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

Effect of nutritionally induced hyperlipidaemia on in vitro bovine embryo quality depends on the type of major fatty acid in the diet

Waleed F. A. Marei A B D , Maria Arias Alvarez A , Veerle Van Hoeck A , Alfonso Gutierrez-Adan C , Peter E. J. Bols A and Jo L. M. R. Leroy A
+ Author Affiliations
- Author Affiliations

A Gamete Research Centre, Laboratory for Veterinary Physiology and Biochemistry, Department of Veterinary Sciences, University of Antwerp, Universiteitsplein 1, B-2610 Wilrijk, Belgium.

B Department of Theriogenology, Faculty of Veterinary Medicine, Cairo University, Giza 12211, Egypt.

C Departamento de Reproduccion Animal, Instituto nacional de Investigacion y Tecnologia Agraria y Alimentaria (INIA), Ctra. De la Coruna Km 5.9, Madrid 28040, Spain.

D Corresponding author. Email: waleed.marei@uantwerpen.be

Reproduction, Fertility and Development 29(9) 1856-1867 https://doi.org/10.1071/RD16297
Submitted: 1 August 2016  Accepted: 10 October 2016   Published: 11 November 2016

Abstract

The present study examined whether the effects of dietary-induced hyperlipidaemia on preimplantation embryo development depend on the predominant fatty acid (FA) type in the diet. In a combined in vivoin vitro bovine model, two groups of cows (n = 3 in each group) were fed with three diets consecutively (4 weeks feeding for each): (1) a maintenance control diet (CONT); (2) a high-starch diet rich in saturated fat (SAT); and (3) a high-starch diet rich in omega-3 unsaturated fat (UNSAT). Two feeding sequences were used to test for carry-over effects: Group A was fed CONT, SAT1 and then UNSAT2, whereas Group B was fed CONT, UNSAT1 and then SAT2. Serum was collected after each dietary period, analysed and tested in bovine in vitro embryo culture. Introducing SAT and UNSAT diets induced hyperlipidaemia (specifically hypercholesterolaemia and elevated free FAs) and reduced insulin sensitivity. Carry-over effects in serum metabolites and FA profile were dependent on the diet and feeding sequence. SAT1 and SAT2 serum decreased blastocyst rates and altered blastocyst mRNA expression related to apoptosis and oxidative stress. UNSAT1 and UNSAT2 serum resulted in normal embryo development and quality. Other in vitro effects depended on the sequence of feeding. In conclusion, substitution of saturated fat with omega-3 fat in a high-caloric diet induced hyperlipidaemia with an FA profile yielding similar rates and quality of blastocysts compared with normolipidaemic controls.

Additional keywords: embryo development, linolenic acid, omega 3, palmitic acid, Western-type diet.


References

Abayasekara, D. R., and Wathes, D. C. (1999). Effects of altering dietary fatty acid composition on prostaglandin synthesis and fertility. Prostaglandins Leukot. Essent. Fatty Acids 61, 275–287.
Effects of altering dietary fatty acid composition on prostaglandin synthesis and fertility.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DC%2BD3cXltF2gtw%3D%3D&md5=f3de2e2ec271010ad480877a385aab17CAS |

Artwohl, M., Lindenmair, A., Roden, M., Waldhausl, W. K., Freudenthaler, A., Klosner, G., Ilhan, A., Luger, A., and Baumgartner-Parzer, S. M. (2009). Fatty acids induce apoptosis in human smooth muscle cells depending on chain length, saturation, and duration of exposure. Atherosclerosis 202, 351–362.
Fatty acids induce apoptosis in human smooth muscle cells depending on chain length, saturation, and duration of exposure.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DC%2BD1MXhtFehu7w%3D&md5=34e82e34358ebe105c55e1a1bb2d9453CAS |

Barnard, R. J., Roberts, C. K., Varon, S. M., and Berger, J. J. (1998). Diet-induced insulin resistance precedes other aspects of the metabolic syndrome. J. Appl. Physiol. (1985) 84, 1311–1315.
| 1:CAS:528:DyaK1cXisVKhurc%3D&md5=4501b87e984257275c232e14f65b57d4CAS |

Bermejo-Álvarez, P., Lonergan, P., Rath, D., Gutiérrez-Adan, A., and Rizos, D. (2010). Developmental kinetics and gene expression in male and female bovine embryos produced in vitro with sex-sorted spermatozoa. Reprod. Fertil. Dev. 22, 426–436.
Developmental kinetics and gene expression in male and female bovine embryos produced in vitro with sex-sorted spermatozoa.Crossref | GoogleScholarGoogle Scholar |

