Acessibilidade / Reportar erro

Cell differentiation events in pre-implantation mouse and bovine embryos

Abstract

Early mammal embryogenesis starts with oocyte fertilization, giving rise to the zygote. The events that the newly formed zygote surpasses are crucial to the embryo developmental success. Shortly after activation of its genome, cells of the embryo segregate into the inner cell mass (ICM) or the trophectoderm (TE). The first will give rise to the embryo while the latter will become the placenta. This first segregation involves cellular and molecular processes that include cell polarity linked to intracellular pathway activation, which will regulate the transcription of trophectoderm-related genes. Then, cells of the ICM undergo the second event of mammalian cell differentiation, which consists of the separation between epiblast (EPI) and hypoblast or primitive endoderm (PrE). This second segregation involves paracrine signaling, leading to differential expression of key genes that will dictate the fate of the cell. Although these processes are described in detail in the mouse, recent studies suggest that the bovine embryo could also be an interesting model for early development, since there are differences to the mouse and similarities with early human embryogenesis. In this review, we gathered the main data available in the literature upon bovine and mouse early development events, suggesting that both models should be analyzed and studied in a complementary way, to better model early events occurring in human development.

Keywords:
early development; embryogenesis; inner cell mass; trophectoderm; blastocyst

Introduction

In mammals, the formation of the zygote initiates a series of events, known as early embryogenesis. The success of this process depends mainly on the first cell cleavages and the differentiation of the first cell lineages, mechanisms that are essential for the embryo implantation at the uterus and further fetal development (as reviewed by Alberio, 2020Alberio R. Regulation of cell fate decisions in early mammalian embryos. Annu Rev Anim Biosci. 2020;8(1):377-93. http://dx.doi.org/10.1146/annurev-animal-021419-083841. PMid:31730400.
http://dx.doi.org/10.1146/annurev-animal...
).

Present since oogenesis, maternal RNAs and proteins orchestrate the beginning of embryonic development. This maternal material will support basic biosynthetic processes and coordinate the first mitotic divisions. As embryo development progresses, embryonic genome activation (EGA) begins. From this moment on, embryos can develop independently of the maternal genome, until the formation of a blastocyst composed of three distinct cell lineages: the trophectoderm (TE), an outer epithelium made of polarized cells that surrounds a group of nonpolar cells, called the inner cell mass (ICM). The ICM will later differentiate into two distinct cell types, the epiblast (EPI) and the primitive endoderm (PrE) cells (Cockburn and Rossant, 2010Cockburn K, Rossant J. Making the blastocyst: lessons from the mouse. J Clin Invest. 2010;120(4):995-1003. http://dx.doi.org/10.1172/JCI41229. PMid:20364097.
http://dx.doi.org/10.1172/JCI41229...
; Underhill and Robins, 2016Underhill LA, Robins JC. Trophoblast development in the murine preimplantation embryo. Semin Reprod Med. 2016;34(1):57-62. http://dx.doi.org/10.1055/s-0035-1570025. PMid:26757060.
http://dx.doi.org/10.1055/s-0035-1570025...
).

The TE is responsible for giving rise to the placental trophoblasts (Underhill and Robins, 2016Underhill LA, Robins JC. Trophoblast development in the murine preimplantation embryo. Semin Reprod Med. 2016;34(1):57-62. http://dx.doi.org/10.1055/s-0035-1570025. PMid:26757060.
http://dx.doi.org/10.1055/s-0035-1570025...
). On the other hand, the EPI will later differentiate into three intraembryonic tissues, known as endoderm, mesoderm, and ectoderm, which subsequently become the most diverse tissues and organs of the developing fetus (Sheng, 2015Sheng G. Epiblast morphogenesis before gastrulation. Dev Biol. 2015;401(1):17-24. http://dx.doi.org/10.1016/j.ydbio.2014.10.003. PMid:25446532.
http://dx.doi.org/10.1016/j.ydbio.2014.1...
). The PrE forms the yolk sac, composed of two extraembryonic cell lineages: visceral and parietal endoderm (Bassalert et al., 2018Bassalert C, Valverde-Estrella L, Chazaud C. Primitive endoderm differentiation: from specification to epithelialization. Curr Top Dev Biol. 2018;128:81-104. http://dx.doi.org/10.1016/bs.ctdb.2017.12.001. PMid:29477172.
http://dx.doi.org/10.1016/bs.ctdb.2017.1...
).

The first cell differentiation of the embryo is widely studied in mice, an animal model that provides the largest source of information about the theme. Although there are some crucial differences in the regulation of early developmental events, some evidence shows that bovine embryos can also be great models for understanding human embryonic development (Chen et al., 2009aChen AE, Egli D, Niakan K, Deng J, Akutsu H, Yamaki M, Cowan C, Fitz-Gerald C, Zhang K, Melton DA, Eggan K. Optimal timing of inner cell mass isolation increases the efficiency of human embryonic stem cell derivation and allows generation of sibling cell lines. Cell Stem Cell. 2009a;4(2):103-6. http://dx.doi.org/10.1016/j.stem.2008.12.001. PMid:19200798.
http://dx.doi.org/10.1016/j.stem.2008.12...
; reviewed by Wrenzycki, 2018Wrenzycki C. Gene expression analysis and in vitro production procedures for bovine preimplantation embryos: past highlights, present concepts and future prospects. Reprod Domest Anim. 2018;53(Suppl 2):14-9. http://dx.doi.org/10.1111/rda.13260. PMid:30238652.
http://dx.doi.org/10.1111/rda.13260...
). Studies indicate that transcription factors such as OCT4 and CDX2, important for the first cell specification, have similar temporal and spatial expression patterns in human (Chen et al., 2009aChen AE, Egli D, Niakan K, Deng J, Akutsu H, Yamaki M, Cowan C, Fitz-Gerald C, Zhang K, Melton DA, Eggan K. Optimal timing of inner cell mass isolation increases the efficiency of human embryonic stem cell derivation and allows generation of sibling cell lines. Cell Stem Cell. 2009a;4(2):103-6. http://dx.doi.org/10.1016/j.stem.2008.12.001. PMid:19200798.
http://dx.doi.org/10.1016/j.stem.2008.12...
; Niakan and Eggan, 2013Niakan KK, Eggan K. Analysis of human embryos from zygote to blastocyst reveals distinct gene expression patterns relative to the mouse. Dev Biol. 2013;375(1):54-64. http://dx.doi.org/10.1016/j.ydbio.2012.12.008. PMid:23261930.
http://dx.doi.org/10.1016/j.ydbio.2012.1...
) and bovine (Berg et al., 2011Berg DK, Smith CS, Pearton DJ, Wells DN, Broadhurst R, Donnison M, Pfeffer PL. Trophectoderm lineage determination in cattle. Dev Cell. 2011;20(2):244-55. http://dx.doi.org/10.1016/j.devcel.2011.01.003. PMid:21316591.
http://dx.doi.org/10.1016/j.devcel.2011....
) embryos, while mouse embryos present substantial differences to these species (Niwa et al., 2005Niwa H, Toyooka Y, Shimosato D, Strumpf D, Takahashi K, Yagi R, Rossant J. Interaction between Oct3/4 and Cdx2 determines trophectoderm differentiation. Cell. 2005;123(5):917-29. http://dx.doi.org/10.1016/j.cell.2005.08.040. PMid:16325584.
http://dx.doi.org/10.1016/j.cell.2005.08...
; Strumpf et al., 2005Strumpf D, Mao CA, Yamanaka Y, Ralston A, Chawengsaksophak K, Beck F, Rossant J. Cdx2 is required for correct cell fate specification and differentiation of trophectoderm in the mouse blastocyst. Development. 2005;132(9):2093-102. http://dx.doi.org/10.1242/dev.01801. PMid:15788452.
http://dx.doi.org/10.1242/dev.01801...
; Niakan and Eggan, 2013Niakan KK, Eggan K. Analysis of human embryos from zygote to blastocyst reveals distinct gene expression patterns relative to the mouse. Dev Biol. 2013;375(1):54-64. http://dx.doi.org/10.1016/j.ydbio.2012.12.008. PMid:23261930.
http://dx.doi.org/10.1016/j.ydbio.2012.1...
). In addition, the biological mechanisms that interfere with the regulation of gene expression seem to be different in bovine and mouse embryos (Sakurai et al., 2017Sakurai N, Takahashi K, Emura N, Hashizume T, Sawai K. Effects of downregulating TEAD4 transcripts by RNA interference on early development of bovine embryos. J Reprod Dev. 2017;63(2):135-42. http://dx.doi.org/10.1262/jrd.2016-130. PMid:27941302.
http://dx.doi.org/10.1262/jrd.2016-130...
; Gerri et al., 2020Gerri C, McCarthy A, Alanis-Lobato G, Demtschenko A, Bruneau A, Loubersac S, Fogarty NME, Hampshire D, Elder K, Snell P, Christie L, David L, van de Velde H, Fouladi-Nashta AA, Niakan KK. Initiation of a conserved trophectoderm program in human, cow and mouse embryos. Nature. 2020;587(7834):443-7. http://dx.doi.org/10.1038/s41586-020-2759-x. PMid:32968278.
http://dx.doi.org/10.1038/s41586-020-275...
; Ortega et al., 2020Ortega MS, Kelleher AM, O’Neil E, Benne J, Cecil R, Spencer TE. NANOG is required to form the epiblast and maintain pluripotency in the bovine embryo. Mol Reprod Dev. 2020;87(1):152-60. http://dx.doi.org/10.1002/mrd.23304. PMid:31803983.
http://dx.doi.org/10.1002/mrd.23304...
; Anjos et al., 2021Anjos SAA, Costa CP, Assumpção MEOA, Visintin JA, Goissis MD. Inhibition of apical domain formation does not block blastocyst development in bovine embryos. Reprod Fertil Dev. 2021;33(10):665. http://dx.doi.org/10.1071/RD20339. PMid:34092280.
http://dx.doi.org/10.1071/RD20339...
).

Considering the importance of the correct specialization of these cell lines for embryo developmental success, this study aims to gather the main data available in the current literature regarding the events of cell differentiation during pre-implantation development, proposing a comparative analysis between the early development of mouse and bovine embryos.

Developmental dynamics

In mammals, the newly fertilized zygote undergoes symmetrical cell divisions, called cleavages, which is a relatively conserved mechanism between species. In the early stages, the blastomeres are relatively spherical, similar to each other and they become smaller with each cleavage round (Johnson and Ziomek, 1981Johnson MH, Ziomek CA. The foundation of two distinct cell lineages within the mouse morula. Cell. 1981;24(1):71-80. http://dx.doi.org/10.1016/0092-8674(81)90502-X. PMid:7237545.
http://dx.doi.org/10.1016/0092-8674(81)9...
; Johnson and McConnell, 2004Johnson MH, McConnell JM. Lineage allocation and cell polarity during mouse embryogenesis. Semin Cell Dev Biol. 2004;15(5):583-97. http://dx.doi.org/10.1016/j.semcdb.2004.04.002. PMid:15271304.
http://dx.doi.org/10.1016/j.semcdb.2004....
). The events that result in the greatest morphological changes in pre-implantation embryonic development happen during the embryonic compaction, when there is a great increase in intercellular contact followed by the blastocyst formation (Johnson, 2009Johnson MH. From mouse egg to mouse embryo: polarities, axes, and tissues. Annu Rev Cell Dev Biol. 2009;25(1):483-512. http://dx.doi.org/10.1146/annurev.cellbio.042308.113348. PMid:19575654.
http://dx.doi.org/10.1146/annurev.cellbi...
). Despite the existence of relatively conserved genetic regulatory mechanisms in mammals, the initial development in cattle and mice has relevant differences, mainly in chronological aspects.

In murine embryos, compaction begins at the 8-cell stage, blastomeres flatten their apical surfaces, forming a cluster of cells with indistinct cell boundaries (Lo and Gilula, 1979Lo CW, Gilula NB. Gap junctional communication in the preimplantation mouse embryo. Cell. 1979;18(2):399-409. http://dx.doi.org/10.1016/0092-8674(79)90059-X. PMid:498274.
http://dx.doi.org/10.1016/0092-8674(79)9...
; Becker and Davies, 1995Becker DL, Davies CS. Role of gap junctions in the development of the preimplantation mouse embryo. Microsc Res Tech. 1995;31(5):364-74. http://dx.doi.org/10.1002/jemt.1070310506. PMid:8534898.
http://dx.doi.org/10.1002/jemt.107031050...
). At this moment, the apical and basolateral membrane domains are formed, in which the apical domain presents proteins like Ezrin (Louvet et al., 1996Louvet S, Aghion J, Santa-Maria A, Mangeat P, Maro B. Ezrin becomes restricted to outer cells following asymmetrical division in the preimplantation mouse embryo. Dev Biol. 1996;177(2):568-79. http://dx.doi.org/10.1006/dbio.1996.0186. PMid:8806832.
http://dx.doi.org/10.1006/dbio.1996.0186...
) and, the cell adhesion caused by zonula adherent epithelial junction (ZA) characteristic proteins, forms the contact region of basolateral membrane domain between epithelial cells (Aberle et al., 1996Aberle H, Schwartz H, Kemler R. Cadherin-catenin complex: protein interactions and their implications for cadherin function. J Cell Biochem. 1996;61(4):514-23. http://dx.doi.org/10.1002/(SICI)1097-4644(19960616)61:4<514::AID-JCB4>3.0.CO;2-R. PMid:8806074.
http://dx.doi.org/10.1002/(SICI)1097-464...
).

Furthermore, between 16 and 32-cell stages, Na/K ATPases located at basolateral membranes of mice TE cells transport fluids and ions, forming the blastocoel cavity, filled with blastocoel fluid, characterizing the embryonic cavitation process, which will culminate in the blastocyst formation and an apparent cellular differentiation (Kidder and Watson, 2005Kidder GM, Watson AJ. Roles of Na,K-ATPase in early development and trophectoderm differentiation. Semin Nephrol. 2005;25(5):352-5. http://dx.doi.org/10.1016/j.semnephrol.2005.03.011. PMid:16139691.
http://dx.doi.org/10.1016/j.semnephrol.2...
).

In vivo produced bovine embryos, however, start compaction near the 32-cell stage, 5 days after insemination, and the cavitation process begins after the 64-cell stage and the early blastocyst contained in average 105 cells (van Soom et al., 1997van Soom A, Boerjan ML, Bols PE, Vanroose G, Lein A, Coryn M, de Kruif A. Timing of compaction and inner cell allocation in bovine embryos produced in vivo after superovulation. Biol Reprod. 1997;57(5):1041-9. http://dx.doi.org/10.1095/biolreprod57.5.1041. PMid:9369168.
http://dx.doi.org/10.1095/biolreprod57.5...
). Bovine in vitro produced (IVP) embryos initiate cavitation earlier than in vivo ones, after the 32-cell stage. This difference could occur due to observations suggesting that in vivo derived embryos spend more time in compaction (van Soom et al., 1992van Soom A, van Vlaenderen I, Mahmoudzadeh AR, Deluyker H, de Kruif A. Compaction rate of in vitro fertilized bovine embryos related to the interval from insemination to first cleavage. Theriogenology. 1992;38(5):905-19. http://dx.doi.org/10.1016/0093-691X(92)90165-N. PMid:16727189.
http://dx.doi.org/10.1016/0093-691X(92)9...
; van Soom et al., 1997van Soom A, Boerjan ML, Bols PE, Vanroose G, Lein A, Coryn M, de Kruif A. Timing of compaction and inner cell allocation in bovine embryos produced in vivo after superovulation. Biol Reprod. 1997;57(5):1041-9. http://dx.doi.org/10.1095/biolreprod57.5.1041. PMid:9369168.
http://dx.doi.org/10.1095/biolreprod57.5...
).

