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

Peroxisome proliferator-activated receptor delta-PPARδ agonist (L-165041) enhances bovine embryo survival and post vitrification viability

Jesús Alfonso Sánchez Viafara https://orcid.org/0000-0001-7527-3300 A B * , Gisvani Lopes de Vasconcelos C , Renata Maculan D , Nadja Gomes Alves C , Marcos Brandao Dias Ferreira E , Mateus José Sudano https://orcid.org/0000-0002-7699-4449 F , Gisele Zoccal Mingoti https://orcid.org/0000-0002-3059-4458 G , Giovana Barros Nunes G , Renato Ribeiro de Lima H , Roberti Martins Drumond I , Raphael Nunes dos Santos I , Marcos Nogueira Eberlin J , Fernanda Negrão J , Jasmin K , Mariana Aragão M. Donato L , Christina A. Peixoto L and José Camisão de Souza C *
+ Author Affiliations
- Author Affiliations

A Departamento de Medicina Veterinária, Universidade Federal de Lavras, Lavras, Minas Gerais, Brasil.

B Universidad de Santander, Facultad de Ciencias Agrícolas y Veterinarias, Valledupar, Colombia.

C Departamento de Zootecnia, Universidade Federal de Lavras, Lavras, Minas Gerais, Brasil.

D Instituto Federal do Sul de Minas, Machado, Minas Gerais, Brasil.

E EPAMIG, Belo Horizonte, Minas Gerais, Brasil.

F Universidade Federal do ABC, Santo André, São Paulo, Brasil.

G Escola de Medicina Veterinária, Laboratório de Fisiologia da Reprodução, Universidade Estadual Paulista, Campus Araçatuba, São Paulo, Brasil.

H Departamento de Estatística, Universidade Federal de Lavras, Lavras, Minas Gerais, Brasil.

I Cenatte Embriões, Pedro Leopoldo, Minas Gerais, Brasil.

J Universidade Estadual de Campinas, Laboratório ThoMSon de Espectrometria de Massas, Campinas, São Paulo, Brasil.

K NUMPEX-Bio, Universidade Federal do Rio de Janeiro, Campus Duque de Caxias, Rio de Janeiro, Brasil.

L Centro de Pesquisas Aggeu Magalhães, Fiocruz/Pernambuco, Brasil.


Handling Editor: Ye Yuan

Reproduction, Fertility and Development 34(9) 658-668 https://doi.org/10.1071/RD21245
Published online: 26 April 2022

© 2022 The Author(s) (or their employer(s)). Published by CSIRO Publishing

Abstract

The effect of L-165041 (PPARδ-agonist) on decreasing apoptosis and intracellular lipid content was assessed in fresh and vitrified–warmed in vitro-produced bovine embryos. It was hypothesised that the addition of L-165041 to the culture medium enhances development and cryopreservation. Oocytes were allocated to one of two treatments: control-standard culture medium, or L-165041 added to the medium on day 1 with no media change. Ultrastructure, cleavage, and blastocyst rates were evaluated in fresh, and in post-vitrification cultured embryos by optical and electronic microscopy. A subset of fresh embryos were fixed for TUNEL assay and for Sudan-Black-B histochemical staining. Vitrified–warmed embryos were assessed using MALDI-MS technique. Cleavage and blastocyst rates (control 49.4 ± 5.2, L-165041 51.8 ± 4.3) were not influenced by L-165041. The proportion of inner cell mass cells (ICM) was higher in fresh embryos, and the rate of total and ICM apoptosis was lower in L-165041. In warmed-embryos, total and ICM apoptosis was lower in L-165041. The overall hatching rate was higher in L-165041 (66.62 ± 2.83% vs 53.19 ± 2.90%). There was less lipid accumulation in fresh L-165041-embryos. In conclusion, the use of L-165041 is recommended to improve the viability of in vitro-derived bovine embryos.

Keywords: bovine, cryopreservation, culture medium, development, embryo apoptosis, embryos, lipid, vitrification.


