossref | GoogleScholarGoogle Scholar | 19501898PubMed |

Kathayat, RS, Cao, Y, Elvira, PD, Sandoz, PA, Zaballa, ME, Springer, MZ, Drake, LE, Macleod, KF, van der Goot, FG, and Dickinson, BC (2018). Active and dynamic mitochondrial S-depalmitoylation revealed by targeted fluorescent probes. Nature Communications 9, 334.
Active and dynamic mitochondrial S-depalmitoylation revealed by targeted fluorescent probes.Crossref | GoogleScholarGoogle Scholar | 29362370PubMed |

Keerthikumar, S, Chisanga, D, Ariyaratne, D, Al Saffar, H, Anand, S, Zhao, K, Samuel, M, Pathan, M, Jois, M, Chilamkurti, N, Gangoda, L, and Mathivanan, S (2016). ExoCarta: a web-based compendium of exosomal cargo. Journal of Molecular Biology 428, 688–692.
ExoCarta: a web-based compendium of exosomal cargo.Crossref | GoogleScholarGoogle Scholar | 26434508PubMed |

Khan, R, Jiang, X, Hameed, U, and Shi, Q (2021). Role of lipid metabolism and signaling in mammalian oocyte maturation, quality, and acquisition of competence. Frontiers in Cell and Developmental Biology 9, 639704.
Role of lipid metabolism and signaling in mammalian oocyte maturation, quality, and acquisition of competence.Crossref | GoogleScholarGoogle Scholar | 33748128PubMed |

Kim, JY, Kinoshita, M, Ohnishi, M, and Fukui, Y (2001). Lipid and fatty acid analysis of fresh and frozen-thawed immature and in vitro matured bovine oocytes. Reproduction 122, 131–138.
Lipid and fatty acid analysis of fresh and frozen-thawed immature and in vitro matured bovine oocytes.Crossref | GoogleScholarGoogle Scholar | 11425337PubMed |

Ko, PJ, and Dixon, SJ (2018). Protein palmitoylation and cancer. EMBO Reports 19, e46666.
Protein palmitoylation and cancer.Crossref | GoogleScholarGoogle Scholar | 30232163PubMed |

Komar, C (2005). Peroxisome proliferator-activated receptors (PPARs) and ovarian function – implications for regulating steroidogenesis, differentiation, and tissue remodeling. Reproductive Biology and Endocrinology 3, 41.
Peroxisome proliferator-activated receptors (PPARs) and ovarian function – implications for regulating steroidogenesis, differentiation, and tissue remodeling.Crossref | GoogleScholarGoogle Scholar | 16131403PubMed |

Komatsu, J, Yamano, S, Kuwahara, A, Tokumura, A, and Irahara, M (2006). The signaling pathways linking to lysophosphatidic acid-promoted meiotic maturation in mice. Life Sciences 79, 506–511.
The signaling pathways linking to lysophosphatidic acid-promoted meiotic maturation in mice.Crossref | GoogleScholarGoogle Scholar | 16492384PubMed |

Labrecque, R, Fournier, E, and Sirard, MA (2016). Transcriptome analysis of bovine oocytes from distinct follicle sizes: insights from correlation network analysis. Molecular Reproduction and Development 83, 558–569.
Transcriptome analysis of bovine oocytes from distinct follicle sizes: insights from correlation network analysis.Crossref | GoogleScholarGoogle Scholar | 27127921PubMed |

Law, SH, Chan, ML, Marathe, GK, Parveen, F, Chen, CH, and Ke, LY (2019). An updated review of lysophosphatidylcholine metabolism in human diseases. International Journal of Molecular Sciences 20, 1149.
An updated review of lysophosphatidylcholine metabolism in human diseases.Crossref | GoogleScholarGoogle Scholar |

Lazar, I, Clement, E, Dauvillier, S, Milhas, D, Ducoux-Petit, M, LeGonidec, S, Moro, C, Soldan, V, Dalle, S, Balor, S, Golzio, M, Burlet-Schiltz, O, Valet, P, Muller, C, and Nieto, L (2016). Adipocyte exosomes promote melanoma aggressiveness through fatty acid oxidation: a novel mechanism linking obesity and cancer. Cancer Research 76, 4051–4057.
Adipocyte exosomes promote melanoma aggressiveness through fatty acid oxidation: a novel mechanism linking obesity and cancer.Crossref | GoogleScholarGoogle Scholar | 27216185PubMed |

Leroy, JLMR, Vanholder, T, Delanghe, JR, Opsomer, G, Van Soom, A, Bols, PEJ, and de Kruif, A (2004). Metabolite and ionic composition of follicular fluid from different-sized follicles and their relationship to serum concentrations in dairy cows. Animal Reproduction Science 80, 201–211.
Metabolite and ionic composition of follicular fluid from different-sized follicles and their relationship to serum concentrations in dairy cows.Crossref | GoogleScholarGoogle Scholar |

Leroy, JLMR, Vanholder, T, Mateusen, B, Christophe, A, Opsomer, G, de Kruif, A, Genicot, G, and Van Soom, A (2005). Non-esterified fatty acids in follicular fluid of dairy cows and their effect on developmental capacity of bovine oocytes in vitro. Reproduction 130, 485–495.
Non-esterified fatty acids in follicular fluid of dairy cows and their effect on developmental capacity of bovine oocytes in vitro.Crossref | GoogleScholarGoogle Scholar |

Leroy, JLMR, Vanholder, T, Van Knegsel, AT, Garcia-Ispierto, I, and Bols, PE (2008a). Nutrient prioritization in dairy cows early postpartum: mismatch between metabolism and fertility? Reproduction in Domestic Animals 43, 96–103.
Nutrient prioritization in dairy cows early postpartum: mismatch between metabolism and fertility?Crossref | GoogleScholarGoogle Scholar |

Leroy, JLMR, Van Soom, A, Opsomer, G, and Bols, PEJ (2008b). The consequences of metabolic changes in high-yielding dairy cows on oocyte and embryo quality. Animal 2, 1120–1127.
The consequences of metabolic changes in high-yielding dairy cows on oocyte and embryo quality.Crossref | GoogleScholarGoogle Scholar |

Leroy, JLMR, Van Hoeck, V, Clemente, M, Rizos, D, Gutierrez-Adan, A, Van Soom, A, Uytterhoeven, M, and Bols, PEJ (2010). The effect of nutritionally induced hyperlipidaemia on in vitro bovine embryo quality. Human Reproduction 25, 768–778.
The effect of nutritionally induced hyperlipidaemia on in vitro bovine embryo quality.Crossref | GoogleScholarGoogle Scholar |