Boden, G. (2001). Free fatty acids – the link between obesity and insulin resistance. Endocr. Pract. 7, 44–51.
Free fatty acids – the link between obesity and insulin resistance.Crossref | GoogleScholarGoogle Scholar | 1:STN:280:DC%2BD38visV2msA%3D%3D&md5=333a450e6cb19c0e89be950d60743d7eCAS |

Boden, G. (2008). Obesity and free fatty acids. Endocrinol. Metab. Clin. North Am. 37, 635–646.
Obesity and free fatty acids.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DC%2BD1cXht1Kqs77J&md5=5557d9de5f39e8953a2846a37e64a7baCAS |

Borgstahl, G. E., Parge, H. E., Hickey, M. J., Beyer, W. F., Hallewell, R. A., and Tainer, J. A. (1992). The structure of human mitochondrial manganese superoxide dismutase reveals a novel tetrameric interface of two 4-helix bundles. Cell 71, 107–118.
The structure of human mitochondrial manganese superoxide dismutase reveals a novel tetrameric interface of two 4-helix bundles.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DyaK38XmsVOksro%3D&md5=2b652b12ebaafc051e5dc0b16c8c7cdcCAS |

Bustin, S. A., Benes, V., Garson, J. A., Hellemans, J., Huggett, J., Kubista, M., Mueller, R., Nolan, T., Pfaffl, M. W., Shipley, G. L., Vandesompele, J., and Wittwer, C. T. (2009). The MIQE guidelines: minimum information for publication of quantitative real-time PCR experiments. Clin. Chem. 55, 611–622.
The MIQE guidelines: minimum information for publication of quantitative real-time PCR experiments.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DC%2BD1MXktVWqs7g%3D&md5=8d74aecc865b6c5e86b41ab1f4665641CAS |

Cagnone, G., and Sirard, M. A. (2014). The impact of exposure to serum lipids during in vitro culture on the transcriptome of bovine blastocysts. Theriogenology 81, 712–722e1–3.
The impact of exposure to serum lipids during in vitro culture on the transcriptome of bovine blastocysts.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DC%2BC2cXhtlSlsLw%3D&md5=0ac721c0bbee096dd22b24cbfef1d1ccCAS |

Campbell, B. K., Souza, C., Gong, J., Webb, R., Kendall, N., Marsters, P., Robinson, G., Mitchell, A., Telfer, E. E., and Baird, D. T. (2003). Domestic ruminants as models for the elucidation of the mechanisms controlling ovarian follicle development in humans. Reprod. Suppl. 61, 429–443.
| 1:CAS:528:DC%2BD3sXptFKhtrc%3D&md5=5727da2415735d0cf494fb5fa5c5b1eeCAS |

Cerri, R. L., Juchem, S. O., Chebel, R. C., Rutigliano, H. M., Bruno, R. G., Galvao, K. N., Thatcher, W. W., and Santos, J. E. (2009). Effect of fat source differing in fatty acid profile on metabolic parameters, fertilization, and embryo quality in high-producing dairy cows. J. Dairy Sci. 92, 1520–1531.
Effect of fat source differing in fatty acid profile on metabolic parameters, fertilization, and embryo quality in high-producing dairy cows.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DC%2BD1MXjvFamt78%3D&md5=55a4567d0407406c9fe3241b4f1a0ecbCAS |

Chao, D. T., and Korsmeyer, S. J. (1998). BCL-2 family: regulators of cell death. Annu. Rev. Immunol. 16, 395–419.
BCL-2 family: regulators of cell death.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DyaK1cXislyntLs%3D&md5=7cb4ffc73de7beb98d5d409431dfcbf3CAS |

Childs, S., Hennessy, A. A., Sreenan, J. M., Wathes, D. C., Cheng, Z., Stanton, C., Diskin, M. G., and Kenny, D. A. (2008). Effect of level of dietary n-3 polyunsaturated fatty acid supplementation on systemic and tissue fatty acid concentrations and on selected reproductive variables in cattle. Theriogenology 70, 595–611.
Effect of level of dietary n-3 polyunsaturated fatty acid supplementation on systemic and tissue fatty acid concentrations and on selected reproductive variables in cattle.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DC%2BD1cXpsVCrtbs%3D&md5=3a7272130196ff591499a04033ca24f4CAS |

Das, U. N. (2010) Essential fatty acids: biochemistry and physiology. In ‘Metabolic Syndrome Pathophysiology’. pp. 181–200. (Wiley-Blackwell.)

