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

Parameters to identify good quality oocytes and embryos in cattle

Christine Wrenzycki A *
+ Author Affiliations
- Author Affiliations

A Chair for Molecular Reproductive Medicine, Clinic for Veterinary Obstetrics, Gynecology and Andrology of Large and Small Animals, Justus-Liebig-University Giessen, Frankfurter Straße 106, Giessen 35392, Germany.

Reproduction, Fertility and Development 34(2) 190-202 https://doi.org/10.1071/RD21283
Published online: 29 October 2021

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

Abstract

Oocyte/embryo selection methodologies are either invasive or noninvasive and can be applied at various stages of development from the oocyte to cleaved embryos and up to the blastocyst stage. Morphology and the proportion of embryos developing to the blastocyst stage are important criteria to assess developmental competence. Evaluation of morphology remains the method of choice for selecting viable oocytes for IVP or embryos prior to transfer. Although non-invasive approaches are improving, invasive ones have been extremely helpful in finding candidate genes to determine oocyte/embryo quality. There is still a strong need for further refinement of existing oocyte and embryo selection methods and quality parameters. The development of novel, robust and non-invasive procedures will ensure that only embryos with the highest developmental potential are chosen for transfer. In the present review, various methods for assessing the quality of oocytes and preimplantation embryos, particularly in cattle, are considered. These methods include assessment of morphology including different staining procedures, transcriptomic and proteomic analyses, metabolic profiling, as well as the use of artificial intelligence technologies.

Keywords: artificial intelligence, cattle, developmental competence, embryo, metabolomics, morphology, oocyte, proteomics, quality, stainings, transcriptomics.


References

Aardema, H, van Tol, HTA, and Vos, PLAM (2019). An overview on how cumulus cells interact with the oocyte in a condition with elevated NEFA levels in dairy cows. Animal Reproduction Science 207, 131–137.
An overview on how cumulus cells interact with the oocyte in a condition with elevated NEFA levels in dairy cows.Crossref | GoogleScholarGoogle Scholar | 31227325PubMed |

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 |

Aguila, L, Treulen, F, Therrien, J, Felmer, R, Valdivia, M, and Smith, LC (2020). Oocyte selection for in vitro embryo production in bovine species: noninvasive approaches for new challenges of oocyte competence. Animals 10, 2196.
Oocyte selection for in vitro embryo production in bovine species: noninvasive approaches for new challenges of oocyte competence.Crossref | GoogleScholarGoogle Scholar |

Alm, H, Torner, H, Löhrke, B, Viergutz, T, Ghoneim, IM, and Kanitz, W (2005). Bovine blastocyst development rate in vitro is influenced by selection of oocytes by brillant cresyl blue staining before IVM as indicator for glucose-6-phosphate dehydrogenase activity. Theriogenology 63, 2194–2205.
Bovine blastocyst development rate in vitro is influenced by selection of oocytes by brillant cresyl blue staining before IVM as indicator for glucose-6-phosphate dehydrogenase activity.Crossref | GoogleScholarGoogle Scholar | 15826683PubMed |

Banliat, C, Dubuisson, F, Corbin, E, Beurois, J, Tomas, D, Le Bourhis, D, Salvetti, P, Labas, V, Mermillod, P, and Saint-Dizier, M (2019). Intraoviductal concentrations of steroid hormones during in vitro culture changed phospholipid profiles and cryotolerance of bovine embryos. Molecular Reproduction and Development 86, 661–672.
Intraoviductal concentrations of steroid hormones during in vitro culture changed phospholipid profiles and cryotolerance of bovine embryos.Crossref | GoogleScholarGoogle Scholar | 30950150PubMed |

Banliat, C, Le Bourhis, D, Bernardi, O, Tomas, D, Labas, V, Salvetti, P, Guyonnet, B, Mermillod, P, and Saint-Dizier, M (2020). Oviduct fluid extracellular vesicles change the phospholipid composition of bovine embryos developed in vitro. International Journal of Molecular Sciences 21, 5326.
Oviduct fluid extracellular vesicles change the phospholipid composition of bovine embryos developed in vitro.Crossref | GoogleScholarGoogle Scholar |

Barceló-Fimbres, M, and Seidel, GE (2011). Cross-validation of techniques for measuring lipid content of bovine oocytes and blastocysts. Theriogenology 75, 434–444.
Cross-validation of techniques for measuring lipid content of bovine oocytes and blastocysts.Crossref | GoogleScholarGoogle Scholar | 21111465PubMed |

Blaschka, C, Sánchez-Guijo, A, Wudy, SA, and Wrenzycki, C (2020). Profile of bile acid subspecies is similar in blood and follicular fluid of cattle. Veterinary Medicine and Science 6, 167–176.
Profile of bile acid subspecies is similar in blood and follicular fluid of cattle.Crossref | GoogleScholarGoogle Scholar | 31713347PubMed |

Blaschka, C, Schuler, G, Sánchez-Guijo, A, Zimmer, B, Feller, S, Kotarski, F, Wudy, SA, and Wrenzycki, C (2018). Occurrence of sulfonated steroids and ovarian expression of steroid sulfatase and SULT1E1 in cyclic cows. The Journal of Steroid Biochemistry and Molecular Biology 179, 79–87.
Occurrence of sulfonated steroids and ovarian expression of steroid sulfatase and SULT1E1 in cyclic cows.Crossref | GoogleScholarGoogle Scholar | 29262378PubMed |

Bustin, SA, Benes, V, Garson, JA, Hellemans, J, Huggett, J, Kubista, M, Mueller, R, Nolan, T, Pfaffl, MW, Shipley, GL, Vandesompele, J, and Wittwer, CT (2009). The MIQE guidelines: minimum information for publication of quantitative real-time PCR experiments. Clinical Chemistry 55, 611–622.
The MIQE guidelines: minimum information for publication of quantitative real-time PCR experiments.Crossref | GoogleScholarGoogle Scholar | 19246619PubMed |

Caamaño, JN, Maside, C, Gil, MA, Muñoz, M, Cuello, C, Díez, C, Sánchez-Osorio, JR, Martín, D, Gomis, J, Vazquez, JM, Roca, J, Carrocera, S, Martinez, EA, and Gómez, E (2011). Use of polarized light microscopy in porcine reproductive technologies. Theriogenology 76, 669–677.
Use of polarized light microscopy in porcine reproductive technologies.Crossref | GoogleScholarGoogle Scholar | 21601264PubMed |

Caujolle, S, Cernat, R, Silvestri, G, Marques, MJ, Bradu, A, Feuchter, T, Robinson, G, Griffin, DK, and Podoleanu, A (2017). Speckle variance OCT for depth resolved assessment of the viability of bovine embryos. Biomedical Optics Express 8, 5139–5150.
Speckle variance OCT for depth resolved assessment of the viability of bovine embryos.Crossref | GoogleScholarGoogle Scholar | 29188109PubMed |

Choi, YH, Carnevale, EM, Seidel, GE, and Squire, EL (2001). Effects of gonadotropins on bovine oocytes matured in TCM-199. Theriogenology 56, 661–670.
Effects of gonadotropins on bovine oocytes matured in TCM-199.Crossref | GoogleScholarGoogle Scholar | 11572446PubMed |