Leroy, JLMR, Sturmey, RG, Van Hoeck, V, De Bie, J, McKeegan, PJ, and Bols, PEJ (2014). Dietary fat supplementation and the consequences for oocyte and embryo quality: hype or significant benefit for dairy cow reproduction? Reproduction in Domestic Animals 49, 353–361.
Dietary fat supplementation and the consequences for oocyte and embryo quality: hype or significant benefit for dairy cow reproduction?Crossref | GoogleScholarGoogle Scholar |

Leroy, JLMR, Valckx, SDM, Jordaens, L, De Bie, J, Desmet, KLJ, Van Hoeck, V, Britt, JH, Marei, WF, and Bols, PEJ (2015). Nutrition and maternal metabolic health in relation to oocyte and embryo quality: critical views on what we learned from the dairy cow model. Reproduction, Fertility and Development 27, 693–703.
Nutrition and maternal metabolic health in relation to oocyte and embryo quality: critical views on what we learned from the dairy cow model.Crossref | GoogleScholarGoogle Scholar |

Li, H, Huang, X, Chang, X, Yao, J, He, Q, Shen, Z, Ji, Y, and Wang, K (2020). S100-A9 protein in exosomes derived from follicular fluid promotes inflammation via activation of NF-κB pathway in polycystic ovary syndrome. Journal of Cellular and Molecular Medicine 24, 114–125.
S100-A9 protein in exosomes derived from follicular fluid promotes inflammation via activation of NF-κB pathway in polycystic ovary syndrome.Crossref | GoogleScholarGoogle Scholar | 31568644PubMed |

Liang, YS, Qi, WT, Guo, W, Wang, CL, Hu, ZB, and Li, AK (2018). Genistein and daidzein induce apoptosis of colon cancer cells by inhibiting the accumulation of lipid droplets. Food & Nutrition Research 62, 1384.
Genistein and daidzein induce apoptosis of colon cancer cells by inhibiting the accumulation of lipid droplets.Crossref | GoogleScholarGoogle Scholar |

Lolicato, F, Brouwers, JF, van de Lest, CHA, Wubbolts, R, Aardema, H, Priore, P, Roelen, BAJ, Helms, JB, and Gadella, BM (2015). The cumulus cell layer protects the bovine maturing oocyte against fatty acid-induced lipotoxicity. Biology of Reproduction 92, 16.
The cumulus cell layer protects the bovine maturing oocyte against fatty acid-induced lipotoxicity.Crossref | GoogleScholarGoogle Scholar | 25297544PubMed |

Lonergan, P, Monaghan, P, Rizos, D, Boland, MP, and Gordon, I (1994). Effect of follicle size on bovine oocyte quality and developmental competence following maturation, fertilization, and culture in vitro. Molecular Reproduction and Development 37, 48–53.
Effect of follicle size on bovine oocyte quality and developmental competence following maturation, fertilization, and culture in vitro.Crossref | GoogleScholarGoogle Scholar | 8129930PubMed |

Marei, WFA, Van Raemdonck, G, Baggerman, G, Bols, PEJ, and Leroy, JLMR (2019). Proteomic changes in oocytes after in vitro maturation in lipotoxic conditions are different from those in cumulus cells. Scientific Reports 9, 3673.
Proteomic changes in oocytes after in vitro maturation in lipotoxic conditions are different from those in cumulus cells.Crossref | GoogleScholarGoogle Scholar | 30842615PubMed |

Mathias, S, Pena, LA, and Kolesnick, RN (1998). Signal transduction of stress via ceramide [Review]. Biochemical Journal 335, 465–480.
Signal transduction of stress via ceramide [Review].Crossref | GoogleScholarGoogle Scholar |

Matoba, S, O’Hara, L, Carter, F, Kelly, AK, Fair, T, Rizos, D, and Lonergan, P (2012). The association between metabolic parameters and oocyte quality early and late postpartum in Holstein dairy cows. Journal of Dairy Science 95, 1257–1266.
The association between metabolic parameters and oocyte quality early and late postpartum in Holstein dairy cows.Crossref | GoogleScholarGoogle Scholar | 22365209PubMed |

Matoba, S, Bender, K, Fahey, AG, Mamo, S, Brennan, L, Lonergan, P, and Fair, T (2014). Predictive value of bovine follicular components as markers of oocyte developmental potential. Reproduction, Fertility and Development 26, 337–345.
Predictive value of bovine follicular components as markers of oocyte developmental potential.Crossref | GoogleScholarGoogle Scholar |

Matsuda, F, Inoue, N, Manabe, N, and Ohkura, S (2012). Follicular growth and atresia in mammalian ovaries: regulation by survival and death of granulosa cells. Journal of Reproduction and Development 58, 44–50.
Follicular growth and atresia in mammalian ovaries: regulation by survival and death of granulosa cells.Crossref | GoogleScholarGoogle Scholar |

Mazerbourg, S, Bondy, CA, Zhou, J, and Monget, P (2003). The insulin-like growth factor system: a key determinant role in the growth and selection of ovarian follicles? a comparative species study. Reproduction in Domestic Animals 38, 247–258.
The insulin-like growth factor system: a key determinant role in the growth and selection of ovarian follicles? a comparative species study.Crossref | GoogleScholarGoogle Scholar | 12887564PubMed |

McEvoy, TG, Coull, GD, Broadbent, PJ, Hutchinson, JSM, and Speake, BK (2000). Fatty acid composition of lipids in immature cattle, pig and sheep oocytes with intact zona pellucida. Journal of Reproduction and Fertility 118, 163–170.
Fatty acid composition of lipids in immature cattle, pig and sheep oocytes with intact zona pellucida.Crossref | GoogleScholarGoogle Scholar | 10793638PubMed |

McNatty, KP, Heath, DA, Henderson, KM, Lun, S, Hurst, PR, Ellis, LM, Montgomery, GW, Morrison, L, and Thurley, DC (1984). Some aspects of thecal and granulosa cell function during follicular development in the bovine ovary. Journal of Reproduction and Fertility 72, 39–53.
Some aspects of thecal and granulosa cell function during follicular development in the bovine ovary.Crossref | GoogleScholarGoogle Scholar | 6540808PubMed |

Milhas, D, Clarke, CJ, and Hannun, YA (2010). Sphingomyelin metabolism at the plasma membrane: implications for bioactive sphingolipids. NIH FEBS Letters 584, 1887–1894.
Sphingomyelin metabolism at the plasma membrane: implications for bioactive sphingolipids.Crossref | GoogleScholarGoogle Scholar | 19857494PubMed |