References

Abe, H, Yamashita, S, Itoh, T, Satoh, T, and Hoshi, H (1999). Ultrastructure of bovine embryos developed from in vitro–matured and –fertilized oocytes: comparative morphological evaluation of embryos cultured either in serum-free medium or in serum-supplemented medium. Molecular Reproduction and Development 53, 325–335.
Ultrastructure of bovine embryos developed from in vitro–matured and –fertilized oocytes: comparative morphological evaluation of embryos cultured either in serum-free medium or in serum-supplemented medium.Crossref | GoogleScholarGoogle Scholar | 10369393PubMed |

Al Darwich, A, Perreau, C, Petit, MH, Papillier, P, Dupont, J, Guillaume, D, Mermillod, P, and Guignot, F (2010). Effect of PUFA on embryo cryoresistance, gene expression and AMPKα phosphorylation in IVF-derived bovine embryos. Prostaglandins & Other Lipid Mediators 93, 30–36.
Effect of PUFA on embryo cryoresistance, gene expression and AMPKα phosphorylation in IVF-derived bovine embryos.Crossref | GoogleScholarGoogle Scholar |

Baust, JM, Van Buskirk, R, and Baust, JG (2000). Cell viability improves following inhibition of cryopreservation-induced apoptosis. In Vitro Cellular & Developmental Biology – Animal 36, 262–270.
Cell viability improves following inhibition of cryopreservation-induced apoptosis.Crossref | GoogleScholarGoogle Scholar |

Behr, B, Gebhardt, J, Lyon, J, and Milki, AA (2002). Factors relating to a successful cryopreserved blastocyst transfer program. Fertility and Sterility 77, 697–699.
Factors relating to a successful cryopreserved blastocyst transfer program.Crossref | GoogleScholarGoogle Scholar | 11937118PubMed |

Berger, J, Leibowitz, MD, Doebber, TW, Elbrecht, A, Zhang, B, Zhou, G, Biswas, C, Cullinan, CA, Hayes, NS, Li, Y, Tanen, M, Ventre, J, Wu, MS, Berger, GD, Mosley, R, Marquis, R, Santini, C, Sahoo, SP, Tolman, RL, Smith, RG, and Moller, DE (1999). Novel peroxisome proliferator-activated receptor (PPAR) gamma and PPAR delta ligands produce distinct biological effects. Journal of Biological Chemistry 274, 6718–6725.
Novel peroxisome proliferator-activated receptor (PPAR) gamma and PPAR delta ligands produce distinct biological effects.Crossref | GoogleScholarGoogle Scholar |

Betts, DH, and King, WA (2001). Genetic regulation of embryo death and senescence. Theriogenology 55, 171–191.
Genetic regulation of embryo death and senescence.Crossref | GoogleScholarGoogle Scholar | 11198081PubMed |

Bionaz, M, Chen, S, Khan, MJ, and Loor, JJ (2013). Functional role of PPARs in ruminants: potential targets for fine-tuning metabolism during growth and lactation. PPAR Research 2013, 684159.
Functional role of PPARs in ruminants: potential targets for fine-tuning metabolism during growth and lactation.Crossref | GoogleScholarGoogle Scholar | 23737762PubMed |

Blanquart, C, Barbier, O, Fruchart, JC, Staels, B, and Glineur, C (2003). Peroxisome proliferator-activated receptors: regulation of transcriptional activities and roles in inflammation. The Journal of Steroid Biochemistry and Molecular Biology 85, 267–273.
Peroxisome proliferator-activated receptors: regulation of transcriptional activities and roles in inflammation.Crossref | GoogleScholarGoogle Scholar | 12943712PubMed |

Brison, DR, and Schultz, RM (1997). Apoptosis during mouse blastocyst formation: evidence for a role for survival factors including transforming growth factor alpha. Biology of Reproduction 56, 1088–1096.
Apoptosis during mouse blastocyst formation: evidence for a role for survival factors including transforming growth factor alpha.Crossref | GoogleScholarGoogle Scholar | 9160705PubMed |