Da Silveira, JC, Andrade, GM, Del Collado, M, Sampaio, RV, Sangalli, JR, Silva, LA, Pinaffi, FVL, Jardim, IB, Cesar, MC, Nogueira, MFG, Cesar, ASM, Coutinho, LL, Pereira, RW, Perecin, F, and Meirelles, FV (2017). Supplementation with small-extracellular vesicles from ovarian follicular fluid during in vitro production modulates bovine embryo development. PLoS One 12, e0179451.
Supplementation with small-extracellular vesicles from ovarian follicular fluid during in vitro production modulates bovine embryo development.Crossref | GoogleScholarGoogle Scholar | 28617821PubMed |

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 |

de Oliveira Fernandes, G, de Faria, OAC, Sifuentes, DN, Franco, MM, and Dode, MAN (2021). Electrospray mass spectrometry analysis of blastocoel fluid as a potential tool for bovine embryo selection. Journal of Assisted Reproduction and Genetics 38, 2209–2217.
Electrospray mass spectrometry analysis of blastocoel fluid as a potential tool for bovine embryo selection.Crossref | GoogleScholarGoogle Scholar | 33866497PubMed |

de Wit, AA, Wurth, YA, and Kruip, TA (2000). Effect of ovarian phase and follicle quality on morphology and developmental capacity of the bovine cumulus-oocyte complex. Journal of Animal Science 78, 1277–1283.
Effect of ovarian phase and follicle quality on morphology and developmental capacity of the bovine cumulus-oocyte complex.Crossref | GoogleScholarGoogle Scholar | 10834583PubMed |

de Wit, AAC, and Kruip, TAM (2001). Bovine cumulus-oocyte-complex-quality is reflected in sensitivity for alpha-amanitin, oocyte-diameter and developmental capacity. Animal Reproduction Science 65, 51–65.
Bovine cumulus-oocyte-complex-quality is reflected in sensitivity for alpha-amanitin, oocyte-diameter and developmental capacity.Crossref | GoogleScholarGoogle Scholar |

Demant, M, Deutsch, DR, Fröhlich, T, Wolf, E, and Arnold, GJ (2015). Proteome analysis of early lineage specification in bovine embryos. Proteomics 15, 688–701.
Proteome analysis of early lineage specification in bovine embryos.Crossref | GoogleScholarGoogle Scholar | 25143135PubMed |

Deutsch, DR, Fröhlich, T, Otte, KA, Beck, A, Habermann, FA, Wolf, E, and Arnold, GJ (2014). Stage-specific proteome signatures in early bovine embryo development. Journal of Proteome Research 13, 4363–4376.
Stage-specific proteome signatures in early bovine embryo development.Crossref | GoogleScholarGoogle Scholar | 25102770PubMed |

Di Santana, PPB, Da Silva, ALC, Ramos, RTJ, Gonçalves, AA, Da Costa, NN, do Ramos, PCA, Silva, TVG, Da Cordeiro, MS, do Santos, SSD, Ohashi, OM, and do Miranda, MS (2019). Contributions of RNA-seq to improve in vitro embryo production (IVP). Animal Reproduction 16, 249–259.
Contributions of RNA-seq to improve in vitro embryo production (IVP).Crossref | GoogleScholarGoogle Scholar |

Donnay, I, Partridge, RJ, and Leese, HJ (1999). Can embryo metabolism be used for selecting bovine embryos before transfer? Reproduction Nutrition Development 39, 523–533.
Can embryo metabolism be used for selecting bovine embryos before transfer?Crossref | GoogleScholarGoogle Scholar |

Driver, AM, Peñagaricano, F, Huang, W, Ahmad, KR, Hackbart, KS, Wiltbank, MC, and Khatib, H (2012). RNA-Seq analysis uncovers transcriptomic variations between morphologically similar in vivo- and in vitro-derived bovine blastocysts. BMC Genomics 13, 118.
RNA-Seq analysis uncovers transcriptomic variations between morphologically similar in vivo- and in vitro-derived bovine blastocysts.Crossref | GoogleScholarGoogle Scholar | 22452724PubMed |

Duranthon, V, and Renard, JP (2001). The developmental competence of mammalian oocytes: a convenient but biologically fuzzy concept. Theriogenology 55, 1277–1289.
The developmental competence of mammalian oocytes: a convenient but biologically fuzzy concept.Crossref | GoogleScholarGoogle Scholar | 11327684PubMed |

Fair, T (2010). Mammalian oocyte development: checkpoints for competence. Reproduction, Fertility and Development 22, 13–20.
Mammalian oocyte development: checkpoints for competence.Crossref | GoogleScholarGoogle Scholar |

Fair, T, Carter, F, Park, S, Evans, ACO, and Lonergan, P (2007). Global gene expression analysis during bovine oocyte in vitro maturation. Theriogenology 68, S91–S97.
Global gene expression analysis during bovine oocyte in vitro maturation.Crossref | GoogleScholarGoogle Scholar | 17512044PubMed |

Fair, T, Hyttel, P, and Greve, T (1995). Bovine oocyte diameter in relation to maturational competence and transcriptional activity. Molecular Reproduction and Development 42, 437–442.
Bovine oocyte diameter in relation to maturational competence and transcriptional activity.Crossref | GoogleScholarGoogle Scholar | 8607973PubMed |

Farin, PW, Britt, JH, Shaw, DW, and Slenning, BD (1995). Agreement among evaluators of bovine embryos produced in vivo or in vitro. Theriogenology 44, 339–349.
Agreement among evaluators of bovine embryos produced in vivo or in vitro.Crossref | GoogleScholarGoogle Scholar | 16727734PubMed |

Fatehi, AN, Zeinstra, EC, Kooij, RV, Colenbrander, B, and Bevers, MM (2002). Effect of cumulus cell removal of in vitro matured bovine oocytes prior to in vitro fertilization on subsequent cleavage rate. Theriogenology 57, 1347–1355.
Effect of cumulus cell removal of in vitro matured bovine oocytes prior to in vitro fertilization on subsequent cleavage rate.Crossref | GoogleScholarGoogle Scholar | 12013454PubMed |

Fouladi-Nashta, AA, Alberio, R, Kafi, M, Nicholas, B, Campbell, KHS, and Webb, R (2005). Differential staining combined with TUNEL labelling to detect apoptosis in preimplantation bovine embryos. Reproductive Biomedicine Online 10, 497–502.
Differential staining combined with TUNEL labelling to detect apoptosis in preimplantation bovine embryos.Crossref | GoogleScholarGoogle Scholar | 15901458PubMed |

Gegenfurtner, K, Flenkenthaler, F, Fröhlich, 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 |

Gilchrist, RB, Ritter, LJ, and Armstrong, DT (2004). Oocyte-somatic cell interactions during follicle development in mammals. Animal Reproduction Science 82–83, 431–446.
Oocyte-somatic cell interactions during follicle development in mammals.Crossref | GoogleScholarGoogle Scholar | 15271471PubMed |

Gjørret, JO, Fabian, D, Avery, B, and Maddox-Hyttel, P (2007). Active caspase-3 and ultrastructural evidence of apoptosis in spontaneous and induced cell death in bovine in vitro produced pre-implantation embryos. Molecular Reproduction and Development 74, 961–971.
Active caspase-3 and ultrastructural evidence of apoptosis in spontaneous and induced cell death in bovine in vitro produced pre-implantation embryos.Crossref | GoogleScholarGoogle Scholar | 17393434PubMed |