Moallem, U (2018). Invited review: roles of dietary n−3 fatty acids in performance, milk fat composition, and reproductive and immune systems in dairy cattle. Journal of Dairy Science 101, 8641–8661.
Invited review: roles of dietary n−3 fatty acids in performance, milk fat composition, and reproductive and immune systems in dairy cattle.Crossref | GoogleScholarGoogle Scholar | 30100509PubMed |

Moallem, U, Shafran, A, Zachut, M, Dekel, I, Portnick, Y, and Arieli, A (2013). 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 146, 603–614.
Dietary alpha-linolenic acid from flaxseed oil improved folliculogenesis and IVF performance in dairy cows, similar to eicosapentaenoic and docosahexaenoic acids from fish oil.Crossref | GoogleScholarGoogle Scholar | 24062566PubMed |

Monget, P, and Martin, GB (1997). Involvement of insulin-like growth factors in the interactions between nutrition and reproduction in female mammals. Human Reproduction 12, 33–52.
Involvement of insulin-like growth factors in the interactions between nutrition and reproduction in female mammals.Crossref | GoogleScholarGoogle Scholar | 9403320PubMed |

Monniaux, D (2016). Driving folliculogenesis by the oocyte-somatic cell dialog: lessons from genetic models. Theriogenology 86, 41–53.
Driving folliculogenesis by the oocyte-somatic cell dialog: lessons from genetic models.Crossref | GoogleScholarGoogle Scholar | 27155734PubMed |

Monniaux, D, Huet, C, Besnard, N, Clement, F, Bosc, M, Pisselet, C, Monget, P, and Mariana, JC (1997). Follicular growth and ovarian dynamics in mammals. Journal of Reproduction and Fertility. Supplement 51, 3–23.
| 9404267PubMed |

Montani, DA, Cordeiro, FB, Regiani, T, Victorino, AB, Pilau, EJ, Gozzo, FC, Ferreira, CR, Fraietta, R, and Lo Turco, EG (2012). The follicular microenviroment as a predictor of pregnancy: MALDI-TOF MS lipid profile in cumulus cells. Journal of Assisted Reproduction and Genetics 29, 1289–1297.
The follicular microenviroment as a predictor of pregnancy: MALDI-TOF MS lipid profile in cumulus cells.Crossref | GoogleScholarGoogle Scholar | 22968515PubMed |

Montani, DA, Braga, DPAF, Borges, E, Camargo, M, Cordeiro, FB, Pilau, EJ, Gozzo, FC, Fraietta, R, and Turco, EGL (2019). Understanding mechanisms of oocyte development by follicular fluid lipidomics. Journal of Assisted Reproduction and Genetics 36, 1003–1011.
Understanding mechanisms of oocyte development by follicular fluid lipidomics.Crossref | GoogleScholarGoogle Scholar | 31011990PubMed |

Mukherjee, S, and Suresh, SN (2019). Neuron–astrocyte liaison to maintain lipid metabolism of brain. Trends in Endocrinology and Metabolism 30, 573–575.
Neuron–astrocyte liaison to maintain lipid metabolism of brain.Crossref | GoogleScholarGoogle Scholar | 31401016PubMed |

Nadolski, MJ, and Linder, ME (2007). Protein lipidation. FEBS Journal 274, 5202–5210.
Protein lipidation.Crossref | GoogleScholarGoogle Scholar |

Nagano, M (2019). Acquisition of developmental competence and in vitro growth culture of bovine oocytes. Journal of Reproduction and Development 65, 195–201.
Acquisition of developmental competence and in vitro growth culture of bovine oocytes.Crossref | GoogleScholarGoogle Scholar |

Naumenko, VS, and Ponimaskin, E (2018). Palmitoylation as a functional regulator of neurotransmitter receptors. Neural Plasticity 2018, 1.
Palmitoylation as a functional regulator of neurotransmitter receptors.Crossref | GoogleScholarGoogle Scholar |

Navakanitworakul, R, Hung, WT, Gunewardena, S, Davis, JS, Chotigeat, W, and Christenson, LK (2016). Characterization and small RNA content of extracellular vesicles in follicular fluid of developing bovine antral follicles. Scientific Reports 6, 25486.
Characterization and small RNA content of extracellular vesicles in follicular fluid of developing bovine antral follicles.Crossref | GoogleScholarGoogle Scholar | 27158133PubMed |

Nuttinck, F, Gall, L, Ruffini, S, Laffont, L, Clement, L, Reinaud, P, Adenot, P, Grimard, B, Charpigny, G, and Marquant-Le Guienne, B (2011). PTGS2-related PGE2 affects oocyte MAPK phosphorylation and meiosis progression in cattle: late effects on early embryonic development. Biology of Reproduction 84, 1248–1257.
PTGS2-related PGE2 affects oocyte MAPK phosphorylation and meiosis progression in cattle: late effects on early embryonic development.Crossref | GoogleScholarGoogle Scholar | 21293029PubMed |

O’Gorman, A, Wallace, M, Cottell, E, Gibney, MJ, McAuliffe, FM, Wingfield, M, and Brennan, L (2013). Metabolic profiling of human follicular fluid identifies potential biomarkers of oocyte developmental competence. Reproduction 146, 389–95.
| 23886995PubMed |
| 23886995PubMed |

Osz, K, Ross, M, and Petrik, J (2014). The thrombospondin-1 receptor CD36 is an important mediator of ovarian angiogenesis and folliculogenesis. Reproductive Biology and Endocrinology 12, 21.
The thrombospondin-1 receptor CD36 is an important mediator of ovarian angiogenesis and folliculogenesis.Crossref | GoogleScholarGoogle Scholar | 24628875PubMed | 24628875PubMed |

Paulini, F, Silva, RC, de Paula Rolo, JLJ, and Lucci, CM (2014). Ultrastructural changes in oocytes during folliculogenesis in domestic mammals. Journal of Ovarian Research 7, 102.
Ultrastructural changes in oocytes during folliculogenesis in domestic mammals.Crossref | GoogleScholarGoogle Scholar | 25358389PubMed | 25358389PubMed |

Pedram, A, Razandi, M, Deschenes, RJ, and Levin, ER (2012). DHHC-7 and -21 are palmitoylacyltransferases for sex steroid receptors. Molecular Biology of the Cell 23, 188–199.
DHHC-7 and -21 are palmitoylacyltransferases for sex steroid receptors.Crossref | GoogleScholarGoogle Scholar | 22031296PubMed | 22031296PubMed |

Perry, WJ, Patterson, NH, Prentice, BM, Neumann, EK, Caprioli, RM, and Spraggins, JM (2020). Uncovering matrix effects on lipid analyses in MALDI imaging mass spectrometry experiments. Journal of Mass Spectrometry 55, e4491.
| 31860760PubMed |
| 31860760PubMed |