De Wit, A. A., Cesar, M. L., and Kruip, T. A. (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=592fa4e1dc8973f12a418adf1ee4c2e8CAS |

Doreau, M., and Chilliard, Y. (1997). Digestion and metabolism of dietary fat in farm animals. Br. J. Nutr. 78, S15–S35.
Digestion and metabolism of dietary fat in farm animals.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DyaK2sXkvFOrs7k%3D&md5=0ea6a4da1158237a30cd5fc476ca5ec6CAS |

Farsi, P. F., Djazayery, A., Eshraghian, M. R., Koohdani, F., Saboor-Yaraghi, A. A., Derakhshanian, H., Zarei, M., Javanbakht, M. H., and Djalali, M. (2014). Effects of supplementation with omega-3 on insulin sensitivity and non-esterified free fatty acid (NEFA) in type 2 diabetic patients. Arq. Bras. Endocrinol. Metabol. 58, 335–340.
Effects of supplementation with omega-3 on insulin sensitivity and non-esterified free fatty acid (NEFA) in type 2 diabetic patients.Crossref | GoogleScholarGoogle Scholar |

Fenwick, M. A., Fitzpatrick, R., Kenny, D. A., Diskin, M. G., Patton, J., Murphy, J. J., and Wathes, D. C. (2008). Interrelationships between negative energy balance (NEB) and IGF regulation in liver of lactating dairy cows. Domest. Anim. Endocrinol. 34, 31–44.
Interrelationships between negative energy balance (NEB) and IGF regulation in liver of lactating dairy cows.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DC%2BD2sXhtlWkt7jP&md5=f11d342344c8fe5e156c5279d182270bCAS |

Ferguson, E. M., and Leese, H. J. (1999). Triglyceride content of bovine oocytes and early embryos. J. Reprod. Fertil. 116, 373–378.
Triglyceride content of bovine oocytes and early embryos.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DyaK1MXkslOltbc%3D&md5=a3a50038d8b7e9a43a4aadb2a7a6e68dCAS |

Ferguson, J. D., Galligan, D. T., Blanchard, T., and Reeves, M. (1993). Serum urea nitrogen and conception rate: the usefulness of test information. J. Dairy Sci. 76, 3742–3746.
Serum urea nitrogen and conception rate: the usefulness of test information.Crossref | GoogleScholarGoogle Scholar | 1:STN:280:DyaK2c7nvFKitQ%3D%3D&md5=dca11686c6a14054354f568e5451521dCAS |

Ferreira, R. C., Halpern, G., Figueira Rde, C., Braga, D. P., Iaconelli, A., and Borges, E. (2010). Physical activity, obesity and eating habits can influence assisted reproduction outcomes. Womens Health (Lond. Engl.) 6, 517–524.
Physical activity, obesity and eating habits can influence assisted reproduction outcomes.Crossref | GoogleScholarGoogle Scholar |

Field, C. J., Angel, A., and Clandinin, M. T. (1985). Relationship of diet to the fatty acid composition of human adipose tissue structural and stored lipids. Am. J. Clin. Nutr. 42, 1206–1220.
| 1:CAS:528:DyaL28XmvVSjtg%3D%3D&md5=c113fbc6b86c8378aca4a9009cbbe605CAS |

Fredrickson, D. S., and Lees, R. S. (1965). A system for phenotyping hyperlipoproteinemia. Circulation 31, 321–327.
A system for phenotyping hyperlipoproteinemia.Crossref | GoogleScholarGoogle Scholar | 1:STN:280:DyaF2M%2FmslWjsg%3D%3D&md5=bbf822f1b3ec2267ca69379d655f765bCAS |

Gallagher, E. J., Leroith, D., and Karnieli, E. (2010). Insulin resistance in obesity as the underlying cause for the metabolic syndrome. Mt. Sinai J. Med. 77, 511–523.
Insulin resistance in obesity as the underlying cause for the metabolic syndrome.Crossref | GoogleScholarGoogle Scholar |

Gardner, D. K., Wale, P. L., Collins, R., and Lane, M. (2011). Glucose consumption of single post-compaction human embryos is predictive of embryo sex and live birth outcome. Hum. Reprod. 26, 1981–1986.
Glucose consumption of single post-compaction human embryos is predictive of embryo sex and live birth outcome.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DC%2BC3MXptFCnsbw%3D&md5=5b096401107b2e42291ed54db17095abCAS |

Gardner, D. K., Gardner, D. K., Harvey, A. J., and Harvey, A. J. (2015). Blastocyst metabolism. Reprod. Fertil. Dev. 27, 638–654.
Blastocyst metabolism.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DC%2BC2MXntVWntbw%3D&md5=2b534b0712b622aa5aa3338f40f97c69CAS |

Gibney, M. J., and Hunter, B. (1993). The effects of short- and long-term supplementation with fish oil on the incorporation of n-3 polyunsaturated fatty acids into cells of the immune system in healthy volunteers. Eur. J. Clin. Nutr. 47, 255–259.
| 1:STN:280:DyaK3s3lvFSqug%3D%3D&md5=c9abb015e45dc70d32846ad3b6237fb9CAS |