González-Serrano, AF, Pirro, V, Ferreira, CR, Oliveri, P, Eberlin, LS, Heinzmann, J, Lucas-Hahn, A, Niemann, H, and Cooks, RG (2013). Desorption electrospray ionization mass spectrometry reveals lipid metabolism of individual oocytes and embryos. PLoS One 8, e74981.
Desorption electrospray ionization mass spectrometry reveals lipid metabolism of individual oocytes and embryos.Crossref | GoogleScholarGoogle Scholar | 24073231PubMed |

Goodacre, R, Vaidyanathan, S, Dunn, WB, Harrigan, GG, and Kell, DB (2004). Metabolomics by numbers: acquiring and understanding global metabolite data. Trends in Biotechnology 22, 245–252.
Metabolomics by numbers: acquiring and understanding global metabolite data.Crossref | GoogleScholarGoogle Scholar | 15109811PubMed |

Gouveia Nogueira, MF, Bertogna Guilherme, V, Pronunciate, M, dos Santos, PH, Da Silva, DLB, and Rocha, JC (2018). Artificial intelligence-based grading quality of bovine blastocyst digital images: direct capture with juxtaposed lenses of smartphone camera and stereomicroscope ocular lens. Sensors 18, 4440.
Artificial intelligence-based grading quality of bovine blastocyst digital images: direct capture with juxtaposed lenses of smartphone camera and stereomicroscope ocular lens.Crossref | GoogleScholarGoogle Scholar |

Graf, A, Krebs, S, Zakhartchenko, V, Schwalb, B, Blum, H, and Wolf, E (2014). Fine mapping of genome activation in bovine embryos by RNA sequencing. Proceedings of the National Academy of Sciences of the United States of America 111, 4139–4144.
Fine mapping of genome activation in bovine embryos by RNA sequencing.Crossref | GoogleScholarGoogle Scholar | 24591639PubMed |

Harvey, AJ (2007). The role of oxygen in ruminant preimplantation embryo development and metabolism. Animal Reproduction Science 98, 113–128.
The role of oxygen in ruminant preimplantation embryo development and metabolism.Crossref | GoogleScholarGoogle Scholar | 17158002PubMed |

Hazeleger, NL, Hill, DJ, Stubbing, RB, and Walton, JS (1995). Relationship of morphology and follicular fluid environment of bovine oocytes to their developmental potential in vitro. Theriogenology 43, 509–522.
Relationship of morphology and follicular fluid environment of bovine oocytes to their developmental potential in vitro.Crossref | GoogleScholarGoogle Scholar | 16727642PubMed |

Hennet, ML, and Combelles, CMH (2012). The antral follicle: a microenvironment for oocyte differentiation. The International Journal of Developmental Biology 56, 819–831.
The antral follicle: a microenvironment for oocyte differentiation.Crossref | GoogleScholarGoogle Scholar | 23417404PubMed |

Hoelker, M, Held, E, Salilew-Wondim, D, Schellander, K, and Tesfaye, D (2014). Molecular signatures of bovine embryo developmental competence. Reproduction, Fertility and Development 26, 22–36.
Molecular signatures of bovine embryo developmental competence.Crossref | GoogleScholarGoogle Scholar |

Holm, P, Booth, PJ, and Callesen, H (2002). Kinetics of early in vitro development of bovine in vivo- and in vitro-derived zygotes produced and/or cultured in chemically defined or serum-containing media. Reproduction 123, 553–565.
Kinetics of early in vitro development of bovine in vivo- and in vitro-derived zygotes produced and/or cultured in chemically defined or serum-containing media.Crossref | GoogleScholarGoogle Scholar | 11914118PubMed |

Holm, P, Shukri, NN, Vajta, G, Booth, P, Bendixen, C, and Callesen, H (1998). Developmental kinetics of the first cell cycles of bovine in vitro produced embryos in relation to their in vitro viability and sex. Theriogenology 50, 1285–1299.
Developmental kinetics of the first cell cycles of bovine in vitro produced embryos in relation to their in vitro viability and sex.Crossref | GoogleScholarGoogle Scholar | 10734442PubMed |

Hung, W-T, 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 |

Iwasaki, W, Yamanaka, K, Sugiyama, D, Teshima, Y, Briones-Nagata, MP, Maeki, M, Yamashita, K, Takahashi, M, and Miyazaki, M (2018). Simple separation of good quality bovine oocytes using a microfluidic device. Scientific Reports 8, 14273.
Simple separation of good quality bovine oocytes using a microfluidic device.Crossref | GoogleScholarGoogle Scholar | 30250059PubMed |

Jensen, PL, Grøndahl, ML, Beck, HC, Petersen, J, Stroebech, L, Christensen, ST, and Yding Andersen, C (2014). Proteomic analysis of bovine blastocoel fluid and blastocyst cells. Systems Biology in Reproductive Medicine 60, 127–135.
Proteomic analysis of bovine blastocoel fluid and blastocyst cells.Crossref | GoogleScholarGoogle Scholar | 24568550PubMed |

Jiang, Z, Dong, H, Zheng, X, Marjani, SL, Donovan, DM, Chen, J, and Tian, XC (2015). mRNA levels of imprinted genes in bovine in vivo oocytes, embryos and cross species comparisons with humans, mice and pigs. Scientific Reports 5, 17898.
mRNA levels of imprinted genes in bovine in vivo oocytes, embryos and cross species comparisons with humans, mice and pigs.Crossref | GoogleScholarGoogle Scholar | 26638780PubMed |

Jiang, Z, Sun, J, Dong, H, Luo, O, Zheng, X, Obergfell, C, Tang, Y, Bi, J, O’Neill, R, Ruan, Y, Chen, J, and Tian, XC (2014). Transcriptional profiles of bovine in vivo pre-implantation development. BMC Genomics 15, 756.
Transcriptional profiles of bovine in vivo pre-implantation development.Crossref | GoogleScholarGoogle Scholar | 25185836PubMed |

Koester, M, Mohammadi-Sangcheshmeh, A, Montag, M, Rings, F, Schimming, T, Tesfaye, D, Schellander, K, and Hoelker, M (2011). Evaluation of bovine zona pellucida characteristics in polarized light as a prognostic marker for embryonic developmental potential. Reproduction 141, 779–787.
Evaluation of bovine zona pellucida characteristics in polarized light as a prognostic marker for embryonic developmental potential.Crossref | GoogleScholarGoogle Scholar | 21415090PubMed |

Kraupp, BG, Ruttkay-Nedecky, B, Koudelka, H, Bukowska, K, Bursch, W, and Schulte-Hermann, R (1995). In situ detection of fragmented DNA (TUNEL assay) fails to discriminate among apoptosis, necrosis, and autolytic cell death: a cautionary note. Hepatology 21, 1465–1468.
In situ detection of fragmented DNA (TUNEL assay) fails to discriminate among apoptosis, necrosis, and autolytic cell death: a cautionary note.Crossref | GoogleScholarGoogle Scholar |

Krisher, RL (2004). The effect of oocyte quality on development. Journal of Animal Science 82, E14–E23.
The effect of oocyte quality on development.Crossref | GoogleScholarGoogle Scholar | 15471793PubMed |