Prates, EG, Nunes, JT, and Pereira, RM (2014). A role of lipid metabolism during cumulus-oocyte complex maturation: impact of lipid modulators to improve embryo production. Mediators of Inflammation 2014, 692067.
A role of lipid metabolism during cumulus-oocyte complex maturation: impact of lipid modulators to improve embryo production.Crossref | GoogleScholarGoogle Scholar | 24733963PubMed | 24733963PubMed |

Prokazova, NV, Zvezdina, ND, and Korotaeva, AA (1998). Effect of lysophosphatidylcholine on transmembrane signal transduction. Biochemistry 63, 31–37.
| 9526092PubMed |
| 9526092PubMed |

Record, M, Silvente-Poirot, S, Poirot, M, and Wakelam, MJO (2018). Extracellular vesicles: lipids as key components of their biogenesis and functions. Journal of Lipid Research 59, 1316–1324.
Extracellular vesicles: lipids as key components of their biogenesis and functions.Crossref | GoogleScholarGoogle Scholar | 29764923PubMed | 29764923PubMed |

Renaville, B, Bacciu, N, Comin, A, Motta, M, Poli, I, Vanini, G, and Prandi, A (2010). Plasma and follicular fluid fatty acid profiles in dairy cows. Reproduction in Domestic Animals 45, 118–121.
Plasma and follicular fluid fatty acid profiles in dairy cows.Crossref | GoogleScholarGoogle Scholar | 19055554PubMed | 19055554PubMed |

Resh, MD (2016). Fatty acylation of proteins: the long and the short of it. Progress in Lipid Research 63, 120–131.
Fatty acylation of proteins: the long and the short of it.Crossref | GoogleScholarGoogle Scholar | 27233110PubMed | 27233110PubMed |

Richardson, MC, Cameron, IT, Simonis, CD, Das, MC, Hodge, TE, Zhang, J, and Byrne, CD (2005). Insulin and human chorionic gonadotropin cause a shift in the balance of sterol regulatory element-binding protein (SREBP) isoforms toward the SREBP-1c isoform in cultures of human granulosa cells. Journal of Clinical Endocrinology and Metabolism 90, 3738–3746.
Insulin and human chorionic gonadotropin cause a shift in the balance of sterol regulatory element-binding protein (SREBP) isoforms toward the SREBP-1c isoform in cultures of human granulosa cells.Crossref | GoogleScholarGoogle Scholar | 15769984PubMed | 15769984PubMed |

Roche, JF (2006). The effect of nutritional management of the dairy cow on reproductive efficiency. Animal Reproduction Science 96, 282–296.
The effect of nutritional management of the dairy cow on reproductive efficiency.Crossref | GoogleScholarGoogle Scholar | 16996705PubMed | 16996705PubMed |

Rodrigues, TA, Tuna, KM, Alli, AA, Tribulo, P, Hansen, PJ, Koh, J, and Paula-Lopes, FF (2019). Follicular fluid exosomes act on the bovine oocyte to improve oocyte competence to support development and survival to heat shock. Reproduction, Fertility and Development 31, 888–897.
Follicular fluid exosomes act on the bovine oocyte to improve oocyte competence to support development and survival to heat shock.Crossref | GoogleScholarGoogle Scholar |

Russell, DL, Gilchrist, RB, Brown, HM, and Thompson, JG (2016). Bidirectional communication between cumulus cells and the oocyte: old hands and new players? Theriogenology 86, 62–68.
Bidirectional communication between cumulus cells and the oocyte: old hands and new players?Crossref | GoogleScholarGoogle Scholar | 27160446PubMed | 27160446PubMed |

Sanchez-Lazo, L, Brisard, D, Elis, S, Maillard, V, Uzbekov, R, Labas, V, Desmarchais, A, Papillier, P, Monget, P, and Uzbekova, S (2014). Fatty acid synthesis and oxidation in cumulus cells support oocyte maturation in bovine. Molecular Endocrinology 28, 1502–1521.
Fatty acid synthesis and oxidation in cumulus cells support oocyte maturation in bovine.Crossref | GoogleScholarGoogle Scholar | 25058602PubMed | 25058602PubMed |

Sanders, JR, and Jones, KT (2018). Regulation of the meiotic divisions of mammalian oocytes and eggs. Biochemical Society Transactions 46, 797–806.
Regulation of the meiotic divisions of mammalian oocytes and eggs.Crossref | GoogleScholarGoogle Scholar | 29934303PubMed | 29934303PubMed |

Santos, PH, Fontes, PK, Franchi, FF, Nogueira, MFG, Belaz, KRA, Tata, A, Eberlin, MN, Sudano, MJ, Barros, CM, and Castilho, ACS (2017). Lipid profiles of follicular fluid from cows submitted to ovarian superstimulation. Theriogenology 94, 64–70.
Lipid profiles of follicular fluid from cows submitted to ovarian superstimulation.Crossref | GoogleScholarGoogle Scholar | 28407862PubMed | 28407862PubMed |

Sarnyai, F, Donkó, MB, Mátyási, J, Gór-Nagy, Z, Marczi, I, Simon-Szabó, L, Zámbó, V, Somogyi, A, Csizmadia, T, Lőw, P, Szelényi, P, Kereszturi, É, Tóth, B, and Csala, M (2019). Cellular toxicity of dietary trans fatty acids and its correlation with ceramide and diglyceride accumulation. Food and Chemical Toxicology 124, 324–335.
Cellular toxicity of dietary trans fatty acids and its correlation with ceramide and diglyceride accumulation.Crossref | GoogleScholarGoogle Scholar | 30572061PubMed | 30572061PubMed |

Scalici, E, Bechoua, S, Astruc, K, Duvillard, L, Gautier, T, Drouineaud, V, Jimenez, C, and Hamamah, S (2016). Apolipoprotein B is regulated by gonadotropins and constitutes a predictive biomarker of IVF outcomes. Reproductive Biology and Endocrinology 14, 28.
Apolipoprotein B is regulated by gonadotropins and constitutes a predictive biomarker of IVF outcomes.Crossref | GoogleScholarGoogle Scholar | 27209151PubMed | 27209151PubMed |

Schams, D, and Berisha, B (2002). Steroids as local regulators of ovarian activity in domestic animals. Domestic Animal Endocrinology 23, 53–65.
Steroids as local regulators of ovarian activity in domestic animals.Crossref | GoogleScholarGoogle Scholar | 12142226PubMed | 12142226PubMed |

Shemesh, M, Bensadoun, A, and Hansel, W (1976). Lipoprotein lipase activity in the bovine corpus luteum during the estrous cycle and early pregnancy. Proceedings of the Society for Experimental Biology and Medicine 151, 667–669.
Lipoprotein lipase activity in the bovine corpus luteum during the estrous cycle and early pregnancy.Crossref | GoogleScholarGoogle Scholar | 1265048PubMed | 1265048PubMed |

Shen, WJ, Azhar, S, and Kraemer, FB (2016). Lipid droplets and steroidogenic cells. Experimental Cell Research 340, 209–214.
Lipid droplets and steroidogenic cells.Crossref | GoogleScholarGoogle Scholar | 26639173PubMed | 26639173PubMed |

Silva, RC, Báo, SN, Jivago, JLPR, and Lucci, CM (2011). Ultrastructural characterization of porcine oocytes and adjacent follicular cells during follicle development: lipid component evolution. Theriogenology 76, 1647–1657.
Ultrastructural characterization of porcine oocytes and adjacent follicular cells during follicle development: lipid component evolution.Crossref | GoogleScholarGoogle Scholar | 21835450PubMed | 21835450PubMed |

Silva-Santos, KC, Ferreira, CR, Santos, GMG, Eberlin, MN, Siloto, LS, Rosa, CO, Marcantonio, TN, and Seneda, MM (2014). MALDI-MS lipid profiles of oocytes recovered by ovum pickup from Bos indicus and 1/2 indicus × taurus with high vs low oocyte yields. Reproduction in Domestic Animals 49, 711–718.
MALDI-MS lipid profiles of oocytes recovered by ovum pickup from Bos indicus and 1/2 indicus × taurus with high vs low oocyte yields.Crossref | GoogleScholarGoogle Scholar | 25110198PubMed | 25110198PubMed |

Simons, K, and Toomre, D (2000). Lipid rafts and signal transduction. Nature Reviews Molecular Cell Biology 1, 31–39.
Lipid rafts and signal transduction.Crossref | GoogleScholarGoogle Scholar | 11413487PubMed | 11413487PubMed |

Sinderewicz, E, Grycmacher, K, Boruszewska, D, Kowalczyk-Zieba, I, Staszkiewicz, J, Ślężak, T, and Woclawek-Potocka, I (2017). Expression of factors involved in apoptosis and cell survival is correlated with enzymes synthesizing lysophosphatidic acid and its receptors in granulosa cells originating from different types of bovine ovarian follicles. Reproductive Biology and Endocrinology 15, 72.
Expression of factors involved in apoptosis and cell survival is correlated with enzymes synthesizing lysophosphatidic acid and its receptors in granulosa cells originating from different types of bovine ovarian follicles.Crossref | GoogleScholarGoogle Scholar | 28874163PubMed | 28874163PubMed |

Sirard, MA, Richard, F, Blondin, P, and Robert, C (2006). Contribution of the oocyte to embryo quality. Theriogenology 65, 126–136.
Contribution of the oocyte to embryo quality.Crossref | GoogleScholarGoogle Scholar | 16256189PubMed | 16256189PubMed |

Skinner, MK, Schmidt, M, Savenkova, MI, Sadler-Riggleman, I, and Nilsson, EE (2008). Regulation of granulosa and theca cell transcriptomes during ovarian antral follicle development. Molecular Reproduction and Development 75, 1457–1472.
Regulation of granulosa and theca cell transcriptomes during ovarian antral follicle development.Crossref | GoogleScholarGoogle Scholar | 18288646PubMed | 18288646PubMed |

Skotland, T, Sandvig, K, and Llorente, A (2017). Lipids in exosomes: current knowledge and the way forward. Progress in Lipid Research 66, 30–41.
Lipids in exosomes: current knowledge and the way forward.Crossref | GoogleScholarGoogle Scholar | 28342835PubMed | 28342835PubMed |

Skotland, T, Sagini, K, Sandvig, K, and Llorente, A (2020). An emerging focus on lipids in extracellular vesicles. Advanced Drug Delivery Reviews 159, 308–321.
An emerging focus on lipids in extracellular vesicles.Crossref | GoogleScholarGoogle Scholar | 32151658PubMed | 32151658PubMed |

Smathers, RL, and Petersen, DR (2011). The human fatty acid-binding protein family: evolutionary divergences and functions. Human Genomics 5, 170.
The human fatty acid-binding protein family: evolutionary divergences and functions.Crossref | GoogleScholarGoogle Scholar | 21504868PubMed | 21504868PubMed |

Sobinoff, AP, Sutherland, JM, and McLaughlin, EA (2013). Intracellular signalling during female gametogenesis. Molecular Human Reproduction 19, 265–278.
Intracellular signalling during female gametogenesis.Crossref | GoogleScholarGoogle Scholar | 23247812PubMed | 23247812PubMed |

Spinelli, M, Fusco, S, and Grassi, C (2018). Nutrient-dependent changes of protein palmitoylation: impact on nuclear enzymes and regulation of gene expression. International Journal of Molecular Sciences 19, 3820.
Nutrient-dependent changes of protein palmitoylation: impact on nuclear enzymes and regulation of gene expression.Crossref | GoogleScholarGoogle Scholar |

Stahl, A (2004). A current review of fatty acid transport proteins (SLC27). Pflügers Archiv: European Journal of Physiology 447, 722–727.
A current review of fatty acid transport proteins (SLC27).Crossref | GoogleScholarGoogle Scholar | 12856180PubMed | 12856180PubMed |

Sturmey, RG, O’Toole, PJ, and Leese, HJ (2006). Fluorescence resonance energy transfer analysis of mitochondrial: lipid association in the porcine oocyte. Reproduction 132, 829–837.
Fluorescence resonance energy transfer analysis of mitochondrial: lipid association in the porcine oocyte.Crossref | GoogleScholarGoogle Scholar | 17127743PubMed | 17127743PubMed |

Sturmey, RG, Reis, A, Leese, HJ, and McEvoy, TG (2009). Role of fatty acids in energy provision during oocyte maturation and early embryo development. Reproduction in Domestic Animals 44, 50–58.
Role of fatty acids in energy provision during oocyte maturation and early embryo development.Crossref | GoogleScholarGoogle Scholar | 19660080PubMed | 19660080PubMed |

Subra, C, Grand, D, Laulagnier, K, Stella, A, Lambeau, G, Paillasse, M, De Medina, P, Monsarrat, B, Perret, B, Silvente-Poirot, S, Poirot, M, and Record, M (2010). Exosomes account for vesicle-mediated transcellular transport of activatable phospholipases and prostaglandins. Journal of Lipid Research 51, 2105–2120.
Exosomes account for vesicle-mediated transcellular transport of activatable phospholipases and prostaglandins.Crossref | GoogleScholarGoogle Scholar | 20424270PubMed | 20424270PubMed |

Sudano, MJ, Rascado, TDS, Tata, A, Belaz, KRA, Santos, VG, Valente, RS, Mesquita, FS, Ferreira, CR, Araújo, JP, Eberlin, MN, and Landim-Alvarenga, FDC (2016). Lipidome signatures in early bovine embryo development. Theriogenology 86, 472–484.e1.
Lipidome signatures in early bovine embryo development.Crossref | GoogleScholarGoogle Scholar | 27107972PubMed | 27107972PubMed |

Sutton, ML, Gilchrist, RB, and Thompson, JG (2003). Effects of in-vivo and in-vitro environments on the metabolism of the cumulus-oocyte complex and its influence on oocyte developmental capacity. Human Reproduction Update 9, 35–48.
Effects of in-vivo and in-vitro environments on the metabolism of the cumulus-oocyte complex and its influence on oocyte developmental capacity.Crossref | GoogleScholarGoogle Scholar | 12638780PubMed | 12638780PubMed |

Sutton-McDowall, ML, Gilchrist, RB, and Thompson, JG (2010). The pivotal role of glucose metabolism in determining oocyte developmental competence. Reproduction 139, 685–695.
The pivotal role of glucose metabolism in determining oocyte developmental competence.Crossref | GoogleScholarGoogle Scholar | 20089664PubMed | 20089664PubMed |

Sutton-McDowall, ML, Yelland, R, MacMillan, KL, Robker, RL, and Thompson, JG (2014). A study relating the composition of follicular fluid and blood plasma from individual Holstein dairy cows to the in vitro developmental competence of pooled abattoir-derived oocytes. Theriogenology 82, 95–103.
A study relating the composition of follicular fluid and blood plasma from individual Holstein dairy cows to the in vitro developmental competence of pooled abattoir-derived oocytes.Crossref | GoogleScholarGoogle Scholar | 24746097PubMed | 24746097PubMed |

Tabaczar, S, Czogalla, A, Podkalicka, J, Biernatowska, A, and Sikorski, AF (2017). Protein palmitoylation: palmitoyltransferases and their specificity. Experimental Biology and Medicine 242, 1150–1157.
Protein palmitoylation: palmitoyltransferases and their specificity.Crossref | GoogleScholarGoogle Scholar | 28485685PubMed | 28485685PubMed |

Tajima, K, Orisaka, M, Mori, T, and Kotsuji, F (2007). Ovarian theca cells in follicular function. Reproductive BioMedicine Online 15, 591–609.
Ovarian theca cells in follicular function.Crossref | GoogleScholarGoogle Scholar | 18028753PubMed | 18028753PubMed |

Tan, Y, Jin, Y, Wu, X, and Ren, Z (2019). PSMD1 and PSMD2 regulate HepG2 cell proliferation and apoptosis via modulating cellular lipid droplet metabolism. BMC Molecular Biology 20, 24.
PSMD1 and PSMD2 regulate HepG2 cell proliferation and apoptosis via modulating cellular lipid droplet metabolism.Crossref | GoogleScholarGoogle Scholar | 31703613PubMed | 31703613PubMed |

Tanghe, S, Van Soom, A, Nauwynck, H, Coryn, M, and De Kruif, A (2002). Mini review: functions of the cumulus oophorus during oocyte maturation, ovulation and fertilization. Molecular Reproduction and Development 61, 414–424.
Mini review: functions of the cumulus oophorus during oocyte maturation, ovulation and fertilization.Crossref | GoogleScholarGoogle Scholar | 11835587PubMed | 11835587PubMed |

Tesfaye, D, Hailay, T, Salilew-Wondim, D, Hoelker, M, Bitseha, S, and Gebremedhn, S (2020). Extracellular vesicle mediated molecular signaling in ovarian follicle: implication for oocyte developmental competence. Theriogenology 150, 70–74.
Extracellular vesicle mediated molecular signaling in ovarian follicle: implication for oocyte developmental competence.Crossref | GoogleScholarGoogle Scholar | 32088041PubMed | 32088041PubMed |

Thompson, JG, Lane, M, and Gilchrist, RB (2007). Metabolism of the bovine cumulus-oocyte complex and influence on subsequent developmental competence. Society for Reproduction and Fertility 64, 179–90.
Metabolism of the bovine cumulus-oocyte complex and influence on subsequent developmental competence.Crossref | GoogleScholarGoogle Scholar |

Tokumura, A, Miyake, M, Nishioka, Y, Yamano, S, Aono, T, and Fukuzawa, K (1999). Production of lysophosphatidic acids by lysophospholipase D in human follicular fluids of in vitro fertilization patients. Biology of Reproduction 61, 195–199.
Production of lysophosphatidic acids by lysophospholipase D in human follicular fluids of in vitro fertilization patients.Crossref | GoogleScholarGoogle Scholar | 10377049PubMed | 10377049PubMed |

Trigatti, BL, Rigotti, A, and Braun, A (2000). Cellular and physiological roles of SR-BI, a lipoprotein receptor which mediates selective lipid uptake. Biochimica et Biophysica Acta, Molecular and Cell Biology of Lipids 1529, 276–286.
Cellular and physiological roles of SR-BI, a lipoprotein receptor which mediates selective lipid uptake.Crossref | GoogleScholarGoogle Scholar |

Uzbekova, S, Elis, S, Teixeira-Gomes, AP, Desmarchais, A, Maillard, V, and Labas, V (2015). MALDI mass spectrometry imaging of lipids and gene expression reveals differences in fatty acid metabolism between follicular compartments in porcine ovaries. Biology 4, 216–236.
MALDI mass spectrometry imaging of lipids and gene expression reveals differences in fatty acid metabolism between follicular compartments in porcine ovaries.Crossref | GoogleScholarGoogle Scholar | 25756245PubMed | 25756245PubMed |