Gómez, E., Gutiérrez-Adán, A., Díez, C., Bermejo-Alvarez, P., Muñoz, M., Rodriguez, A., Otero, J., Alvarez-Viejo, M., Martín, D., Carrocera, S., and Caamaño, J. N. (2009). Biological differences between in vitro produced bovine embryos and parthenotes. Reproduction 137, 285–295.
Biological differences between in vitro produced bovine embryos and parthenotes.Crossref | GoogleScholarGoogle Scholar |

Guerif, F., McKeegan, P., Leese, H. J., and Sturmey, R. G. (2013). A simple approach for COnsumption and RElease (CORE) analysis of metabolic activity in single mammalian embryos. PLoS One 8, e67834.
A simple approach for COnsumption and RElease (CORE) analysis of metabolic activity in single mammalian embryos.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DC%2BC3sXhtleqsrvF&md5=b54ea1d5b95b9eadac38d815884bf892CAS |

Guretzky, N. A., Carlson, D. B., Garrett, J. E., and Drackley, J. K. (2006). Lipid metabolite profiles and milk production for Holstein and Jersey cows fed rumen-protected choline during the periparturient period. J. Dairy Sci. 89, 188–200.
Lipid metabolite profiles and milk production for Holstein and Jersey cows fed rumen-protected choline during the periparturient period.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DC%2BD28XisVSktw%3D%3D&md5=6c2bcb14fd80f72765e634e46ad58cd7CAS |

Haggarty, P., Wood, M., Ferguson, E., Hoad, G., Srikantharajah, A., Milne, E., Hamilton, M., and Bhattacharya, S. (2006). Fatty acid metabolism in human preimplantation embryos. Hum. Reprod. 21, 766–773.
Fatty acid metabolism in human preimplantation embryos.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DC%2BD28Xhs1Sks7w%3D&md5=20bea91e896e7cbc19b6a26cf13f21b7CAS |

Hawkins, D. E., Niswender, K. D., Oss, G. M., Moeller, C. L., Odde, K. G., Sawyer, H. R., and Niswender, G. D. (1995). An increase in serum lipids increases luteal lipid content and alters the disappearance rate of progesterone in cows. J. Anim. Sci. 73, 541–545.
An increase in serum lipids increases luteal lipid content and alters the disappearance rate of progesterone in cows.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DyaK2MXjsFehs7o%3D&md5=632da47f7b8fdcc279d912eda363fa94CAS |

Hochi, S., Kimura, K., and Hanada, A. (1999). Effect of linoleic acid–albumin in the culture medium on freezing sensitivity of in vitro-produced bovine morulae. Theriogenology 52, 497–504.
Effect of linoleic acid–albumin in the culture medium on freezing sensitivity of in vitro-produced bovine morulae.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DyaK1MXmtlyhuro%3D&md5=31ecf1f1eaa5a89c7e9dcd7cad0aadc6CAS |

Holtenius, P., and Holtenius, K. (2007). A model to estimate insulin sensitivity in dairy cows. Acta Vet. Scand. 49, 29.
A model to estimate insulin sensitivity in dairy cows.Crossref | GoogleScholarGoogle Scholar |

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=9d6a93b5a4108a6d6cdcc36bee8d0c91CAS |

Igosheva, N., Abramov, A. Y., Poston, L., Eckert, J. J., Fleming, T. P., Duchen, M. R., and McConnell, J. (2010). Maternal diet-induced obesity alters mitochondrial activity and redox status in mouse oocytes and zygotes. PLoS One 5, e10074.
Maternal diet-induced obesity alters mitochondrial activity and redox status in mouse oocytes and zygotes.Crossref | GoogleScholarGoogle Scholar |

Jordaens, L., Arias-Alvarez, M., Pintelon, I., Thys, S., Valckx, S., Dezhkam, Y., Bols, P. E., and Leroy, J. L. (2015). Elevated non-esterified fatty acid concentrations hamper bovine oviductal epithelial cell physiology in three different in vitro culture systems. Theriogenology 84, 899–910.
Elevated non-esterified fatty acid concentrations hamper bovine oviductal epithelial cell physiology in three different in vitro culture systems.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DC%2BC2MXhtVKmsb3E&md5=384d3eb345a9fff26afae72a11cc662dCAS |

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=638b7ac72565fcdebd202d30641081c8CAS |

Kim, J. Y., 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 | 1:CAS:528:DC%2BD3MXlsVGis70%3D&md5=f0cf4e744ada4756c8e8e10207580143CAS |

Kletzien, R. F., Harris, P. K., and Foellmi, L. A. (1994). Glucose-6-phosphate dehydrogenase: a ‘housekeeping’ enzyme subject to tissue-specific regulation by hormones, nutrients, and oxidant stress. FASEB J. 8, 174–181.
| 1:CAS:528:DyaK2cXitlyktr4%3D&md5=94c0ce2a3f59d7d9efde83463ddc22b5CAS |

Kris-Etherton, P. M., and Fleming, J. A. (2015). Emerging nutrition science on fatty acids and cardiovascular disease: nutritionists’ perspectives. Adv. Nutr. 6, 326S–337S.
Emerging nutrition science on fatty acids and cardiovascular disease: nutritionists’ perspectives.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DC%2BC2MXhtVyiur%2FN&md5=44096758e6669943bf005e63547c7f9eCAS |

Landau, Z., Forti, E., Alcaly, M., and Birk, R. Z. (2006). Palmitate induced lipoapoptosis of exocrine pancreas AR42J cells. Apoptosis 11, 717–724.
Palmitate induced lipoapoptosis of exocrine pancreas AR42J cells.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DC%2BD28XlsV2ks7c%3D&md5=9f4581f40c4e7f9feceafbafb1226f74CAS |

Larqué, E., Gil-Sánchez, A., Prieto-Sánchez, M. T., and Koletzko, B. (2012). Omega 3 fatty acids, gestation and pregnancy outcomes. Br. J. Nutr. 107, S77–S84.
Omega 3 fatty acids, gestation and pregnancy outcomes.Crossref | GoogleScholarGoogle Scholar |

Leary, C., Leese, H. J., and Sturmey, R. G. (2015). Human embryos from overweight and obese women display phenotypic and metabolic abnormalities. Hum. Reprod. 30, 122–132.
Human embryos from overweight and obese women display phenotypic and metabolic abnormalities.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=84fc6944f81c3f63ef1ef0571e91939cCAS |

Lei, S., Liu, Y., Liu, H., Yu, H., Wang, H., and Xia, Z. (2012). Effects of N-acetylcysteine on nicotinamide dinucleotide phosphate oxidase activation and antioxidant status in heart, lung, liver and kidney in streptozotocin-induced diabetic rats. Yonsei Med. J. 53, 294–303.
Effects of N-acetylcysteine on nicotinamide dinucleotide phosphate oxidase activation and antioxidant status in heart, lung, liver and kidney in streptozotocin-induced diabetic rats.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DC%2BC38XkvVeksrY%3D&md5=38f62feb99f3862f6f5c0dfd92e52356CAS |

Lepage, G., and Roy, C. C. (1986). Direct transesterification of all classes of lipids in a one-step reaction. J. Lipid Res. 27, 114–120.
| 1:CAS:528:DyaL28XhtVGgsLg%3D&md5=fa9042cd5ccfcb348b7a7bf39167b5ffCAS |

Leroy, J. L., Vanholder, T., Delanghe, J. R., Opsomer, G., Van Soom, A., Bols, P. E., Dewulf, 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=1af36118c3f6162125662231130e1d31CAS |

Leroy, J. L., Opsomer, G., De Vliegher, S., Vanholder, T., Goossens, L., Geldhof, A., Bols, P. E., de Kruif, A., and Van Soom, A. (2005). Comparison of embryo quality in high-yielding dairy cows, in dairy heifers and in beef cows. Theriogenology 64, 2022–2036.
Comparison of embryo quality in high-yielding dairy cows, in dairy heifers and in beef cows.Crossref | GoogleScholarGoogle Scholar | 1:STN:280:DC%2BD2MnitlejtQ%3D%3D&md5=94ed974c900fb9ede3c4499f7b9e32d7CAS |

Leroy, J. L., Van Hoeck, V., Clemente, M., Rizos, D., Gutierrez-Adan, A., Van Soom, A., Uytterhoeven, M., and Bols, P. E. (2010). The effect of nutritionally induced hyperlipidaemia on in vitro bovine embryo quality. Hum. Reprod. 25, 768–778.
The effect of nutritionally induced hyperlipidaemia on in vitro bovine embryo quality.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DC%2BC3cXitFeitr8%3D&md5=87af537b11e8d3d7953e6cd90592f945CAS |

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

Lichtenstein, A. H., Appel, L. J., Brands, M., Carnethon, M., Daniels, S., Franch, H. A., Franklin, B., Kris-Etherton, P., Harris, W. S., Howard, B., Karanja, N., Lefevre, M., Rudel, L., Sacks, F., Van Horn, L., Winston, M., and Wylie-Rosett, J. (2006). Diet and lifestyle recommendations revision 2006: a scientific statement from the American Heart Association Nutrition Committee. Circulation 114, 82–96.
Diet and lifestyle recommendations revision 2006: a scientific statement from the American Heart Association Nutrition Committee.Crossref | GoogleScholarGoogle Scholar |

Livak, K. J., and Schmittgen, T. D. (2001). Analysis of relative gene expression data using real-time quantitative PCR and the 2–ΔΔCT method. Methods 25, 402–408.
Analysis of relative gene expression data using real-time quantitative PCR and the 2–ΔΔCT method.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DC%2BD38XhtFelt7s%3D&md5=3d53b7f31b0c4fa91bfc79f22f87effaCAS |

Lonergan, P., Woods, A., Fair, T., Carter, F., Rizos, D., Ward, F., Quinn, K., and Evans, A. (2007). Effect of embryo source and recipient progesterone environment on embryo development in cattle. Reprod. Fertil. Dev. 19, 861–868.
Effect of embryo source and recipient progesterone environment on embryo development in cattle.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DC%2BD2sXhtVWhtrzK&md5=4a5d39480829ab05184959c4b9ed619dCAS |

Luzzo, K. M., Wang, Q., Purcell, S. H., Chi, M., Jimenez, P. T., Grindler, N., Schedl, T., and Moley, K. H. (2012). High fat diet induced developmental defects in the mouse: oocyte meiotic aneuploidy and fetal growth retardation/brain defects. PLoS One 7, e49217.
High fat diet induced developmental defects in the mouse: oocyte meiotic aneuploidy and fetal growth retardation/brain defects.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DC%2BC38XhslKjtrvN&md5=ab8600b6e2c15631c69b14f5f10f5829CAS |

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=156075a750bf6bbdc287f522b75e69f9CAS |

Marei, W. F., Wathes, D. C., and Fouladi-Nashta, A. A. (2010). Impact of linoleic acid on bovine oocyte maturation and embryo development. Reproduction 139, 979–988.
Impact of linoleic acid on bovine oocyte maturation and embryo development.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DC%2BC3cXns12qsLY%3D&md5=c7e8bb2e1cc21c723a53bf547c08ae39CAS |

Marsden, V. S., O’Connor, L., O’Reilly, L. A., Silke, J., Metcalf, D., Ekert, P. G., Huang, D. C., Cecconi, F., Kuida, K., Tomaselli, K. J., Roy, S., Nicholson, D. W., Vaux, D. L., Bouillet, P., Adams, J. M., and Strasser, A. (2002). Apoptosis initiated by Bcl-2-regulated caspase activation independently of the cytochrome c/Apaf-1/caspase-9 apoptosome. Nature 419, 634–637.
Apoptosis initiated by Bcl-2-regulated caspase activation independently of the cytochrome c/Apaf-1/caspase-9 apoptosome.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DC%2BD38Xns1Ont70%3D&md5=00ddf3b794ed3e3d25729e33e641f590CAS |

Mendoza, C., Ruiz-Requena, E., Ortega, E., Cremades, N., Martinez, F., Bernabeu, R., Greco, E., and Tesarik, J. (2002). Follicular fluid markers of oocyte developmental potential. Hum. Reprod. 17, 1017–1022.
Follicular fluid markers of oocyte developmental potential.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DC%2BD38Xjs12isLk%3D&md5=dd9bc9ce941ac73d55667d16bf11b2e0CAS |

Moss, J. I., Garrett, T. J., and Hansen, P. J. (2012). Involvement of free cholesterol and high-density lipoprotein in development and resistance of the preimplantation bovine embryo to heat shock. J. Anim. Sci. 90, 3762–3769.
Involvement of free cholesterol and high-density lipoprotein in development and resistance of the preimplantation bovine embryo to heat shock.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DC%2BC38XhslOit7bM&md5=71f5bbd9677105cafa1e6b3f36574fd3CAS |

National Research Council (U.S.) Subcommittee on Dairy Cattle Nutrition (2001) ‘Nutrient Requirements of Dairy Cattle.’ 7th Revised Edition. (National Academy Press: Washington, DC.)