Byrne, AT, Southgate, J, Brison, DR, and Leese, HJ (1999). Analysis of apoptosis in the preimplantation bovine embryo using TUNEL. Reproduction 117, 97–105.
Analysis of apoptosis in the preimplantation bovine embryo using TUNEL.Crossref | GoogleScholarGoogle Scholar |

Carroll, J, Depypere, H, and Matthews, CD (1990). Freeze–thaw-induced changes of the zona pellucida explains decreased rates of fertilization in frozen–thawed mouse oocytes. Journal of Reproduction and Fertililty 90, 547–553.
Freeze–thaw-induced changes of the zona pellucida explains decreased rates of fertilization in frozen–thawed mouse oocytes.Crossref | GoogleScholarGoogle Scholar |

Chauhan, S, Canet, M, Huang, J-C, Dunn, R, Grunert, G, and Wun, W-S (2013). Peroxisome proliferator activated receptor δ (PPAR δ) activation enhances the development of cryopreserved embryos. Fertility and Sterility 100, S187.
Peroxisome proliferator activated receptor δ (PPAR δ) activation enhances the development of cryopreserved embryos.Crossref | GoogleScholarGoogle Scholar |

Crosier, AE, Farin, PW, Dykstra, MJ, Alexander, JE, and Farin, CE (2001). Ultrastructural morphometry of bovine blastocysts produced in vivo or in vitro. Biology of Reproduction 64, 1375–1385.
Ultrastructural morphometry of bovine blastocysts produced in vivo or in vitro.Crossref | GoogleScholarGoogle Scholar | 11319141PubMed |

Datta, SR, Dudek, H, Tao, X, Masters, S, Fu, H, Gotoh, Y, and Greenberg, ME (1997). Akt phosphorylation of BAD couples survival signals to the cell-intrinsic death machinery. Cell 91, 231–241.
Akt phosphorylation of BAD couples survival signals to the cell-intrinsic death machinery.Crossref | GoogleScholarGoogle Scholar | 9346240PubMed |

Desvergne, B, and Wahli, W (1999). Peroxisome proliferator-activated receptors: nuclear control of metabolism. Endocrine Reviews 20, 649–688.
Peroxisome proliferator-activated receptors: nuclear control of metabolism.Crossref | GoogleScholarGoogle Scholar | 10529898PubMed |

Dobrinsky, JR (1996). Cellular approach to cryopreservation of embryos. Theriogenology 45, 17–26.
Cellular approach to cryopreservation of embryos.Crossref | GoogleScholarGoogle Scholar |

Ferré, LB, Kjelland, ME, Strøbech, LB, Hyttel, P, Mermillod, P, and Ross, PJ (2020). Review: Recent advances in bovine in vitro embryo production: reproductive biotechnology history and methods. Animal 14, 991–1004.
Review: Recent advances in bovine in vitro embryo production: reproductive biotechnology history and methods.Crossref | GoogleScholarGoogle Scholar | 31760966PubMed |

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

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 |

Grunert, G, Dunn, R, Huang, J-C, Schenk, L, Chauhan, S, and Wun, W-S (2014). Prostacyclin agonist (L165041) enhances embryos survival after vitrification and warming. Fertility and Sterility 101, e15–e16.
Prostacyclin agonist (L165041) enhances embryos survival after vitrification and warming.Crossref | GoogleScholarGoogle Scholar |

Hao, CM, Redha, R, Morrow, J, and Breyer, MD (2002). Peroxisome proliferator-activated receptor δ activation promotes cell survival following hypertonic stress. Journal of Biological Chemistry 277, 21341–21345.
Peroxisome proliferator-activated receptor δ activation promotes cell survival following hypertonic stress.Crossref | GoogleScholarGoogle Scholar |

Hardy, K (1997). Cell death in the mammalian blastocyst. Molecular Human Reproduction 3, 919–925.
Cell death in the mammalian blastocyst.Crossref | GoogleScholarGoogle Scholar | 9395266PubMed |