Krisher, RL (2013). In vivo and in vitro environmental effects on mammalian oocyte quality. Annual Review of Animal Biosciences 1, 393–417.
In vivo and in vitro environmental effects on mammalian oocyte quality.Crossref | GoogleScholarGoogle Scholar | 25387025PubMed |

Krisher, RL, Heuberger, AL, Paczkowski, M, Stevens, J, Pospisil, C, Prather, RS, Sturmey, RG, Herrick, JR, and Schoolcraft, WB (2015). Applying metabolomic analyses to the practice of embryology: physiology, development and assisted reproductive technology. Reproduction, Fertility and Development 27, 602–620.
Applying metabolomic analyses to the practice of embryology: physiology, development and assisted reproductive technology.Crossref | GoogleScholarGoogle Scholar |

Kropp, J, and Khatib, H (2015). Characterization of microRNA in bovine in vitro culture media associated with embryo quality and development. Journal of Dairy Science 98, 6552–6563.
Characterization of microRNA in bovine in vitro culture media associated with embryo quality and development.Crossref | GoogleScholarGoogle Scholar | 26142856PubMed |

Kropp, J, Salih, SM, and Khatib, H (2014). Expression of microRNAs in bovine and human pre-implantation embryo culture media. Frontiers in Genetics 5, 91.
Expression of microRNAs in bovine and human pre-implantation embryo culture media.Crossref | GoogleScholarGoogle Scholar | 24795753PubMed |

Kues, WA, Sudheer, S, Herrmann, D, Carnwath, JW, Havlicek, V, Besenfelder, U, Lehrach, H, Adjaye, J, and Niemann, H (2008). Genome-wide expression profiling reveals distinct clusters of transcriptional regulation during bovine preimplantation development in vivo. Proceedings of the National Academy of Sciences of the United States of America 105, 19768–19773.
Genome-wide expression profiling reveals distinct clusters of transcriptional regulation during bovine preimplantation development in vivo.Crossref | GoogleScholarGoogle Scholar | 19064908PubMed |

Kuijk, EW, Du Puy, L, van Tol, HTA, Oei, CHY, Haagsman, HP, Colenbrander, B, and Roelen, BAJ (2008). Differences in early lineage segregation between mammals. Developmental Dynamics 237, 918–927.
Differences in early lineage segregation between mammals.Crossref | GoogleScholarGoogle Scholar | 18330925PubMed |

Labrecque, R, and Sirard, M-A (2014). The study of mammalian oocyte competence by transcriptome analysis: progress and challenges. Molecular Human Reproduction 20, 103–116.
The study of mammalian oocyte competence by transcriptome analysis: progress and challenges.Crossref | GoogleScholarGoogle Scholar | 24233546PubMed |

Lamy, J, Gatien, J, Dubuisson, F, Nadal-Desbarats, L, Salvetti, P, Mermillod, P, and Saint-Dizier, M (2018). Metabolomic profiling of bovine oviductal fluid across the oestrous cycle using proton nuclear magnetic resonance spectroscopy. Reproduction, Fertility and Development 30, 1021–1028.
Metabolomic profiling of bovine oviductal fluid across the oestrous cycle using proton nuclear magnetic resonance spectroscopy.Crossref | GoogleScholarGoogle Scholar |

Leese, HJ (2002). Quiet please, do not disturb: a hypothesis of embryo metabolism and viability. Bioessays 24, 845–849.
Quiet please, do not disturb: a hypothesis of embryo metabolism and viability.Crossref | GoogleScholarGoogle Scholar | 12210521PubMed |

Leese, HJ, Guerif, F, Allgar, V, Brison, DR, Lundin, K, and Sturmey, RG (2016). Biological optimization, the Goldilocks principle, and how much is lagom in the preimplantation embryo. Molecular Reproduction and Development 83, 748–754.
Biological optimization, the Goldilocks principle, and how much is lagom in the preimplantation embryo.Crossref | GoogleScholarGoogle Scholar | 27465801PubMed |

Leibfried-Rutledge, ML, Critser, ES, and First, NL (1986). Effects of fetal calf serum and bovine serum albumin on in vitro maturation and fertilization of bovine and hamster cumulus-oocyte complexes. Biology of Reproduction 35, 850–857.
Effects of fetal calf serum and bovine serum albumin on in vitro maturation and fertilization of bovine and hamster cumulus-oocyte complexes.Crossref | GoogleScholarGoogle Scholar | 3814698PubMed |

Lequarre, AS, Grisart, B, Moreau, B, Schuurbiers, N, Massip, A, and Dessy, F (1997). Glucose metabolism during bovine preimplantation development: analysis of gene expression in single oocytes and embryos. Molecular Reproduction and Development 48, 216–226.
Glucose metabolism during bovine preimplantation development: analysis of gene expression in single oocytes and embryos.Crossref | GoogleScholarGoogle Scholar | 9291471PubMed |

Leroy, JLMR, Genicot, G, Donnay, I, and van Soom, A (2005). Evaluation of the lipid content in bovine oocytes and embryos with Nile red: a practical approach. Reproduction in Domestic Animals 40, 76–78.
Evaluation of the lipid content in bovine oocytes and embryos with Nile red: a practical approach.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 |

Lindner, GM, and Wright, RW (1983). Bovine embryo morphology and evaluation. Theriogenology 20, 407–416.
Bovine embryo morphology and evaluation.Crossref | GoogleScholarGoogle Scholar | 16725857PubMed |

Lonergan, P, and Fair, T (2016). Maturation of oocytes in vitro. Annual Review of Animal Biosciences 4, 255–268.
Maturation of oocytes in vitro.Crossref | GoogleScholarGoogle Scholar | 26566159PubMed |

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 |

Lonergan, P, Rizos, D, Gutiérrez-Adán, A, Fair, T, and Boland, MP (2003). Effect of culture environment on embryo quality and gene expression – experience from animal studies. Reproductive BioMedicine Online 7, 657–663.
Effect of culture environment on embryo quality and gene expression – experience from animal studies.Crossref | GoogleScholarGoogle Scholar | 14748964PubMed |

Lopera-Vasquez, R, Hamdi, M, Maillo, V, Lloreda, V, Coy, P, Gutierrez-Adan, A, Bermejo-Alvarez, P, and Rizos, D (2017). Effect of bovine oviductal fluid on development and quality of bovine embryos produced in vitro. Reproduction, Fertility and Development 29, 621–629.
Effect of bovine oviductal fluid on development and quality of bovine embryos produced in vitro.Crossref | GoogleScholarGoogle Scholar |

Lopera-Vásquez, R, Hamdi, M, Fernandez-Fuertes, B, Maillo, V, Beltrán-Breña, P, Calle, A, Redruello, A, López-Martín, S, Gutierrez-Adán, A, Yañez-Mó, M, Ramirez, MÁ, Rizos, D, and Sturmey, R (2016). Extracellular vesicles from BOEC in in vitro embryo development and quality. PLoS One 11, e0148083.
Extracellular vesicles from BOEC in in vitro embryo development and quality.Crossref | GoogleScholarGoogle Scholar | 26845570PubMed |

