<<

Insights & Perspectives

Cell death and morphogenesis

Think again during early mouse development: Are they interconnected?

Ivan Bedzhov1)2) and Magdalena Zernicka-Goetz1)2)*

Shortly after implantation the embryonic lineage transforms from a coherent ball of the vertebrate limb [1] and until of cells into polarized cup shaped . Recently we elucidated a recently was considered responsible previously unknown apoptosis-independent morphogenic event that reorgan- for the establishment of the hollow tube of the egg cylinder [2, 3]. Alternatively, izes the pluripotent lineage. Polarization cues from the surrounding basement central luminal space can be formed in a membrane rearrange the into a polarized rosette-like structure, where solid cluster by apoptosis-indepen- subsequently a central lumen is established. Thus, we provided a new model dent mechanism if strong polarization revising the current concept of apoptosis-dependent epiblast morphogenesis. cues are provided [4, 5]. In early Cell death however has to be tightly regulated during embryogenesis to ensure such signals originate from the base- ment membrane that surrounds the developmental success. Here, we follow the stages of early mouse development epiblast, and they drive polarization and take a glimpse at the critical signaling and morphogenic events that and lumenogenesis in the embryonic determine cells destiny and reshape the embryonic lineage. lineage during the peri-implantation stages [6]. This concept is contrary to Keywords: the apoptosis-dependent mechanism .apoptosis; ; egg cylinder; epiblast; implantation; morphogenesis of the textbook model describing epi- blast morphogenesis from pre- to post- implantation stages [2, 3].

Introduction diversity of tissues that undergo mor- phogenetic transformations, reshaping Apoptosis and necrosis - Sperm entry triggers the “big bang” that the developing . The first two cell transforms a fertilized egg into a new fate decisions set up the embryonic and major mechanisms of cell organism. Cell fate choices generate a extraembryonic lineages. The embryonic death lineage or the epiblast (EPI) contains pluripotent progenitors that give rise to Multiple cell generations maintain and DOI 10.1002/bies.201400147 all tissues of the foetus. The signals that expand the embryonic and extraembry- organise and support the development onic structures throughout embryogene- 1) Department of Physiology, Development and of the EPI are provided by derivatives of sis. This continuity relies on the tightly Neuroscience, University of Cambridge, Cambridge, UK the two extraembryonic lineages - the balanced processes of cell death and 2) Wellcome Trust/Cancer Research UK Gurdon trophectoderm (TE) and the primitive survival. Cell death can occur following Institute, University of Cambridge, Cambridge, UK (PE) (Fig. 1A). The fitness of the paths of necrosis or apoptosis. *Corresponding author: these early lineages is essential for Necrosis is not a regulated process and Magdalena Zernicka-Goetz embryo survival and development to is usually a result of physical damage or E-mail: [email protected] term. Cells that fail to segregate into ischemia that compromises cell integrity appropriate positions according to their leading to the release of cell contents into Abbreviations: BM, basement membrane; EB, ; EC cell fate program, or lack survival the external environment. In contrast, cell, embryonic cell; ECM, extracellular signals, have to be eliminated to main- (PCD) or apopto- matrix; EPI, epiblast; ES cell, embryonic ; tain integrity and function. Cell sis is a multilevel, heavily regulated ExE, extraembryonic ; ICM, ; PCD, programmed cell death; PE, primitive endo- death also directs morphogenic process- process, activated by external or cell derm; TE, trophectoderm; VE, visceral endoderm. es such as the sculpting of the digits intrinsic stimuli. The intrinsic apoptotic

372 www.bioessays-journal.com Bioessays 37: 372–378, ß 2015 The Authors. Bioessays published by WILEY Periodicals, Inc. This is an open access article under the terms of the Creative Commons Attribution License, which permits use, distribution and reproduction in any medium, provided the original work is properly cited...... Insights & Perspectives I. Bedzhov and M. Zernicka-Goetz hn again Think

Figure 1. Determinants of embryo viability during early mouse development.A:Overview of the pre-, peri- and the early post-implantation stages of mouse embryogenesis. B: Patterns of cytoplasmic movements predict the developmental potential of the . C: Cell division patterns of 2- to 4-cell stage transition in relation to the developmental success to term. D: Paracrine and autocrine pro-survival factors during pre-implantation embryogenesis. E: Cell sorting and elimination of mis-positioned cells by apoptosis during the second cell lineage segregation. F: Activation of the mTOR pathway downstream of the PI3K/Akt signalling cascade. G: Mdm2–p53 regulatory loop regulating embryo survival during the peri-implantation stages of development.

Bioessays 37: 372–378, ß 2015 The Authors. Bioessays published by WILEY Periodicals, Inc. 373 I. Bedzhov and M. Zernicka-Goetz Insights & Perspectives.....

program can be triggered as a response signals can be provided via integrin- and downstream signaling components to DNA damage, high calcium and extracellular matrix interactions [10], oftheIgf,Egf,Tgf-b and Pdgf families are oxidant levels or lack of survival signals, cadherin mediated cell-cell contacts [11] expressed throughout pre-implantation whereas the extrinsic apoptotic cascade or by soluble factors. Thus, a crosstalk of development [14] (Fig. 1D). In addition, E- is downstream of ligand activated cell multiple signalling pathways tightly cadherin (E-cad) mediated adhesion, surface receptors. Membrane blebbing, regulates the balance between cell death which is essential for proper blastocyst nuclear condensation and formation of and survival. Embryo viability directly formation, also provides pro-survival apoptotic bodies are the typical morpho- depends on the fitness of the zygote and cues. The E-cad extracellular domain logical characteristic of cells undergoing embryo’s capacity for full development to interacts with Igf1r, mediating efficient apoptosis. Subsequently, the dying cells term can be predicted as early as the first activation of the receptor [15, 16]. In turn,

Think again and debris are rapidly cleared from the hours post fertilization (Box 1). Observa- Igf1r triggers anti-apoptotic, metabolic tissue by efferocytosis [7, 8]. tions of in vitro and mitogenic responses in developing show that embryos cultured in larger embryo through the PI3K/Akt pathway groups or in smaller volumes of medium [17, 18]. Activated Akt phosphorylates develop to the blastocyst stage with and sequesters pro-apoptotic factors How are the processes of higher rates and contain fewer cells such as BAD, thus keeping the intrinsic cell death and survival undergoing apoptosis in comparison to apoptotic program at bay [19]. balanced during pre- individually cultured embryos [12, 13]. The observed incidence of apoptosis The commonly used media are relatively at the blastocyst stage differs between implantation simple, chemically defined solutions, the TE and the inner cell mass (ICM). development? containing no additional growth factors. PCD in the TE is a relatively rare event, Thus, embryos themselves produce solu- whereastheratesofapoptosisintheICM Apoptosis is suggested as a default cell ble ligands acting in auto- and paracrine are significantly higher [20, 21]. It is destiny that has to be continuously manner, and in larger volumes these proposed that the apoptotic process in suppressed by survival signals [9]. Such factors are diluted out. Ligands, receptors the ICM eliminates cells that failed to

Box 1 How can early embryo survival be predicted? PCD is involved in the process of oogenesis, where most of to 4- cell stage, single lose capacity for full the oocytes are lost during the first stages of meiotic development to term [62]. How can this be explained? On prophase I [57]. Thus, the females are born with a the one hand the 4-cell stage blastomeres are smaller, since substantially lower number of oocytes in comparison to the divisions during the pre-implantation period the early stages of oogenesis. The mature oocytes can increase the cell number, but the total cell volume remains activate multiple signaling cascades, driving cell death constant. These individual blastomeres are still able to form through apoptosis, necrosis and autophagy [58]. After small blastocyst-like structures, known as trophoblastic fertilization the first typical features of PCD can be vesicles, but they fail to develop further. This could be observed in the polar bodies [59]. Since embryogenesis because a minimum of four EPI cells needs to be present in directly depends on the fitness of the zygote, can the the blastocyst before implantation for successful establish- potential for full development be predicted at that stage? ment of the embryo proper, and this number is never Fertilization of the egg triggers cytoplasmic Ca2þ oscil- reached in embryos derived from individual 4-cell stage lations that induce contractions of the actomyosin blastomeres [63]. On the other hand, the cleavage pattern of network, manifested as rhythmic cytoplasmic movements. the 2- to 4-cell stage transition was also found to influence Using high-speed time-lapse imaging the patterns of developmental success. The zygote and 2-cell stage cytoplasmic movements have recently been related to the blastomeres can divide either meridionally (M) or equato- developmental potential of the zygote. Frequent speed rially (E). These sequential divisions generate four different peaks, as well as a low mean basal speed of cytoplasmic kinds of embryos: ME, EM, MM or EE (Fig. 1C). In contrast movement, were found to be associated with a low rate of to the other three classes, the development of EE embryos development to the blastocyst stage in vitro and to birth in to term is significantly reduced [64]. To elucidate the vivo (Fig. 1B). Thus, the dynamics of cytoplasmic flows underlying molecular mechanism and potentially identify a triggered by the sperm are predictive of the embryo’s unique EE transcriptional signature, it will be important to potential to develop to term [60]. correlate the transcriptional profile of individual 4-cell stage The regulative development of pre-implantation embry- blastomeres with their division pattern. It also remains to os enables adaptation to reductions in cell number. If one of be determined whether the history of the EE divisions can the 2-cell stage blastomeres is destroyed, an individual be traced back and correlated with the cytoplasmic totipotent can still complete embryonic devel- movements of that have predicted reduced opment [61]. Within one round of cell divisions, from the 2- developmental potential.

374 Bioessays 37: 372–378, ß 2015 The Authors. Bioessays published by WILEY Periodicals, Inc...... Insights & Perspectives I. Bedzhov and M. Zernicka-Goetz

translocate to the correct position for Interestingly, inactivation of genes as- ment. Only embryos that successfully their fate during the segregation of the sociated with neoplastic transforma- attach and implant stand a chance of PE and EPI cell populations [22–24] tions in adult tissues, such as Brca1 completing embryogenesis. The process hn again Think (Fig. 1E). However, the mechanism by and 2, lead to growth arrest of the early of implantation also boosts cell prolif- which cells sense their “incorrect” posi- egg cylinders [34–36]. Another example eration and, for the first time, embryo tion remains unknown. A potential is the loss of function of Mdm2 oncogene growth is initiated. As the egg cylinder autocrine loop of Egfr activation by the that results in a complete elimination of emerges the EPI dramatically changes Tgf-a ligand is proposed to modulate the all cells in the embryo shortly after its morphology from a simple ball of levels of PCD in the ICM [25]. This is also implantation. Mdm2 binds directly to unpolarised cells into a cup-shaped indicated by the depletion of Egfr, which p53 and inhibits the expression of p53 pseudostratified epithelium, surround- results in complete ICM degeneration in target genes (Fig. 1G). In the absence of ing the proamniotic cavity. the CF-1 mouse genetic background [26]. Mdm2, p53 activity is not regulated and Cavitation and hollowing are the Maintenance of the PE depends on pro- the intrinsic apoptotic program is trig- two major paths through which a solid survival cues downstream of Pdgfra. gered, killing the embryo by E5.5. The cohort of cells can be transformed into Genetic ablation or pharmacological Mdm2–p53 regulatory circuit can be a tube by the generation of a central inhibition of this receptor result in bypassed by combined deletion of both luminal space. Cavitation is an apopto- increased Caspase-3 activity and deple- genes, rescuing the double knockout sis-dependent mechanism of cell elimi- tion of the PE layer [27]. embryos [37, 38]. Deletion of p53 alone nation in the core of a coherent mass of After the second lineage segregation does not affect embryonic development cells. Hollowing is apoptosis-indepen- is complete, the mature blastocyst in general, although a subset of later dent, but requires separation of apical hatches out of the zona and initiates (E13.5–E16.5) embryos exhibit exence- membranes in a radially polarized implantation. The mural TE mediates phaly (location of the brain outside structure [4, 5]. In vitro, the same type the first interactions of the implanting the cranial cavity) [39]. Thus, hyper- of cells are able to follow either of these blastocyst with the maternal environ- activation of p53 leads to global PCD and alternative paths, depending on cell ment. The direct contact between the peri-implantation lethality, whereas in- density and the efficiency of establish- TE cells and the uterine epithelium activation of p53-dependent apoptosis ing apical-basal polarity. For example, induces apoptosis at the attachment has no effect on early embryogenesis. MDCK cells grown at low density and site, allowing penetration of the embryo receiving strong polarization cues po- into the underlying stroma. This apo- larize rapidly and form a central lumen ptotic process is suggested to be a result via hollowing. However, when MDCK of TNF-receptor I activation that triggers Does peri-implantation cells are grown at high density, in the Caspase-3 mediated local PCD of the morphogenesis depend on absence of strong polarization signals, luminal epithelium [28, 29]. the process of apoptosis the central space is gradually formed by apoptosis [44]. Which mechanism is or is there an alternative utilized by the embryo to establish the mechanism? cup-shaped EPI after implantation? Multiple signaling According to a long-standing simple pathways regulate Following implantation the polar TE and elegant two-step model, the EPI was apoptosis and survival forms the extraembryonic ectoderm thought to be reshaped by an apoptosis during the peri- (ExE) at the proximal region of the driven process of cavitation (Fig. 2A). egg cylinder. The ExE contains the This model proposes that at E5.0 the EPI implantation and early multipotent progenitors of the tropho- of the early egg cylinder is a coherent post-implantation stages blast lineage that form the embryonic mass of pluripotent cells. As the egg portion of the placenta. The PE layer cylinder elongates, the VE provides an As the embryo invades the maternal differentiates into parietal endoderm apoptotic signal to eliminate the cells in environment the surrounding stroma (PE) that migrates over the mural TE the core. The EPI cells in direct contact proliferates and transforms into the surface and visceral endoderm (VE) that with the surrounding basement mem- decidua that supports the growth of engulfs the developing egg cylinder. brane (BM) are rescued and form the developing egg cylinder and ensures Signalling centers of the VE pattern the polarized epithelium at the same time foetomaternal immune tolerance [30]. underlying EPI, breaking its symmetry as the cavity is established [2, 3]. The Following implantation, multiple signal- to establish the anterior – posterior evidence supporting this model comes ing cascades regulate the processes of axis [40–43]. from studies using embryoid bodies cell death, survival and proliferation in The process of EPI re-organization (EBs) composed of embryonic stem the embryo. The Igf pathway promotes during peri-implantation stages has (ES) or embryonic carcinoma (EC) cells. survival and cell proliferation by acti- been a long-standing mystery. Embryos Cavitation in EBs never starts from the vating mTOR downstream of the PI3K/ at those stages are no longer free- center, instead multiple peripheral cav- Akt cascade [31] (Fig. 1F). Genetic floating and as they invade the maternal ities are established that coalesce as the inactivation of class IA or class 3 PI3K tissues they become relatively inacces- core is gradually eliminated by PCD. EBs results in embryonic lethality at the time sible. The time of implantation is one of contain hundreds to thousands of cells or shortly after implantation [32, 33]. the most critical periods of develop- and the processes of cavity formation

Bioessays 37: 372–378, ß 2015 The Authors. Bioessays published by WILEY Periodicals, Inc. 375 I. Bedzhov and M. Zernicka-Goetz Insights & Perspectives..... Think again

Figure 2. Models of peri-implantation morphogenesis. A: Apoptosis-dependent two-step signalling mechanism of cavitation in the early embryo. B: Self-organizing mechanism of EPI re-organization into a rosette-like structure, orchestrated by BM-provided polarization cues.

and establishment of epithelial polarity cluster in the center. Although apoptotic the basket-shaped BM can act as a mold, are slow, over the course of several cells and cell debris can be found in transforming the symmetric rosette into days [2, 45]. Does the embryo follow the some embryos, a single central lumen a cup [6, 48]. same path of EPI re-organization? emerges independently of PCD through The BM function in the establish- Using embryos directly isolated from hollowing, most likely via charge repul- ment of apical basal polarity can be the or cultured in sion of apical membranes. Fluid filling substituted in vitro by culturing isolated vitro throughout the corresponding peri- mechanisms such as exocytosis and ICMs in 3D ECM. The EPI cells polarize implantation stages we revealed a pumping are likely to contribute to and initiate lumenogenesis in the ab- strikingly different sequence of events further enlarging the lumen. Similar sence of a surrounding VE layer, (Fig. 2B). The polar TE and the PE secrete morphogenic changes occur in the indicating that a death signal from the ECM proteins that establish a basement ExE, where the processes of polarization VE is not required for this process. membrane (BM) that wraps around the and lumenogenesis generate small inter- Similar morphogenetic changes can be EPI of the late E4.5 blastocyst [46, 47]. membranous spaces that later contrib- induced in ES cells when they are Within 24 hours, polarization cues pro- ute to the mature proamniotic cavity. embedded into 3D ECM at low density. vided by the BM establish apical basal During the differentiation of the polar The ES cell spheres formed in these polarity in the EPI cells, through integrin TE into ExE, the BM that separates conditions efficiently polarize and es- mediated signalling. As a result, at this lineage from the EPI is no longer tablish central lumen, mimicking the the time of implantation the EPI globally maintained (Fig. 1A). Thus, at the early pre- to post-implantation transition re-organizes into a radially polarized egg cylinder stage a common BM of the EPI lineage [6]. Together these rosette-like structure. Constriction of the provided by the VE surrounds both the observations lead to a new model actomyosin network reshapes the ini- EPI and the ExE. Since the EPI cells of peri-implantation morphogenesis, tially round cells, as the apical domains are anchored to the ECM by integrins, where the BM established by the

376 Bioessays 37: 372–378, ß 2015 The Authors. Bioessays published by WILEY Periodicals, Inc...... Insights & Perspectives I. Bedzhov and M. Zernicka-Goetz

extraembryonic tissues acts as niche Acknowledgments mouse preimplantation embryo. J Endocrinol providing instructive signals for the self- We are grateful to Sarah Graham for the 172: 221–36. organization of the EPI independently of valuable feedback on the manuscript 18. Riley JK, Carayannopoulos MO, Wyman AH, Chi M. et al. 2006. Phosphatidylinositol again Think apoptosis. and to the Wellcome Trust for support- 3-kinase activity is critical for glucose metab- ing the research in our group. olism and embryo survival in murine blasto- cysts. J Biol Chem 281: 6010–9. 19. Datta SR, Dudek H, Tao X, Masters S. et al. Conclusions and outlook The authors have declared no conflict of 1997. Akt phosphorylation of BAD couples interest. survival signals to the cell-intrinsic death Cytoplasmic movements in the zygote machinery. Cell 91: 231–41. 20. Brison DR, Schultz RM. 1998. Increased and cell division patterns of the 2-cell incidence of apoptosis in transforming growth stage embryo are the earliest indicators References factor alpha-deficient mouse blastocysts. of an embryo’s potential for full devel- Biol Reprod 59: 136–44. opment to term. As cleavage divisions 1. Zaleske DJ. 1985. Development of the upper 21. Pampfer S. 2000. Apoptosis in rodent peri- limb. Hand clinics 1: 383–90. implantation embryos: Differential suscepti- progressively generate smaller blasto- bility of inner cell mass and trophectoderm 2. Coucouvanis E, Martin GR. 1995. Signals for cell lineages - A review. Placenta 21: S3–10. meres, multiple paracrine and autocrine death and survival: a two-step mechanism for 22. Meilhac SM, Adams RJ, Morris SA, Danck- signals promote embryo viability. The cavitation in the vertebrate embryo. Cell 83: aert A. et al. 2009. Active cell movements 279–87. balance between cell survival and coupled to positional induction are involved in 3. Wolpert L. 2011. Principles of Development. apoptosis is tightly regulated to main- lineage segregation in the mouse blastocyst. Oxford University Press. Dev Biol 331: 210–21. tain the functional integrity of the early 4. Bryant DM, Mostov KE. 2008. From cells to 23. Morris SA, Teo RTY, Li H, Robson P,etal. organs: building polarized tissue. Nat Rev Mol lineages. A burst of cell proliferation in 2010. Origin and formation of the first two Cell Biol 9: 887–901. the implanting embryo expands all cell distinct cell types of the inner cell mass in the 5. Lubarsky B, Krasnow MA. 2003. Tube mouse embryo. Proc Natl Acad Sci USA 107: lineages, alongside a dramatic morpho- morphogenesis: making and shaping biologi- 6364–9. genetic transformation of the EPI. Cells cal tubes. Cell 112: 19–28. 24. Plusa B, Piliszek A, Frankenberg S, 6. Bedzhov I, Zernicka-Goetz M. 2014. Self- that are damaged or miss-positioned are Artus J,etal. 2008. Distinct sequential cell organizing properties of mouse pluripotent behaviours direct primitive endoderm forma- most likely eliminated through apopto- cells initiate morphogenesis upon implanta- tion in the mouse blastocyst. Development sis; however the process of PCD is tion. Cell 156: 1032–44. 135: 3081–91. 7. Vandivier RW, Henson PM, Douglas IS. dispensable for the morphogenetic 25. Brison DR, Schultz RM. 1997. Apoptosis 2006. Burying the dead: the impact of failed changes that take place at that time. during mouse blastocyst formation: evidence apoptotic cell removal (efferocytosis) on for a role for survival factors including trans- The BM components secreted by the chronic inflammatory disease. Chest forming growth factor alpha. Biol Reprod 56: extraembryonic lineages establish a 129: 1673–82. 1088–96. 8. Wyllie AH. 1997. Apoptosis: an overview. Brit niche that provides polarization cues 26. Threadgill DW, Dlugosz AA, Hansen LA, Med Bull 53: 451–65. Tennenbaum T,etal. 1995. Targeted dis- to the pluripotent cells of the implanting 9. Raff MC, Barres BA, Burne JF, Coles HS, ruption of mouse EGF receptor: effect of embryo. In line with this, embryos that et al. 1993. Programmed cell death and the genetic background on mutant phenotype. b control of cell survival: lessons from the fail to assemble BM or lack 1-integrin Science 269: 230–4. . Science 262: 695–700. receptors die during the implantation 27. Artus J, Kang MJ, Cohen-Tannoudji M, 10. Frisch SM, Francis H. 1994. Disruption of Hadjantonakis AK. 2013. PDGF signaling is stages [47, 49, 50]. Similarly, genetic epithelial cell-matrix interactions induces required for primitive endoderm cell survival in apoptosis. J Cell Biol 124: 619–26. ablation of ILK and Pinch1 adaptor the inner cell mass of the mouse blastocyst. proteins result in peri-implantation 11. Hermiston ML, Gordon JI. 1995. In vivo Stem Cells 31: 1932–41. lethality [51–53]. Thus, the integrin analysis of cadherin function in the mouse 28. Joswig A, Gabriel HD, Kibschull M, Win- intestinal epithelium: essential roles in adhe- adaptor complex can serve as a media- terhager E. 2003. Apoptosis in uterine sion, maintenance of differentiation, and epithelium and decidua in response to tor, transmitting signals for polarization regulation of programmed cell death. J Cell implantation: evidence for two different path- in the EPI from the surrounding BM. The Biol 129: 489–506. ways. Reprod Biol Endocrinol 1: 44. 12. Lane M, Gardner DK. 1992. Effect of small GTPase Cdc42 appears as a central 29. Parr EL, Tung HN, Parr MB. 1987. Apoptosis incubation volume and embryo density on as the mode of uterine epithelial cell death player in this process. In epithelial cells the development and viability of mouse during embryo implantation in mice and rats. Cdc42 regulates actin polymerisation, embryos in vitro. Hum Reprod 7: 558–62. Biol Reprod 36: 211–25. cell junctions assembly and signals to 13. Paria BC, Dey SK. 1990. Preimplantation 30. Nancy P, Tagliani E, Tay CS, Asp P, embryo development in vitro: cooperative the myosin light-chain kinase and the et al. 2012. Chemokine gene silencing in interactions among embryos and role of decidual stromal cells limits T cell access to apical determinants of the Par com- growth factors. Proc Natl Acad Sci USA 87: the maternal-fetal interface. Science 336: plex [54]. During early embryogenesis 4756–60. 1317–21. Cdc42 is indispensable for the establish- 14. Kaye PL. 1997. Preimplantation growth factor 31. Yang Q, Guan KL. 2007. