Register      Login
Reproduction, Fertility and Development Reproduction, Fertility and Development Society
Vertebrate reproductive science and technology
REVIEW

Nutrition and maternal metabolic health in relation to oocyte and embryo quality: critical views on what we learned from the dairy cow model

Jo L. M. R. Leroy A D , Sara D. M. Valckx A , Lies Jordaens A , Jessie De Bie A , Karolien L. J. Desmet A , Veerle Van Hoeck B , Jack H. Britt C , Waleed F. Marei A and Peter E. J. Bols A
+ Author Affiliations
- Author Affiliations

A Gamete Research Centre, Veterinary Physiology and Biochemistry, Departement of Veterinary Sciences, University of Antwerp, Universiteitsplein 1, B2610 Wilrijk, Belgium.

B Laboratório de Fisiologia e Endocrinologia Molecular, University SaoPaulo Pirassununga, CEP 13-3565-4220 Pirassununga, Sao Paulo, Brasil.

C Department of Animal Science, North Carolina State University, Raleigh, NC 27695-7621, USA.

D Corresponding author. Email: jo.leroy@uantwerpen.be

Reproduction, Fertility and Development 27(4) 693-703 https://doi.org/10.1071/RD14363
Submitted: 26 September 2014  Accepted: 8 January 2015   Published: 18 February 2015

Abstract

Although fragmented and sometimes inconsistent, the proof of a vital link between the importance of the physiological status of the mother and her subsequent reproductive success is building up. High-yielding dairy cows are suffering from a substantial decline in fertility outcome over past decades. For many years, this decrease in reproductive output has correctly been considered multifactorial, with factors including farm management, feed ratios, breed and genetics and, last, but not least, ever-rising milk production. Because the problem is complex and requires a multidisciplinary approach, it is hard to formulate straightforward conclusions leading to improvements on the ‘work floor’. However, based on remarkable similarities on the preimplantation reproductive side between cattle and humans, there is a growing tendency to consider the dairy cow’s negative energy balance and accompanying fat mobilisation as an interesting model to study the impact of maternal metabolic disorders on human fertility and, more specifically, on oocyte and preimplantation embryo quality. Considering the mutual interest of human and animal scientists studying common reproductive problems, this review has several aims. First, we briefly introduce the ‘dairy cow case’ by describing the state of the art of research into metabolic imbalances and their possible effects on dairy cow reproduction. Second, we try to define relevant in vitro models that can clarify certain mechanisms by which aberrant metabolite levels may influence embryonic health. We report on recent advances in the assessment of embryo metabolism and meantime critically elaborate on advantages and major limitations of in vitro models used so far. Finally, we discuss hurdles to be overcome to successfully translate the scientific data to the field.

Additional keyword: research models.


References

Aardema, H., Vos, P. L., Lolicato, F., Roelen, B. A., Knijn, H. M., Vaandrager, A. B., Helms, J. B., and Gadella, B. M. (2011). Oleic acid prevents detrimental effects of saturated fatty acids on bovine oocyte developmental competence. Biol. Reprod. 85, 62–69.
Oleic acid prevents detrimental effects of saturated fatty acids on bovine oocyte developmental competence.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DC%2BC3MXotFOls74%3D&md5=91c35c1502e6480acc58b76f6a2faff4CAS | 21311036PubMed |

Abe, H., Yamashita, S., Satoh, T., and Hoshi, H. (2002). Accumulation of cytoplasmic lipid droplets in bovine embryos and cryotolerance of embryos developed in different culture systems using serum-free or serum-containing media. Mol. Reprod. Dev. 61, 57–66.
Accumulation of cytoplasmic lipid droplets in bovine embryos and cryotolerance of embryos developed in different culture systems using serum-free or serum-containing media.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DC%2BD3MXptVejur4%3D&md5=9f309a13a31a61fae851812e002e6e0eCAS | 11774376PubMed |

Adamiak, S. J., Powell, K., Rooke, J. A., Webb, R., and Sinclair, K. D. (2006). Body composition, dietary carbohydrates and fatty acids determine post-fertilisation development of bovine oocytes in vitro. Reproduction 131, 247–258.
Body composition, dietary carbohydrates and fatty acids determine post-fertilisation development of bovine oocytes in vitro.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DC%2BD28XisFalsL8%3D&md5=4387bb5a6237925289f117ee4a43d24fCAS | 16452718PubMed |

Akar, Y., and Gazioglu, A. (2006). Relationship between vitamin A and β-carotene levels during the postpartum period and fertility parameters in cows with and without retained placenta. Bull. Vet. Inst. Pulawy 50, 93–96.

Aréchiga, C. F., Vázquez-Flores, S., Ortiz, O., Hernández-Ceron, J., Porras, A., McDowell, L. R., and Hansen, P. J. (1998). Effect of injection of beta-carotene or vitamin E and selenium on fertility of lactating dairy cows. Theriogenology 50, 65–76.
Effect of injection of beta-carotene or vitamin E and selenium on fertility of lactating dairy cows.Crossref | GoogleScholarGoogle Scholar | 10734475PubMed |

Barker, D. J., Osmond, C., and Law, C. M. (1989). The intrauterine and early postnatal origins of cardiovascular disease and chronic bronchitis. J. Epidemiol. Community Health 43, 237–240.
The intrauterine and early postnatal origins of cardiovascular disease and chronic bronchitis.Crossref | GoogleScholarGoogle Scholar | 1:STN:280:DyaK3c7htFGqsQ%3D%3D&md5=86216f941f7846468312440d5facade6CAS | 2607302PubMed |

Beam, S. W., and Butler, W. R. (1997). Energy balance and ovarian follicle development prior to the first ovulation postpartum in dairy cows receiving three levels of dietary fat. Biol. Reprod. 56, 133–142.
Energy balance and ovarian follicle development prior to the first ovulation postpartum in dairy cows receiving three levels of dietary fat.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DyaK2sXis1Kguw%3D%3D&md5=b54e7f0c9e835cac9ec3bb9e6d701aa3CAS | 9002642PubMed |

Beaujean, N., Hartshorne, G., Cavilla, J., Taylor, J., Gardner, J., Wilmut, I., Meehan, R., and Young, L. (2004). Non-conservation of mammalian pre-implantation methylation dynamics. Curr. Biol. 14, R266–R267.
Non-conservation of mammalian pre-implantation methylation dynamics.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DC%2BD2cXivVKltb4%3D&md5=b980d430d29fdab5cdf1ab7ecabf14b5CAS | 15062117PubMed |

Bender, R. W., Hackbart, K. S., Dresch, A. R., Carvalho, P. D., Vieira, L. M., Crump, P. M., Guenther, J. N., Fricke, P. M., Shaver, R. D., Combs, D. K., and Wiltbank, M. C. (2014). Effects of acute feed restriction combined with targeted use of increasing luteinizing hormone content of follicle-stimulating hormone preparations on ovarian superstimulation, fertilization, and embryo quality in lactating dairy cows. J. Dairy Sci. 97, 764–778.
Effects of acute feed restriction combined with targeted use of increasing luteinizing hormone content of follicle-stimulating hormone preparations on ovarian superstimulation, fertilization, and embryo quality in lactating dairy cows.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DC%2BC3sXhvFOhur3P&md5=f323c9af10df3e5c7dde25d1dab6c7feCAS | 24359829PubMed |

Bilodeau-Goeseels, S. (2011). Cows are not mice: the role of cyclic AMP, phosphodiesterases, and adenosine monophosphate-activated protein kinase in the maintenance of meiotic arrest in bovine oocytes. Mol. Reprod. Dev. 78, 734–743.
Cows are not mice: the role of cyclic AMP, phosphodiesterases, and adenosine monophosphate-activated protein kinase in the maintenance of meiotic arrest in bovine oocytes.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DC%2BC3MXhtlagu7vM&md5=e3dc6197eaffeb7e820d14422579d66bCAS | 21688336PubMed |

Bilodeau-Goeseels, S., and Kastelic, J. P. (2003). Factors affecting embryo survival and strategies to reduce embryonic mortality in cattle. Can. J. Anim. Sci. 83, 659–671.
Factors affecting embryo survival and strategies to reduce embryonic mortality in cattle.Crossref | GoogleScholarGoogle Scholar |

Bossaert, P., Fransen, E., Langbeen, A., Stalpaert, M., Vandenbroeck, I., Bols, P. E., and Leroy, J. L. (2014). Effects of parity and periconceptional metabolic state of Holstein-Friesian dams on the glucose metabolism and conformation in their newborn calves. Animal 8, 975–981.
Effects of parity and periconceptional metabolic state of Holstein-Friesian dams on the glucose metabolism and conformation in their newborn calves.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DC%2BC2cXotFCrsrg%3D&md5=2956ab7af3e201a37a96af283423e3a9CAS | 24690495PubMed |

Bousquet, D., Bouchard, E., and DuTremblay, D. (2004). Decrease in fertility in dairy cows: myth or reality? Le Médecin Vétérinaire de Quebec 34, 59–61.

Britt, J. H. (1992). Impacts of early postpartum metabolism on follicular development and fertility. Bovine Practitioner 24, 39–43.

Bunel, A., Jorssen, E. P. A., Merckx, E., Leroy, J. L. M. R., Bols, P. E. J., and Sirard, M. A. (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 | 1:CAS:528:DC%2BC2cXhs1ymu7bE&md5=0bae41b0d758f727dfe41aa95a6d30e4CAS | 25442391PubMed |

Carvalho, P. D., Souza, A. H., Amundson, M. C., Hackbart, K. S., Fuenzalida, M. J., Herlihy, M. M., Ayres, H., Dresch, A. R., Vieira, L. M., Guenther, J. N., Grummer, R. R., Fricke, P. M., Shaver, R. D., and Wiltbank, M. C. (2014). Relationships between fertility and postpartum changes in body condition and body weight in lactating dairy cows. J. Dairy Sci. 97, 3666–3683.
Relationships between fertility and postpartum changes in body condition and body weight in lactating dairy cows.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DC%2BC2cXmtFahsbo%3D&md5=5c9d686b2c36f2d1e6e52b64c1a31a04CAS | 24731646PubMed |

Chauveau-Duriot, B., Thomas, D., Portelli, J., and Doreau, M. (2005). Effet du mode de conservation sur la teneur en caroténoïdes des fourrages. Rencontres autour des Recherches sur les ruminants 12, 117.

Chavatte-Palmer, P., Al Gubory, K., Picone, O., and Heyman, Y. (2008). Maternal nutrition: effects on offspring fertility and importance of the periconceptional period on long-term development. Gynecol. Obstet. Fertil. 36, 920–929.
Maternal nutrition: effects on offspring fertility and importance of the periconceptional period on long-term development.Crossref | GoogleScholarGoogle Scholar | 1:STN:280:DC%2BD1cnjsVyjtg%3D%3D&md5=11f77e6dc64300cea1f8afa97cbd1f7cCAS | 18693060PubMed |

Chavatte-Palmer, P., Dupont, C., Debus, N., and Camous, S. (2014). Nutritional programming and the reproductive function of the offspring. Anim. Prod. Sci. 54, 1166–1176.
Nutritional programming and the reproductive function of the offspring.Crossref | GoogleScholarGoogle Scholar |

Cnop, M., Hannaert, J. C., Hoorens, A., Eizirik, D. L., and Pipeleers, D. G. (2001). Inverse relationship between cytotoxicity of free fatty acids in pancreatic islet cells and cellular triglyceride accumulation. Diabetes 50, 1771–1777.
Inverse relationship between cytotoxicity of free fatty acids in pancreatic islet cells and cellular triglyceride accumulation.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DC%2BD3MXlslKnu74%3D&md5=0bbc3cc7af778fb97fa9e7dd19a750cbCAS | 11473037PubMed |

Cortvrindt, R., and Smitz, J. (1998). Early preantral mouse follicle in vitro maturation: oocyte growth, meiotic maturation and granulosa-cell proliferation. Theriogenology 49, 845–859.
Early preantral mouse follicle in vitro maturation: oocyte growth, meiotic maturation and granulosa-cell proliferation.Crossref | GoogleScholarGoogle Scholar | 1:STN:280:DC%2BD3c7ps1KnsA%3D%3D&md5=9d0df684a9f9e9a010d8e9f0012b195eCAS | 10732093PubMed |

Cortvrindt, R., and Smitz, J. (2001). In vitro follicle growth: achievements in mammalian species. Reprod. Domest. Anim. 36, 3–9.
In vitro follicle growth: achievements in mammalian species.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DC%2BD3MXitlSju70%3D&md5=84c37ea58d918f8cf6ee27713555a08dCAS | 11305484PubMed |

Cortvrindt, R. G., and Smitz, J. E. J. (2002). Follicle culture in reproductive toxicology: a tool for in vitro testing of ovarian function? Hum. Reprod. Update 8, 243–254.
Follicle culture in reproductive toxicology: a tool for in vitro testing of ovarian function?Crossref | GoogleScholarGoogle Scholar | 1:STN:280:DC%2BD38zjs1ygsA%3D%3D&md5=b894f3d1b28fc192264c6fcb4d3b49c4CAS | 12078835PubMed |

De Bie, J., Langbeen, A., Verlaet, A., Florizoone, F., Immig, I., Hermans, N., Bols, P. E. J., and Leroy, J. L. M. R. (2014). β-Carotene supplementation to non-lactating dairy cows can restore b-carotene availability in the follicular environment under negative energy balance conditions. Adv. Anim. Biosci. 5, 247–255.
β-Carotene supplementation to non-lactating dairy cows can restore b-carotene availability in the follicular environment under negative energy balance conditions.Crossref | GoogleScholarGoogle Scholar |

de Ondarza, M. B., and Engstrom, M. (2009). Can beta-carotene help dairy reproduction? Feedstuffs 81, 16–22.

De Wit, A. A. C., Cesar, M. L. F., and Kruip, T. A. M. (2001). Effect of urea during in vitro maturation on nuclear maturation and embryo development of bovine cumulus–oocyte-complexes. J. Dairy Sci. 84, 1800–1804.
Effect of urea during in vitro maturation on nuclear maturation and embryo development of bovine cumulus–oocyte-complexes.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DC%2BD3MXmtFaqurg%3D&md5=e3246809ec4096cce0acac50e73e40b2CAS |

Desmet, K., Van Hoeck, V., Merckx, E., Sirard, M. A., Bols, P. E. J., and Leroy, J. L. M. R. (2014a). Elevated non-esterified fatty acid concentrations during bovine oocyte maturation influences DNA methylation in blastocysts. Adv. Anim. Biosci. 5, 262.

Desmet, K., Van Hoeck, V., Merckx, E., Sirard, M. A., Bols, P. E. J., and Leroy, J. L. M. R. (2014b). Elevated non-esterified fatty acid concentrations during bovine embryo culture influences DNA methylation in blastocysts. In ‘Proceedings of the EPICONCEPT Workshop 2014 Epigenomic Toolbox: From Methods to Models, Las Palmas’. 7–9 May 2014. (Eds M. Izquierdo, F. Otero-Ferrer, T. Fair, A. Gutiérrez-Adán, A. Fazeli and A. Van Soom.) p. 28 (Abstract). (EPICONCEPT: Las Palmas.)

Dobrinski, I., Smith, T. T., Suarez, S. S., and Ball, B. A. (1997). Membrane contact with oviductal epithelium modulates the intracellular calcium concentration of equine spermatozoa in vitro. Biol. Reprod. 56, 861–869.
Membrane contact with oviductal epithelium modulates the intracellular calcium concentration of equine spermatozoa in vitro.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DyaK2sXitFGrtrg%3D&md5=fdc6c16072e0fafdd097ea44a9dad6e1CAS | 9096866PubMed |

Dunning, K. R., Cashman, K., Russell, D. L., Thompson, J. G., Norman, R. J., and Robker, R. L. (2010). Beta-oxidation is essential for mouse oocyte developmental competence and early embryo development. Biol. Reprod. 83, 909–918.
Beta-oxidation is essential for mouse oocyte developmental competence and early embryo development.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DC%2BC3cXhsFahurfN&md5=d2b87b938fd1b9207ccd0d890a41f1beCAS | 20686180PubMed |

Dupont, J., Scaramuzzi, R. J., and Reverchon, M. (2014). The effect of nutrition and metabolic status on the development of follicles, oocytes and embryos in ruminants. animal 8, 1031–1044.
The effect of nutrition and metabolic status on the development of follicles, oocytes and embryos in ruminants.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DC%2BC2cXpvVWktrc%3D&md5=e1a3acadbf2aef918cfc8beb8d10ea1bCAS |

Edwards, L. J., Batt, P. A., Gandolfi, F., and Gardner, D. K. (1997). Modifications made to culture medium by bovine oviduct epithelial cells: changes to carbohydrates stimulate bovine embryo development. Mol. Reprod. Dev. 46, 146–154.
Modifications made to culture medium by bovine oviduct epithelial cells: changes to carbohydrates stimulate bovine embryo development.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DyaK2sXoslWhug%3D%3D&md5=de02dc4d27d05727afe512f8e56f7a32CAS | 9021746PubMed |

Ellington, J. E. (1991). The bovine oviduct and its role in reproduction: a review of the literature. Cornell Vet. 81, 313–328.
| 1:STN:280:DyaK3MzkslKlsg%3D%3D&md5=f6594597897944a7b311b3eaea574e24CAS | 1879144PubMed |

Fazeli, A., Elliott, R. M. A., Duncan, A. E., Moore, A., Watson, P. F., and Holt, W. V. (2003). In vitro maintenance of boar sperm viability by a soluble fraction obtained from oviductal apical plasma membrane preparations. Reproduction 125, 509–517.
In vitro maintenance of boar sperm viability by a soluble fraction obtained from oviductal apical plasma membrane preparations.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DC%2BD3sXjvFOqt7c%3D&md5=7a0e9b7a584d37a9bafb42e0d6f1fb78CAS | 12683921PubMed |

Ferguson, E. M., and Leese, H. J. (2006). A potential role for triglyceride as an energy source during bovine oocyte maturation and early embryo development. Mol. Reprod. Dev. 73, 1195–1201.
A potential role for triglyceride as an energy source during bovine oocyte maturation and early embryo development.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DC%2BD28XnvFahtr0%3D&md5=f2578182c56ab88a367fbfe61db95607CAS | 16804881PubMed |

Forde, N., Carter, F., Fair, T., Crowe, M. A., Evans, A. C., Spencer, T. E., Bazer, F. W., McBride, R., Boland, M. P., O’Gaora, P., Lonergan, P., and Roche, J. F. (2009). Progesterone-regulated changes in endometrial gene expression contribute to advanced conceptus development in cattle. Biol. Reprod. 81, 784–794.
Progesterone-regulated changes in endometrial gene expression contribute to advanced conceptus development in cattle.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DC%2BD1MXhtFyhsLbM&md5=fdff0efe89c1877c45b80279b217564cCAS | 19553605PubMed |

Fulka, H., Mrazek, M., Tepla, O., and Fulka, J. (2004). DNA methylation pattern in human zygotes and developing embryos. Reproduction 128, 703–708.
DNA methylation pattern in human zygotes and developing embryos.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DC%2BD2MXhtFSlug%3D%3D&md5=e282e5cf0dcade60d3332df6f81dc066CAS | 15579587PubMed |

Gardner, D. K., and Lane, M. (1996). Alleviation of the ‘2-cell block’ and development to the blastocyst of CF1 mouse embryos: role of amino acids, EDTA and physical parameters. Hum. Reprod. 11, 2703–2712.
Alleviation of the ‘2-cell block’ and development to the blastocyst of CF1 mouse embryos: role of amino acids, EDTA and physical parameters.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DyaK2sXhs1OjtL8%3D&md5=edbdd173b3a62065b45be32db1af9d70CAS | 9021376PubMed |

Genicot, G., Leroy, J. L. M. R., Van Soom, A., and Donnay, I. (2005). The use of a fluorescent dye, Nile red, to evaluate the lipid content of single mammalian oocytes. Theriogenology 63, 1181–1194.
The use of a fluorescent dye, Nile red, to evaluate the lipid content of single mammalian oocytes.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DC%2BD2MXhtlSltb8%3D&md5=8fd6bfd07bb205746f3eca368c7564adCAS | 15710202PubMed |

Gossen, N., and Hoedemaker, M. (2005). [Effect of beta-carotin serum concentration on the reproductive performance in dairy cows.] Berl. Munch. Tierarztl. Wochenschr. 118, 326–333.
| 1:CAS:528:DC%2BD2MXhtVGit73P&md5=0cad24d21068c7e97b8af3ec35f8ac49CAS | 16048045PubMed |

Graf, A., Krebs, S., Heininen-Brown, M., Zakhartchenco, V., Blum, H., and Wolf, E. (2014). Genome activation in bovine embryos: review of the literature and new insights from RNA sequencing experiments. Anim. Reprod. Sci. 149, 46–58.
Genome activation in bovine embryos: review of the literature and new insights from RNA sequencing experiments.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DC%2BC2cXhtVymu7bF&md5=15ee4fa5e714aef756805eaea687603aCAS | 24975847PubMed |

Heijmans, B. T., Tobi, E. W., Stein, A. D., Putter, H., Blauw, G. J., Susser, E. S., Slagboom, P. E., and Lumey, L. H. (2008). Persistent epigenetic differences associated with prenatal exposure to famine in humans. Proc. Natl Acad. Sci. USA 105, 17 046–17 049.
Persistent epigenetic differences associated with prenatal exposure to famine in humans.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DC%2BD1cXhtlKqt7%2FP&md5=8421cd8de311a098eb9ce0ee365340d6CAS |

Houghton, F. D., Hawkhead, J. A., Humpherson, P. G., Hogg, J. E., Balen, A. H., Rutherford, A. J., and Leese, H. J. (2002). Non-invasive amino acid turnover predicts human embryo developmental capacity. Hum. Reprod. 17, 999–1005.
Non-invasive amino acid turnover predicts human embryo developmental capacity.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DC%2BD38Xjs12isLg%3D&md5=adfd3bb337f670f6d799e1d8fe73da21CAS | 11925397PubMed |

Hughes, J., Kwong, W. Y., Li, D., Salter, A. M., Lea, R. G., and Sinclair, K. D. (2011). Effects of omega-3 and -6 polyunsaturated fatty acids on ovine follicular cell steroidogenesis, embryo development and molecular markers of fatty acid metabolism. Reproduction 141, 105–118.
Effects of omega-3 and -6 polyunsaturated fatty acids on ovine follicular cell steroidogenesis, embryo development and molecular markers of fatty acid metabolism.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DC%2BC3MXisVehs7k%3D&md5=4d83bc08eae4b06abd4123a646860befCAS | 21045166PubMed |

Hunter, R. H. (2003). Reflections upon sperm-endosalpingeal and sperm-zona pellucida interactions in vivo and in vitro. Reprod. Domest. Anim. 38, 147–154.
Reflections upon sperm-endosalpingeal and sperm-zona pellucida interactions in vivo and in vitro.Crossref | GoogleScholarGoogle Scholar | 1:STN:280:DC%2BD3s7nvFyhsg%3D%3D&md5=bd45e5a964e3aa44bb88ea45cac17d46CAS | 12654026PubMed |

Iwańska, S., and Strusińska, D. (1997). The effect of beta-carotene and vitamins A, D3 and E on some reproductive parameters in cows. Acta Vet. Hung. 45, 95–107.
| 9270133PubMed |

Jungheim, E. S., Macones, G. A., Odem, R. R., Patterson, B. W., Lanzendorf, S. E., Ratts, V. S., and Moley, K. H. (2011). Associations between free fatty acids, cumulus oocyte complex morphology and ovarian function during in vitro fertilization. Fertil. Steril. 95, 1970–1974.
Associations between free fatty acids, cumulus oocyte complex morphology and ovarian function during in vitro fertilization.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DC%2BC3MXltFymt7s%3D&md5=76d9b420ab9daf852b82db55d5d51d58CAS | 21353671PubMed |

Kidson, A., Schoevers, E., Langendijk, P., Verheijden, J., Colenbrander, B., and Bevres, M. (2003). The effect of oviductal epithelial cell co-culture during in vitro maturation on sow oocyte morphology, fertilization and embryo development. Theriogenology 59, 1889–1903.
The effect of oviductal epithelial cell co-culture during in vitro maturation on sow oocyte morphology, fertilization and embryo development.Crossref | GoogleScholarGoogle Scholar | 12600727PubMed |

Krisher, R. L., and Bavister, B. D. (1998). Responses of oocytes and embryos to the culture environment. Theriogenology 49, 103–114.
Responses of oocytes and embryos to the culture environment.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DyaK1cXnsFCrtQ%3D%3D&md5=7313a95018e89eac1d456d7c670dd2a5CAS | 10732124PubMed |

Lane, M., and Gardner, D. K. (1998). Amino acids and vitamins prevent culture-induced metabolic perturbations and associated loss of viability of mouse blastocysts. Hum. Reprod. 13, 991–997.
Amino acids and vitamins prevent culture-induced metabolic perturbations and associated loss of viability of mouse blastocysts.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DyaK1cXjs12guro%3D&md5=0c4a71a225bc6ac5f0bb294af32ede91CAS | 9619560PubMed |

Latham, K. E. (1998). Epigenetic modification and imprinting of the mammalian genome during development. Curr. Top. Dev. Biol. 43, 1–49.
Epigenetic modification and imprinting of the mammalian genome during development.Crossref | GoogleScholarGoogle Scholar |

Leese, H. J. (1988). The formation and function of oviduct fluid. J. Reprod. Fertil. 82, 843–856.
The formation and function of oviduct fluid.Crossref | GoogleScholarGoogle Scholar | 1:STN:280:DyaL1c3gtVWitA%3D%3D&md5=3c0758c4e403edde24c76153b7b0d2aaCAS | 3283349PubMed |

Leese, H. J., Baumann, C. G., Brison, D. R., McEvoy, T. G., and Sturmey, R. G. (2008). Metabolism of the viable mammalian embryo: quietness revisited. Mol. Hum. Reprod. 14, 667–672.
Metabolism of the viable mammalian embryo: quietness revisited.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DC%2BD1MXltV2gsA%3D%3D&md5=9034204c84df6b71f9e2d5645a866619CAS | 19019836PubMed |

Leroy, J. L. M. R., Vanholder, T., Delanghe, J. R., Opsomer, G., Van Soom, A., Bols, P. E. J., De Wulf, J., and de Kruif, A. (2004). Metabolic changes in follicular fluid of the dominant follicle in high-yielding dairy cows early post partum. Theriogenology 62, 1131–1143.
Metabolic changes in follicular fluid of the dominant follicle in high-yielding dairy cows early post partum.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DC%2BD2cXmt1Kmsr8%3D&md5=d19cad5500d79274e7278a65dbb6687cCAS |

Leroy, J. L. M. R., Vanholder, T., Mateusen, B., Christophe, A., Opsomer, G., de Kruif, A., Genicot, G., and Van Soom, A. (2005a). 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 | 1:CAS:528:DC%2BD2MXhtFent7rJ&md5=16ddfb8af1de3a55f051d7e88b361557CAS |

Leroy, J. L. M. R., Genicot, G., Donnay, I., and Van Soom, A. (2005b). Evaluation of the lipid content in bovine oocytes and embryos with Nile red: a practical approach. Reprod. Domest. Anim. 40, 76–78.
Evaluation of the lipid content in bovine oocytes and embryos with Nile red: a practical approach.Crossref | GoogleScholarGoogle Scholar | 1:STN:280:DC%2BD2M%2FivVyjsg%3D%3D&md5=8ecae84743f1b5739477d4482d13d05fCAS |

Leroy, J. L., Opsomer, G., Van Soom, A., Goovaerts, I. G., and Bols, P. E. (2008a). Reduced fertility in high-yielding dairy cows: are the oocyte and embryo in danger? Part I. The importance of negative energy balance and altered corpus luteum function to the reduction of oocyte and embryo quality in high-yielding dairy cows. Reprod. Domest. Anim. 43, 612–622.
Reduced fertility in high-yielding dairy cows: are the oocyte and embryo in danger? Part I. The importance of negative energy balance and altered corpus luteum function to the reduction of oocyte and embryo quality in high-yielding dairy cows.Crossref | GoogleScholarGoogle Scholar | 1:STN:280:DC%2BD1cnksVeqsg%3D%3D&md5=ba3f190a9dddb3914fd8ad29487cf0feCAS | 18384499PubMed |

Leroy, J. L., Van Soom, A., Opsomer, G., Goovaerts, I. G., and Bols, P. E. (2008b). Reduced fertility in high-yielding dairy cows: are the oocyte and embryo in danger? Part II. Mechanisms linking nutrition and reduced oocyte and embryo quality in high-yielding dairy cows. Reprod. Domest. Anim. 43, 623–632.
Reduced fertility in high-yielding dairy cows: are the oocyte and embryo in danger? Part II. Mechanisms linking nutrition and reduced oocyte and embryo quality in high-yielding dairy cows.Crossref | GoogleScholarGoogle Scholar | 1:STN:280:DC%2BD1cnksVeruw%3D%3D&md5=b83ac25496933f646bb74ff82a4c6c54CAS | 18384498PubMed |

Leroy, J. L. M. R., Van Hoeck, V., Rizos, D., Guttierez-Adan, A., Clemente, M., Van Soom, A., Uyterhoeven, M., and Bols, P. E. J. (2010). The effect of nutritionally induced hyperlipidemia on in vitro embryo quality. Hum. Reprod. 25, 768–778.
The effect of nutritionally induced hyperlipidemia on in vitro embryo quality.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DC%2BC3cXitFeitr8%3D&md5=4dee32b7251ffc0edff01d2f80e5b2eaCAS |

Leroy, J. L. M. R., Rizos, D., Sturmey, R. G. S., Bossaert, P., Gutierrez-Adan, A., Van Hoeck, V., Valckx, S., and Bols, P. E. (2012). Intrafollicular conditions as a major link between maternal metabolic disorders and oocyte quality: a focus on dairy cow fertility. Reprod. Fertil. Dev. 24, 1–12.
Intrafollicular conditions as a major link between maternal metabolic disorders and oocyte quality: a focus on dairy cow fertility.Crossref | GoogleScholarGoogle Scholar |

Leroy, J. L., Sturmey, R. G., Van Hoeck, V., De Bie, J., McKeegan, P. J., and Bols, P. E. (2014). Dietary fat supplementation and the consequences for oocyte and embryo quality: hype or significant benefit for dairy cow reproduction? Reprod. Domest. Anim. 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 | 1:CAS:528:DC%2BC2cXns12murk%3D&md5=80a302876ebf685784cfb4e1186992b9CAS | 24697981PubMed |

Lillycrop, K. A., and Burdge, G. C. (2012). Epigenetic mechanisms linking early nutrition to long term health. Best Pract. Res. Clin. Endocrinol. Metab. 26, 667–676.
Epigenetic mechanisms linking early nutrition to long term health.Crossref | GoogleScholarGoogle Scholar | 22980048PubMed |

Lim, A. L., and Ferguson-Smith, A. C. (2010). Genomic imprinting effects in a compromised in utero environment: implications for a healthy pregnancy. Semin. Cell Dev. Biol. 21, 201–208.
Genomic imprinting effects in a compromised in utero environment: implications for a healthy pregnancy.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DC%2BC3cXisFGrt7s%3D&md5=9107a06109045138b991e6cc460343a5CAS | 19879952PubMed |

Lonergan, P. (2011). Influence of progesterone on oocyte quality and embryo development in cows. Theriogenology 76, 1594–1601.
Influence of progesterone on oocyte quality and embryo development in cows.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DC%2BC3MXhsVanurzI&md5=5172acb9ba1bba15fe4277d07b92efe2CAS | 21855985PubMed |

Lucy, M. C. (2003). Mechanisms linking nutrition and reproduction in postpartum cows. Reprod. Suppl. 61, 415–427.
| 1:CAS:528:DC%2BD3sXptFKhtrY%3D&md5=6d2baff7eac5e4259103bc7e319ca290CAS | 14635952PubMed |

Macaulay, A. D., Gilbert, I., Caballero, J., Barreto, R., Fournier, E., Tossou, P., Sirard, M. A., Clarke, H. J., Khandijan, E. W., Richard, F. J., Hyttel, P., and Robert, C. (2014). The gametic synapse; RNA transfer to the bovine oocyte. Biol. Reprod. 91, 90.
The gametic synapse; RNA transfer to the bovine oocyte.Crossref | GoogleScholarGoogle Scholar | 25143353PubMed |

Mahmood, T., and Arulkumaran, S. (Eds) (2012). ‘Obesity: A ticking time bomb for reproductive health’. (Elsevier: London.)

Maillo, V., Rizos, D., Besenfelder, U., Havlicek, V., Kelly, A. K., Garrett, M., and Lonergan, P. (2012). Influence of lactation on metabolic characteristics and embryo development in postpartum Holstein dairy cows. J. Dairy Sci. 95, 3865–3876.
Influence of lactation on metabolic characteristics and embryo development in postpartum Holstein dairy cows.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DC%2BC38XoslGlsb4%3D&md5=0b775da8a7862b04a0f2df4e6fe69683CAS | 22720941PubMed |

Mamo, S., Mehta, J. P., Forde, N., McGettigan, P., and Lonergan, P. (2012). Conceptus–endometrium crosstalk during maternal recognition of pregnancy in cattle. Biol. Reprod. 87, 6.
Conceptus–endometrium crosstalk during maternal recognition of pregnancy in cattle.Crossref | GoogleScholarGoogle Scholar | 22517619PubMed |

Mann, G. E., Fray, M. D., and Lamming, G. E. (2006). Effects of time of progesterone supplementation on embryo development and interferon-tau production in the cow. Vet. J. 171, 500–503.
Effects of time of progesterone supplementation on embryo development and interferon-tau production in the cow.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DC%2BD28Xjs1Kis70%3D&md5=0979dcd567b6a8eddb36aa3f252c2af7CAS | 16624716PubMed |

Matoba, S., O’Hara, L., Carter, F., Kelly, A. K., Fair, T., Rizos, D., and Lonergan, P. (2012). The association between metabolic parameters and oocyte quality early and late post-partum in Holstein dairy cows. J. Dairy Sci. 95, 1257–1266.
The association between metabolic parameters and oocyte quality early and late post-partum in Holstein dairy cows.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DC%2BC38XjtFWgsLY%3D&md5=75b858208e51abfee957dc025d3b28b0CAS | 22365209PubMed |

McArthur, M. J., Atshaves, B. P., Frolov, A., Foxworth, W. D., Kier, A. B., and Schroeder, F. (1999). Cellular uptake and intracellular trafficking of long chain fatty acids. J. Lipid Res. 40, 1371–1383.
| 1:CAS:528:DyaK1MXlt1Clsb4%3D&md5=82bebf56cefb9eb22e4401ee17629098CAS | 10428973PubMed |

Ménézo, Y. J., and Hérubel, F. (2002). Mouse and bovine models for human IVF. Reprod. Biomed. Online 4, 170–175.
Mouse and bovine models for human IVF.Crossref | GoogleScholarGoogle Scholar | 12470581PubMed |

Mesquita, F. S., Pugliesi, G., Scolari, S. C., França, M. R., Ramos, R. S., Oliveira, M., Papa, P. C., Bressan, F. F., Meirelles, F. V., Silva, L. A., Noquiera, G. P., Membrive, C. M., and Binelli, M. (2014). Manipulation of the periovulatory sex steroidal milieu affects endometrial but not luteal gene expression in early diestrus Nelore cows. Theriogenology 81, 861–869.
Manipulation of the periovulatory sex steroidal milieu affects endometrial but not luteal gene expression in early diestrus Nelore cows.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DC%2BC2cXitVyrtbw%3D&md5=de3e087618b896bb76ba85f0e5254ce6CAS | 24507960PubMed |

Niemann, H., Carnwath, J. W., and Kues, W. (2007). Application of DNA array technology to mammalian embryos. Theriogenology 68, S165–S177.
Application of DNA array technology to mammalian embryos.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DC%2BD2sXotlaitb8%3D&md5=4be0f99e54b8a7ae88e9b1e9758e3fe1CAS | 17582485PubMed |

O’Doherty, A. M., O’Gorman, A., al Naib, A., Brennan, L., Daly, E., Duffy, P., and Fair, T. (2014). Negative energy balance affects imprint stability in oocytes recovered from postpartum dairy cows. Genomics 104, 177–185.
Negative energy balance affects imprint stability in oocytes recovered from postpartum dairy cows.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DC%2BC2cXht1Kkur7L&md5=4aac0fb13eef087311719fde4fc4c76cCAS | 25084396PubMed |

Opsomer, G., Coryn, M., Deluyker, H., and de Kruif, A. (1998). An analysis of ovarian dysfunction in high yielding dairy cows after calving based on progesterone profiles. Reprod. Domest. Anim. 33, 193–204.
An analysis of ovarian dysfunction in high yielding dairy cows after calving based on progesterone profiles.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DyaK1cXksVaiurw%3D&md5=3e212a21f6ecb67972b480d68b716a84CAS |

Paczkowski, M., Silva, E., Schoolcraft, W. B., and Krisher, R. L. (2013). Comparative importance of fatty acid beta-oxidation to nuclear maturation, gene expression, and glucose metabolism in mouse, bovine, and porcine cumulus oocyte complexes. Biol. Reprod. 88, 111.
Comparative importance of fatty acid beta-oxidation to nuclear maturation, gene expression, and glucose metabolism in mouse, bovine, and porcine cumulus oocyte complexes.Crossref | GoogleScholarGoogle Scholar | 23536372PubMed |

Radford, E. J., Ito, M., Shi, H., Corish, J. A., Yamazawa, K., Isganaitis, E., Seisenberger, S., Hore, T. A., Reik, W., Erkek, S., Peters, A. H., Patti, M. E., and Ferguson-Smith, A. C. (2014). In utero undernourishment perturbs the adult sperm methylome and intergenerational metabolism. Science 345, 1255903.
In utero undernourishment perturbs the adult sperm methylome and intergenerational metabolism.Crossref | GoogleScholarGoogle Scholar | 25011554PubMed |

Rhodes, F. M., McDougall, S., Burke, C. R., Verkerk, G. A., and Macmillan, K. L. (2003). Treatment of cows with an extended postpartum anestrous interval. J. Dairy Sci. 86, 1876–1894.
Treatment of cows with an extended postpartum anestrous interval.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DC%2BD3sXks1Git74%3D&md5=1664e8489fd9ce6f9c3a0b0d65a54c84CAS | 12836922PubMed |

Richieri, G. V., and Kleinfeld, A. M. (1995). Unbound free fatty acid levels in human serum. J. Lipid Res. 36, 229–240.
| 1:CAS:528:DyaK2MXjvVWnsL4%3D&md5=726c45d197ae1676e161f3ff55a52637CAS | 7751810PubMed |

Rizos, D., Gutierrez-Adan, A., Perez-Garnelo, S., de la Fuente, J., Boland, M. P., and Lonergan, P. (2003). Bovine embryo culture in the presence or absence of serum: implications for blastocyst development, cryotolerance, and messenger RNA expression. Biol. Reprod. 68, 236–243.
Bovine embryo culture in the presence or absence of serum: implications for blastocyst development, cryotolerance, and messenger RNA expression.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DC%2BD3sXjtFWj&md5=7eac6d4191c91ddad2567e3bfaf367f2CAS | 12493719PubMed |

Rizos, D., Carter, F., Besenfelder, U., Havlicek, V., and Lonergan, P. (2010). Contribution of the female reproductive tract to low fertility in postpartum lactating dairy cows. J. Dairy Sci. 93, 1022–1029.
Contribution of the female reproductive tract to low fertility in postpartum lactating dairy cows.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DC%2BC3cXitlOjurc%3D&md5=b3637df12efaa2f7d45f6f0f537bd0b0CAS | 20172222PubMed |

Robker, R. L., Akison, L. K., Bennett, B. D., Thrupp, P. N., Chura, L. R., Russell, D. L., Lane, M., and Norman, R. J. (2009). Obese women exhibit differences in ovarian metabolites, hormones, and gene expression compared with moderate-weight women. J. Clin. Endocrinol. Metab. 94, 1533–1540.
Obese women exhibit differences in ovarian metabolites, hormones, and gene expression compared with moderate-weight women.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DC%2BD1MXlvVCqtrw%3D&md5=ad9fc9f21f34fed2964925dd65571412CAS | 19223519PubMed |

Rooke, J. A., Watt, R. G., Ashworth, C. J., and McEvoy, T. G. (2012). Inclusion of bovine lipoproteins and the vitamin E analogue, trolox, during in vitro culture of bovine embryos changes both embryo and fetal development. Reprod. Fertil. Dev. 24, 309–316.
Inclusion of bovine lipoproteins and the vitamin E analogue, trolox, during in vitro culture of bovine embryos changes both embryo and fetal development.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DC%2BC38Xht1Oitbw%3D&md5=ef96a8bcb447ecc89c9a0eb5b6782f62CAS | 22281076PubMed |

Roth, Z. (2008). Heat stress, the follicle, and its enclosed oocyte: mechanisms and potential strategies to improve fertility in dairy cows. Reprod. Domest. Anim. 43, 238–244.
Heat stress, the follicle, and its enclosed oocyte: mechanisms and potential strategies to improve fertility in dairy cows.Crossref | GoogleScholarGoogle Scholar | 18638130PubMed |

Santos, J. E. P., Rutigliano, H. M., and SaFilho, M. F. (2009). Risk factors for resumption of postpartum estrous cycles and embryonic survival in lactating dairy cows. Anim. Reprod. Sci. 110, 207–221.
Risk factors for resumption of postpartum estrous cycles and embryonic survival in lactating dairy cows.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DC%2BD1cXhsVeru73N&md5=56f5aa6cb3615865405ff21663aeef7bCAS |

Santos, J. E., Bisinotto, R. S., Ribeiro, E. S., Lima, F. S., Greco, L. F., Staples, C. R., and Thatcher, W. W. (2010). Applying nutrition and physiology to improve reproduction in dairy cattle. Soc. Reprod. Fertil. Suppl. 67, 387–403.
| 1:STN:280:DC%2BC3MnosFemtA%3D%3D&md5=7aa28d7284a38a5f5b0d45b01497f8eaCAS | 21755686PubMed |

Scaramuzzi, R. J., Baird, D. T., Campbell, B. K., Driancourt, M.-A., Dupont, J., Fortune, J. E., Gilchrist, R. B., Martin, G. B., McNatty, K. P., McNeilly, A. S., Monget, P., Monniaux, D., Vinõles, C., and Webb, R. (2011). Regulation of folliculogenesis and the determination of ovulation rate in ruminants. Reprod. Fertil. Dev. 23, 444–467.
Regulation of folliculogenesis and the determination of ovulation rate in ruminants.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DC%2BC3MXjt12ntLg%3D&md5=d250734c3e9c9baef1d84ebc61c9998eCAS | 21426863PubMed |

Schneider, J. E. (2004). Energy balance and reproduction. Physiol. Behav. 81, 289–317.
Energy balance and reproduction.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DC%2BD2cXkt1Srtrg%3D&md5=3c969c1a377224f7f9f17f82c3af5425CAS | 15159173PubMed |

Shehab-El-Deen, M. A. M. M., Leroy, J. L. M. R., Fadel, M. S., Saleh, S. Y. A., Maes, D., and Van Soom, A. (2010). Biochemical changes in the follicular fluid of the dominant follicle of high producing dairy cows exposed to heat stress early post-partum. Anim. Reprod. Sci. 117, 189–200.
Biochemical changes in the follicular fluid of the dominant follicle of high producing dairy cows exposed to heat stress early post-partum.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DC%2BD1MXhsVGltrnJ&md5=371b6c6b266d0975aeb543227361b280CAS |

Sinclair, K. D., and Singh, R. (2007). Modelling the developmental origins of health and disease in the early embryo. Theriogenology 67, 43–53.
Modelling the developmental origins of health and disease in the early embryo.Crossref | GoogleScholarGoogle Scholar | 17049592PubMed |

Smith, T. T., and Nothnick, W. B. (1997). Role of direct contact between spermatozoa and oviductal epithelial cells in maintaining rabbit sperm viability. Biol. Reprod. 56, 83–89.
Role of direct contact between spermatozoa and oviductal epithelial cells in maintaining rabbit sperm viability.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DyaK2sXis1Gqsg%3D%3D&md5=1abbb6e237c4ac228ac58a00986e8eceCAS | 9002636PubMed |

Sorrentino, D., Stump, D., Potter, B. J., Robinson, R. B., White, R., Kiang, C. L., and Berk, P. D. (1988). Oleate uptake by cardiac myocytes is carrier mediated and involves a 40-kD plasma membrane fatty acid binding protein similar to that in liver adipose tissue and gut. J. Clin. Invest. 82, 928–935.
Oleate uptake by cardiac myocytes is carrier mediated and involves a 40-kD plasma membrane fatty acid binding protein similar to that in liver adipose tissue and gut.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DyaL1cXls1OrtbY%3D&md5=b9773096c5db946f11335c5f843c05f7CAS | 3417874PubMed |

Spencer, T. E. (2014). Biological roles of uterine glands in pregnancy. Semin. Reprod. Med. 32, 346–357.
Biological roles of uterine glands in pregnancy.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DC%2BC2cXhvVeku7jP&md5=aac19ba06536815d2cffe012e6489766CAS | 24959816PubMed |

Sturmey, R. G., Brison, D. R., and Leese, H. J. (2008). Assessing embryo viability by measurement of amino acid turnover. Reprod. Biomed. Online 17, 486–496.
Assessing embryo viability by measurement of amino acid turnover.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DC%2BD1cXht1Kqtb7M&md5=949dc4b46a29ef66f6ef14e4d8deb53aCAS | 18854101PubMed |

Sturmey, R. G., Hawkhead, J. A., Barker, E. A., and Leese, H. J. (2009). DNA damage and metabolic activity in the preimplantation embryo. Hum. Reprod. 24, 81–91.
DNA damage and metabolic activity in the preimplantation embryo.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DC%2BD1cXhsFWjtbnF&md5=aac4d0f1e406b0c3f2e10ce7c5cae58cCAS | 18835872PubMed |

Sturmey, R. G., Bermejo-Alvarez, P., Gutierrez-Adan, A., Rizos, D., Leese, H. J., and Lonergan, P. (2010). Amino acid metabolism of bovine blastocysts: a biomarker of sex and viability. Mol. Reprod. Dev. 77, 285–296.
Amino acid metabolism of bovine blastocysts: a biomarker of sex and viability.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DC%2BC3cXotFSmuw%3D%3D&md5=e95dd21b2c1d7b3b02318a8323bd5bd9CAS | 20058302PubMed |

Suarez, S. S. (2002). Formation of a reservoir of sperm in the oviduct. Reprod. Domest. Anim. 37, 140–143.
Formation of a reservoir of sperm in the oviduct.Crossref | GoogleScholarGoogle Scholar | 12071887PubMed |

Sutton-McDowall, M. L., Gilchrist, R. B., and Thompson, J. G. (2004). Cumulus expansion and glucose utilisation by bovine cumulus–oocyte complexes during in vitro maturation: the influence of glucosamine and follicle-stimulating hormone. Reproduction 128, 313–319.
Cumulus expansion and glucose utilisation by bovine cumulus–oocyte complexes during in vitro maturation: the influence of glucosamine and follicle-stimulating hormone.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DC%2BD2cXot1KitLk%3D&md5=076f1fdaf03198182b986cc18f80b06bCAS | 15333782PubMed |

Synak, M., Gorecka, M., Langfort, J., Smol, E., and Zernicka, E. (2003). Palmitic acid incorporation into intramuscular acylglycerols depends on both total and unbound to albumin palmitic acid concentration. Biochem. Cell Biol. 81, 35–41.
Palmitic acid incorporation into intramuscular acylglycerols depends on both total and unbound to albumin palmitic acid concentration.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DC%2BD3sXitlOkurs%3D&md5=c156b68649d236badfa5a20f7b195da5CAS | 12683634PubMed |

Trigatti, B. L., Mangroo, D., and Gerber, G. E. (1995). A direct role for serum albumin in the cellular uptake of long-chain fatty acids. Biochem. J. 308, 155–159.
| 1:CAS:528:DyaK2MXlvFantrw%3D&md5=cf46e5857fe3c25f9c771be759e6072eCAS | 7755560PubMed |

Tse, P. K., Lee, Y. L., Chow, W. N., Luk, J. M., Lee, K. F., and Yeung, W. S. (2008). Preimplantation embryos cooperate with oviductal cells to produce embryothrophic inactivated complement-3b. Endocrinology 149, 1268–1276.
Preimplantation embryos cooperate with oviductal cells to produce embryothrophic inactivated complement-3b.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DC%2BD1cXisVaqtLs%3D&md5=e06967a804bfa752c9e3a1a88da976b0CAS | 18039777PubMed |

Ulloth, J. E., Casiano, C. A., and De Leon, M. (2003). Palmitic and stearic fatty acids induce caspase-dependent and -independent cell death in nerve growth factor differentiated PC12 cells. J. Neurochem. 84, 655–668.
Palmitic and stearic fatty acids induce caspase-dependent and -independent cell death in nerve growth factor differentiated PC12 cells.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DC%2BD3sXht1amsb4%3D&md5=cb0a3bb26cbeae3a2f7ea8bda999f6e3CAS | 12562510PubMed |

Valckx, S. D., De Pauw, I., De Neubourg, D., Inion, I., Berth, M., Fransen, E., Bols, P. E., and Leroy, J. L. (2012). BMI-related metabolic composition of the follicular fluid of women undergoing assisted reproductive treatment and the consequences for oocyte and embryo quality. Hum. Reprod. 27, 3531–3539.
BMI-related metabolic composition of the follicular fluid of women undergoing assisted reproductive treatment and the consequences for oocyte and embryo quality.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DC%2BC38XhslKrsLrL&md5=36e8e2c48e7445163a84e1ab94e8a470CAS | 23019302PubMed |

Valckx, S. D. M., Van Hoeck, V., Arias-Alvarez, M., Maillo, V., Lopez-Cardona, A. P., Gutierrez-Adan, A., Berth, M., Cortvrindt, R., Bols, P. E. J., and Leroy, J. L. M. R. (2014a). Elevated non-esterified fatty acid concentrations during in vitro murine follicle growth alter follicular physiology and reduce oocyte developmental competence. Fertil. Steril. 102, 1769–1776.e1.
Elevated non-esterified fatty acid concentrations during in vitro murine follicle growth alter follicular physiology and reduce oocyte developmental competence.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DC%2BC2cXhs1Wiu7bN&md5=5f545b256df9936d01e8f29e8753e5edCAS |

Valckx, S. D., Arias-Alvarez, M., De Pauw, I., Fievez, V., Vlaeminck, B., Fransen, E., Bols, P. E., and Leroy, J. L. (2014b). Fatty acid composition of the follicular fluid of normal weight, overweight and obese women undergoing assisted reproductive treatment: a descriptive cross-sectional study. Reprod. Biol. Endocrinol. 12, 13.
Fatty acid composition of the follicular fluid of normal weight, overweight and obese women undergoing assisted reproductive treatment: a descriptive cross-sectional study.Crossref | GoogleScholarGoogle Scholar | 24498875PubMed |

Valckx, S. D. M., De Bie, J., Michiels, E. D., Goovaerts, I. G., Punjabi, U., Ramos-Ibeas, P., Gutierrez-Adan, A., Bols, P. E., and Leroy, J. L. (2014c). The effect of human follicular fluid on bovine oocyte developmental competence and embryo quality. RBM Online 30, 203–207.
The effect of human follicular fluid on bovine oocyte developmental competence and embryo quality.Crossref | GoogleScholarGoogle Scholar |

Van Hoeck, V., Sturmey, R. G., Bermejo-Alvarez, P., Rizos, D., Gutierrez-Adan, A., Leese, H. J., Bols, P. E., and Leroy, J. L. (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 | 1:CAS:528:DC%2BC3MXhtFKmsLjM&md5=b6eb69e3072d782b967fdaef51eb2625CAS | 21858021PubMed |

Van Hoeck, V., Leroy, J. L., Arias-Alvarez, M., Rizos, D., Gutierrez-Adan, A., Schnorbusch, K., Bols, P. E., Leese, H. J., and Sturmey, R. G. (2013a). Oocyte developmental failure in response to elevated nonesterified fatty acid concentrations: mechanistic insights. Reproduction 145, 33–44.
Oocyte developmental failure in response to elevated nonesterified fatty acid concentrations: mechanistic insights.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DC%2BC3sXhslGisb8%3D&md5=2e22f5e936a1f5d16f80b6bbb94a824bCAS | 23108110PubMed |

Van Hoeck, V., Rizos, D., Gutierrez-Adan, A., Pintelon, I., Jorssen, E., Dufort, I., Sirard, M. A., Verlaet, A., Hermans, N., Bols, P. E., and Leroy, J. L. (2013b). Interaction between differential gene expression profile and phenotype in bovine blastocysts originating from oocytes exposed to elevated non-esterified fatty acid concentrations. Reprod. Fertil. Dev. , .
Interaction between differential gene expression profile and phenotype in bovine blastocysts originating from oocytes exposed to elevated non-esterified fatty acid concentrations.Crossref | GoogleScholarGoogle Scholar |

Van Hoeck, V., Bols, P. E., Binelli, M., and Leroy, J. L. (2014). Reduced oocyte and embryo quality in response to elevated non-esterified fatty acid concentrations: a possible pathway to subfertility? Anim. Reprod. Sci. 149, 19–29.
Reduced oocyte and embryo quality in response to elevated non-esterified fatty acid concentrations: a possible pathway to subfertility?Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DC%2BC2cXhtlGnu7jI&md5=c8590464e7bfea21b8ca3fc4b777bd2dCAS | 25129636PubMed |

Vanholder, T., Leroy, J. L., Soom, A. V., Opsomer, G., Maes, D., Coryn, M., and de Kruif, A. (2005). Effect of non-esterified fatty acids on bovine granulosa cell steroidogenesis and proliferation in vitro. Anim. Reprod. Sci. 87, 33–44.
Effect of non-esterified fatty acids on bovine granulosa cell steroidogenesis and proliferation in vitro.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DC%2BD2MXktVOju74%3D&md5=f58b57b6abf5aad4e2afae8498969245CAS | 15885439PubMed |

Velazquez, M. A., Parrilla, I., Van Soom, A., Verberckmoes, S., Kues, W., and Niemann, H. (2010). Sampling techniques for oviductal and uterine luminal fluid in cattle. Theriogenology 73, 758–767.
Sampling techniques for oviductal and uterine luminal fluid in cattle.Crossref | GoogleScholarGoogle Scholar | 1:STN:280:DC%2BC3c7ot1ahtw%3D%3D&md5=b1ffad7193ed341ffc2c8aac23592c0eCAS | 19682731PubMed |

Wathes, D. C., Fenwick, M., Cheng, Z., Bourne, N., Llewellyn, S., Morris, D. G., 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 | 1:CAS:528:DC%2BD2sXotlaitbg%3D&md5=f974c5b9e9ebbc6ba502f6cf9b4b75fcCAS | 17475319PubMed |

Watson, A. J., Westhusin, M. E., and Winger, Q. A. (1999). IGF paracrine and autocrine interactions between conceptus and oviduct. J. Reprod. Fertil. Suppl. 54, 303–315.
| 1:CAS:528:DyaK1MXnslSitrY%3D&md5=5219e0f1b0e836ef1826f47270667ec4CAS | 10692863PubMed |

Wiltbank, M. C., Garcia-Guerra, A., Carvalho, P. D., Hackbart, K. S., Bender, R. W., Souza, A. H., Toledo, M. Z., Baez, G. M., Surjus, R. S., and Sartori, R. (2014). Effects of energy and protein nutrition in the dam on embryonic development. Anim. Reprod. 11, 168–182.

Wonnacott, K. E., Kwong, W. Y., Hughes, J., Salter, A. M., Lea, R. G., Gamsworthy, P. C., and Sinclair, K. D. (2010). Dietary omega-3 and -6 polyunsaturated fatty acids affect the composition and development of sheep granulosa cells, oocytes and embryos. Reproduction 139, 57–69.
Dietary omega-3 and -6 polyunsaturated fatty acids affect the composition and development of sheep granulosa cells, oocytes and embryos.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DC%2BC3cXovVWnug%3D%3D&md5=52e2c73819d802a94d383c79e8e0bb57CAS | 19789173PubMed |

Wrenzycki, C., Herrmann, D., and Niemann, H. (2007). Messenger RNA in oocytes and embryos in relation to embryo viability. Theriogenology 68, S77–S83.
Messenger RNA in oocytes and embryos in relation to embryo viability.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DC%2BD2sXotlaiurk%3D&md5=135bf3c4556bed96c9419f79723d3ae0CAS | 17524469PubMed |

Yung, M. C., VandeHaar, M. J., Fogwell, R. L., and Sharma, B. K. (1996). Effect of energy balance and somatotropin on insulin-like growth factor I in serum and on weight and progesterone of corpus luteum in heifers. J. Anim. Sci. 74, 2239–2244.
| 1:CAS:528:DyaK28Xls1OmtLk%3D&md5=11289c6615aee321bbd9396c10c2ec7bCAS | 8880428PubMed |