Hardy, K, Handyside, AH, and Winston, RM (1989). The human blastocyst: cell number, death and allocation during late preimplantation development in vitro. Development 107, 597–604.
The human blastocyst: cell number, death and allocation during late preimplantation development in vitro.Crossref | GoogleScholarGoogle Scholar | 2612378PubMed |

Heng, BC, Clement, MV, and Cao, T (2007). Caspase inhibitor Z-VAD-FMK enhances the freeze-thaw survival rate of human embryonic stem cells. Bioscience Reports 27, 257–264.
Caspase inhibitor Z-VAD-FMK enhances the freeze-thaw survival rate of human embryonic stem cells.Crossref | GoogleScholarGoogle Scholar | 17594512PubMed |

Holm, P, Booth, PJ, Schmidt, MH, Greve, T, and Callesen, H (1999). High bovine blastocyst development in a static in vitro production system using sofaa medium supplemented with sodium citrate and myo-inositol with or without serum-proteins. Theriogenology 52, 683–700.
High bovine blastocyst development in a static in vitro production system using sofaa medium supplemented with sodium citrate and myo-inositol with or without serum-proteins.Crossref | GoogleScholarGoogle Scholar | 10734366PubMed |

Huang, J-C, Goldsby, JS, and Wun, W-SA (2004). Prostacyclin enhances the implantation and live birth potentials of mouse embryos. Human Reproduction 19, 1856–1860.
Prostacyclin enhances the implantation and live birth potentials of mouse embryos.Crossref | GoogleScholarGoogle Scholar | 15205402PubMed |

Huang, J-C, Wun, W-SA, Goldsby, JS, Wun, IC, Falconi, SM, and Wu, KK (2003). Prostacyclin enhances embryo hatching but not sperm motility. Human Reproduction 18, 2582–2589.
Prostacyclin enhances embryo hatching but not sperm motility.Crossref | GoogleScholarGoogle Scholar | 14645174PubMed |

Huang, J-C, Wun, W-SA, Goldsby, JS, Wun, IC, Noorhasan, D, and Wu, KK (2007). Stimulation of embryo hatching and implantation by prostacyclin and peroxisome proliferator-activated receptor δ activation: implication in IVF. Human Reproduction 22, 807–814.
Stimulation of embryo hatching and implantation by prostacyclin and peroxisome proliferator-activated receptor δ activation: implication in IVF.Crossref | GoogleScholarGoogle Scholar | 17114194PubMed |

Imai, K, Matoba, S, Dochi, O, and Shimohira, I (2002). Different factors affect developmental competence and cryotolerance in in vitro produced bovine embryo. Journal of Veterinary Medical Science 64, 887–891.
Different factors affect developmental competence and cryotolerance in in vitro produced bovine embryo.Crossref | GoogleScholarGoogle Scholar |

Iwasaki, S, Yoshiba, N, Ushijima, H, Watanabe, S, and Nakahara, T (1990). Morphology and proportion of inner cell mass of bovine blastocysts fertilized in vitro and in vivo. Journal of Reproduction and Fertility 90, 279–284.
Morphology and proportion of inner cell mass of bovine blastocysts fertilized in vitro and in vivo.Crossref | GoogleScholarGoogle Scholar | 2231548PubMed |

Kim, HJ, Park, SB, Yang, JB, Choi, YB, and Lee, KH (2017). Effects of laser-assisted hatching and exposure time to vitrification solution on mouse embryo development. Clinical Experimental Reproductive Medicine 44, 193–200.
Effects of laser-assisted hatching and exposure time to vitrification solution on mouse embryo development.Crossref | GoogleScholarGoogle Scholar | 29376016PubMed |

Kliewer, SA, Forman, BM, Blumberg, B, Ong, ES, Borgmeyer, U, Mangelsdorf, DJ, Umesono, K, and Evans, RM (1994). Differential expression and activation of a family of murine peroxisome proliferator-activated receptors. Proceedings of the National Academy of Sciences of the United States of America 91, 7355–7359.
Differential expression and activation of a family of murine peroxisome proliferator-activated receptors.Crossref | GoogleScholarGoogle Scholar | 8041794PubMed |