At this stage of development, the morphological aspect is the most indicative factor on embryo quality for embryo transfer (Watson et al., 1999Watson AJ, Westhusin ME, De Sousa PA, Betts DH, Barcroft LC. Gene expression regulating blastocyst formation. Theriogenology. 1999;51(1):117-33. http://dx.doi.org/10.1016/S0093-691X(98)00236-2. PMid:10729067.
http://dx.doi.org/10.1016/S0093-691X(98)...
). Although the developmental stages present a very similar morphology in bovine and mouse embryos, there are divergences in the duration of each stage and the occurrence timing of remarkable morphological events, such as the compaction and formation of the blastocoel (Figure 1). Still, there are differences in expression pattern and function of proteins related to TE cells specification (such as CDX2 and YAP) and ICM (OCT4, SOX2, NANOG, GATA4, GATA6, and SOX17) (Yamanaka et al., 2006Yamanaka Y, Ralston A, Stephenson RO, Rossant J. Cell and molecular regulation of the mouse blastocyst. Dev Dyn. 2006;235(9):2301-14. http://dx.doi.org/10.1002/dvdy.20844. PMid:16773657.
http://dx.doi.org/10.1002/dvdy.20844...
; Wicklow et al., 2014Wicklow E, Blij S, Frum T, Hirate Y, Lang RA, Sasaki H, Ralston A. HIPPO pathway members restrict SOX2 to the inner cell mass where it promotes ICM fates in the mouse blastocyst. PLoS Genet. 2014;10(10):e1004618. http://dx.doi.org/10.1371/journal.pgen.1004618. PMid:25340657.
http://dx.doi.org/10.1371/journal.pgen.1...
; Wei et al., 2017Wei Q, Zhong L, Zhang S, Mu H, Xiang J, Yue L, Dai Y, Han J. Bovine lineage specification revealed by single-cell gene expression analysis from zygote to blastocyst. Biol Reprod. 2017;97(1):5-17. http://dx.doi.org/10.1093/biolre/iox071. PMid:28859285.
http://dx.doi.org/10.1093/biolre/iox071...
; Sharma and Madan, 2020Sharma J, Madan P. Characterisation of the Hippo signalling pathway during bovine preimplantation embryo development. Reprod Fertil Dev. 2020;32(4):392-401. http://dx.doi.org/10.1071/RD18320. PMid:31718770.
http://dx.doi.org/10.1071/RD18320...
). The genetic regulation mechanisms, protein expression profiles, and cell signaling pathways related to cell differentiation events are going to be presented next.

Figure 1
Timeline of morphology and lineage markers expression during early development of bovine and murine embryos. During early development, the main divergences between bovine and mouse embryos occur in the duration of each stage and the occurrence timing of morphological events. For example, compaction gives rise to the morula at 5dpf in bovine and 3dpf in mouse, and the blastocyst formation occurs rapidly in mouse, as soon as 18 hours after morula stage, while bovine blastocyst only forms at 7dpf. Unlike in mouse embryos, bovine morulae do not express trophectoderm marker CDX2 (gold) in outer cells and SOX2 (lilac) is present in all cells. At this point, embryos express epiblast marker NANOG and GATA6 in all cells. After blastocyst formation, NANOG (red) and primitive endoderm markers such as GATA4, GATA6, SOX17 (blue) are mutually exclusive, form a salt-and-pepper expression pattern in the inner cell mass in both bovine and mouse embryos. At the late blastocyst stage in mouse (4.0 dpf), blastomeres expressing either epiblast markers or primitive endoderm markers form different layers while in the hatched blastocyst stage in bovine (9.0 dpf) these two lineages are not still separated into layers. Embryos are represented bidimensionally for ease of understanding.

First cell differentiation event

In mammals, the first cleavages will give rise to an embryo composed of totipotent blastomeres, with full differentiation potential in all cell lineages (Fleming and Johnson, 1988Fleming TP, Johnson MH. From egg to epithelium. Annu Rev Cell Biol. 1988;4(1):459-85. http://dx.doi.org/10.1146/annurev.cb.04.110188.002331. PMid:3058163.
http://dx.doi.org/10.1146/annurev.cb.04....
). During development, the first cell differentiation leads to the formation of TE epithelial cells or ICM cells. In the post-implantation stages, the TE will give rise to the extra-embryonic ectoderm and the placental trophoblasts, while the ICM will be responsible for developing the embryo itself and the extra-embryonic endoderm (Strumpf et al., 2005Strumpf D, Mao CA, Yamanaka Y, Ralston A, Chawengsaksophak K, Beck F, Rossant J. Cdx2 is required for correct cell fate specification and differentiation of trophectoderm in the mouse blastocyst. Development. 2005;132(9):2093-102. http://dx.doi.org/10.1242/dev.01801. PMid:15788452.
http://dx.doi.org/10.1242/dev.01801...
).

Two models that seek to explain the mechanisms involved in the first cell differentiation in the mouse are currently accepted. The first one, called the inside-outside model (Tarkowski and Wróblewska, 1967Tarkowski AK, Wróblewska J. Development of blastomeres of mouse eggs isolated at the 4- and 8-cell stage. J Embryol Exp Morphol. 1967;18(1):155-80. http://dx.doi.org/10.1242/dev.18.1.155. PMid:6048976.
http://dx.doi.org/10.1242/dev.18.1.155...
), proposes that the cell position in the embryo is the main factor that will define the cell type fate. The second model, which does not exclude the first one, argues that cell polarization defines cell lineage (Johnson and Ziomek, 1981Johnson MH, Ziomek CA. The foundation of two distinct cell lineages within the mouse morula. Cell. 1981;24(1):71-80. http://dx.doi.org/10.1016/0092-8674(81)90502-X. PMid:7237545.
http://dx.doi.org/10.1016/0092-8674(81)9...
). According to this model, polarization and the beginning of asymmetric cell divisions are crucial events for cell specifying process initiation (Graham and Deussen, 1978Graham CF, Deussen ZA. Features of cell lineage in preimplantation mouse development. J Embryol Exp Morphol. 1978;48(1):53-72. http://dx.doi.org/10.1242/dev.48.1.53. PMid:581769.
http://dx.doi.org/10.1242/dev.48.1.53...
; Johnson and Ziomek, 1981Johnson MH, Ziomek CA. The foundation of two distinct cell lineages within the mouse morula. Cell. 1981;24(1):71-80. http://dx.doi.org/10.1016/0092-8674(81)90502-X. PMid:7237545.
http://dx.doi.org/10.1016/0092-8674(81)9...
; Fleming, 1987Fleming TP. A quantitative analysis of cell allocation to trophectoderm and inner cell mass in the mouse blastocyst. Dev Biol. 1987;119(2):520-31. http://dx.doi.org/10.1016/0012-1606(87)90055-8. PMid:3803716.
http://dx.doi.org/10.1016/0012-1606(87)9...
).

Despite these studies, there is no consensus on the exact moment when blastomeres determine their cell lineage fate (Posfai et al., 2017Posfai E, Petropoulos S, Barros FRO, Schell JP, Jurisica I, Sandberg R, Lanner F, Rossant J. Position- and Hippo signaling-dependent plasticity during lineage segregation in the early mouse embryo. eLife. 2017;6:e22906. http://dx.doi.org/10.7554/eLife.22906. PMid:28226240.
http://dx.doi.org/10.7554/eLife.22906...
). In mice, before these two cell lines segregate, evidence shows that gene expression pattern starts to change between internal and external cells at the 8-cell stage. As a result, genes initially expressed in all blastomeres start to have their expressions regulated according to the cell location (Pesce and Scholer, 2001Pesce M, Schöler HR. Oct-4: gatekeeper in the beginnings of mammalian development. Stem Cells. 2001;19(4):271-8. http://dx.doi.org/10.1634/stemcells.19-4-271. PMid:11463946.
http://dx.doi.org/10.1634/stemcells.19-4...
).

Embryonic compaction promotes an increase in cell-cell interactions and cell adhesion proteins, resulting in cell polarization (Chen and Zhang, 2013Chen J, Zhang M. The Par3/Par6/aPKC complex and epithelial cell polarity. Exp Cell Res. 2013;319(10):1357-64. http://dx.doi.org/10.1016/j.yexcr.2013.03.021. PMid:23535009.
http://dx.doi.org/10.1016/j.yexcr.2013.0...
). The appearance of polarity-related proteins, such as PARD3, PARD6, and PKCs, was already reported in the mouse morula stage (Plusa et al., 2005Plusa B, Frankenberg S, Chalmers A, Hadjantonakis AK, Moore CA, Papalopulu N, Papaioannou VE, Glover DM, Zernicka-Goetz M. Downregulation of Par3 and aPKC function directs cells towards the ICM in the preimplantation mouse embryo. J Cell Sci. 2005;118(Pt 3):505-15. http://dx.doi.org/10.1242/jcs.01666. PMid:15657073.
http://dx.doi.org/10.1242/jcs.01666...
). The organization of these proteins during compaction leads to an apical domain formation. Once cells express two distinct intercellular junction complexes, tight and adherent, an apical region and a basolateral region establish cellular polarity (Plusa et al., 2005Plusa B, Frankenberg S, Chalmers A, Hadjantonakis AK, Moore CA, Papalopulu N, Papaioannou VE, Glover DM, Zernicka-Goetz M. Downregulation of Par3 and aPKC function directs cells towards the ICM in the preimplantation mouse embryo. J Cell Sci. 2005;118(Pt 3):505-15. http://dx.doi.org/10.1242/jcs.01666. PMid:15657073.
http://dx.doi.org/10.1242/jcs.01666...
; Vinot et al., 2005Vinot S, Le T, Ohno S, Pawson T, Maro B, Louvet-Vallée S. Asymmetric distribution of PAR proteins in the mouse embryo begins at the 8-cell stage during compaction. Dev Biol. 2005;282(2):307-19. http://dx.doi.org/10.1016/j.ydbio.2005.03.001. PMid:15950600.
http://dx.doi.org/10.1016/j.ydbio.2005.0...
; Ralston and Rossant, 2008Ralston A, Rossant J. Cdx2 acts downstream of cell polarization to cell-autonomously promote trophectoderm fate in the early mouse embryo. Dev Biol. 2008;313(2):614-29. http://dx.doi.org/10.1016/j.ydbio.2007.10.054. PMid:18067887.
http://dx.doi.org/10.1016/j.ydbio.2007.1...
).

At this stage of development, asymmetric cell divisions are responsible for generating two types of daughter cells: an externally located polar cell, which will inherit both apical and basolateral domains, and an internally positioned cell, which will acquire only the basolateral domain proteins, distributed homogeneously, becoming then nonpolar cells (Figure 2A). In bovine embryos, recent research reported a similar cellular polarity pattern to murine embryos, essential for the beginning of TE specification; however, polarization inhibition did not show as drastic effects as in mouse embryos (Gerri et al., 2020Gerri C, McCarthy A, Alanis-Lobato G, Demtschenko A, Bruneau A, Loubersac S, Fogarty NME, Hampshire D, Elder K, Snell P, Christie L, David L, van de Velde H, Fouladi-Nashta AA, Niakan KK. Initiation of a conserved trophectoderm program in human, cow and mouse embryos. Nature. 2020;587(7834):443-7. http://dx.doi.org/10.1038/s41586-020-2759-x. PMid:32968278.
http://dx.doi.org/10.1038/s41586-020-275...
) and another study showed that inhibition of apical domain formation using blebbistatin did not impair bovine blastocyst formation (Anjos et al., 2021Anjos SAA, Costa CP, Assumpção MEOA, Visintin JA, Goissis MD. Inhibition of apical domain formation does not block blastocyst development in bovine embryos. Reprod Fertil Dev. 2021;33(10):665. http://dx.doi.org/10.1071/RD20339. PMid:34092280.
http://dx.doi.org/10.1071/RD20339...
), suggesting that polarization is not the only process controlling this differentiation in bovine embryos.

Figure 2
Influence of cell polarity and Hippo signaling pathway in TE and ICM cell differentiation in mouse embryos. (A) At the 8-cell stage, the tight junctions (red) and the adherent junctions (purple) establish a polarization pattern in the embryo. The asymmetric cell division axis (dotted line) is responsible for generating two types of daughter cells; (B) The first, an externally located polar cell, which will inherit both apical and basolateral domains, and the second one, internally positioned, which will acquire only the basolateral domain proteins, distributed homogeneously, becoming then nonpolar cells. The apical domain blocks the Hippo pathway activation in polar cells, allowing YAP1 translocation into the nucleus, and then resulting in Cdx2 transcription. With the absence of tight junctions, Hippo pathway is activated at nonpolar cells.

In the mouse, cell division asymmetry is also responsible for generating blastomeres with different cell contractility. Polar cells maintain low contractility in the apical domain, while nonpolar cells, which have an increase in cell contractility, compete for internal positions (Maître et al., 2016Maître JL, Turlier H, Illukkumbura R, Eismann B, Niwayama R, Nédélec F, Hiiragi T. Asymmetric division of contractile domains couples cell positioning and fate specification. Nature. 2016;536(7616):344-8. http://dx.doi.org/10.1038/nature18958. PMid:27487217.
http://dx.doi.org/10.1038/nature18958...
). Studies indicate that differences in the organization of cytoskeleton proteins occur before the spatial organization of these cells, contributing to cell differentiation. Mouse blastomeres with apical domain show an increase in cell surface keratins expression, which prevents the internalization of these cells and consequently precedes the expression of TE markers (Lim et al., 2020Lim HYG, Alvarez YD, Gasnier M, Wang Y, Tetlak P, Bissiere S, Wang H, Biro M, Plachta N. Keratins are asymmetrically inherited fate determinants in the mammalian embryo. Nature. 2020;585(7825):404-9. http://dx.doi.org/10.1038/s41586-020-2647-4. PMid:32848249.
http://dx.doi.org/10.1038/s41586-020-264...
).

Polarity establishment and the asymmetric cell divisions influence Hippo pathway activity, which behaves differently in polar and nonpolar cells, controlling cellular fate (Yamanaka et al., 2006Yamanaka Y, Ralston A, Stephenson RO, Rossant J. Cell and molecular regulation of the mouse blastocyst. Dev Dyn. 2006;235(9):2301-14. http://dx.doi.org/10.1002/dvdy.20844. PMid:16773657.
http://dx.doi.org/10.1002/dvdy.20844...
; Nishioka et al., 2009Nishioka N, Inoue K, Adachi K, Kiyonari H, Ota M, Ralston A, Yabuta N, Hirahara S, Stephenson RO, Ogonuki N, Makita R, Kurihara H, Morin-Kensicki EM, Nojima H, Rossant J, Nakao K, Niwa H, Sasaki H. The Hippo signaling pathway components Lats and Yap pattern Tead4 activity to distinguish mouse trophectoderm from inner cell mass. Dev Cell. 2009;16(3):398-410. http://dx.doi.org/10.1016/j.devcel.2009.02.003. PMid:19289085.
http://dx.doi.org/10.1016/j.devcel.2009....
) as externally positioned cells acquire an apical domain, which influences the Hippo pathway, giving rise to TE in mouse embryos (Korotkevich et al., 2017Korotkevich E, Niwayama R, Courtois A, Friese S, Berger N, Buchholz F, Hiiragi T. The apical domain is required and sufficient for the first lineage segregation in the mouse embryo. Dev Cell. 2017;40(3):235-247.e7. http://dx.doi.org/10.1016/j.devcel.2017.01.006. PMid:28171747.
http://dx.doi.org/10.1016/j.devcel.2017....
). The Hippo pathway activation in the internal nonpolar cells causes LATS1/2 serine/threonine kinases to promote YAP1 protein phosphorylation, which prevents its transport to the nucleus. Once phosphorylated, YAP is not able to act as a transcription cofactor together with TEAD4. In the polar apical cells, the Hippo pathway remains inactive and YAP1 is translocated to the nucleus, where it will function as a TEAD4 co-activator (Figure 2B). TEAD4 is a transcription factor member of the TEA domain family, essential for the maintenance and/or positive regulation of Cdx2 in mouse, which expression directly relates to further development of TE (Yagi et al., 2007Yagi R, Kohn MJ, Karavanova I, Kaneko KJ, Vullhorst D, DePamphilis ML, Buonanno A. Transcription factor TEAD4 specifies the trophectoderm lineage at the beginning of mammalian development. Development. 2007;134(21):3827-36. http://dx.doi.org/10.1242/dev.010223. PMid:17913785.
http://dx.doi.org/10.1242/dev.010223...
; Nishioka et al., 2008Nishioka N, Yamamoto S, Kiyonari H, Sato H, Sawada A, Ota M, Nakao K, Sasaki H. Tead4 is required for specification of trophectoderm in pre-implantation mouse embryos. Mech Dev. 2008;125(3-4):270-83. http://dx.doi.org/10.1016/j.mod.2007.11.002. PMid:18083014.
http://dx.doi.org/10.1016/j.mod.2007.11....
; Nishioka et al., 2009Nishioka N, Inoue K, Adachi K, Kiyonari H, Ota M, Ralston A, Yabuta N, Hirahara S, Stephenson RO, Ogonuki N, Makita R, Kurihara H, Morin-Kensicki EM, Nojima H, Rossant J, Nakao K, Niwa H, Sasaki H. The Hippo signaling pathway components Lats and Yap pattern Tead4 activity to distinguish mouse trophectoderm from inner cell mass. Dev Cell. 2009;16(3):398-410. http://dx.doi.org/10.1016/j.devcel.2009.02.003. PMid:19289085.
http://dx.doi.org/10.1016/j.devcel.2009....
; Misra and Irvine, 2018Misra JR, Irvine KD. The hippo signaling network and its biological functions. Annu Rev Genet. 2018;52(1):65-87. http://dx.doi.org/10.1146/annurev-genet-120417-031621. PMid:30183404.
http://dx.doi.org/10.1146/annurev-genet-...
).