Uzbekova, S, Almiñana, C, Labas, V, Teixeira-Gomes, AP, Combes-Soia, L, Tsikis, G, Carvalho, AV, Uzbekov, R, and Singina, G (2020). Protein cargo of extracellular vesicles from bovine follicular fluid and analysis of their origin from different ovarian cells. Frontiers in Veterinary Science 7, 584948.
Protein cargo of extracellular vesicles from bovine follicular fluid and analysis of their origin from different ovarian cells.Crossref | GoogleScholarGoogle Scholar | 33330709PubMed |

van der Veen, JN, Lingrell, S, da Silva, RP, Jacobs, RL, and Vance, DE (2014). The concentration of phosphatidylethanolamine in mitochondria can modulate ATP production and glucose metabolism in mice. Diabetes 63, 2620–2630.
The concentration of phosphatidylethanolamine in mitochondria can modulate ATP production and glucose metabolism in mice.Crossref | GoogleScholarGoogle Scholar | 24677714PubMed | 24677714PubMed |

van der Veen, JN, Kennelly, JP, Wan, S, Vance, JE, Vance, DE, and Jacobs, RL (2017). The critical role of phosphatidylcholine and phosphatidylethanolamine metabolism in health and disease. Biochimica et Biophysica Acta – Biomembranes 1859, 1558–1572.
The critical role of phosphatidylcholine and phosphatidylethanolamine metabolism in health and disease.Crossref | GoogleScholarGoogle Scholar | 28411170PubMed | 28411170PubMed |

Van Hoeck, V, Sturmey, RG, Bermejo-Alvarez, P, Rizos, D, Gutierrez-Adan, A, Leese, HJ, Bols, PEJ, and Leroy, JLMR (2011). Elevated non-esterified fatty acid concentrations during bovine oocyte maturation compromise early embryo physiology. PLoS One 6, e23183.
Elevated non-esterified fatty acid concentrations during bovine oocyte maturation compromise early embryo physiology.Crossref | GoogleScholarGoogle Scholar | 21858021PubMed | 21858021PubMed |

Van Hoeck, V, Leroy, JLMR, Arias-Alvarez, M, Rizos, D, Gutierrez-Adan, A, Schnorbusch, K, Bols, PEJ, Leese, HJ, and Sturmey, RG (2013). Oocyte developmental failure in response to elevated non-esterified fatty acid concentrations: mechanistic insights. Reproduction 145, 33–44.
Oocyte developmental failure in response to elevated non-esterified fatty acid concentrations: mechanistic insights.Crossref | GoogleScholarGoogle Scholar | 23108110PubMed | 23108110PubMed |

van Montfoort, APA, Plösch, T, Hoek, A, and Tietge, UJF (2014). Impact of maternal cholesterol metabolism on ovarian follicle development and fertility. Journal of Reproductive Immunology 104–105, 32–36.
Impact of maternal cholesterol metabolism on ovarian follicle development and fertility.Crossref | GoogleScholarGoogle Scholar |

Vance, JE (2015). Phospholipid synthesis and transport in mammalian cells. Traffic 16, 1–18.
Phospholipid synthesis and transport in mammalian cells.Crossref | GoogleScholarGoogle Scholar | 25243850PubMed | 25243850PubMed |

Vitti, M, Di Emidio, G, Di Carlo, M, Carta, G, Antonosante, A, Artini, PG, Cimini, A, Tatone, C, and Benedetti, E (2016). Peroxisome proliferator-activated receptors in female reproduction and fertility. PPAR Research 2016, 4612306.
Peroxisome proliferator-activated receptors in female reproduction and fertility.Crossref | GoogleScholarGoogle Scholar | 27559343PubMed | 27559343PubMed |

Volpe, A, Coukos, G, Uccelli, E, Droghini, F, Adamo, R, and Artini, PG (1991). Follicular fluid lipoproteins in preovulatory period and their relationship with follicular maturation and progesterone production by human granulosa-luteal cells in vivo and in vitro. Journal of Endocrinological Investigation 14, 737–742.
Follicular fluid lipoproteins in preovulatory period and their relationship with follicular maturation and progesterone production by human granulosa-luteal cells in vivo and in vitro.Crossref | GoogleScholarGoogle Scholar | 1761808PubMed | 1761808PubMed |

Walter, J, Monthoux, C, Fortes, C, Grossmann, J, Roschitzki, B, Meili, T, Riond, B, Hofmann-Lehmann, R, Naegeli, H, and Bleul, U (2020). The bovine cumulus proteome is influenced by maturation condition and maturational competence of the oocyte. Scientific Reports 10, 9880.
The bovine cumulus proteome is influenced by maturation condition and maturational competence of the oocyte.Crossref | GoogleScholarGoogle Scholar | 32555221PubMed | 32555221PubMed |

Wang, Q, and Sun, QY (2007). Evaluation of oocyte quality: morphological, cellular and molecular predictors. Reproduction, Fertility and Development 19, 1–12.
Evaluation of oocyte quality: morphological, cellular and molecular predictors.Crossref | GoogleScholarGoogle Scholar |

Wang, J, Chen, Q, Zhou, J, Wen, J, Bian, F, Li, G, Mu, X, Han, Y, Xia, G, and Zhang, M (2012). Specific protein kinase C isoforms α and βI are involved in follicle-stimulating hormone-induced mouse follicle-enclosed oocytes meiotic resumption. PLoS ONE 7, e45043.
Specific protein kinase C isoforms α and βI are involved in follicle-stimulating hormone-induced mouse follicle-enclosed oocytes meiotic resumption.Crossref | GoogleScholarGoogle Scholar | 23028752PubMed | 23028752PubMed |

Wang, W, Zhu, N, Yan, T, Shi, YN, Chen, J, Zhang, CJ, Xie, XJ, Liao, DF, and Qin, L (2020). The crosstalk: exosomes and lipid metabolism. Cell Communication and Signaling 18, 119.
The crosstalk: exosomes and lipid metabolism.Crossref | GoogleScholarGoogle Scholar | 32746850PubMed | 32746850PubMed |

Warzych, E, and Lipinska, P (2020). Energy metabolism of follicular environment during oocyte growth and maturation. Journal of Reproduction and Development 66, 1–7.
Energy metabolism of follicular environment during oocyte growth and maturation.Crossref | GoogleScholarGoogle Scholar |

Warzych, E, Pawlak, P, Pszczola, M, Cieslak, A, and Lechniak, D (2017a). Prepubertal heifers versus cows—the differences in the follicular environment. Theriogenology 87, 36–47.
Prepubertal heifers versus cows—the differences in the follicular environment.Crossref | GoogleScholarGoogle Scholar | 27634396PubMed | 27634396PubMed |

Warzych, E, Pawlak, P, Pszczola, M, Cieslak, A, Madeja, ZE, and Lechniak, D (2017b). Interactions of bovine oocytes with follicular elements with respect to lipid metabolism. Animal Science Journal 88, 1491–1497.
Interactions of bovine oocytes with follicular elements with respect to lipid metabolism.Crossref | GoogleScholarGoogle Scholar | 28402007PubMed | 28402007PubMed |

Wathes, DC, Fenwick, M, Cheng, Z, Bourne, N, Llewellyn, S, Morris, DG, Kenny, D, Murphy, J, and Fitzpatrick, R (2007). Influence of negative energy balance on cyclicity and fertility in the high producing dairy cow. Theriogenology 68, S232–S241.
Influence of negative energy balance on cyclicity and fertility in the high producing dairy cow.Crossref | GoogleScholarGoogle Scholar | 17475319PubMed | 17475319PubMed |

Wathes, DC, Clempson, AM, and Pollott, GE (2013). Associations between lipid metabolism and fertility in the dairy cow. Reproduction, Fertility and Development 25, 48–61.
Associations between lipid metabolism and fertility in the dairy cow.Crossref | GoogleScholarGoogle Scholar |

Wei, X, Song, H, and Semenkovich, CF (2014). Insulin-regulated protein palmitoylation impacts endothelial cell function. Arteriosclerosis, Thrombosis, and Vascular Biology 34, 346–354.
Insulin-regulated protein palmitoylation impacts endothelial cell function.Crossref | GoogleScholarGoogle Scholar | 24357059PubMed | 24357059PubMed |

Wen, X, Kuang, Y, Zhou, L, Yu, B, Chen, Q, Fu, Y, Yan, Z, Guo, H, Lyu, Q, Xie, J, and Chai, W (2018). Lipidomic components alterations of human follicular fluid reveal the relevance of improving clinical outcomes in women using progestin-primed ovarian stimulation compared to short-term protocol. Medical Science Monitor 24, 3357–3365.
Lipidomic components alterations of human follicular fluid reveal the relevance of improving clinical outcomes in women using progestin-primed ovarian stimulation compared to short-term protocol.Crossref | GoogleScholarGoogle Scholar | 29783268PubMed | 29783268PubMed |

Yáñez-Mó, M, Siljander, PRM, Andreu, Z, Zavec, AB, Borràs, FE, Buzas, EI, Buzas, K, Casal, E, Cappello, F, Carvalho, J, Colás, E, Cordeiro-da Silva, A, Fais, S, Falcon-Perez, JM, Ghobrial, IM, Giebel, B, Gimona, M, Graner, M, Gursel, I, Gursel, M, Heegaard, NHH, Hendrix, A, Kierulf, P, Kokubun, K, Kosanovic, M, Kralj-Iglic, V, Krämer-Albers, EM, Laitinen, S, Lässer, C, Lener, T, Ligeti, E, Linē, A, Lipps, G, Llorente, A, Lötvall, J, Manček-Keber, M, Marcilla, A, Mittelbrunn, M, Nazarenko, I, Nolte-’t Hoen, ENM, Nyman, TA, O’Driscoll, L, Olivan, M, Oliveira, C, Pállinger, É, Del Portillo, HA, Reventós, J, Rigau, M, Rohde, E, Sammar, M, Sánchez-Madrid, F, Santarém, N, Schallmoser, K, Ostenfeld, MS, Stoorvogel, W, Stukelj, R, Van der Grein, SG, Vasconcelos, MH, Wauben, MHM, and De Wever, O (2015). Biological properties of extracellular vesicles and their physiological functions. Journal of Extracellular Vesicles 4, 27066.
Biological properties of extracellular vesicles and their physiological functions.Crossref | GoogleScholarGoogle Scholar | 25979354PubMed | 25979354PubMed |

Yang, Y, Lee, M, and Fairn, GD (2018). Phospholipid subcellular localization and dynamics. Journal of Biological Chemistry 293, 6230–6240.
Phospholipid subcellular localization and dynamics.Crossref | GoogleScholarGoogle Scholar |

Yenuganti, VR, Viergutz, T, and Vanselow, J (2016). Oleic acid induces specific alterations in the morphology, gene expression and steroid hormone production of cultured bovine granulosa cells. General and Comparative Endocrinology 232, 134–144.
Oleic acid induces specific alterations in the morphology, gene expression and steroid hormone production of cultured bovine granulosa cells.Crossref | GoogleScholarGoogle Scholar | 27118706PubMed | 27118706PubMed |

Yonezawa, T, Yonekura, S, Kobayashi, Y, Hagino, A, Katoh, K, and Obara, Y (2004). Effects of long-chain fatty acids on cytosolic triacylglycerol accumulation and lipid droplet formation in primary cultured bovine mammary epithelial cells. Journal of Dairy Science 87, 2527–2534.
Effects of long-chain fatty acids on cytosolic triacylglycerol accumulation and lipid droplet formation in primary cultured bovine mammary epithelial cells.Crossref | GoogleScholarGoogle Scholar | 15328276PubMed | 15328276PubMed |

Young, JM, and McNeilly, AS (2010). Theca: the forgotten cell of the ovarian follicle. Reproduction 140, 489–504.
Theca: the forgotten cell of the ovarian follicle.Crossref | GoogleScholarGoogle Scholar | 20628033PubMed | 20628033PubMed |

Zhang, JY, Jiang, Y, Lin, T, Kang, JW, Lee, JE, and Jin, DI (2015). Lysophosphatidic acid improves porcine oocyte maturation and embryo development in vitro. Molecular Reproduction and Development 82, 66–77.
Lysophosphatidic acid improves porcine oocyte maturation and embryo development in vitro.Crossref | GoogleScholarGoogle Scholar | 25564987PubMed | 25564987PubMed |

Zhang, X, Wang, T, Song, J, Deng, J, and Sun, Z (2020). Study on follicular fluid metabolomics components at different ages based on lipid metabolism. Reproductive Biology and Endocrinology 18, 42.
Study on follicular fluid metabolomics components at different ages based on lipid metabolism.Crossref | GoogleScholarGoogle Scholar | 32398082PubMed | 32398082PubMed |

Zheng, W, Kollmeyer, J, Symolon, H, Momin, A, Munter, E, Wang, E, Kelly, S, Allegood, JC, Liu, Y, Peng, Q, Ramaraju, H, Sullards, MC, Cabot, M, and Merrill, AH (2006). Ceramides and other bioactive sphingolipid backbones in health and disease: lipidomic analysis, metabolism and roles in membrane structure, dynamics, signaling and autophagy. Biochimica et Biophysica Acta (BBA) – Biomembranes 1758, 1864–1884.

Zheng, W, Nagaraju, G, Liu, Z, and Liu, K (2012). Functional roles of the phosphatidylinositol 3-kinases (PI3Ks) signaling in the mammalian ovary. Molecular and Cellular Endocrinology 356, 24–30.
Functional roles of the phosphatidylinositol 3-kinases (PI3Ks) signaling in the mammalian ovary.Crossref | GoogleScholarGoogle Scholar | 21684319PubMed | 21684319PubMed |