Knijn, HM (2003). Consequences of in vivo development and subsequent culture on apoptosis, cell number, and blastocyst formation in bovine embryos. Biology of Reproduction 69, 1371–1378.
Consequences of in vivo development and subsequent culture on apoptosis, cell number, and blastocyst formation in bovine embryos.Crossref | GoogleScholarGoogle Scholar | 12826584PubMed |

Koo, D-B, Kang, Y-K, Choi, Y-H, Park, JS, Kim, H-N, Oh, KB, Son, D-S, Park, H, Lee, K-K, and Han, Y-M (2002). Aberrant allocations of inner cell mass and trophectoderm cells in bovine nuclear transfer blastocysts. Biology of Reproduction 67, 487–492.
Aberrant allocations of inner cell mass and trophectoderm cells in bovine nuclear transfer blastocysts.Crossref | GoogleScholarGoogle Scholar | 12135886PubMed |

Korhonen, K, Kananen, K, Ketoja, E, Matomäki, J, Halmekytö, M, and Peippo, J (2010). Effects of serum-free in vitro maturation of bovine oocytes on subsequent embryo development and cell allocation in two developmental stages of day 7 blastocysts. Reproduction in Domestic Animals 45, 42–49.
Effects of serum-free in vitro maturation of bovine oocytes on subsequent embryo development and cell allocation in two developmental stages of day 7 blastocysts.Crossref | GoogleScholarGoogle Scholar | 19144013PubMed |

Kota, BP, Huang, TH-W, and Roufogalis, BD (2005). An overview on biological mechanisms of PPARs. Pharmacological Research 51, 85–94.
An overview on biological mechanisms of PPARs.Crossref | GoogleScholarGoogle Scholar | 15629253PubMed |

Leão, BCS, Rocha-Frigoni, NAS, Cabral, EC, Franco, MF, Ferreira, CR, Eberlin, MN, Filgueiras, PR, and Mingoti, GZ (2015). Membrane lipid profile monitored by mass spectrometry detected differences between fresh and vitrified in vitro-produced bovine embryos. Zygote 23, 732–741.
Membrane lipid profile monitored by mass spectrometry detected differences between fresh and vitrified in vitro-produced bovine embryos.Crossref | GoogleScholarGoogle Scholar |

Lim, H, Gupta, RA, Ma, W-g, Paria, BC, Moller, DE, Morrow, JD, DuBois, RN, Trzaskos, JM, and Dey, SK (1999). Cyclo-oxygenase-2-derived prostacyclin mediates embryo implantation in the mouse via PPARδ. Genes & Development 13, 1561–1574.
Cyclo-oxygenase-2-derived prostacyclin mediates embryo implantation in the mouse via PPARδ.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, Rizos, D, Gutierrez-Adan, A, Fair, T, and Boland, MP (2003). Oocyte and embryo quality: effect of origin, culture conditions and gene expression patterns. Reproduction in Domestic Animals 38, 259–267.
Oocyte and embryo quality: effect of origin, culture conditions and gene expression patterns.Crossref | GoogleScholarGoogle Scholar | 12887565PubMed |

Márquez-Alvarado, YC, Galina, CS, Castilla, B, León, H, and Moreno-Mendoza, N (2004). Evidence of damage in cryopreserved and fresh bovine embryos using the tunel technique. Reproduction in Domestic Animals 39, 141–145.
Evidence of damage in cryopreserved and fresh bovine embryos using the tunel technique.Crossref | GoogleScholarGoogle Scholar | 15182289PubMed |

Matson, PL, Graefling, J, Junk, SM, Yovich, JL, and Edirisinghe, WR (1997). Cryopreservation of oocytes and embryos: use of a mouse model to investigate effects upon zona hardness and formulate treatment strategies in an in-vitro fertilization programme. Human Reproduction 12, 1550–1553.
Cryopreservation of oocytes and embryos: use of a mouse model to investigate effects upon zona hardness and formulate treatment strategies in an in-vitro fertilization programme.Crossref | GoogleScholarGoogle Scholar | 9262294PubMed |