Tead4 deletion in mouse embryos increased the expression of specific ICM factors, such as Oct4, Nanog, and Sox2, suggesting that Tead4 is not essential for ICM formation (Nishioka et al., 2008Nishioka N, Yamamoto S, Kiyonari H, Sato H, Sawada A, Ota M, Nakao K, Sasaki H. Tead4 is required for specification of trophectoderm in pre-implantation mouse embryos. Mech Dev. 2008;125(3-4):270-83. http://dx.doi.org/10.1016/j.mod.2007.11.002. PMid:18083014.
http://dx.doi.org/10.1016/j.mod.2007.11....
). Bovine embryos have low TEAD4 mRNA levels in the early stages of development, increasing their expression from the 16-cell stage. During the 8 to 16-cell stage, TEAD4 is detected in the cytoplasm, while in the morula and blastocyst it is located in the nucleus of most blastomeres. Bovine embryos in which TEAD4 was knocked down using small interfering RNA (siRNA) present blastocyst formation and normal TE cell differentiation and expansion. Unlike in mice, TEAD4 knockdown did not influence the gene expression of OCT4, NANOG, CDX2, and GATA3, suggesting that TEAD4 is not essential for TE formation in the bovine embryo (Sakurai et al., 2017Sakurai N, Takahashi K, Emura N, Hashizume T, Sawai K. Effects of downregulating TEAD4 transcripts by RNA interference on early development of bovine embryos. J Reprod Dev. 2017;63(2):135-42. http://dx.doi.org/10.1262/jrd.2016-130. PMid:27941302.
http://dx.doi.org/10.1262/jrd.2016-130...
).

Deletion of Yap1 and Wwtr1 (TAZ), a functional homolog of YAP1, led to the death of mouse embryos before reaching the morula stage. By contrast, overexpression of Yap1 and Taz promotes the expression of Cdx2, confirming the hypothesis that YAP1 together with TAZ and TEAD4 regulates the expression of Cdx2 (Nishioka et al., 2009Nishioka N, Inoue K, Adachi K, Kiyonari H, Ota M, Ralston A, Yabuta N, Hirahara S, Stephenson RO, Ogonuki N, Makita R, Kurihara H, Morin-Kensicki EM, Nojima H, Rossant J, Nakao K, Niwa H, Sasaki H. The Hippo signaling pathway components Lats and Yap pattern Tead4 activity to distinguish mouse trophectoderm from inner cell mass. Dev Cell. 2009;16(3):398-410. http://dx.doi.org/10.1016/j.devcel.2009.02.003. PMid:19289085.
http://dx.doi.org/10.1016/j.devcel.2009....
).

Recently, differences in YAP1 and TAZ patterns were reported in bovine embryos, suggesting that in these animals, Hippo signaling may be established by different mechanisms. Despite the pattern shown in murine embryos, phosphorylated YAP1 is found both in the nucleus and in the cytoplasm of bovine embryos, from 2-cell to the 16-cell stage, becoming mostly nuclear from the morula onwards (Sharma and Madan, 2020Sharma J, Madan P. Characterisation of the Hippo signalling pathway during bovine preimplantation embryo development. Reprod Fertil Dev. 2020;32(4):392-401. http://dx.doi.org/10.1071/RD18320. PMid:31718770.
http://dx.doi.org/10.1071/RD18320...
). The TAZ coactivator exhibited nuclear localization in the external cells of the blastocyst, whereas, in mice, TAZ localization becomes exclusively nuclear from the morula stage onwards (Sharma and Madan, 2020Sharma J, Madan P. Characterisation of the Hippo signalling pathway during bovine preimplantation embryo development. Reprod Fertil Dev. 2020;32(4):392-401. http://dx.doi.org/10.1071/RD18320. PMid:31718770.
http://dx.doi.org/10.1071/RD18320...
).

The Hippo signaling pathway is also related to glucose metabolism, which in turn may be related to this first cell differentiation event. In the mouse, it was first demonstrated that transcription factor TEAD4 was not necessary for the formation of the blastocoel when embryos were cultured in conditions with lower concentrations of oxygen. However, Tead4 knockout embryos cultured in the absence of glucose did not initiate blastocoel formation, unlike wild-type embryos, suggesting that the role of TEAD4 during pre-implantation development is to establish the essential energy homeostasis for the transition from the morula to blastocyst (Kaneko and DePamphilis, 2013Kaneko KJ, DePamphilis ML. TEAD4 establishes the energy homeostasis essential for blastocoel formation. Development. 2013;140(17):3680-90. http://dx.doi.org/10.1242/dev.093799. PMid:23903192.
http://dx.doi.org/10.1242/dev.093799...
).

The relationship between glucose metabolism and blastocyst formation is known for some time. Shortly before the formation of the blastocoel, the mouse embryo abruptly changes its energy sources from pyruvate and lactate to glucose, concomitantly with a 2.7-fold increase in oxygen consumption (Gardner, 1998Gardner DK. Changes in requirements and utilization of nutrients during mammalian preimplantation embryo development and their significance in embryo culture. Theriogenology. 1998;49(1):83-102. http://dx.doi.org/10.1016/S0093-691X(97)00404-4. PMid:10732123.
http://dx.doi.org/10.1016/S0093-691X(97)...
; Johnson et al., 2003Johnson MT, Mahmood S, Patel MS. Intermediary metabolism and energetics during murine early embryogenesis. J Biol Chem. 2003;278(34):31457-60. http://dx.doi.org/10.1074/jbc.R300002200. PMid:12788927.
http://dx.doi.org/10.1074/jbc.R300002200...
). Brown and Whittingham (1991)Brown JJ, Whittingham DG. The roles of pyruvate, lactate and glucose during preimplantation development of embryos from F1 hybrid mice in vitro. Development. 1991;112(1):99-105. http://dx.doi.org/10.1242/dev.112.1.99. PMid:1769345.
http://dx.doi.org/10.1242/dev.112.1.99...
established a specific glucose requirement during the transition from morula to blastocyst, and unlike the early stages of mouse embryonic development in which lactate and pyruvate meet the needs of the embryo; glucose is required during the first cell differentiation.

In mice, when glucose was not available in the medium, a developmental block occurred in the eight-cell stage and there was no expression of the transcription factor Cdx2, ensuing just the expression of ICM-related transcription factors (Chi et al., 2020Chi F, Sharpley MS, Nagaraj R, Roy SS, Banerjee U. Glycolysis-independent glucose metabolism distinguishes TE from ICM fate during mammalian embryogenesis. Dev Cell. 2020;53(1):9-26.e4. http://dx.doi.org/10.1016/j.devcel.2020.02.015. PMid:32197068.
http://dx.doi.org/10.1016/j.devcel.2020....
). It was also shown that the expression of Cdx2 was influenced by the modulation of the hexosamine biosynthetic pathway and pentose phosphate pathway (Chi et al., 2020Chi F, Sharpley MS, Nagaraj R, Roy SS, Banerjee U. Glycolysis-independent glucose metabolism distinguishes TE from ICM fate during mammalian embryogenesis. Dev Cell. 2020;53(1):9-26.e4. http://dx.doi.org/10.1016/j.devcel.2020.02.015. PMid:32197068.
http://dx.doi.org/10.1016/j.devcel.2020....
).

These pathways would be involved in the O-glycosylation of YAP1, allowing nuclear translocation of YAP1, directly interfering in the first cell differentiation process and playing a decisive role in the specification of the trophectoderm (Chi et al., 2020Chi F, Sharpley MS, Nagaraj R, Roy SS, Banerjee U. Glycolysis-independent glucose metabolism distinguishes TE from ICM fate during mammalian embryogenesis. Dev Cell. 2020;53(1):9-26.e4. http://dx.doi.org/10.1016/j.devcel.2020.02.015. PMid:32197068.
http://dx.doi.org/10.1016/j.devcel.2020....
). More specifically, pharmacological modulation of the O-GlcNAcylation process in pre-implantation stages proved to be important for future differentiation of trophoblast cells, interfering with the expression of transcription factors such as CDX2 and GATA3 (Ruane et al., 2020Ruane PT, Tan CMJ, Adlam DJ, Kimber SJ, Brison DR, Aplin JD, Westwood M. Protein O-GlcNAcylation promotes trophoblast differentiation at implantation. Cells. 2020;9(10):2246. http://dx.doi.org/10.3390/cells9102246. PMid:33036308.
http://dx.doi.org/10.3390/cells9102246...
).

It is known that the Cdx2 transcription factor is an important regulator and is abundantly present in TE cells (Beck et al., 1995Beck F, Erler T, Russell A, James R. Expression of Cdx-2 in the mouse embryo and placenta: possible role in patterning of the extra-embryonic membranes. Dev Dyn. 1995;204(3):219-27. http://dx.doi.org/10.1002/aja.1002040302. PMid:8573715.
http://dx.doi.org/10.1002/aja.1002040302...
). In mouse embryos, since the 8-cell stage, Cdx2 mRNA traces are identified, and Cdx2 expression is observed in the morula stage (approximately from 16-cells) in the nucleus and the cytoplasm of outer cells. At the expanded blastocyst stage, Cdx2 is restricted to the nucleus of these cells (Strumpf et al., 2005Strumpf D, Mao CA, Yamanaka Y, Ralston A, Chawengsaksophak K, Beck F, Rossant J. Cdx2 is required for correct cell fate specification and differentiation of trophectoderm in the mouse blastocyst. Development. 2005;132(9):2093-102. http://dx.doi.org/10.1242/dev.01801. PMid:15788452.
http://dx.doi.org/10.1242/dev.01801...
; Ralston and Rossant, 2005Ralston A, Rossant J. Genetic regulation of stem cell origins in the mouse embryo. Clin Genet. 2005;68(2):106-12. http://dx.doi.org/10.1111/j.1399-0004.2005.00478.x. PMid:15996204.
http://dx.doi.org/10.1111/j.1399-0004.20...
). In bovine embryos, CDX2 is present only at the blastocyst stage (Madeja et al., 2013Madeja ZE, Sosnowski J, Hryniewicz K, Warzych E, Pawlak P, Rozwadowska N, Plusa B, Lechniak D. Changes in sub-cellular localization of trophoblast and inner cell mass-specific transcription factors during bovine preimplantation development. BMC Dev Biol. 2013;13(1):32. http://dx.doi.org/10.1186/1471-213X-13-32. PMid:23941255.
http://dx.doi.org/10.1186/1471-213X-13-3...
; Goissis and Cibelli, 2014bGoissis MD, Cibelli JB. Functional characterization of CDX2 during bovine preimplantation development in vitro. Mol Reprod Dev. 2014b;81(10):962-70. http://dx.doi.org/10.1002/mrd.22415. PMid:25251051.
http://dx.doi.org/10.1002/mrd.22415...
). The deletion of Cdx2 in mouse and bovine embryos influences the epithelium integrity, which becomes more permeable with CDX2 silencing, but does not impair TE or blastocyst formation, reinforcing that TE differentiation is regulated by factors or pathways hierarchically superior to CDX2 (Strumpf et al., 2005Strumpf D, Mao CA, Yamanaka Y, Ralston A, Chawengsaksophak K, Beck F, Rossant J. Cdx2 is required for correct cell fate specification and differentiation of trophectoderm in the mouse blastocyst. Development. 2005;132(9):2093-102. http://dx.doi.org/10.1242/dev.01801. PMid:15788452.
http://dx.doi.org/10.1242/dev.01801...
; Wu et al., 2010Wu G, Gentile L, Fuchikami T, Sutter J, Psathaki K, Esteves TC, Araúzo-Bravo MJ, Ortmeier C, Verberk G, Abe K, Schöler HR. Initiation of trophectoderm lineage specification in mouse embryos is independent of Cdx2. Development. 2010;137(24):4159-69. http://dx.doi.org/10.1242/dev.056630. PMid:21098565.
http://dx.doi.org/10.1242/dev.056630...
; Goissis and Cibelli, 2014bGoissis MD, Cibelli JB. Functional characterization of CDX2 during bovine preimplantation development in vitro. Mol Reprod Dev. 2014b;81(10):962-70. http://dx.doi.org/10.1002/mrd.22415. PMid:25251051.
http://dx.doi.org/10.1002/mrd.22415...
).

In the internal apolar cells, transcription factors related to the ICM pluripotency maintenance such as Oct4 (also known as Pou5f1), Sox2, and Nanog, are initially expressed (Schöler et al., 1990Schöler HR, Dressler GR, Balling R, Rohdewohld H, Gruss P. Oct-4: a germline-specific transcription factor mapping to the mouse t-complex. EMBO J. 1990;9(7):2185-95. http://dx.doi.org/10.1002/j.1460-2075.1990.tb07388.x. PMid:2357966.
http://dx.doi.org/10.1002/j.1460-2075.19...
; Niwa et al., 2000Niwa H, Miyazaki J, Smith AG. Quantitative expression of Oct-3/4 defines differentiation, dedifferentiation or self-renewal of ES cells. Nat Genet. 2000;24(4):372-6. http://dx.doi.org/10.1038/74199. PMid:10742100.
http://dx.doi.org/10.1038/74199...
; Chambers et al., 2003Chambers I, Colby D, Robertson M, Nichols J, Lee S, Tweedie S, Smith A. Functional expression cloning of Nanog, a pluripotency sustaining factor in embryonic stem cells. Cell. 2003;113(5):643-55. http://dx.doi.org/10.1016/S0092-8674(03)00392-1. PMid:12787505.
http://dx.doi.org/10.1016/S0092-8674(03)...
; Nishioka et al., 2008Nishioka N, Yamamoto S, Kiyonari H, Sato H, Sawada A, Ota M, Nakao K, Sasaki H. Tead4 is required for specification of trophectoderm in pre-implantation mouse embryos. Mech Dev. 2008;125(3-4):270-83. http://dx.doi.org/10.1016/j.mod.2007.11.002. PMid:18083014.
http://dx.doi.org/10.1016/j.mod.2007.11....
). In the mouse morula early stages, blastomeres have both Cdx2 and Oct4 expression, but gradually these factors develop a mutual exclusive regulation. Cdx2 or Oct4 exclusive expression is essential for the establishment of TE and ICM strains, respectively (Niwa et al., 2005Niwa H, Toyooka Y, Shimosato D, Strumpf D, Takahashi K, Yagi R, Rossant J. Interaction between Oct3/4 and Cdx2 determines trophectoderm differentiation. Cell. 2005;123(5):917-29. http://dx.doi.org/10.1016/j.cell.2005.08.040. PMid:16325584.
http://dx.doi.org/10.1016/j.cell.2005.08...
; Ralston and Rossant, 2005Ralston A, Rossant J. Genetic regulation of stem cell origins in the mouse embryo. Clin Genet. 2005;68(2):106-12. http://dx.doi.org/10.1111/j.1399-0004.2005.00478.x. PMid:15996204.
http://dx.doi.org/10.1111/j.1399-0004.20...
; Strumpf et al., 2005Strumpf D, Mao CA, Yamanaka Y, Ralston A, Chawengsaksophak K, Beck F, Rossant J. Cdx2 is required for correct cell fate specification and differentiation of trophectoderm in the mouse blastocyst. Development. 2005;132(9):2093-102. http://dx.doi.org/10.1242/dev.01801. PMid:15788452.
http://dx.doi.org/10.1242/dev.01801...
; Guo et al., 2010Guo G, Huss M, Tong GQ, Wang C, Li Sun L, Clarke ND, Robson P. Resolution of cell fate decisions revealed by single-cell gene expression analysis from zygote to blastocyst. Dev Cell. 2010;18(4):675-85. http://dx.doi.org/10.1016/j.devcel.2010.02.012. PMid:20412781.
http://dx.doi.org/10.1016/j.devcel.2010....
). This same mutual suppression mechanism has also been observed between Nanog and Cdx2, indicating Cdx2 role in restricting these transcripts to the ICM (Chen et al., 2009bChen L, Yabuuchi A, Eminli S, Takeuchi A, Lu CW, Hochedlinger K, Daley GQ. Cross-regulation of the Nanog and Cdx2 promoters. Cell Res. 2009b;19(9):1052-61. http://dx.doi.org/10.1038/cr.2009.79. PMid:19564890.
http://dx.doi.org/10.1038/cr.2009.79...
).

In cattle, however, OCT4 is detected in TE and ICM cells, even in the blastocyst stage (Kirchhof et al., 2000Kirchhof N, Carnwath JW, Lemme E, Anastassiadis K, Schöler H, Niemann H. Expression pattern of Oct-4 in preimplantation embryos of different species. Biol Reprod. 2000;63(6):1698-705. http://dx.doi.org/10.1095/biolreprod63.6.1698. PMid:11090438.
http://dx.doi.org/10.1095/biolreprod63.6...
; Kuijk et al., 2008Kuijk EW, Du Puy L, van Tol HT, Oei CH, Haagsman HP, Colenbrander B, Roelen BA. Differences in early lineage segregation between mammals. Dev Dyn. 2008;237(4):918-27. http://dx.doi.org/10.1002/dvdy.21480. PMid:18330925.
http://dx.doi.org/10.1002/dvdy.21480...
), suggesting that in these animals, OCT4 may be involved in other regulatory mechanisms. There is evidence that, unlike what has already been reported in mice, CDX2 does not negatively regulate OCT4 transcription in bovine embryos, due to OCT4 gene promoter region differences in cattle and other species, including human, when compared to mouse (Berg et al., 2011Berg DK, Smith CS, Pearton DJ, Wells DN, Broadhurst R, Donnison M, Pfeffer PL. Trophectoderm lineage determination in cattle. Dev Cell. 2011;20(2):244-55. http://dx.doi.org/10.1016/j.devcel.2011.01.003. PMid:21316591.
http://dx.doi.org/10.1016/j.devcel.2011....
). Still, other studies have demonstrated that CDX2 silencing does not affect OCT4 detection (Goissis and Cibelli, 2014bGoissis MD, Cibelli JB. Functional characterization of CDX2 during bovine preimplantation development in vitro. Mol Reprod Dev. 2014b;81(10):962-70. http://dx.doi.org/10.1002/mrd.22415. PMid:25251051.
http://dx.doi.org/10.1002/mrd.22415...
; Sakurai et al., 2017Sakurai N, Takahashi K, Emura N, Hashizume T, Sawai K. Effects of downregulating TEAD4 transcripts by RNA interference on early development of bovine embryos. J Reprod Dev. 2017;63(2):135-42. http://dx.doi.org/10.1262/jrd.2016-130. PMid:27941302.
http://dx.doi.org/10.1262/jrd.2016-130...
).

Oct4 knockout murine embryos are able to form blastocysts, although these embryos ICM is not pluripotent, and the cells differentiate into extraembryonic trophoblastic strains only (Nichols et al., 1998Nichols J, Zevnik B, Anastassiadis K, Niwa H, Klewe-Nebenius D, Chambers I, Schöler H, Smith A. Formation of pluripotent stem cells in the mammalian embryo depends on the POU transcription factor Oct4. Cell. 1998;95(3):379-91. http://dx.doi.org/10.1016/S0092-8674(00)81769-9. PMid:9814708.
http://dx.doi.org/10.1016/S0092-8674(00)...
), suggesting a requirement for OCT4 in order to achieve ICM development (Niwa et al., 2000Niwa H, Miyazaki J, Smith AG. Quantitative expression of Oct-3/4 defines differentiation, dedifferentiation or self-renewal of ES cells. Nat Genet. 2000;24(4):372-6. http://dx.doi.org/10.1038/74199. PMid:10742100.
http://dx.doi.org/10.1038/74199...
). OCT4 deletion using CRISPR/Cas9 in bovine embryos demonstrated that blastocyst initial formation depends on OCT4 expression since knockout embryos were blocked at the morula stage, prior to the first cell specification, similar to human embryos (Fogarty et al., 2017Fogarty NME, McCarthy A, Snijders KE, Powell BE, Kubikova N, Blakeley P, Lea R, Elder K, Wamaitha SE, Kim D, Maciulyte V, Kleinjung J, Kim JS, Wells D, Vallier L, Bertero A, Turner JMA, Niakan KK. Genome editing reveals a role for OCT4 in human embryogenesis. Nature. 2017;550(7674):67-73. http://dx.doi.org/10.1038/nature24033. PMid:28953884.
http://dx.doi.org/10.1038/nature24033...
; Daigneault et al., 2018Daigneault BW, Rajput S, Smith GW, Ross PJ. Embryonic POU5F1 is required for expanded bovine blastocyst formation. Sci Rep. 2018;8(1):7753. http://dx.doi.org/10.1038/s41598-018-25964-x. PMid:29773834.
http://dx.doi.org/10.1038/s41598-018-259...
). Studies have also indicated that possibly OCT4 has a regulatory role in cattle TE differentiation, probably through CDX2 regulation (Daigneault et al., 2018Daigneault BW, Rajput S, Smith GW, Ross PJ. Embryonic POU5F1 is required for expanded bovine blastocyst formation. Sci Rep. 2018;8(1):7753. http://dx.doi.org/10.1038/s41598-018-25964-x. PMid:29773834.
http://dx.doi.org/10.1038/s41598-018-259...
).

Also necessary for pluripotency maintenance (Avilion et al., 2003Avilion AA, Nicolis SK, Pevny LH, Perez L, Vivian N, Lovell-Badge R. Multipotent cell lineages in early mouse development depend on SOX2 function. Genes Dev. 2003;17(1):126-40. http://dx.doi.org/10.1101/gad.224503. PMid:12514105.
http://dx.doi.org/10.1101/gad.224503...
), SOX2 expression is ICM-restricted at murine blastocysts (Guo et al., 2010Guo G, Huss M, Tong GQ, Wang C, Li Sun L, Clarke ND, Robson P. Resolution of cell fate decisions revealed by single-cell gene expression analysis from zygote to blastocyst. Dev Cell. 2010;18(4):675-85. http://dx.doi.org/10.1016/j.devcel.2010.02.012. PMid:20412781.
http://dx.doi.org/10.1016/j.devcel.2010....
). In mice, maternal SOX2 is located in the nucleus already in 2-cell embryos, while Sox2 zygotic transcription begins lately, at the 16-cell stage, in ICM precursor cells (Avilion et al., 2003Avilion AA, Nicolis SK, Pevny LH, Perez L, Vivian N, Lovell-Badge R. Multipotent cell lineages in early mouse development depend on SOX2 function. Genes Dev. 2003;17(1):126-40. http://dx.doi.org/10.1101/gad.224503. PMid:12514105.
http://dx.doi.org/10.1101/gad.224503...
; Guo et al., 2010Guo G, Huss M, Tong GQ, Wang C, Li Sun L, Clarke ND, Robson P. Resolution of cell fate decisions revealed by single-cell gene expression analysis from zygote to blastocyst. Dev Cell. 2010;18(4):675-85. http://dx.doi.org/10.1016/j.devcel.2010.02.012. PMid:20412781.
http://dx.doi.org/10.1016/j.devcel.2010....
). Despite the early NANOG, OCT4, and CDX2 co-localization pattern, there is no evidence of SOX2 expression in external CDX2-positive cells, being the first exclusive marker of ICM-precursors. Studies reveal that SOX2 is restricted to the ICM by CDX2-independent mechanisms but it requires Tead4 and Lats2 (Wicklow et al., 2014Wicklow E, Blij S, Frum T, Hirate Y, Lang RA, Sasaki H, Ralston A. HIPPO pathway members restrict SOX2 to the inner cell mass where it promotes ICM fates in the mouse blastocyst. PLoS Genet. 2014;10(10):e1004618. http://dx.doi.org/10.1371/journal.pgen.1004618. PMid:25340657.
http://dx.doi.org/10.1371/journal.pgen.1...
). Maternal YAP1 and TAZ and zygotic TEAD4 directly prevent SOX2 expression in the early stages of development, a mechanism that ceases in cells that have positioned themselves internally, by the 16-cell stage (Frum et al., 2019Frum T, Watts JL, Ralston A. TEAD4, YAP1 and WWTR1 prevent the premature onset of pluripotency prior to the 16-cell stage. Development. 2019;146(17):dev179861. http://dx.doi.org/10.1242/dev.179861. PMid:31444221.
http://dx.doi.org/10.1242/dev.179861...
). Interestingly, mouse blastomeres showing SOX2 long-term binding to DNA, at the stage of 4-cells, were predisposed to differentiate into ICM (White et al., 2016White MD, Angiolini JF, Alvarez YD, Kaur G, Zhao ZW, Mocskos E, Bruno L, Bissiere S, Levi V, Plachta N. Long-lived binding of Sox2 to DNA predicts cell fate in the four-cell mouse embryo. Cell. 2016;165(1):75-87. http://dx.doi.org/10.1016/j.cell.2016.02.032. PMid:27015308.
http://dx.doi.org/10.1016/j.cell.2016.02...
).

The silencing of maternal and zygotic SOX2 protein impairs mouse blastocyst formation. Most of these embryos have their development stagnated at the morula stage, failing to cavitate. In these modified embryos, the lack of SOX2 does not affect Oct4 and Nanog expression but results in a large reduction in TE transcripts, such as TEAD4, CDX2, Eomes, and YAP, but not GATA3 (Keramari et al., 2010Keramari M, Razavi J, Ingman KA, Patsch C, Edenhofer F, Ward CM, Kimber SJ. Sox2 is essential for formation of trophectoderm in the preimplantation embryo. PLoS One. 2010;5(11):e13952. http://dx.doi.org/10.1371/journal.pone.0013952. PMid:21103067.
http://dx.doi.org/10.1371/journal.pone.0...
). Other studies suggest that Sox2 knockout does not prevent blastocyst formation, but ICM cells differentiate into TE lineage or extraembryonic endoderm (Avilion et al., 2003Avilion AA, Nicolis SK, Pevny LH, Perez L, Vivian N, Lovell-Badge R. Multipotent cell lineages in early mouse development depend on SOX2 function. Genes Dev. 2003;17(1):126-40. http://dx.doi.org/10.1101/gad.224503. PMid:12514105.
http://dx.doi.org/10.1101/gad.224503...
).

In bovine embryos, expression of SOX2 is observed in all cells from the 16-cell stage to the morula stage, while restricted to the ICM in the blastocyst. The knockdown of SOX2 in these embryos reduced NANOG expression in blastocysts, suggesting a regulatory relationship between these transcripts. Unlike murine embryos, OCT4 levels were not compromised, suggesting that SOX2 and OCT4 may not establish a direct regulatory relationship in cattle (Goissis and Cibelli, 2014aGoissis MD, Cibelli JB. Functional characterization of SOX2 in bovine preimplantation embryos. Biol Reprod. 2014a;90(2):30. http://dx.doi.org/10.1095/biolreprod.113.111526. PMid:24389873.
http://dx.doi.org/10.1095/biolreprod.113...
; Xie et al., 2010Xie D, Chen CC, Ptaszek LM, Xiao S, Cao X, Fang F, Ng HH, Lewin HA, Cowan C, Zhong S. Rewirable gene regulatory networks in the preimplantation embryonic development of three mammalian species. Genome Res. 2010;20(6):804-15. http://dx.doi.org/10.1101/gr.100594.109. PMid:20219939.
http://dx.doi.org/10.1101/gr.100594.109...
).

Second cell differentiation event

Initially, it was believed that the decisive factor for cell differentiation within the ICM of murine embryos would be the cell location. The cells near the blastocoel would differentiate into PrE, while the innermost cells of the ICM would give rise to EPI (Rossant, 1975Rossant J. Investigation of the determinative state of the mouse inner cell mass. II. The fate of isolated inner cell masses transferred to the oviduct. J Embryol Exp Morphol. 1975;33(4):991-1001. PMid:1176885.). This hypothesis, however, was challenged by demonstration of differential expression of transcription factors in ICM cells, such as Nanog and Gata6, which are essential for the establishment of a blastocyst composed of three cell lineages (Mitsui et al., 2003Mitsui K, Tokuzawa Y, Itoh H, Segawa K, Murakami M, Takahashi K, Maruyama M, Maeda M, Yamanaka S. The homeoprotein Nanog is required for maintenance of pluripotency in mouse epiblast and ES cells. Cell. 2003;113(5):631-42. http://dx.doi.org/10.1016/S0092-8674(03)00393-3. PMid:12787504.
http://dx.doi.org/10.1016/S0092-8674(03)...
; Chazaud et al., 2006Chazaud C, Yamanaka Y, Pawson T, Rossant J. Early lineage segregation between epiblast and primitive endoderm in mouse blastocysts through the Grb2-MAPK pathway. Dev Cell. 2006;10(5):615-24. http://dx.doi.org/10.1016/j.devcel.2006.02.020. PMid:16678776.
http://dx.doi.org/10.1016/j.devcel.2006....
; Plusa et al., 2008Plusa B, Piliszek A, Frankenberg S, Artus J, Hadjantonakis AK. Distinct sequential cell behaviours direct primitive endoderm formation in the mouse blastocyst. Development. 2008;135(18):3081-91. http://dx.doi.org/10.1242/dev.021519. PMid:18725515.
http://dx.doi.org/10.1242/dev.021519...
).

In the late blastocyst, EPI cells are characterized by the expression of pluripotency factors such as Nanog (Mitsui et al., 2003Mitsui K, Tokuzawa Y, Itoh H, Segawa K, Murakami M, Takahashi K, Maruyama M, Maeda M, Yamanaka S. The homeoprotein Nanog is required for maintenance of pluripotency in mouse epiblast and ES cells. Cell. 2003;113(5):631-42. http://dx.doi.org/10.1016/S0092-8674(03)00393-3. PMid:12787504.
http://dx.doi.org/10.1016/S0092-8674(03)...
; Chambers et al., 2003Chambers I, Colby D, Robertson M, Nichols J, Lee S, Tweedie S, Smith A. Functional expression cloning of Nanog, a pluripotency sustaining factor in embryonic stem cells. Cell. 2003;113(5):643-55. http://dx.doi.org/10.1016/S0092-8674(03)00392-1. PMid:12787505.
http://dx.doi.org/10.1016/S0092-8674(03)...
) and Sox2 (Avilion et al., 2003Avilion AA, Nicolis SK, Pevny LH, Perez L, Vivian N, Lovell-Badge R. Multipotent cell lineages in early mouse development depend on SOX2 function. Genes Dev. 2003;17(1):126-40. http://dx.doi.org/10.1101/gad.224503. PMid:12514105.
http://dx.doi.org/10.1101/gad.224503...
), while PrE cells have a sequential expression activation of Gata6, Gata4 (Fujikura et al., 2002 Fujikura J, Yamato E, Yonemura S, Hosoda K, Masui S, Nakao K, Miyazaki J, Niwa H. Differentiation of embryonic stem cells is induced by GATA factors. Genes Dev. 2002;16(7):784-9. http://dx.doi.org/10.1101/gad.968802. PMid:11937486.
http://dx.doi.org/10.1101/gad.968802...
), Sox17 and Sox7 (Artus et al., 2011Artus J, Piliszek A, Hadjantonakis AK. The primitive endoderm lineage of the mouse blastocyst: sequential transcription factor activation and regulation of differentiation by Sox17. Dev Biol. 2011;350(2):393-404. http://dx.doi.org/10.1016/j.ydbio.2010.12.007. PMid:21146513.
http://dx.doi.org/10.1016/j.ydbio.2010.1...
). In mice, NANOG (Mitsui et al., 2003Mitsui K, Tokuzawa Y, Itoh H, Segawa K, Murakami M, Takahashi K, Maruyama M, Maeda M, Yamanaka S. The homeoprotein Nanog is required for maintenance of pluripotency in mouse epiblast and ES cells. Cell. 2003;113(5):631-42. http://dx.doi.org/10.1016/S0092-8674(03)00393-3. PMid:12787504.
http://dx.doi.org/10.1016/S0092-8674(03)...
) and GATA6 (Fujikura et al., 2002 Fujikura J, Yamato E, Yonemura S, Hosoda K, Masui S, Nakao K, Miyazaki J, Niwa H. Differentiation of embryonic stem cells is induced by GATA factors. Genes Dev. 2002;16(7):784-9. http://dx.doi.org/10.1101/gad.968802. PMid:11937486.
http://dx.doi.org/10.1101/gad.968802...
) are co-expressed in all cells from the 8-cell stage to the blastocyst. At the blastocyst stage, these transcription factors become mutually exclusive in ICM cells, characterizing the “salt-and-pepper pattern”, before the establishment of EPI and PrE lineages (Rossant et al., 2003Rossant J, Chazaud C, Yamanaka Y. Lineage allocation and asymmetries in the early mouse embryo. Philos Trans R Soc Lond B Biol Sci. 2003;358(1436):1341-8, discussion 1349. http://dx.doi.org/10.1098/rstb.2003.1329. PMid:14511480.
http://dx.doi.org/10.1098/rstb.2003.1329...
; Chazaud et al., 2006Chazaud C, Yamanaka Y, Pawson T, Rossant J. Early lineage segregation between epiblast and primitive endoderm in mouse blastocysts through the Grb2-MAPK pathway. Dev Cell. 2006;10(5):615-24. http://dx.doi.org/10.1016/j.devcel.2006.02.020. PMid:16678776.
http://dx.doi.org/10.1016/j.devcel.2006....
; Plusa et al., 2008Plusa B, Piliszek A, Frankenberg S, Artus J, Hadjantonakis AK. Distinct sequential cell behaviours direct primitive endoderm formation in the mouse blastocyst. Development. 2008;135(18):3081-91. http://dx.doi.org/10.1242/dev.021519. PMid:18725515.
http://dx.doi.org/10.1242/dev.021519...
; Guo et al., 2010Guo G, Huss M, Tong GQ, Wang C, Li Sun L, Clarke ND, Robson P. Resolution of cell fate decisions revealed by single-cell gene expression analysis from zygote to blastocyst. Dev Cell. 2010;18(4):675-85. http://dx.doi.org/10.1016/j.devcel.2010.02.012. PMid:20412781.
http://dx.doi.org/10.1016/j.devcel.2010....
).

In cattle, NANOG expression in the ICM begins at the early blastocyst stage, increasing its expression at days 7 and 8 of development (Kuijk et al., 2012Kuijk EW, van Tol LTA, Van de Velde H, Wubbolts R, Welling M, Geijsen N, Roelen BAJ. The roles of FGF and MAP kinase signaling in the segregation of the epiblast and hypoblast cell lineages in bovine and human embryos. Development. 2012;139(5):871–82. http://dx.doi.org /10.1242/dev.071688. PMid: 22278923.
http://dx.doi.org /10.1242/dev.071688...
), although there is some evidence of the onset of NANOG expression at the 8-cell stage (Canizo et al., 2019Canizo JR, Ynsaurralde Rivolta AE, Vazquez Echegaray C, Suvá M, Alberio V, Aller JF, Guberman AS, Salamone DF, Alberio RH, Alberio R. A dose-dependent response to MEK inhibition determines hypoblast fate in bovine embryos. BMC Dev Biol. 2019;19(1):13. http://dx.doi.org/10.1186/s12861-019-0193-9. PMid:31272387.
http://dx.doi.org/10.1186/s12861-019-019...
). GATA6 expression was detected in all embryo cells from the morula stage and its expression was restricted to ICM only after developmental day 8, already at the blastocyst stage. As in murine embryos, blastomeres initially express both NANOG and GATA6, and they progressively become restricted to EPI and PrE precursor cells (Kuijk et al., 2012Kuijk EW, van Tol LTA, Van de Velde H, Wubbolts R, Welling M, Geijsen N, Roelen BAJ. The roles of FGF and MAP kinase signaling in the segregation of the epiblast and hypoblast cell lineages in bovine and human embryos. Development. 2012;139(5):871–82. http://dx.doi.org /10.1242/dev.071688. PMid: 22278923.
http://dx.doi.org /10.1242/dev.071688...
).

Knockout of Nanog in mouse embryos did not alter GATA6 expression, indicating that the initiation of PrE differentiation is independent of Nanog (Frankenberg et al., 2011Frankenberg S, Gerbe F, Bessonnard S, Belville C, Pouchin P, Bardot O, Chazaud C. Primitive endoderm differentiates via a three-step mechanism involving Nanog and RTK signaling. Dev Cell. 2011;21(6):1005-13. http://dx.doi.org/10.1016/j.devcel.2011.10.019. PMid:22172669.
http://dx.doi.org/10.1016/j.devcel.2011....
). Nanog knockdown in embryos using siRNA also have shown GATA6-positive across the ICM, however, these cells failed to activate the PrE differentiation program, evidenced by the absence of Gata4 and Sox17 expression (Frankenberg et al., 2011Frankenberg S, Gerbe F, Bessonnard S, Belville C, Pouchin P, Bardot O, Chazaud C. Primitive endoderm differentiates via a three-step mechanism involving Nanog and RTK signaling. Dev Cell. 2011;21(6):1005-13. http://dx.doi.org/10.1016/j.devcel.2011.10.019. PMid:22172669.
http://dx.doi.org/10.1016/j.devcel.2011....
). Contrary to in the mouse, knockout of NANOG in bovine embryos reduced GATA6 expression (Ortega et al., 2020Ortega MS, Kelleher AM, O’Neil E, Benne J, Cecil R, Spencer TE. NANOG is required to form the epiblast and maintain pluripotency in the bovine embryo. Mol Reprod Dev. 2020;87(1):152-60. http://dx.doi.org/10.1002/mrd.23304. PMid:31803983.
http://dx.doi.org/10.1002/mrd.23304...
), suggesting different PrE differentiation dynamics in the bovine embryo.

One of the signaling pathways that regulate the second differentiation is the FGF/ERK pathway, activated by FGF4 binding to its receptors (FGFRs) (Yamanaka et al., 2010Yamanaka Y, Lanner F, Rossant J. FGF signal-dependent segregation of primitive endoderm and epiblast in the mouse blastocyst. Development. 2010;137(5):715-24. http://dx.doi.org/10.1242/dev.043471. PMid:20147376.
http://dx.doi.org/10.1242/dev.043471...
; Kang et al., 2013Kang M, Piliszek A, Artus J, Hadjantonakis, AK. FGF4 is required for lineage restriction and salt-and-pepper distribution of primitive endoderm factors but not their initial expression in the mouse. Development. 2013;140(2): 267–79. http://dx.doi.org /10.1242/dev.084996. PMid: 23193166.
http://dx.doi.org /10.1242/dev.084996...
; Molotkov et al., 2017Molotkov A, Mazot P, Brewer JR, Cinalli RM, Soriano P. Distinct Requirements for FGFR1 and FGFR2 in Primitive Endoderm Development and Exit from Pluripotency. Dev Cell. 2017;41(5):511-526.e4. http://dx.doi.org/10.1016/j.devcel.2017.05.004. PMid:28552557.
http://dx.doi.org/10.1016/j.devcel.2017....
; Soszynska et al., 2019Soszyńska A, Klimczewska K, Suwińska A. FGF/ERK signaling pathway: how it operates in mammalian preimplantation embryos and embryo-derived stem cells. Int J Dev Biol. 2019;63(3-5):171-86. PMid:31058295.). In mouse embryos, Fgf4 expression could be observed in EPI precursor cells, directly regulated by NANOG (Frankenberg et al., 2011Frankenberg S, Gerbe F, Bessonnard S, Belville C, Pouchin P, Bardot O, Chazaud C. Primitive endoderm differentiates via a three-step mechanism involving Nanog and RTK signaling. Dev Cell. 2011;21(6):1005-13. http://dx.doi.org/10.1016/j.devcel.2011.10.019. PMid:22172669.
http://dx.doi.org/10.1016/j.devcel.2011....
) and, in the neighboring cells, FGF4 binds to its receptors, activating the PrE differentiation program (Yamanaka et al., 2010Yamanaka Y, Lanner F, Rossant J. FGF signal-dependent segregation of primitive endoderm and epiblast in the mouse blastocyst. Development. 2010;137(5):715-24. http://dx.doi.org/10.1242/dev.043471. PMid:20147376.
http://dx.doi.org/10.1242/dev.043471...
; Frankenberg et al., 2011Frankenberg S, Gerbe F, Bessonnard S, Belville C, Pouchin P, Bardot O, Chazaud C. Primitive endoderm differentiates via a three-step mechanism involving Nanog and RTK signaling. Dev Cell. 2011;21(6):1005-13. http://dx.doi.org/10.1016/j.devcel.2011.10.019. PMid:22172669.
http://dx.doi.org/10.1016/j.devcel.2011....
) (Figure 3).

Figure 3
FGF/ERK role at EPI and PrE cell specification in mouse embryos. In the early blastocyst stage, NANOG expression directly regulates Fgf4 transcription, while it downregulates GATA6 expression in EPI precursor cells (red). FGF4, in a paracrine manner, binds to the receptors (FGFRs) in neighboring cells, activating the ERK cascade in PrE precursor cells (blue), leading to GATA6 expression and further inhibition of NANOG.

Mouse embryos with Fgf4 deletion showed NANOG and GATA6 labeling in most ICM cells, from the morula stage to early blastocysts, suggesting that the initial expression of these proteins does not depend on the signaling triggered by FGF4. However, with further development of blastocysts, these embryos formed a predominantly NANOG-positive ICM, suggesting that FGF4 is necessary for the maintenance of GATA6 expression in ICM. The GATA4 and SOX17 expression was not detected in these Fgf4-/- embryos, indicating the need for an active PrE cell differentiation program (Kang et al., 2013Kang M, Piliszek A, Artus J, Hadjantonakis, AK. FGF4 is required for lineage restriction and salt-and-pepper distribution of primitive endoderm factors but not their initial expression in the mouse. Development. 2013;140(2): 267–79. http://dx.doi.org /10.1242/dev.084996. PMid: 23193166.
http://dx.doi.org /10.1242/dev.084996...
).

Bovine embryos treated with exogenous FGF4 formed blastocysts with an ICM composed exclusively of GATA6-positive cells, demonstrating that FGF4 presence can induce gene expression pattern of the PrE precursors and block differentiation into EPI cells (Kuijk et al., 2012Kuijk EW, van Tol LTA, Van de Velde H, Wubbolts R, Welling M, Geijsen N, Roelen BAJ. The roles of FGF and MAP kinase signaling in the segregation of the epiblast and hypoblast cell lineages in bovine and human embryos. Development. 2012;139(5):871–82. http://dx.doi.org /10.1242/dev.071688. PMid: 22278923.
http://dx.doi.org /10.1242/dev.071688...
). Studies have also shown that exogenous FGF2 was also able to induce the formation and proliferation of PrE in bovine cells in vitro, indicating that other resources can be used to trigger the activation of the cell differentiation program in these animals (Yang et al., 2011Yang QE, Fields SD, Zhang K, Ozawa M, Johnson SE, Ealy AD. Fibroblast growth factor 2 promotes primitive endoderm development in bovine blastocyst outgrowths. Biol Reprod. 2011;85(5):946-53. http://dx.doi.org/10.1095/biolreprod.111.093203. PMid:21778141.
http://dx.doi.org/10.1095/biolreprod.111...
).

Studies indicate that the activity of the FGF/ERK pathway depends on the types of FGFR expressed in the cells. Studies in mice embryos have shown that both EPI and PrE precursors present Fgfr1 expression, and Fgfr2 is preferentially expressed in PrE precursor cells only. Fgfr1 or Fgfr2 knockouts demonstrated that FGFR1 has a fundamental role in PrE differentiation, while FGFR2 played a secondary role (Molotkov et al., 2017Molotkov A, Mazot P, Brewer JR, Cinalli RM, Soriano P. Distinct Requirements for FGFR1 and FGFR2 in Primitive Endoderm Development and Exit from Pluripotency. Dev Cell. 2017;41(5):511-526.e4. http://dx.doi.org/10.1016/j.devcel.2017.05.004. PMid:28552557.
http://dx.doi.org/10.1016/j.devcel.2017....
). There are reports describing FGFR1 and FGFR3 expression in the ICM of bovine embryos, but without functional information (Ozawa et al., 2013Ozawa M, Yang QE, Ealy AD. The expression of fibroblast growth factor receptors during early bovine conceptus development and pharmacological analysis of their actions on trophoblast growth in vitro. Reproduction. 2013;145(2):191-201. http://dx.doi.org/10.1530/REP-12-0220. PMid:23241344.
http://dx.doi.org/10.1530/REP-12-0220...
). The location of bovine FGFR2 is controversial, as studies have indicated its presence restricted to TE cells (Ozawa et al., 2013Ozawa M, Yang QE, Ealy AD. The expression of fibroblast growth factor receptors during early bovine conceptus development and pharmacological analysis of their actions on trophoblast growth in vitro. Reproduction. 2013;145(2):191-201. http://dx.doi.org/10.1530/REP-12-0220. PMid:23241344.
http://dx.doi.org/10.1530/REP-12-0220...
), but ICM expression has also been described (Akizawa et al., 2016Akizawa H, Nagatomo H, Odagiri H, Kohri N, Yamauchi N, Yanagawa Y, Nagano M, Takahashi M, Kawahara M. Conserved roles of fibroblast growth factor receptor 2 signaling in the regulation of inner cell mass development in bovine blastocysts. Mol Reprod Dev. 2016;83(6):516-25. http://dx.doi.org/10.1002/mrd.22646. PMid:27060901.
http://dx.doi.org/10.1002/mrd.22646...
) and mainly in PrE cells (Negrón-Pérez and Hansen, 2018Negrón-Pérez VM, Hansen PJ. Role of yes-associated protein 1, angiomotin, and mitogen-activated kinase kinase 1/2 in development of the bovine blastocyst. Biol Reprod. 2018;98(2):170-83. http://dx.doi.org/10.1093/biolre/iox172. PMid:29228123.
http://dx.doi.org/10.1093/biolre/iox172...
).

FGFRs activation causes the recruitment of adapter proteins that will connect FGF signaling and the ERK pathway (Wang et al., 2004Wang Y, Wang F, Sun T, Trostinskaia A, Wygle D, Puscheck E, Rappolee DA. Entire mitogen activated protein kinase (MAPK) pathway is present in preimplantation mouse embryos. Dev Dyn. 2004;231(1):72-87. http://dx.doi.org/10.1002/dvdy.20114. PMid:15305288.
http://dx.doi.org/10.1002/dvdy.20114...
; Soszynska et al., 2019Soszyńska A, Klimczewska K, Suwińska A. FGF/ERK signaling pathway: how it operates in mammalian preimplantation embryos and embryo-derived stem cells. Int J Dev Biol. 2019;63(3-5):171-86. PMid:31058295.). ERKs activation negatively regulates NANOG, since the ERK1-mediated NANOG phosphorylation subsequently caused NANOG degradation (Kim et al., 2014Kim SH, Kim MO, Cho YY, Yao K, Kim DJ, Jeong CH, Yu DH, Bae KB, Cho EJ, Jung SK, Lee MH, Chen H, Kim JY, Bode AM, Dong Z. ERK1 phosphorylates Nanog to regulate protein stability and stem cell self-renewal. Stem Cell Res (Amst). 2014;13(1):1-11. http://dx.doi.org/10.1016/j.scr.2014.04.001. PMid:24793005.
http://dx.doi.org/10.1016/j.scr.2014.04....
), generating an imbalance in the early coexpression pattern of NANOG and GATA6 and contributing to cellular specification into PrE (Plusa et al., 2008Plusa B, Piliszek A, Frankenberg S, Artus J, Hadjantonakis AK. Distinct sequential cell behaviours direct primitive endoderm formation in the mouse blastocyst. Development. 2008;135(18):3081-91. http://dx.doi.org/10.1242/dev.021519. PMid:18725515.
http://dx.doi.org/10.1242/dev.021519...
; Guo et al., 2010Guo G, Huss M, Tong GQ, Wang C, Li Sun L, Clarke ND, Robson P. Resolution of cell fate decisions revealed by single-cell gene expression analysis from zygote to blastocyst. Dev Cell. 2010;18(4):675-85. http://dx.doi.org/10.1016/j.devcel.2010.02.012. PMid:20412781.
http://dx.doi.org/10.1016/j.devcel.2010....
). Manipulation of FGF/ERK downstream pathway elements has evidenced its importance for the initial development success. The deletion of Grb2 in mouse embryos did not impair ICM compaction, but inhibited Gata6 and the expression of other PrE markers, resulting in the absence of PrE formation. However, these embryos maintained Nanog expression in all ICM cells, which developed into EPI, suggesting the role of the FGF/ERK pathway in PrE development (Chazaud et al., 2006Chazaud C, Yamanaka Y, Pawson T, Rossant J. Early lineage segregation between epiblast and primitive endoderm in mouse blastocysts through the Grb2-MAPK pathway. Dev Cell. 2006;10(5):615-24. http://dx.doi.org/10.1016/j.devcel.2006.02.020. PMid:16678776.
http://dx.doi.org/10.1016/j.devcel.2006....
).

Treatment of mouse embryos with FGFR or MEK inhibitors caused the same phenotype observed in blastocysts presenting Grb2 or Gata6 deletion. However, when the treatment was interrupted at the 16-cell stage, there were no changes observed in the expression pattern of ICM transcription factors, suggesting that the FGF/ERK signaling pathway activation occurs in the early blastocyst stages (Yamanaka et al., 2010Yamanaka Y, Lanner F, Rossant J. FGF signal-dependent segregation of primitive endoderm and epiblast in the mouse blastocyst. Development. 2010;137(5):715-24. http://dx.doi.org/10.1242/dev.043471. PMid:20147376.
http://dx.doi.org/10.1242/dev.043471...
).

Bovine embryos treated with MEK inhibitors had a significant decrease in TE and ICM cells numbers (Canizo et al., 2019Canizo JR, Ynsaurralde Rivolta AE, Vazquez Echegaray C, Suvá M, Alberio V, Aller JF, Guberman AS, Salamone DF, Alberio RH, Alberio R. A dose-dependent response to MEK inhibition determines hypoblast fate in bovine embryos. BMC Dev Biol. 2019;19(1):13. http://dx.doi.org/10.1186/s12861-019-0193-9. PMid:31272387.
http://dx.doi.org/10.1186/s12861-019-019...
), as well as increased expression of NANOG and FGF4, and decreased expression of GATA4 and SOX17, at the blastocyst stage, indicating the direct ERK pathway influence at the ICM differentiation process. However, GATA6 labeling was observed in some blastomeres, showing that some cells maintain PrE differentiation program regardless of ERK activation (Kuijk et al., 2012Kuijk EW, van Tol LTA, Van de Velde H, Wubbolts R, Welling M, Geijsen N, Roelen BAJ. The roles of FGF and MAP kinase signaling in the segregation of the epiblast and hypoblast cell lineages in bovine and human embryos. Development. 2012;139(5):871–82. http://dx.doi.org /10.1242/dev.071688. PMid: 22278923.
http://dx.doi.org /10.1242/dev.071688...
; Harris et al., 2013Harris D, Huang B, Oback B. Inhibition of MAP2K and GSK3 signaling promotes bovine blastocyst development and epiblast-associated expression of pluripotency factors. Biol Reprod. 2013;88(3):74. http://dx.doi.org/10.1095/biolreprod.112.103390. PMid:23390165.
http://dx.doi.org/10.1095/biolreprod.112...
; McLean et al., 2014McLean Z, Meng F, Henderson H, Turner P, Oback B. Increased MAP kinase inhibition enhances epiblast-specific gene expression in bovine blastocysts. Biol Reprod. 2014;91(2):49. http://dx.doi.org/10.1095/biolreprod.114.120832. PMid:25009207.
http://dx.doi.org/10.1095/biolreprod.114...
; Negrón-Pérez and Hansen, 2018Negrón-Pérez VM, Hansen PJ. Role of yes-associated protein 1, angiomotin, and mitogen-activated kinase kinase 1/2 in development of the bovine blastocyst. Biol Reprod. 2018;98(2):170-83. http://dx.doi.org/10.1093/biolre/iox172. PMid:29228123.
http://dx.doi.org/10.1093/biolre/iox172...
; Canizo et al., 2019Canizo JR, Ynsaurralde Rivolta AE, Vazquez Echegaray C, Suvá M, Alberio V, Aller JF, Guberman AS, Salamone DF, Alberio RH, Alberio R. A dose-dependent response to MEK inhibition determines hypoblast fate in bovine embryos. BMC Dev Biol. 2019;19(1):13. http://dx.doi.org/10.1186/s12861-019-0193-9. PMid:31272387.
http://dx.doi.org/10.1186/s12861-019-019...
).

This differentiation process is essentially GATA6-dependent in mice since Gata6 knockout embryos did not form PrE lineage, expressing NANOG in all ICM cells (Schrode et al., 2014Schrode N, Saiz N, Di Talia S, Hadjantonakis AK. GATA6 levels modulate primitive endoderm cell fate choice and timing in the mouse blastocyst. Dev Cell. 2014;29(4):454-67. http://dx.doi.org/10.1016/j.devcel.2014.04.011. PMid:24835466.
http://dx.doi.org/10.1016/j.devcel.2014....
). Late expression of transcription factors related to PrE, such as SOX17 and GATA4, seemed dependent on Gata6 and Fgf4-activated signaling (Bessonnard et al., 2014Bessonnard S, De Mot L, Gonze D, Barriol M, Dennis C, Goldbeter A, Dupont G, Chazaud C. Gata6, Nanog and Erk signaling control cell fate in the inner cell mass through a tristable regulatory network. Development. 2014;141(19):3637-48. http://dx.doi.org/10.1242/dev.109678. PMid:25209243.
http://dx.doi.org/10.1242/dev.109678...
). Still, mouse wild type and Gata6 knockout embryos treated with exogenous FGF4 in the 8 cell stage, did not express NANOG, showing that the FGF/ERK pathway can inhibit NANOG independently of Gata6. However, when they were treated in later stages, as in the early blastocyst, NANOG expression was maintained (Bessonnard et al., 2014Bessonnard S, De Mot L, Gonze D, Barriol M, Dennis C, Goldbeter A, Dupont G, Chazaud C. Gata6, Nanog and Erk signaling control cell fate in the inner cell mass through a tristable regulatory network. Development. 2014;141(19):3637-48. http://dx.doi.org/10.1242/dev.109678. PMid:25209243.
http://dx.doi.org/10.1242/dev.109678...
). Currently, no studies downregulated or ablated GATA6 in bovine embryos.

Conclusion

The main knowledge of early embryonic development in mammals derived mostly from studies in mouse models. Research in bovine embryos shows that the biological processes involved in early cell differentiation are not as conserved (summarized in Table 1), for example, the timing and molecular control of CDX2 expression, or the molecular control of genes related to the second cell differentiation. There are still significant literature information gaps about pre-implantation cell differentiation events in humans and previously presented evidence indicates that human embryos have some patterns and regulatory mechanisms that are more similar to bovine embryos. Thus, we suggest the that bovine and mouse models should be studied in parallel for further understanding of the biological mechanisms involved in early embryonic morphological events.

Table 1
Summary of key events and biological mechanisms ocurring in mouse and bovine embryos. EPI: Epiblast; FGFR: Fibroblast Growth Factor Receptor; ICM: Inner Cell Mass; PrE: primitive endoderm; TE: Trophectoderm.
  • Financial support: LEC received funding from FAPESP (grant number 2019/22649-5). GSS received funding from FAPESP (grant number 2019/21303-8), FEL received funding from FAPESP (grant number 2019/03014-9), MDG received funding from FAPESP (grant number 2017/09576-3) and CNPq (grant number 408634/20189).
  • How to cite: Carreiro LE, Santos GS, Luedke FE, Goissis MD. Cell differentiation events in pre-implantation mouse and bovine embryos. Anim Reprod. 2021;18(4):e20210054. https://doi.org/10.1590/1984-3143-AR2021-0054

References

  • Aberle H, Schwartz H, Kemler R. Cadherin-catenin complex: protein interactions and their implications for cadherin function. J Cell Biochem. 1996;61(4):514-23. http://dx.doi.org/10.1002/(SICI)1097-4644(19960616)61:4<514::AID-JCB4>3.0.CO;2-R PMid:8806074.
    » http://dx.doi.org/10.1002/(SICI)1097-4644(19960616)61:4<514::AID-JCB4>3.0.CO;2-R
  • Akizawa H, Nagatomo H, Odagiri H, Kohri N, Yamauchi N, Yanagawa Y, Nagano M, Takahashi M, Kawahara M. Conserved roles of fibroblast growth factor receptor 2 signaling in the regulation of inner cell mass development in bovine blastocysts. Mol Reprod Dev. 2016;83(6):516-25. http://dx.doi.org/10.1002/mrd.22646 PMid:27060901.
    » http://dx.doi.org/10.1002/mrd.22646
  • Alberio R. Regulation of cell fate decisions in early mammalian embryos. Annu Rev Anim Biosci. 2020;8(1):377-93. http://dx.doi.org/10.1146/annurev-animal-021419-083841 PMid:31730400.
    » http://dx.doi.org/10.1146/annurev-animal-021419-083841
  • Anjos SAA, Costa CP, Assumpção MEOA, Visintin JA, Goissis MD. Inhibition of apical domain formation does not block blastocyst development in bovine embryos. Reprod Fertil Dev. 2021;33(10):665. http://dx.doi.org/10.1071/RD20339 PMid:34092280.
    » http://dx.doi.org/10.1071/RD20339
  • Artus J, Piliszek A, Hadjantonakis AK. The primitive endoderm lineage of the mouse blastocyst: sequential transcription factor activation and regulation of differentiation by Sox17. Dev Biol. 2011;350(2):393-404. http://dx.doi.org/10.1016/j.ydbio.2010.12.007 PMid:21146513.
    » http://dx.doi.org/10.1016/j.ydbio.2010.12.007
  • Avilion AA, Nicolis SK, Pevny LH, Perez L, Vivian N, Lovell-Badge R. Multipotent cell lineages in early mouse development depend on SOX2 function. Genes Dev. 2003;17(1):126-40. http://dx.doi.org/10.1101/gad.224503 PMid:12514105.
    » http://dx.doi.org/10.1101/gad.224503
  • Bassalert C, Valverde-Estrella L, Chazaud C. Primitive endoderm differentiation: from specification to epithelialization. Curr Top Dev Biol. 2018;128:81-104. http://dx.doi.org/10.1016/bs.ctdb.2017.12.001 PMid:29477172.
    » http://dx.doi.org/10.1016/bs.ctdb.2017.12.001
  • Beck F, Erler T, Russell A, James R. Expression of Cdx-2 in the mouse embryo and placenta: possible role in patterning of the extra-embryonic membranes. Dev Dyn. 1995;204(3):219-27. http://dx.doi.org/10.1002/aja.1002040302 PMid:8573715.
    » http://dx.doi.org/10.1002/aja.1002040302
  • Becker DL, Davies CS. Role of gap junctions in the development of the preimplantation mouse embryo. Microsc Res Tech. 1995;31(5):364-74. http://dx.doi.org/10.1002/jemt.1070310506 PMid:8534898.
    » http://dx.doi.org/10.1002/jemt.1070310506
  • Berg DK, Smith CS, Pearton DJ, Wells DN, Broadhurst R, Donnison M, Pfeffer PL. Trophectoderm lineage determination in cattle. Dev Cell. 2011;20(2):244-55. http://dx.doi.org/10.1016/j.devcel.2011.01.003 PMid:21316591.
    » http://dx.doi.org/10.1016/j.devcel.2011.01.003
  • Bessonnard S, De Mot L, Gonze D, Barriol M, Dennis C, Goldbeter A, Dupont G, Chazaud C. Gata6, Nanog and Erk signaling control cell fate in the inner cell mass through a tristable regulatory network. Development. 2014;141(19):3637-48. http://dx.doi.org/10.1242/dev.109678 PMid:25209243.
    » http://dx.doi.org/10.1242/dev.109678
  • Brown JJ, Whittingham DG. The roles of pyruvate, lactate and glucose during preimplantation development of embryos from F1 hybrid mice in vitro. Development. 1991;112(1):99-105. http://dx.doi.org/10.1242/dev.112.1.99 PMid:1769345.
    » http://dx.doi.org/10.1242/dev.112.1.99
  • Canizo JR, Ynsaurralde Rivolta AE, Vazquez Echegaray C, Suvá M, Alberio V, Aller JF, Guberman AS, Salamone DF, Alberio RH, Alberio R. A dose-dependent response to MEK inhibition determines hypoblast fate in bovine embryos. BMC Dev Biol. 2019;19(1):13. http://dx.doi.org/10.1186/s12861-019-0193-9 PMid:31272387.
    » http://dx.doi.org/10.1186/s12861-019-0193-9
  • Chambers I, Colby D, Robertson M, Nichols J, Lee S, Tweedie S, Smith A. Functional expression cloning of Nanog, a pluripotency sustaining factor in embryonic stem cells. Cell. 2003;113(5):643-55. http://dx.doi.org/10.1016/S0092-8674(03)00392-1 PMid:12787505.
    » http://dx.doi.org/10.1016/S0092-8674(03)00392-1
  • Chazaud C, Yamanaka Y, Pawson T, Rossant J. Early lineage segregation between epiblast and primitive endoderm in mouse blastocysts through the Grb2-MAPK pathway. Dev Cell. 2006;10(5):615-24. http://dx.doi.org/10.1016/j.devcel.2006.02.020 PMid:16678776.
    » http://dx.doi.org/10.1016/j.devcel.2006.02.020
  • Chen AE, Egli D, Niakan K, Deng J, Akutsu H, Yamaki M, Cowan C, Fitz-Gerald C, Zhang K, Melton DA, Eggan K. Optimal timing of inner cell mass isolation increases the efficiency of human embryonic stem cell derivation and allows generation of sibling cell lines. Cell Stem Cell. 2009a;4(2):103-6. http://dx.doi.org/10.1016/j.stem.2008.12.001 PMid:19200798.
    » http://dx.doi.org/10.1016/j.stem.2008.12.001
  • Chen L, Yabuuchi A, Eminli S, Takeuchi A, Lu CW, Hochedlinger K, Daley GQ. Cross-regulation of the Nanog and Cdx2 promoters. Cell Res. 2009b;19(9):1052-61. http://dx.doi.org/10.1038/cr.2009.79 PMid:19564890.
    » http://dx.doi.org/10.1038/cr.2009.79
  • Chen J, Zhang M. The Par3/Par6/aPKC complex and epithelial cell polarity. Exp Cell Res. 2013;319(10):1357-64. http://dx.doi.org/10.1016/j.yexcr.2013.03.021 PMid:23535009.
    » http://dx.doi.org/10.1016/j.yexcr.2013.03.021
  • Chi F, Sharpley MS, Nagaraj R, Roy SS, Banerjee U. Glycolysis-independent glucose metabolism distinguishes TE from ICM fate during mammalian embryogenesis. Dev Cell. 2020;53(1):9-26.e4. http://dx.doi.org/10.1016/j.devcel.2020.02.015 PMid:32197068.
    » http://dx.doi.org/10.1016/j.devcel.2020.02.015
  • Cockburn K, Rossant J. Making the blastocyst: lessons from the mouse. J Clin Invest. 2010;120(4):995-1003. http://dx.doi.org/10.1172/JCI41229 PMid:20364097.
    » http://dx.doi.org/10.1172/JCI41229
  • Daigneault BW, Rajput S, Smith GW, Ross PJ. Embryonic POU5F1 is required for expanded bovine blastocyst formation. Sci Rep. 2018;8(1):7753. http://dx.doi.org/10.1038/s41598-018-25964-x PMid:29773834.
    » http://dx.doi.org/10.1038/s41598-018-25964-x
  • Fleming TP, Johnson MH. From egg to epithelium. Annu Rev Cell Biol. 1988;4(1):459-85. http://dx.doi.org/10.1146/annurev.cb.04.110188.002331 PMid:3058163.
    » http://dx.doi.org/10.1146/annurev.cb.04.110188.002331
  • Fleming TP. A quantitative analysis of cell allocation to trophectoderm and inner cell mass in the mouse blastocyst. Dev Biol. 1987;119(2):520-31. http://dx.doi.org/10.1016/0012-1606(87)90055-8 PMid:3803716.
    » http://dx.doi.org/10.1016/0012-1606(87)90055-8
  • Fogarty NME, McCarthy A, Snijders KE, Powell BE, Kubikova N, Blakeley P, Lea R, Elder K, Wamaitha SE, Kim D, Maciulyte V, Kleinjung J, Kim JS, Wells D, Vallier L, Bertero A, Turner JMA, Niakan KK. Genome editing reveals a role for OCT4 in human embryogenesis. Nature. 2017;550(7674):67-73. http://dx.doi.org/10.1038/nature24033 PMid:28953884.
    » http://dx.doi.org/10.1038/nature24033
  • Frankenberg S, Gerbe F, Bessonnard S, Belville C, Pouchin P, Bardot O, Chazaud C. Primitive endoderm differentiates via a three-step mechanism involving Nanog and RTK signaling. Dev Cell. 2011;21(6):1005-13. http://dx.doi.org/10.1016/j.devcel.2011.10.019 PMid:22172669.
    » http://dx.doi.org/10.1016/j.devcel.2011.10.019
  • Frum T, Watts JL, Ralston A. TEAD4, YAP1 and WWTR1 prevent the premature onset of pluripotency prior to the 16-cell stage. Development. 2019;146(17):dev179861. http://dx.doi.org/10.1242/dev.179861 PMid:31444221.
    » http://dx.doi.org/10.1242/dev.179861
  • Fujikura J, Yamato E, Yonemura S, Hosoda K, Masui S, Nakao K, Miyazaki J, Niwa H. Differentiation of embryonic stem cells is induced by GATA factors. Genes Dev. 2002;16(7):784-9. http://dx.doi.org/10.1101/gad.968802 PMid:11937486.
    » http://dx.doi.org/10.1101/gad.968802
  • Gardner DK. Changes in requirements and utilization of nutrients during mammalian preimplantation embryo development and their significance in embryo culture. Theriogenology. 1998;49(1):83-102. http://dx.doi.org/10.1016/S0093-691X(97)00404-4 PMid:10732123.
    » http://dx.doi.org/10.1016/S0093-691X(97)00404-4
  • Gerri C, McCarthy A, Alanis-Lobato G, Demtschenko A, Bruneau A, Loubersac S, Fogarty NME, Hampshire D, Elder K, Snell P, Christie L, David L, van de Velde H, Fouladi-Nashta AA, Niakan KK. Initiation of a conserved trophectoderm program in human, cow and mouse embryos. Nature. 2020;587(7834):443-7. http://dx.doi.org/10.1038/s41586-020-2759-x PMid:32968278.
    » http://dx.doi.org/10.1038/s41586-020-2759-x
  • Goissis MD, Cibelli JB. Functional characterization of SOX2 in bovine preimplantation embryos. Biol Reprod. 2014a;90(2):30. http://dx.doi.org/10.1095/biolreprod.113.111526 PMid:24389873.
    » http://dx.doi.org/10.1095/biolreprod.113.111526
  • Goissis MD, Cibelli JB. Functional characterization of CDX2 during bovine preimplantation development in vitro. Mol Reprod Dev. 2014b;81(10):962-70. http://dx.doi.org/10.1002/mrd.22415 PMid:25251051.
    » http://dx.doi.org/10.1002/mrd.22415
  • Graham CF, Deussen ZA. Features of cell lineage in preimplantation mouse development. J Embryol Exp Morphol. 1978;48(1):53-72. http://dx.doi.org/10.1242/dev.48.1.53 PMid:581769.
    » http://dx.doi.org/10.1242/dev.48.1.53
  • Guo G, Huss M, Tong GQ, Wang C, Li Sun L, Clarke ND, Robson P. Resolution of cell fate decisions revealed by single-cell gene expression analysis from zygote to blastocyst. Dev Cell. 2010;18(4):675-85. http://dx.doi.org/10.1016/j.devcel.2010.02.012 PMid:20412781.
    » http://dx.doi.org/10.1016/j.devcel.2010.02.012
  • Harris D, Huang B, Oback B. Inhibition of MAP2K and GSK3 signaling promotes bovine blastocyst development and epiblast-associated expression of pluripotency factors. Biol Reprod. 2013;88(3):74. http://dx.doi.org/10.1095/biolreprod.112.103390 PMid:23390165.
    » http://dx.doi.org/10.1095/biolreprod.112.103390
  • Johnson MH, McConnell JM. Lineage allocation and cell polarity during mouse embryogenesis. Semin Cell Dev Biol. 2004;15(5):583-97. http://dx.doi.org/10.1016/j.semcdb.2004.04.002 PMid:15271304.
    » http://dx.doi.org/10.1016/j.semcdb.2004.04.002
  • Johnson MH, Ziomek CA. The foundation of two distinct cell lineages within the mouse morula. Cell. 1981;24(1):71-80. http://dx.doi.org/10.1016/0092-8674(81)90502-X PMid:7237545.
    » http://dx.doi.org/10.1016/0092-8674(81)90502-X
  • Johnson MH. From mouse egg to mouse embryo: polarities, axes, and tissues. Annu Rev Cell Dev Biol. 2009;25(1):483-512. http://dx.doi.org/10.1146/annurev.cellbio.042308.113348 PMid:19575654.
    » http://dx.doi.org/10.1146/annurev.cellbio.042308.113348
  • Johnson MT, Mahmood S, Patel MS. Intermediary metabolism and energetics during murine early embryogenesis. J Biol Chem. 2003;278(34):31457-60. http://dx.doi.org/10.1074/jbc.R300002200 PMid:12788927.
    » http://dx.doi.org/10.1074/jbc.R300002200
  • Kaneko KJ, DePamphilis ML. TEAD4 establishes the energy homeostasis essential for blastocoel formation. Development. 2013;140(17):3680-90. http://dx.doi.org/10.1242/dev.093799 PMid:23903192.
    » http://dx.doi.org/10.1242/dev.093799
  • Kang M, Piliszek A, Artus J, Hadjantonakis, AK. FGF4 is required for lineage restriction and salt-and-pepper distribution of primitive endoderm factors but not their initial expression in the mouse. Development. 2013;140(2): 267–79. http://dx.doi.org /10.1242/dev.084996 PMid: 23193166.
    » http://dx.doi.org /10.1242/dev.084996
  • Keramari M, Razavi J, Ingman KA, Patsch C, Edenhofer F, Ward CM, Kimber SJ. Sox2 is essential for formation of trophectoderm in the preimplantation embryo. PLoS One. 2010;5(11):e13952. http://dx.doi.org/10.1371/journal.pone.0013952 PMid:21103067.
    » http://dx.doi.org/10.1371/journal.pone.0013952
  • Kidder GM, Watson AJ. Roles of Na,K-ATPase in early development and trophectoderm differentiation. Semin Nephrol. 2005;25(5):352-5. http://dx.doi.org/10.1016/j.semnephrol.2005.03.011 PMid:16139691.
    » http://dx.doi.org/10.1016/j.semnephrol.2005.03.011
  • Kim SH, Kim MO, Cho YY, Yao K, Kim DJ, Jeong CH, Yu DH, Bae KB, Cho EJ, Jung SK, Lee MH, Chen H, Kim JY, Bode AM, Dong Z. ERK1 phosphorylates Nanog to regulate protein stability and stem cell self-renewal. Stem Cell Res (Amst). 2014;13(1):1-11. http://dx.doi.org/10.1016/j.scr.2014.04.001 PMid:24793005.
    » http://dx.doi.org/10.1016/j.scr.2014.04.001
  • Kirchhof N, Carnwath JW, Lemme E, Anastassiadis K, Schöler H, Niemann H. Expression pattern of Oct-4 in preimplantation embryos of different species. Biol Reprod. 2000;63(6):1698-705. http://dx.doi.org/10.1095/biolreprod63.6.1698 PMid:11090438.
    » http://dx.doi.org/10.1095/biolreprod63.6.1698
  • Korotkevich E, Niwayama R, Courtois A, Friese S, Berger N, Buchholz F, Hiiragi T. The apical domain is required and sufficient for the first lineage segregation in the mouse embryo. Dev Cell. 2017;40(3):235-247.e7. http://dx.doi.org/10.1016/j.devcel.2017.01.006 PMid:28171747.
    » http://dx.doi.org/10.1016/j.devcel.2017.01.006
  • Kuijk EW, Du Puy L, van Tol HT, Oei CH, Haagsman HP, Colenbrander B, Roelen BA. Differences in early lineage segregation between mammals. Dev Dyn. 2008;237(4):918-27. http://dx.doi.org/10.1002/dvdy.21480 PMid:18330925.
    » http://dx.doi.org/10.1002/dvdy.21480
  • Kuijk EW, van Tol LTA, Van de Velde H, Wubbolts R, Welling M, Geijsen N, Roelen BAJ. The roles of FGF and MAP kinase signaling in the segregation of the epiblast and hypoblast cell lineages in bovine and human embryos. Development. 2012;139(5):871–82. http://dx.doi.org /10.1242/dev.071688 PMid: 22278923.
    » http://dx.doi.org /10.1242/dev.071688
  • Lim HYG, Alvarez YD, Gasnier M, Wang Y, Tetlak P, Bissiere S, Wang H, Biro M, Plachta N. Keratins are asymmetrically inherited fate determinants in the mammalian embryo. Nature. 2020;585(7825):404-9. http://dx.doi.org/10.1038/s41586-020-2647-4 PMid:32848249.
    » http://dx.doi.org/10.1038/s41586-020-2647-4
  • Lo CW, Gilula NB. Gap junctional communication in the preimplantation mouse embryo. Cell. 1979;18(2):399-409. http://dx.doi.org/10.1016/0092-8674(79)90059-X PMid:498274.
    » http://dx.doi.org/10.1016/0092-8674(79)90059-X
  • Louvet S, Aghion J, Santa-Maria A, Mangeat P, Maro B. Ezrin becomes restricted to outer cells following asymmetrical division in the preimplantation mouse embryo. Dev Biol. 1996;177(2):568-79. http://dx.doi.org/10.1006/dbio.1996.0186 PMid:8806832.
    » http://dx.doi.org/10.1006/dbio.1996.0186
  • Madeja ZE, Sosnowski J, Hryniewicz K, Warzych E, Pawlak P, Rozwadowska N, Plusa B, Lechniak D. Changes in sub-cellular localization of trophoblast and inner cell mass-specific transcription factors during bovine preimplantation development. BMC Dev Biol. 2013;13(1):32. http://dx.doi.org/10.1186/1471-213X-13-32 PMid:23941255.
    » http://dx.doi.org/10.1186/1471-213X-13-32
  • Maître JL, Turlier H, Illukkumbura R, Eismann B, Niwayama R, Nédélec F, Hiiragi T. Asymmetric division of contractile domains couples cell positioning and fate specification. Nature. 2016;536(7616):344-8. http://dx.doi.org/10.1038/nature18958 PMid:27487217.
    » http://dx.doi.org/10.1038/nature18958
  • McLean Z, Meng F, Henderson H, Turner P, Oback B. Increased MAP kinase inhibition enhances epiblast-specific gene expression in bovine blastocysts. Biol Reprod. 2014;91(2):49. http://dx.doi.org/10.1095/biolreprod.114.120832 PMid:25009207.
    » http://dx.doi.org/10.1095/biolreprod.114.120832
  • Misra JR, Irvine KD. The hippo signaling network and its biological functions. Annu Rev Genet. 2018;52(1):65-87. http://dx.doi.org/10.1146/annurev-genet-120417-031621 PMid:30183404.
    » http://dx.doi.org/10.1146/annurev-genet-120417-031621
  • Mitsui K, Tokuzawa Y, Itoh H, Segawa K, Murakami M, Takahashi K, Maruyama M, Maeda M, Yamanaka S. The homeoprotein Nanog is required for maintenance of pluripotency in mouse epiblast and ES cells. Cell. 2003;113(5):631-42. http://dx.doi.org/10.1016/S0092-8674(03)00393-3 PMid:12787504.
    » http://dx.doi.org/10.1016/S0092-8674(03)00393-3
  • Molotkov A, Mazot P, Brewer JR, Cinalli RM, Soriano P. Distinct Requirements for FGFR1 and FGFR2 in Primitive Endoderm Development and Exit from Pluripotency. Dev Cell. 2017;41(5):511-526.e4. http://dx.doi.org/10.1016/j.devcel.2017.05.004 PMid:28552557.
    » http://dx.doi.org/10.1016/j.devcel.2017.05.004
  • Negrón-Pérez VM, Hansen PJ. Role of yes-associated protein 1, angiomotin, and mitogen-activated kinase kinase 1/2 in development of the bovine blastocyst. Biol Reprod. 2018;98(2):170-83. http://dx.doi.org/10.1093/biolre/iox172 PMid:29228123.
    » http://dx.doi.org/10.1093/biolre/iox172
  • Niakan KK, Eggan K. Analysis of human embryos from zygote to blastocyst reveals distinct gene expression patterns relative to the mouse. Dev Biol. 2013;375(1):54-64. http://dx.doi.org/10.1016/j.ydbio.2012.12.008 PMid:23261930.
    » http://dx.doi.org/10.1016/j.ydbio.2012.12.008
  • Nichols J, Zevnik B, Anastassiadis K, Niwa H, Klewe-Nebenius D, Chambers I, Schöler H, Smith A. Formation of pluripotent stem cells in the mammalian embryo depends on the POU transcription factor Oct4. Cell. 1998;95(3):379-91. http://dx.doi.org/10.1016/S0092-8674(00)81769-9 PMid:9814708.
    » http://dx.doi.org/10.1016/S0092-8674(00)81769-9
  • Nishioka N, Inoue K, Adachi K, Kiyonari H, Ota M, Ralston A, Yabuta N, Hirahara S, Stephenson RO, Ogonuki N, Makita R, Kurihara H, Morin-Kensicki EM, Nojima H, Rossant J, Nakao K, Niwa H, Sasaki H. The Hippo signaling pathway components Lats and Yap pattern Tead4 activity to distinguish mouse trophectoderm from inner cell mass. Dev Cell. 2009;16(3):398-410. http://dx.doi.org/10.1016/j.devcel.2009.02.003 PMid:19289085.
    » http://dx.doi.org/10.1016/j.devcel.2009.02.003
  • Nishioka N, Yamamoto S, Kiyonari H, Sato H, Sawada A, Ota M, Nakao K, Sasaki H. Tead4 is required for specification of trophectoderm in pre-implantation mouse embryos. Mech Dev. 2008;125(3-4):270-83. http://dx.doi.org/10.1016/j.mod.2007.11.002 PMid:18083014.
    » http://dx.doi.org/10.1016/j.mod.2007.11.002
  • Niwa H, Miyazaki J, Smith AG. Quantitative expression of Oct-3/4 defines differentiation, dedifferentiation or self-renewal of ES cells. Nat Genet. 2000;24(4):372-6. http://dx.doi.org/10.1038/74199 PMid:10742100.
    » http://dx.doi.org/10.1038/74199
  • Niwa H, Toyooka Y, Shimosato D, Strumpf D, Takahashi K, Yagi R, Rossant J. Interaction between Oct3/4 and Cdx2 determines trophectoderm differentiation. Cell. 2005;123(5):917-29. http://dx.doi.org/10.1016/j.cell.2005.08.040 PMid:16325584.
    » http://dx.doi.org/10.1016/j.cell.2005.08.040
  • Ortega MS, Kelleher AM, O’Neil E, Benne J, Cecil R, Spencer TE. NANOG is required to form the epiblast and maintain pluripotency in the bovine embryo. Mol Reprod Dev. 2020;87(1):152-60. http://dx.doi.org/10.1002/mrd.23304 PMid:31803983.
    » http://dx.doi.org/10.1002/mrd.23304
  • Ozawa M, Yang QE, Ealy AD. The expression of fibroblast growth factor receptors during early bovine conceptus development and pharmacological analysis of their actions on trophoblast growth in vitro. Reproduction. 2013;145(2):191-201. http://dx.doi.org/10.1530/REP-12-0220 PMid:23241344.
    » http://dx.doi.org/10.1530/REP-12-0220
  • Pesce M, Schöler HR. Oct-4: gatekeeper in the beginnings of mammalian development. Stem Cells. 2001;19(4):271-8. http://dx.doi.org/10.1634/stemcells.19-4-271 PMid:11463946.
    » http://dx.doi.org/10.1634/stemcells.19-4-271
  • Plusa B, Frankenberg S, Chalmers A, Hadjantonakis AK, Moore CA, Papalopulu N, Papaioannou VE, Glover DM, Zernicka-Goetz M. Downregulation of Par3 and aPKC function directs cells towards the ICM in the preimplantation mouse embryo. J Cell Sci. 2005;118(Pt 3):505-15. http://dx.doi.org/10.1242/jcs.01666 PMid:15657073.
    » http://dx.doi.org/10.1242/jcs.01666
  • Plusa B, Piliszek A, Frankenberg S, Artus J, Hadjantonakis AK. Distinct sequential cell behaviours direct primitive endoderm formation in the mouse blastocyst. Development. 2008;135(18):3081-91. http://dx.doi.org/10.1242/dev.021519 PMid:18725515.
    » http://dx.doi.org/10.1242/dev.021519
  • Posfai E, Petropoulos S, Barros FRO, Schell JP, Jurisica I, Sandberg R, Lanner F, Rossant J. Position- and Hippo signaling-dependent plasticity during lineage segregation in the early mouse embryo. eLife. 2017;6:e22906. http://dx.doi.org/10.7554/eLife.22906 PMid:28226240.
    » http://dx.doi.org/10.7554/eLife.22906
  • Ralston A, Rossant J. Cdx2 acts downstream of cell polarization to cell-autonomously promote trophectoderm fate in the early mouse embryo. Dev Biol. 2008;313(2):614-29. http://dx.doi.org/10.1016/j.ydbio.2007.10.054 PMid:18067887.
    » http://dx.doi.org/10.1016/j.ydbio.2007.10.054
  • Ralston A, Rossant J. Genetic regulation of stem cell origins in the mouse embryo. Clin Genet. 2005;68(2):106-12. http://dx.doi.org/10.1111/j.1399-0004.2005.00478.x PMid:15996204.
    » http://dx.doi.org/10.1111/j.1399-0004.2005.00478.x
  • Rossant J, Chazaud C, Yamanaka Y. Lineage allocation and asymmetries in the early mouse embryo. Philos Trans R Soc Lond B Biol Sci. 2003;358(1436):1341-8, discussion 1349. http://dx.doi.org/10.1098/rstb.2003.1329 PMid:14511480.
    » http://dx.doi.org/10.1098/rstb.2003.1329
  • Rossant J. Investigation of the determinative state of the mouse inner cell mass. II. The fate of isolated inner cell masses transferred to the oviduct. J Embryol Exp Morphol. 1975;33(4):991-1001. PMid:1176885.
  • Ruane PT, Tan CMJ, Adlam DJ, Kimber SJ, Brison DR, Aplin JD, Westwood M. Protein O-GlcNAcylation promotes trophoblast differentiation at implantation. Cells. 2020;9(10):2246. http://dx.doi.org/10.3390/cells9102246 PMid:33036308.
    » http://dx.doi.org/10.3390/cells9102246
  • Sakurai N, Takahashi K, Emura N, Hashizume T, Sawai K. Effects of downregulating TEAD4 transcripts by RNA interference on early development of bovine embryos. J Reprod Dev. 2017;63(2):135-42. http://dx.doi.org/10.1262/jrd.2016-130 PMid:27941302.
    » http://dx.doi.org/10.1262/jrd.2016-130
  • Schöler HR, Dressler GR, Balling R, Rohdewohld H, Gruss P. Oct-4: a germline-specific transcription factor mapping to the mouse t-complex. EMBO J. 1990;9(7):2185-95. http://dx.doi.org/10.1002/j.1460-2075.1990.tb07388.x PMid:2357966.
    » http://dx.doi.org/10.1002/j.1460-2075.1990.tb07388.x
  • Schrode N, Saiz N, Di Talia S, Hadjantonakis AK. GATA6 levels modulate primitive endoderm cell fate choice and timing in the mouse blastocyst. Dev Cell. 2014;29(4):454-67. http://dx.doi.org/10.1016/j.devcel.2014.04.011 PMid:24835466.
    » http://dx.doi.org/10.1016/j.devcel.2014.04.011
  • Sharma J, Madan P. Characterisation of the Hippo signalling pathway during bovine preimplantation embryo development. Reprod Fertil Dev. 2020;32(4):392-401. http://dx.doi.org/10.1071/RD18320 PMid:31718770.
    » http://dx.doi.org/10.1071/RD18320
  • Sheng G. Epiblast morphogenesis before gastrulation. Dev Biol. 2015;401(1):17-24. http://dx.doi.org/10.1016/j.ydbio.2014.10.003 PMid:25446532.
    » http://dx.doi.org/10.1016/j.ydbio.2014.10.003
  • Soszyńska A, Klimczewska K, Suwińska A. FGF/ERK signaling pathway: how it operates in mammalian preimplantation embryos and embryo-derived stem cells. Int J Dev Biol. 2019;63(3-5):171-86. PMid:31058295.
  • Strumpf D, Mao CA, Yamanaka Y, Ralston A, Chawengsaksophak K, Beck F, Rossant J. Cdx2 is required for correct cell fate specification and differentiation of trophectoderm in the mouse blastocyst. Development. 2005;132(9):2093-102. http://dx.doi.org/10.1242/dev.01801 PMid:15788452.
    » http://dx.doi.org/10.1242/dev.01801
  • Tarkowski AK, Wróblewska J. Development of blastomeres of mouse eggs isolated at the 4- and 8-cell stage. J Embryol Exp Morphol. 1967;18(1):155-80. http://dx.doi.org/10.1242/dev.18.1.155 PMid:6048976.
    » http://dx.doi.org/10.1242/dev.18.1.155
  • Underhill LA, Robins JC. Trophoblast development in the murine preimplantation embryo. Semin Reprod Med. 2016;34(1):57-62. http://dx.doi.org/10.1055/s-0035-1570025 PMid:26757060.
    » http://dx.doi.org/10.1055/s-0035-1570025
  • van Soom A, Boerjan ML, Bols PE, Vanroose G, Lein A, Coryn M, de Kruif A. Timing of compaction and inner cell allocation in bovine embryos produced in vivo after superovulation. Biol Reprod. 1997;57(5):1041-9. http://dx.doi.org/10.1095/biolreprod57.5.1041 PMid:9369168.
    » http://dx.doi.org/10.1095/biolreprod57.5.1041
  • van Soom A, van Vlaenderen I, Mahmoudzadeh AR, Deluyker H, de Kruif A. Compaction rate of in vitro fertilized bovine embryos related to the interval from insemination to first cleavage. Theriogenology. 1992;38(5):905-19. http://dx.doi.org/10.1016/0093-691X(92)90165-N PMid:16727189.
    » http://dx.doi.org/10.1016/0093-691X(92)90165-N
  • Vinot S, Le T, Ohno S, Pawson T, Maro B, Louvet-Vallée S. Asymmetric distribution of PAR proteins in the mouse embryo begins at the 8-cell stage during compaction. Dev Biol. 2005;282(2):307-19. http://dx.doi.org/10.1016/j.ydbio.2005.03.001 PMid:15950600.
    » http://dx.doi.org/10.1016/j.ydbio.2005.03.001
  • Wang Y, Wang F, Sun T, Trostinskaia A, Wygle D, Puscheck E, Rappolee DA. Entire mitogen activated protein kinase (MAPK) pathway is present in preimplantation mouse embryos. Dev Dyn. 2004;231(1):72-87. http://dx.doi.org/10.1002/dvdy.20114 PMid:15305288.
    » http://dx.doi.org/10.1002/dvdy.20114
  • Watson AJ, Westhusin ME, De Sousa PA, Betts DH, Barcroft LC. Gene expression regulating blastocyst formation. Theriogenology. 1999;51(1):117-33. http://dx.doi.org/10.1016/S0093-691X(98)00236-2 PMid:10729067.
    » http://dx.doi.org/10.1016/S0093-691X(98)00236-2
  • Wei Q, Zhong L, Zhang S, Mu H, Xiang J, Yue L, Dai Y, Han J. Bovine lineage specification revealed by single-cell gene expression analysis from zygote to blastocyst. Biol Reprod. 2017;97(1):5-17. http://dx.doi.org/10.1093/biolre/iox071 PMid:28859285.
    » http://dx.doi.org/10.1093/biolre/iox071
  • White MD, Angiolini JF, Alvarez YD, Kaur G, Zhao ZW, Mocskos E, Bruno L, Bissiere S, Levi V, Plachta N. Long-lived binding of Sox2 to DNA predicts cell fate in the four-cell mouse embryo. Cell. 2016;165(1):75-87. http://dx.doi.org/10.1016/j.cell.2016.02.032 PMid:27015308.
    » http://dx.doi.org/10.1016/j.cell.2016.02.032
  • Wicklow E, Blij S, Frum T, Hirate Y, Lang RA, Sasaki H, Ralston A. HIPPO pathway members restrict SOX2 to the inner cell mass where it promotes ICM fates in the mouse blastocyst. PLoS Genet. 2014;10(10):e1004618. http://dx.doi.org/10.1371/journal.pgen.1004618 PMid:25340657.
    » http://dx.doi.org/10.1371/journal.pgen.1004618
  • Wrenzycki C. Gene expression analysis and in vitro production procedures for bovine preimplantation embryos: past highlights, present concepts and future prospects. Reprod Domest Anim. 2018;53(Suppl 2):14-9. http://dx.doi.org/10.1111/rda.13260 PMid:30238652.
    » http://dx.doi.org/10.1111/rda.13260
  • Wu G, Gentile L, Fuchikami T, Sutter J, Psathaki K, Esteves TC, Araúzo-Bravo MJ, Ortmeier C, Verberk G, Abe K, Schöler HR. Initiation of trophectoderm lineage specification in mouse embryos is independent of Cdx2. Development. 2010;137(24):4159-69. http://dx.doi.org/10.1242/dev.056630 PMid:21098565.
    » http://dx.doi.org/10.1242/dev.056630
  • Xie D, Chen CC, Ptaszek LM, Xiao S, Cao X, Fang F, Ng HH, Lewin HA, Cowan C, Zhong S. Rewirable gene regulatory networks in the preimplantation embryonic development of three mammalian species. Genome Res. 2010;20(6):804-15. http://dx.doi.org/10.1101/gr.100594.109 PMid:20219939.
    » http://dx.doi.org/10.1101/gr.100594.109
  • Yagi R, Kohn MJ, Karavanova I, Kaneko KJ, Vullhorst D, DePamphilis ML, Buonanno A. Transcription factor TEAD4 specifies the trophectoderm lineage at the beginning of mammalian development. Development. 2007;134(21):3827-36. http://dx.doi.org/10.1242/dev.010223 PMid:17913785.
    » http://dx.doi.org/10.1242/dev.010223
  • Yamanaka Y, Lanner F, Rossant J. FGF signal-dependent segregation of primitive endoderm and epiblast in the mouse blastocyst. Development. 2010;137(5):715-24. http://dx.doi.org/10.1242/dev.043471 PMid:20147376.
    » http://dx.doi.org/10.1242/dev.043471
  • Yamanaka Y, Ralston A, Stephenson RO, Rossant J. Cell and molecular regulation of the mouse blastocyst. Dev Dyn. 2006;235(9):2301-14. http://dx.doi.org/10.1002/dvdy.20844 PMid:16773657.
    » http://dx.doi.org/10.1002/dvdy.20844
  • Yang QE, Fields SD, Zhang K, Ozawa M, Johnson SE, Ealy AD. Fibroblast growth factor 2 promotes primitive endoderm development in bovine blastocyst outgrowths. Biol Reprod. 2011;85(5):946-53. http://dx.doi.org/10.1095/biolreprod.111.093203 PMid:21778141.
    » http://dx.doi.org/10.1095/biolreprod.111.093203

Publication Dates

  • Publication in this collection
    07 Jan 2022
  • Date of issue
    2021

History

  • Received
    24 Aug 2021
  • Accepted
    02 Dec 2021
Colégio Brasileiro de Reprodução Animal Coronel José dias Bicalho, 1224, CEP: , 31275-050, Belo Horizonte, MG - Brasil, Tel.: 55-31-3491 7122 - Belo Horizonte - MG - Brazil
E-mail: animreprod.journal@gmail.com