Newman, A., Mann, S., Nydam, D. V., Overton, T. R., and Behling-Kelly, E. (2016). Impact of dietary plane of energy during the dry period on lipoprotein parameters in the transition period in dairy cattle. J. Anim. Physiol. Anim. Nutr. (Berl.) 100, 118–126.
Impact of dietary plane of energy during the dry period on lipoprotein parameters in the transition period in dairy cattle.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DC%2BC28XpsFCluw%3D%3D&md5=bdc9e345cd48832624fa402d2194bc98CAS |

O’Hara, L., Forde, N., Kelly, A. K., and Lonergan, P. (2014). Effect of bovine blastocyst size at embryo transfer on Day 7 on conceptus length on Day 14: can supplementary progesterone rescue small embryos? Theriogenology 81, 1123–1128.
Effect of bovine blastocyst size at embryo transfer on Day 7 on conceptus length on Day 14: can supplementary progesterone rescue small embryos?Crossref | GoogleScholarGoogle Scholar |

Palmquist, D. L. (1976). A kinetic concepto of lipid transport in ruminants. J. Dairy Sci. 59, 355–363.
A kinetic concepto of lipid transport in ruminants.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DyaE28Xht1Sju7c%3D&md5=d3803b89dcaf675a8bacd5e2253e3bc0CAS |

Panchal, S. K., Poudyal, H., Iyer, A., Nazer, R., Alam, A., Diwan, V., Kauter, K., Sernia, C., Campbell, F., Ward, L., Gobe, G., Fenning, A., and Brown, L. (2011). High-carbohydrate high-fat diet-induced metabolic syndrome and cardiovascular remodeling in rats. J. Cardiovasc. Pharmacol. 57, 51–64.
| 1:CAS:528:DC%2BC3MXlsFSisQ%3D%3D&md5=5ba6f8fe6e5674291ea26eeb141b6082CAS |

Perk, J., De Backer, G., Gohlke, H., Graham, I., Reiner, Z., Verschuren, M., Albus, C., Benlian, P., Boysen, G., Cifkova, R., et al. (2012). European Guidelines on cardiovascular disease prevention in clinical practice (version 2012). The Fifth Joint Task Force of the European Society of Cardiology and Other Societies on Cardiovascular Disease Prevention in Clinical Practice (constituted by representatives of nine societies and by invited experts). Eur. Heart J. 33, 1635–1701.
European Guidelines on cardiovascular disease prevention in clinical practice (version 2012). The Fifth Joint Task Force of the European Society of Cardiology and Other Societies on Cardiovascular Disease Prevention in Clinical Practice (constituted by representatives of nine societies and by invited experts).Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DC%2BC38XhtVeitr3P&md5=234c0a27ca3aaf1a2b59624ba37e3ed8CAS |

Perseghin, G., Caumo, A., Caloni, M., Testolin, G., and Luzi, L. (2001). Incorporation of the fasting plasma FFA concentration into QUICKI improves its association with insulin sensitivity in nonobese individuals. J. Clin. Endocrinol. Metab. 86, 4776–4781.
Incorporation of the fasting plasma FFA concentration into QUICKI improves its association with insulin sensitivity in nonobese individuals.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DC%2BD3MXptVars7k%3D&md5=a1a7b777e70448c7d73174e20413dda6CAS |

Petit, H. V., Cavalieri, F. B., Santos, G. T., Morgan, J., and Sharpe, P. (2008). Quality of embryos produced from dairy cows fed whole flaxseed and the success of embryo transfer. J. Dairy Sci. 91, 1786–1790.
Quality of embryos produced from dairy cows fed whole flaxseed and the success of embryo transfer.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DC%2BD1cXltlWmtLw%3D&md5=f0e4c106cc4c54edbbc99f508831b5dbCAS |

Popp-Snijders, C., Schouten, J. A., Heine, R. J., van der Meer, J., and van der Veen, E. A. (1987). Dietary supplementation of omega-3 polyunsaturated fatty acids improves insulin sensitivity in non-insulin-dependent diabetes. Diabetes Res. 4, 141–147.
| 1:STN:280:DyaL2s3nsVGrsw%3D%3D&md5=75ed69d3f5cef96fd2fef02faa2f5d1fCAS |

Rabasa-Lhoret, R., Bastard, J. P., Jan, V., Ducluzeau, P. H., Andreelli, F., Guebre, F., Bruzeau, J., Louche-Pellissier, C., Maîtrepierre, C., Peyrat, J., Chagné, J., Vidal, H., and Laville, M. (2003). Modified quantitative insulin sensitivity check index is better correlated to hyperinsulinemic glucose clamp than other fasting-based index of insulin sensitivity in different insulin-resistant states. J. Clin. Endocrinol. Metab. 88, 4917–4923.
Modified quantitative insulin sensitivity check index is better correlated to hyperinsulinemic glucose clamp than other fasting-based index of insulin sensitivity in different insulin-resistant states.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DC%2BD3sXoslemurg%3D&md5=d12ce62306a479adf721163dd928320aCAS |

Ruxton, C. H. S., Reed, S. C., Simpson, M. J. A., and Millington, K. J. (2007). The health benefits of omega-3 polyunsaturated fatty acids: a review of the evidence. J. Hum. Nutr. Diet. 20, 275–285.
The health benefits of omega-3 polyunsaturated fatty acids: a review of the evidence.Crossref | GoogleScholarGoogle Scholar | 1:STN:280:DC%2BD2szislygsQ%3D%3D&md5=d86b3551e92308c0ebb3518fe479e040CAS |

Sánchez, I., Reynoso-Camacho, R., and Salgado, L. M. (2015). The diet-induced metabolic syndrome is accompanied by whole-genome epigenetic changes. Genes Nutr. 10, 21.
The diet-induced metabolic syndrome is accompanied by whole-genome epigenetic changes.Crossref | GoogleScholarGoogle Scholar |

Santos, J. E., Bilby, T. R., Thatcher, W. W., Staples, C. R., and Silvestre, F. T. (2008). Long chain fatty acids of diet as factors influencing reproduction in cattle. Reprod. Domest. Anim. 43, 23–30.
Long chain fatty acids of diet as factors influencing reproduction in cattle.Crossref | GoogleScholarGoogle Scholar |

Schulz, K., Frahm, J., Meyer, U., Kersten, S., Reiche, D., Rehage, J., and Danicke, S. (2014). Effects of prepartal body condition score and peripartal energy supply of dairy cows on postpartal lipolysis, energy balance and ketogenesis: an animal model to investigate subclinical ketosis. J. Dairy Res. 81, 257–266.
Effects of prepartal body condition score and peripartal energy supply of dairy cows on postpartal lipolysis, energy balance and ketogenesis: an animal model to investigate subclinical ketosis.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DC%2BC2cXht1SqsLnI&md5=da1cf53e8467e4b6062c4c6d140b33d2CAS |

Styne-Gross, A., Elkind-Hirsch, K., and Scott, R. T. (2005). Obesity does not impact implantation rates or pregnancy outcome in women attempting conception through oocyte donation. Fertil. Steril. 83, 1629–1634.
Obesity does not impact implantation rates or pregnancy outcome in women attempting conception through oocyte donation.Crossref | GoogleScholarGoogle Scholar |

Valckx, S. D., Arias-Alvarez, M., De Pauw, I., Fievez, V., Vlaeminck, B., Fransen, E., Bols, P. E., and Leroy, J. L. (2014). 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 |

van der Steeg, J. W., Steures, P., Eijkemans, M. J., Habbema, J. D., Hompes, P. G., Burggraaff, J. M., Oosterhuis, G. J., Bossuyt, P. M., van der Veen, F., and Mol, B. W. (2008). Obesity affects spontaneous pregnancy chances in subfertile, ovulatory women. Hum. Reprod. 23, 324–328.
Obesity affects spontaneous pregnancy chances in subfertile, ovulatory women.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=a0b552d20a6393da836bc87c594fdd9eCAS |

Wakefield, S. L., Lane, M., Schulz, S. J., Hebart, M. L., Thompson, J. G., and Mitchell, M. (2008). Maternal supply of omega-3 polyunsaturated fatty acids alter mechanisms involved in oocyte and early embryo development in the mouse. Am. J. Physiol. Endocrinol. Metab. 294, E425–E434.
Maternal supply of omega-3 polyunsaturated fatty acids alter mechanisms involved in oocyte and early embryo development in the mouse.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DC%2BD1cXit1ykur0%3D&md5=6ae9b241ab593d26af9789d5e0bfa8b8CAS |

Waterman, R. A., and Wall, R. J. (1988). Lipid interactions with in vitro development of mammalian zygotes. Gamete Res. 21, 243–254.
Lipid interactions with in vitro development of mammalian zygotes.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DyaL1MXlt1Sk&md5=22e7ca10a9354330014a6cc1bf9b0854CAS |

Wehrman, M. E., Welsh, T. H., and Williams, G. L. (1991). Diet-induced hyperlipidemia in cattle modifies the intrafollicular cholesterol environment, modulates ovarian follicular dynamics, and hastens the onset of postpartum luteal activity. Biol. Reprod. 45, 514–522.
Diet-induced hyperlipidemia in cattle modifies the intrafollicular cholesterol environment, modulates ovarian follicular dynamics, and hastens the onset of postpartum luteal activity.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DyaK3MXlvVWqu7s%3D&md5=bc3f19c00d5b0bb00cc662a3d2a0d29dCAS |

Zraika, S., Dunlop, M., Proietto, J., and Andrikopoulos, S. (2002). Effects of free fatty acids on insulin secretion in obesity. Obes. Rev. 3, 103–112.
Effects of free fatty acids on insulin secretion in obesity.Crossref | GoogleScholarGoogle Scholar | 1:CAS:528:DC%2BD38XksFyrsLc%3D&md5=f7ea7aaeb248cd2f8ead2e77a9943d0cCAS |