Lopes, AS, Greve, T, and Callesen, H (2007). Quantification of embryo quality by respirometry. Theriogenology 67, 21–31.
Quantification of embryo quality by respirometry.Crossref | GoogleScholarGoogle Scholar | 17109946PubMed |

Machado, MF, Caixeta, ES, Sudiman, J, Gilchrist, RB, Thompson, JG, Lima, PF, Price, CA, and Buratini, J (2015). Fibroblast growth factor 17 and bone morphogenetic protein 15 enhance cumulus expansion and improve quality of in vitro-produced embryos in cattle. Theriogenology 84, 390–398.
Fibroblast growth factor 17 and bone morphogenetic protein 15 enhance cumulus expansion and improve quality of in vitro-produced embryos in cattle.Crossref | GoogleScholarGoogle Scholar | 25930732PubMed |

Mangia, F, and Epstein, CJ (1975). Biochemical studies of growing mouse oocytes: preparation of oocytes and analysis of glucose-6-phosphate dehydrogenase and lactate dehydrogenase activities. Developmental Biology 45, 211–220.
Biochemical studies of growing mouse oocytes: preparation of oocytes and analysis of glucose-6-phosphate dehydrogenase and lactate dehydrogenase activities.Crossref | GoogleScholarGoogle Scholar | 1242707PubMed |

Masuda, Y, Hasebe, R, Kuromi, Y, Kobayashi, M, Iwamoto, M, Hishinuma, M, Ohbayashi, T, and Nishimura, R (2021a). Three-dimensional live imaging of bovine embryos by optical coherence tomography. The Journal of Reproduction and Development 67, 149–154.
Three-dimensional live imaging of bovine embryos by optical coherence tomography.Crossref | GoogleScholarGoogle Scholar | 33487605PubMed |

Masuda, Y, Hasebe, R, Kuromi, Y, Kobayashi, M, Urataki, K, Hishinuma, M, Ohbayashi, T, and Nishimura, R (2021b). Three-dimensional live imaging of bovine preimplantation embryos: a new method for IVF embryo evaluation. Frontiers in Veterinary Science 8, 639249.
Three-dimensional live imaging of bovine preimplantation embryos: a new method for IVF embryo evaluation.Crossref | GoogleScholarGoogle Scholar | 33981741PubMed |

Matos, FD, Rocha, JC, and Nogueira, MFG (2014). A method using artificial neural networks to morphologically assess mouse blastocyst quality. Journal of Animal Science and Technology 56, 15.
A method using artificial neural networks to morphologically assess mouse blastocyst quality.Crossref | GoogleScholarGoogle Scholar | 26290704PubMed |

McLennan, HJ, Saini, A, Dunning, KR, and Thompson, JG (2020). Oocyte and embryo evaluation by AI and multi-spectral auto-fluorescence imaging: livestock embryology needs to catch-up to clinical practice. Theriogenology 150, 255–262.
Oocyte and embryo evaluation by AI and multi-spectral auto-fluorescence imaging: livestock embryology needs to catch-up to clinical practice.Crossref | GoogleScholarGoogle Scholar | 32088032PubMed |

Milewski, R, and Ajduk, A (2017). Time-lapse imaging of cleavage divisions in embryo quality assessment. Reproduction 154, R37–R53.
Time-lapse imaging of cleavage divisions in embryo quality assessment.Crossref | GoogleScholarGoogle Scholar | 28408705PubMed |

Ming, H, Sun, J, Pasquariello, R, Gatenby, L, Herrick, JR, Yuan, Y, Pinto, CR, Bondioli, KR, Krisher, RL, and Jiang, Z (2021). The landscape of accessible chromatin in bovine oocytes and early embryos. Epigenetics 16, 300–312.
The landscape of accessible chromatin in bovine oocytes and early embryos.Crossref | GoogleScholarGoogle Scholar | 32663104PubMed |

Mondou, E, Dufort, I, Gohin, M, Fournier, E, and Sirard, M-A (2012). Analysis of microRNAs and their precursors in bovine early embryonic development. Molecular Human Reproduction 18, 425–434.
Analysis of microRNAs and their precursors in bovine early embryonic development.Crossref | GoogleScholarGoogle Scholar | 22491901PubMed |

Muñoz, M, Uyar, A, Correia, E, Díez, C, Fernandez-Gonzalez, A, Caamaño, JN, Martínez-Bello, D, Trigal, B, Humblot, P, Ponsart, C, Guyader-Joly, C, Carrocera, S, Martin, D, Marquant Le Guienne, B, Seli, E, and Gomez, E (2014). Prediction of pregnancy viability in bovine in vitro-produced embryos and recipient plasma with Fourier transform infrared spectroscopy. Journal of Dairy Science 97, 5497–5507.
Prediction of pregnancy viability in bovine in vitro-produced embryos and recipient plasma with Fourier transform infrared spectroscopy.Crossref | GoogleScholarGoogle Scholar | 24997663PubMed |

Nagano, M, Katagiri, S, and Takahashi, Y (2006). Relationship between bovine oocyte morphology and in vitro developmental potential. Zygote 14, 53–61.
Relationship between bovine oocyte morphology and in vitro developmental potential.Crossref | GoogleScholarGoogle Scholar | 16700976PubMed |

Nagyova, E (2012). Regulation of cumulus expansion and hyaluronan synthesis in porcine oocyte-cumulus complexes during in vitro maturation. Endocrine Regulations 46, 225–235.
Regulation of cumulus expansion and hyaluronan synthesis in porcine oocyte-cumulus complexes during in vitro maturation.Crossref | GoogleScholarGoogle Scholar | 23127506PubMed |

Nel-Themaat, L, and Nagy, ZP (2011). A review of the promises and pitfalls of oocyte and embryo metabolomics. Placenta 32, S257–S263.
A review of the promises and pitfalls of oocyte and embryo metabolomics.Crossref | GoogleScholarGoogle Scholar | 21703683PubMed |

Niemann, H, and Wrenzycki, C (2000). Alterations of expression of developmentally important genes in preimplantation bovine embryos by in vitro culture conditions: implications for subsequent development. Theriogenology 53, 21–34.
Alterations of expression of developmentally important genes in preimplantation bovine embryos by in vitro culture conditions: implications for subsequent development.Crossref | GoogleScholarGoogle Scholar | 10735059PubMed |

Nõmm, M, Porosk, R, Pärn, P, Kilk, K, Soomets, U, Kõks, S, and Jaakma, Ü (2019). In vitro culture and non-invasive metabolic profiling of single bovine embryos. Reproduction, Fertility and Development 31, 306–314.
In vitro culture and non-invasive metabolic profiling of single bovine embryos.Crossref | GoogleScholarGoogle Scholar |

Opiela, J, and Kątska-Książkiewicz, L (2013). The utility of Brilliant Cresyl Blue (BCB) staining of mammalian oocytes used for in vitro embryo production (IVP). Reproductive Biology 13, 177–183.
The utility of Brilliant Cresyl Blue (BCB) staining of mammalian oocytes used for in vitro embryo production (IVP).Crossref | GoogleScholarGoogle Scholar | 24011188PubMed |

Partridge, RJ, and Leese, HJ (1996). Consumption of amino acids by bovine preimplantation embryos. Reproduction, Fertility and Development 8, 945–950.
Consumption of amino acids by bovine preimplantation embryos.Crossref | GoogleScholarGoogle Scholar |