Meneghel, M, Dall’Acqua, PC, Ambrogi, M, Leão, BCS, Rocha-Frigoni, NAS, and Mingoti, GZ (2017). Lipid content and cryotolerance of in vitro-produced bovine embryos treated with forskolin before vitrification. Pesquisa Veterinária Brasileira 37, 395–400.
Lipid content and cryotolerance of in vitro-produced bovine embryos treated with forskolin before vitrification.Crossref | GoogleScholarGoogle Scholar |

Mohan, M, Malayer, JR, Geisert, RD, and Morgan, GL (2002). Expression patterns of retinoid X receptors, retinaldehyde dehydrogenase, and peroxisome proliferator activated receptor gamma in bovine preattachment embryos. Biology of Reproduction 66, 692–700.
Expression patterns of retinoid X receptors, retinaldehyde dehydrogenase, and peroxisome proliferator activated receptor gamma in bovine preattachment embryos.Crossref | GoogleScholarGoogle Scholar | 11870076PubMed |

Montag, M, Koll, B, Holmes, P, and van der Ven, H (2000). Significance of the number of embryonic cells and the state of the zona pellucida for hatching of mouse blastocysts in vitro versus in vivo. Biology of Reproduction 62, 1738–1744.
Significance of the number of embryonic cells and the state of the zona pellucida for hatching of mouse blastocysts in vitro versus in vivo.Crossref | GoogleScholarGoogle Scholar | 10819778PubMed |

Pakrasi, PL, and Jain, AK (2007). Evaluation of cyclooxygenase 2 derived endogenous prostacyclin in mouse preimplantation embryo development in vitro. Life Sciences 80, 1503–1507.
Evaluation of cyclooxygenase 2 derived endogenous prostacyclin in mouse preimplantation embryo development in vitro.Crossref | GoogleScholarGoogle Scholar | 17328923PubMed |

Park, SY, Kim, EY, Cui, XS, Tae, JC, Lee, WD, Kim, NH, Park, SP, and Lim, JH (2006). Increase in DNA fragmentation and apoptosis-related gene expression in frozen-thawed bovine blastocysts. Zygote 14, 125–131.
Increase in DNA fragmentation and apoptosis-related gene expression in frozen-thawed bovine blastocysts.Crossref | GoogleScholarGoogle Scholar | 16719948PubMed |

Parrish, JJ, Krogenaes, A, and Susko-Parrish, JL (1995). Effect of bovine sperm separation by either swim-up or Percoll method on success of in vitro fertilization and early embryonic development. Theriogenology 44, 859–869.
Effect of bovine sperm separation by either swim-up or Percoll method on success of in vitro fertilization and early embryonic development.Crossref | GoogleScholarGoogle Scholar | 16727781PubMed |

Paschoal, DM, Sudano, MJ, Schwarz, KRL, Maziero, RRD, Guastali, MD, Crocomo, LF, Magalhães, LCO, Martins, A, Leal, CLV, and Landim-Alvarenga, FdC (2017). Cell apoptosis and lipid content of in vitro–produced, vitrified bovine embryos treated with forskolin. Theriogenology 87, 108–114.
Cell apoptosis and lipid content of in vitro–produced, vitrified bovine embryos treated with forskolin.Crossref | GoogleScholarGoogle Scholar | 27634395PubMed |

Rizos, D, Gutiérrez-Adán, A, Pérez-Garnelo, S, de la Fuente, J, Boland, MP, and Lonergan, P (2003). Bovine embryo culture in the presence or absence of serum: implications for blastocyst development, cryotolerance, and messenger RNA expression. Biology of Reproduction 68, 236–243.
Bovine embryo culture in the presence or absence of serum: implications for blastocyst development, cryotolerance, and messenger RNA expression.Crossref | GoogleScholarGoogle Scholar | 12493719PubMed |