Pavlok, A, Lucas-Hahn, A, and Niemann, H (1992). Fertilization and developmental competence of bovine oocytes derived from different categories of antral follicles. Molecular Reproduction and Development 31, 63–67.
Fertilization and developmental competence of bovine oocytes derived from different categories of antral follicles.Crossref | GoogleScholarGoogle Scholar | 1562328PubMed |

Pawlak, P, Pers-Kamczyc, E, Renska, N, Kubickova, S, and Lechniak, D (2011). Disturbances of nuclear maturation in BCB positive oocytes collected from peri-pubertal gilts. Theriogenology 75, 832–840.
Disturbances of nuclear maturation in BCB positive oocytes collected from peri-pubertal gilts.Crossref | GoogleScholarGoogle Scholar | 21144571PubMed |

Pérez-Cerezales, S, Ramos-Ibeas, P, Acuña, OS, Avilés, M, Coy, P, Rizos, D, and Gutiérrez-Adán, A (2018). The oviduct: from sperm selection to the epigenetic landscape of the embryo. Biology of Reproduction 98, 262–276.
The oviduct: from sperm selection to the epigenetic landscape of the embryo.Crossref | GoogleScholarGoogle Scholar | 29228115PubMed |

Perkel, KJ, and Madan, P (2017). Spent culture medium analysis from individually cultured bovine embryos demonstrates metabolomic differences. Zygote 25, 662–674.
Spent culture medium analysis from individually cultured bovine embryos demonstrates metabolomic differences.Crossref | GoogleScholarGoogle Scholar | 29032784PubMed |

Pillai, VV, Weber, DM, Phinney, BS, Selvaraj, V, and Kues, WA (2017). Profiling of proteins secreted in the bovine oviduct reveals diverse functions of this luminal microenvironment. PLoS One 12, e0188105.
Profiling of proteins secreted in the bovine oviduct reveals diverse functions of this luminal microenvironment.Crossref | GoogleScholarGoogle Scholar | 29155854PubMed |

Ramanujam, N (2000). Fluorescence spectroscopy of neoplastic and non-neoplastic tissues. Neoplasia 2, 89–117.
Fluorescence spectroscopy of neoplastic and non-neoplastic tissues.Crossref | GoogleScholarGoogle Scholar | 10933071PubMed |

Raposo, G, and Stoorvogel, W (2013). Extracellular vesicles: exosomes, microvesicles, and friends. The Journal of Cell Biology 200, 373–383.
Extracellular vesicles: exosomes, microvesicles, and friends.Crossref | GoogleScholarGoogle Scholar | 23420871PubMed |

Reyes, JM, Chitwood, JL, and Ross, PJ (2015). RNA-Seq profiling of single bovine oocyte transcript abundance and its modulation by cytoplasmic polyadenylation. Molecular Reproduction and Development 82, 103–114.
RNA-Seq profiling of single bovine oocyte transcript abundance and its modulation by cytoplasmic polyadenylation.Crossref | GoogleScholarGoogle Scholar | 25560149PubMed |

Ribeiro, ES (2018). Symposium review: Lipids as regulators of conceptus development: implications for metabolic regulation of reproduction in dairy cattle. Journal of Dairy Science 101, 3630–3641.
Symposium review: Lipids as regulators of conceptus development: implications for metabolic regulation of reproduction in dairy cattle.Crossref | GoogleScholarGoogle Scholar | 29174158PubMed |

Richani, D, Dunning, KR, Thompson, JG, and Gilchrist, RB (2021). Metabolic co-dependence of the oocyte and cumulus cells: essential role in determining oocyte developmental competence. Human Reproduction Update 27, 27–47.
Metabolic co-dependence of the oocyte and cumulus cells: essential role in determining oocyte developmental competence.Crossref | GoogleScholarGoogle Scholar | 33020823PubMed |

Rieger, D, Loskutoff, NM, and Betteridge, KJ (1992). Developmentally related changes in the uptake and metabolism of glucose, glutamine and pyruvate by cattle embryos produced in vitro. Reproduction, Fertility and Development 4, 547–557.
Developmentally related changes in the uptake and metabolism of glucose, glutamine and pyruvate by cattle embryos produced in vitro.Crossref | GoogleScholarGoogle Scholar |

Riegler, MA, Stensen, MH, Witczak, O, Andersen, JM, Hicks, SA, Hammer, HL, Delbarre, E, Halvorsen, P, Yazidi, A, Holst, N, and Haugen, TB (2021). Artificial intelligence in the fertility clinic: status, pitfalls and possibilities. Human Reproduction 36, 2442.
Artificial intelligence in the fertility clinic: status, pitfalls and possibilities.Crossref | GoogleScholarGoogle Scholar | 34324672PubMed |

Rivera, RM (2020). Consequences of assisted reproductive techniques on the embryonic epigenome in cattle. Reproduction, Fertility and Development 32, 65–81.
Consequences of assisted reproductive techniques on the embryonic epigenome in cattle.Crossref | GoogleScholarGoogle Scholar |

Robertson I, Nelson RE (2009) Certification and identification of embryos. In ‘Manual of the International Embryo Transfer Society’. Chapter 9. pp. 86–105. (International Embryo Transfer Society)

Rocha, JC, Passalia, F, Matos, FD, Maserati, MP, Alves, MF, de Almeida, TG, Cardoso, BL, Basso, AC, and Nogueira, MFG (2016). Methods for assessing the quality of mammalian embryos: how far we are from the gold standard? JBRA Assisted Reproduction 20, 150–158.
Methods for assessing the quality of mammalian embryos: how far we are from the gold standard?Crossref | GoogleScholarGoogle Scholar | 27584609PubMed |

Rocha, JC, Passalia, FJ, Matos, FD, Takahashi, MB, de Ciniciato, DS, Maserati, MP, Alves, MF, de Almeida, TG, Cardoso, BL, Basso, AC, and Nogueira, MFG (2017a). A method based on artificial intelligence to fully automatize the evaluation of bovine blastocyst images. Scientific Reports 7, 7659.
A method based on artificial intelligence to fully automatize the evaluation of bovine blastocyst images.Crossref | GoogleScholarGoogle Scholar | 28794478PubMed |

Rocha, JC, Passalia, FJ, Matos, FD, Takahashi, MB, Maserati, MP, Alves, MF, de Almeida, TG, Cardoso, BL, Basso, AC, and Nogueira, MFG (2017b). Automatized image processing of bovine blastocysts produced in vitro for quantitative variable determination. Scientific Data 4, 344.
Automatized image processing of bovine blastocysts produced in vitro for quantitative variable determination.Crossref | GoogleScholarGoogle Scholar |

Rødgaard, T, Heegaard, PMH, and Callesen, H (2015). Non-invasive assessment of in-vitro embryo quality to improve transfer success. Reproductive Biomedicine Online 31, 585–592.
Non-invasive assessment of in-vitro embryo quality to improve transfer success.Crossref | GoogleScholarGoogle Scholar | 26380864PubMed |

Sánchez-Guijo, A, Blaschka, C, Hartmann, MF, Wrenzycki, C, and Wudy, SA (2016). Profiling of bile acids in bovine follicular fluid by fused-core-LC-MS/MS. The Journal of Steroid Biochemistry and Molecular Biology 162, 117–125.
Profiling of bile acids in bovine follicular fluid by fused-core-LC-MS/MS.Crossref | GoogleScholarGoogle Scholar | 26924583PubMed |

Scholkamy, TH, Darwish, SF, and Mahmoud, KGM (2015). Effect of vitrification by straw and cryotop on DNA integrity using comet assay with reference to brilliant cresyl blue exposure in buffalo oocytes. Alexandria Journal of Veterinary Sciences 46, 117.
Effect of vitrification by straw and cryotop on DNA integrity using comet assay with reference to brilliant cresyl blue exposure in buffalo oocytes.Crossref | GoogleScholarGoogle Scholar |

Sirard, M-A (2019). Folliculogenesis and acquisition of oocyte competence in cows. Animal Reproduction 16, 449–454.
Folliculogenesis and acquisition of oocyte competence in cows.Crossref | GoogleScholarGoogle Scholar | 32435288PubMed |

Sirard, M-A, 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 |

Sohel, MMH, Hoelker, M, Noferesti, SS, Salilew-Wondim, D, Tholen, E, Looft, C, Rings, F, Uddin, MJ, Spencer, TE, Schellander, K, Tesfaye, D, and Busson, P (2013). Exosomal and non-exosomal transport of extra-cellular microRNAs in follicular fluid: implications for bovine oocyte developmental competence. PLoS One 8, e78505.
Exosomal and non-exosomal transport of extra-cellular microRNAs in follicular fluid: implications for bovine oocyte developmental competence.Crossref | GoogleScholarGoogle Scholar |

Sturmey, RG, Bermejo-Alvarez, P, Gutierrez-Adan, A, Rizos, D, Leese, HJ, and Lonergan, P (2010). Amino acid metabolism of bovine blastocysts: a biomarker of sex and viability. Molecular Reproduction and Development 77, 285–296.
Amino acid metabolism of bovine blastocysts: a biomarker of sex and viability.Crossref | GoogleScholarGoogle Scholar | 20058302PubMed |

Sudano, MJ, Santos, VG, Tata, A, Ferreira, CR, Paschoal, DM, Machado, R, Buratini, J, Eberlin, MN, and Landim-Alvarenga, FDC (2012). Phosphatidylcholine and sphingomyelin profiles vary in Bos taurus indicus and Bos taurus taurusin vitro- and in vivo-produced blastocysts. Biology of Reproduction 87, 130.
Phosphatidylcholine and sphingomyelin profiles vary in Bos taurus indicus and Bos taurus taurusin vitro- and in vivo-produced blastocysts.Crossref | GoogleScholarGoogle Scholar | 23053436PubMed |

Sugimura, S, Akai, T, Hashiyada, Y, Somfai, T, Inaba, Y, Hirayama, M, Yamanouchi, T, Matsuda, H, Kobayashi, S, Aikawa, Y, Ohtake, M, Kobayashi, E, Konishi, K, Imai, K, and Tian, XC (2012). Promising system for selecting healthy in vitro-fertilized embryos in cattle. PLoS One 7, e36627.
Promising system for selecting healthy in vitro-fertilized embryos in cattle.Crossref | GoogleScholarGoogle Scholar | 22590579PubMed |

Sugimura, S, Akai, T, and Imai, K (2017). Selection of viable in vitro-fertilized bovine embryos using time-lapse monitoring in microwell culture dishes. The Journal of Reproduction and Development 63, 353–357.
Selection of viable in vitro-fertilized bovine embryos using time-lapse monitoring in microwell culture dishes.Crossref | GoogleScholarGoogle Scholar | 28552887PubMed |

Sugimura, S, Akai, T, Somfai, T, Hirayama, M, Aikawa, Y, Ohtake, M, Hattori, H, Kobayashi, S, Hashiyada, Y, Konishi, K, and Imai, K (2010). Time-lapse cinematography-compatible polystyrene-based microwell culture system: a novel tool for tracking the development of individual bovine embryos. Biology of Reproduction 83, 970–978.
Time-lapse cinematography-compatible polystyrene-based microwell culture system: a novel tool for tracking the development of individual bovine embryos.Crossref | GoogleScholarGoogle Scholar | 20739661PubMed |

Sugiyama, D, Teshima, Y, Yamanaka, K, Briones-Nagata, MP, Maeki, M, Yamashita, K, Takahashi, M, and Miyazaki, M (2014). Simple density-based particle separation in a microfluidic chip. Analytical Methods 6, 308–311.
Simple density-based particle separation in a microfluidic chip.Crossref | GoogleScholarGoogle Scholar |

Sutton-McDowall, ML, Gilchrist, RB, and Thompson, JG (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 | 15333782PubMed |

Sutton-McDowall, ML, Gosnell, M, Anwer, AG, White, M, Purdey, M, Abell, AD, Goldys, EM, and Thompson, JG (2017). Hyperspectral microscopy can detect metabolic heterogeneity within bovine post-compaction embryos incubated under two oxygen concentrations (7% versus 20%). Human Reproduction 32, 2016–2025.
Hyperspectral microscopy can detect metabolic heterogeneity within bovine post-compaction embryos incubated under two oxygen concentrations (7% versus 20%).Crossref | GoogleScholarGoogle Scholar | 28938734PubMed |

Sutton-McDowall, ML, Mottershead, DG, Gardner, DK, Gilchrist, RB, and Thompson, JG (2012). Metabolic differences in bovine cumulus-oocyte complexes matured in vitro in the presence or absence of follicle-stimulating hormone and bone morphogenetic protein 15. Biology of Reproduction 87, 87.
Metabolic differences in bovine cumulus-oocyte complexes matured in vitro in the presence or absence of follicle-stimulating hormone and bone morphogenetic protein 15.Crossref | GoogleScholarGoogle Scholar | 22895854PubMed |

Sutton-McDowall, ML, Purdey, M, Brown, HM, Abell, AD, Mottershead, DG, Cetica, PD, Dalvit, GC, Goldys, EM, Gilchrist, RB, Gardner, DK, and Thompson, JG (2015). Redox and anti-oxidant state within cattle oocytes following in vitro maturation with bone morphogenetic protein 15 and follicle stimulating hormone. Molecular Reproduction and Development 82, 281–294.
Redox and anti-oxidant state within cattle oocytes following in vitro maturation with bone morphogenetic protein 15 and follicle stimulating hormone.Crossref | GoogleScholarGoogle Scholar | 25721374PubMed |

Takahashi, H, Kato, K, Ueyama, K, Kobayashi, M, Baik, G, Yukawa, Y, Suehiro, J-I, and Matsunaga, YT (2017). Visualizing dynamics of angiogenic sprouting from a three-dimensional microvasculature model using stage-top optical coherence tomography. Scientific Reports 7, 42426.
Visualizing dynamics of angiogenic sprouting from a three-dimensional microvasculature model using stage-top optical coherence tomography.Crossref | GoogleScholarGoogle Scholar | 28186184PubMed |

Takahashi, MB, Rocha, JC, and Núñez, EGF (2016). Optimization of artificial neural network by genetic algorithm for describing viral production from uniform design data. Process Biochemistry 51, 422–430.
Optimization of artificial neural network by genetic algorithm for describing viral production from uniform design data.Crossref | GoogleScholarGoogle Scholar |

Tesfaye, D, Worku, D, Rings, F, Phatsara, C, Tholen, E, Schellander, K, and Hoelker, M (2009). Identification and expression profiling of microRNAs during bovine oocyte maturation using heterologous approach. Molecular Reproduction and Development 76, 665–677.
Identification and expression profiling of microRNAs during bovine oocyte maturation using heterologous approach.Crossref | GoogleScholarGoogle Scholar | 19170227PubMed |

Thélie, A, Papillier, P, Pennetier, S, Perreau, C, Traverso, JM, Uzbekova, S, Mermillod, P, Joly, C, Humblot, P, and Dalbiès-Tran, R (2007). Differential regulation of abundance and deadenylation of maternal transcripts during bovine oocyte maturation in vitro and in vivo. BMC Developmental Biology 7, 125.
Differential regulation of abundance and deadenylation of maternal transcripts during bovine oocyte maturation in vitro and in vivo.Crossref | GoogleScholarGoogle Scholar | 17988387PubMed |

Thompson, JG, Brown, HM, and Sutton-McDowall, ML (2016). Measuring embryo metabolism to predict embryo quality. Reproduction, Fertility and Development 28, 41–50.
Measuring embryo metabolism to predict embryo quality.Crossref | GoogleScholarGoogle Scholar |

Valadi, H, Ekström, K, Bossios, A, Sjöstrand, M, Lee, JJ, and Lötvall, JO (2007). Exosome-mediated transfer of mRNAs and microRNAs is a novel mechanism of genetic exchange between cells. Nature Cell Biology 9, 654–659.
Exosome-mediated transfer of mRNAs and microRNAs is a novel mechanism of genetic exchange between cells.Crossref | GoogleScholarGoogle Scholar | 17486113PubMed |

van Soom, A, Boerjan, M, Ysebaert, MT, and de Kruif, A (1996). Cell allocation to the inner cell mass and the trophectoderm in bovine embryos cultured in two different media. Molecular Reproduction and Development 45, 171–182.
Cell allocation to the inner cell mass and the trophectoderm in bovine embryos cultured in two different media.Crossref | GoogleScholarGoogle Scholar | 8914075PubMed |

van Soom, A, Ysebaert, MT, and de Kruif, A (1997). Relationship between timing of development, morula morphology, and cell allocation to inner cell mass and trophectoderm in in vitro-produced bovine embryos. Molecular Reproduction and Development 47, 47–56.
Relationship between timing of development, morula morphology, and cell allocation to inner cell mass and trophectoderm in in vitro-produced bovine embryos.Crossref | GoogleScholarGoogle Scholar | 9110314PubMed |

Virant-Klun, I, and Krijgsveld, J (2014). Proteomes of animal oocytes: what can we learn for human oocytes in the in vitro fertilization programme? BioMed Research International 2014, 856907.
Proteomes of animal oocytes: what can we learn for human oocytes in the in vitro fertilization programme?Crossref | GoogleScholarGoogle Scholar | 24804254PubMed |

Wang, Q, and Sun, Q-Y (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, Z, Gerstein, M, and Snyder, M (2009). RNA-Seq: a revolutionary tool for transcriptomics. Nature Reviews. Genetics 10, 57–63.
RNA-Seq: a revolutionary tool for transcriptomics.Crossref | GoogleScholarGoogle Scholar | 19015660PubMed |

Wathes, DC, Clempson, AM, and Pollott, GE (2012). 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 |

Wrenzycki, C (2018). Gene expression analysis and in vitro production procedures for bovine preimplantation embryos: past highlights, present concepts and future prospects. Reproduction in Domestic Animals 53, 14–19.
Gene expression analysis and in vitro production procedures for bovine preimplantation embryos: past highlights, present concepts and future prospects.Crossref | GoogleScholarGoogle Scholar | 30238652PubMed |

Wrenzycki, C, Herrmann, D, Carnwath, JW, and Niemann, H (1996). Expression of the gap junction gene connexin43 (Cx43) in preimplantation bovine embryos derived in vitro or in vivo. Journal of Reproduction and Fertility 108, 17–24.
Expression of the gap junction gene connexin43 (Cx43) in preimplantation bovine embryos derived in vitro or in vivo.Crossref | GoogleScholarGoogle Scholar | 8958823PubMed |

Wrenzycki, C, Herrmann, D, Carnwath, JW, and Niemann, H (1999). Alterations in the relative abundance of gene transcripts in preimplantation bovine embryos cultured in medium supplemented with either serum or PVA. Molecular Reproduction Development 53, 8–18.
Alterations in the relative abundance of gene transcripts in preimplantation bovine embryos cultured in medium supplemented with either serum or PVA.Crossref | GoogleScholarGoogle Scholar | 10230812PubMed |

Wrenzycki, C, Herrmann, D, Lucas-Hahn, A, Korsawe, K, Lemme, E, and Niemann, H (2005). Messenger RNA expression patterns in bovine embryos derived from in vitro procedures and their implications for development. Reproduction, Fertility and Development 17, 23–35.
Messenger RNA expression patterns in bovine embryos derived from in vitro procedures and their implications for development.Crossref | GoogleScholarGoogle Scholar |

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 | 17524469PubMed |

Wrenzycki, C, and Stinshoff, H (2013). Maturation environment and impact on subsequent developmental competence of bovine oocytes. Reproduction in Domestic Animals 48, 38–43.
Maturation environment and impact on subsequent developmental competence of bovine oocytes.Crossref | GoogleScholarGoogle Scholar | 23962213PubMed |

Wydooghe, E, Vandaele, L, Beek, J, Favoreel, H, Heindryckx, B, De Sutter, P, and Van Soom, A (2011). Differential apoptotic staining of mammalian blastocysts based on double immunofluorescent CDX2 and active caspase-3 staining. Analytical Biochemistry 416, 228–230.
Differential apoptotic staining of mammalian blastocysts based on double immunofluorescent CDX2 and active caspase-3 staining.Crossref | GoogleScholarGoogle Scholar | 21684250PubMed |

Yao, T, Suzuki, R, Furuta, N, Suzuki, Y, Kabe, K, Tokoro, M, Sugawara, A, Yajima, A, Nagasawa, T, Matoba, S, Yamagata, K, and Sugimura, S (2018). Live-cell imaging of nuclear-chromosomal dynamics in bovine in vitro fertilised embryos. Scientific Reports 8, 7460.
Live-cell imaging of nuclear-chromosomal dynamics in bovine in vitro fertilised embryos.Crossref | GoogleScholarGoogle Scholar | 29748644PubMed |

Yotsushima, K, Shimizu, M, Kon, H, and Izaike, Y (2007). A simple method for selection of cumulus-oocyte complexes from bovine ovaries by sedimentation with percoll. The Journal of Reproduction and Development 53, 971–976.
A simple method for selection of cumulus-oocyte complexes from bovine ovaries by sedimentation with percoll.Crossref | GoogleScholarGoogle Scholar | 17519521PubMed |

Zuccotti, M, Merico, V, Cecconi, S, Redi, CA, and Garagna, S (2011). What does it take to make a developmentally competent mammalian egg? Human Reproduction Update 17, 525–540.
What does it take to make a developmentally competent mammalian egg?Crossref | GoogleScholarGoogle Scholar | 21444328PubMed |