Rubio Pomar, FJ, Teerds, KJ, Kidson, A, Colenbrander, B, Tharasanit, T, Aguilar, B, and Roelen, BAJ (2005). Differences in the incidence of apoptosis between in vivo and in vitro produced blastocysts of farm animal species: a comparative study. Theriogenology 63, 2254–2268.
Differences in the incidence of apoptosis between in vivo and in vitro produced blastocysts of farm animal species: a comparative study.Crossref | GoogleScholarGoogle Scholar |

Sala, P, Pötz, S, Brunner, M, Trötzmüller, M, Fauland, A, Triebl, A, Hartler, J, Lankmayr, E, and Köfeler, HC (2015). Determination of oxidized phosphatidylcholines by hydrophilic interaction liquid chromatography coupled to Fourier transform mass spectrometry. International Journal of Molecular Sciences 16, 8351–8363.
Determination of oxidized phosphatidylcholines by hydrophilic interaction liquid chromatography coupled to Fourier transform mass spectrometry.Crossref | GoogleScholarGoogle Scholar | 25874761PubMed |

Sawada, H, Yamazaki, K, and Hoshi, M (1990). Trypsin-like hatching protease from mouse embryos: Evidence for the presence in culture medium and its enzymatic properties. Journal of Experimental Zoology 254, 83–87.
Trypsin-like hatching protease from mouse embryos: Evidence for the presence in culture medium and its enzymatic properties.Crossref | GoogleScholarGoogle Scholar |

Schratt, G, Philippar, U, Hockemeyer, D, Schwarz, H, Alberti, S, and Nordheim, A (2004). SRF regulates Bcl-2 expression and promotes cell survival during murine embryonic development. The EMBO Journal 23, 1834–1844.
SRF regulates Bcl-2 expression and promotes cell survival during murine embryonic development.Crossref | GoogleScholarGoogle Scholar | 15057274PubMed |

Seimandi, M, Lemaire, G, Pillon, A, Perrin, A, Carlavan, I, Voegel, JJ, Vignon, F, Nicolas, J-C, and Balaguer, P (2005). Differential responses of PPARα, PPARδ, and PPARγ reporter cell lines to selective PPAR synthetic ligands. Analytical Biochemistry 344, 8–15.
Differential responses of PPARα, PPARδ, and PPARγ reporter cell lines to selective PPAR synthetic ligands.Crossref | GoogleScholarGoogle Scholar | 16038868PubMed |

Stinshoff, H, Wilkening, S, Hanstedt, A, Bollwein, H, and Wrenzycki, C (2014). Dimethylsulfoxide and conjugated linoleic acids affect bovine embryo development in vitro. Reproduction, Fertility and Development 26, 502–510.
Dimethylsulfoxide and conjugated linoleic acids affect bovine embryo development in vitro.Crossref | GoogleScholarGoogle Scholar |

Stroh, C, Cassens, U, Samraj, AK, Sibrowski, W, Schulze-Osthoff, K, and Los, M (2002). The role of caspases in cryoinjury: caspase inhibition strongly improves the recovery of cryopreserved hematopoietic and other cells. The FASEB Journal 16, 1651–1653.
The role of caspases in cryoinjury: caspase inhibition strongly improves the recovery of cryopreserved hematopoietic and other cells.Crossref | GoogleScholarGoogle Scholar | 12207004PubMed |

Subramaniam, HN, and Chaubal, KA (1990). Evaluation of intracellular lipids by standardized staining with a Sudan black B fraction. Journal of Biochemical and Biophysical Methods 21, 9–16.
Evaluation of intracellular lipids by standardized staining with a Sudan black B fraction.Crossref | GoogleScholarGoogle Scholar | 1698849PubMed |

Sudano, MJ, Paschoal, DM, da Silva Rascado, T, Magalhães, LCO, Crocomo, LF, de Lima-Neto, JF, and da Cruz Landim-Alvarenga, F (2011). Lipid content and apoptosis of in vitro-produced bovine embryosas determinants of susceptibility to vitrification. Theriogenology 75, 1211–1220.
Lipid content and apoptosis of in vitro-produced bovine embryosas determinants of susceptibility to vitrification.Crossref | GoogleScholarGoogle Scholar | 21247620PubMed |

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 taurus in vitro- and in vivo-produced blastocysts. Biology of Reproduction 87, 130.
Phosphatidylcholine and sphingomyelin profiles vary in Bos taurus indicus and Bos taurus taurus in vitro- and in vivo-produced blastocysts.Crossref | GoogleScholarGoogle Scholar | 23053436PubMed |

Tata, A, Sudano, MJ, Santos, VG, Landim-Alvarenga, FDC, Ferreira, CR, and Eberlin, MN (2013). Optimal single-embryo mass spectrometry fingerprinting. Journal of Mass Spectrometry 48, 844–849.
Optimal single-embryo mass spectrometry fingerprinting.Crossref | GoogleScholarGoogle Scholar | 23832940PubMed |

Tzivion, G, and Avruch, J (2002). 14-3-3 Proteins: active cofactors in cellular regulation by serine/threonine phosphorylation. Journal of Biological Chemistry 277, 3061–3064.
14-3-3 Proteins: active cofactors in cellular regulation by serine/threonine phosphorylation.Crossref | GoogleScholarGoogle Scholar |

van Meer, G, Voelker, DR, and Feigenson, GW (2008). Membrane lipids: where they are and how they behave. Nature Reviews Molecular Cell Biology 9, 112–124.
Membrane lipids: where they are and how they behave.Crossref | GoogleScholarGoogle Scholar | 18216768PubMed |

van Soom, A, Ysebaert, M-T, 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 |

Wahli, W, Braissant, O, and Desvergne, B (1995). Peroxisome proliferator activated receptors: transcriptional regulators of adipogenesis, lipid metabolism and more…. Chemistry & Biology 2, 261–266.
Peroxisome proliferator activated receptors: transcriptional regulators of adipogenesis, lipid metabolism and more….Crossref | GoogleScholarGoogle Scholar |

Watanabe, YF, de Souza, AH, Mingoti, RD, Ferreira, RM, Batista, EOS, Dayan, A, Watanabe, O, Meirelles, FV, Nogueira, MFG, Ferraz, JBS, and Baruselli, PS (2017). Number of oocytes retrieved per donor during OPU and its relationship with in vitro embryo production and field fertility following embryo transfer. Animal Reproduction 14, 635–644.
Number of oocytes retrieved per donor during OPU and its relationship with in vitro embryo production and field fertility following embryo transfer.Crossref | GoogleScholarGoogle Scholar |

Wolfe, J, and Bryant, G (1999). Freezing, drying, and/or vitrification of membrane–solute–water systems. Cryobiology 39, 103–129.
Freezing, drying, and/or vitrification of membrane–solute–water systems.Crossref | GoogleScholarGoogle Scholar | 10529304PubMed |

Xia, J, Mandal, R, Sinelnikov, IV, Broadhurst, D, and Wishart, DS (2012). MetaboAnalyst 2.0 – a comprehensive server for metabolomic data analysis. Nucleic Acids Research 40, W127–W133.
MetaboAnalyst 2.0 – a comprehensive server for metabolomic data analysis.Crossref | GoogleScholarGoogle Scholar | 22553367PubMed |

Yeung, WSB, Ho, PC, Lau, EYL, and Chan, STH (1992). Improved development of human embryos in vitro by a human oviductal cell co-culture system. Human Reproduction 7, 1144–11499.
Improved development of human embryos in vitro by a human oviductal cell co-culture system.Crossref | GoogleScholarGoogle Scholar |

Zha, J, Harada, H, Yang, E, Jockel, J, and Korsmeyer, SJ (1996). Serine phosphorylation of death agonist BAD in response to survival factor results in binding to 14-3-3 not BCL-X(L). Cell 87, 619–628.
Serine phosphorylation of death agonist BAD in response to survival factor results in binding to 14-3-3 not BCL-X(L).Crossref | GoogleScholarGoogle Scholar | 8929531PubMed |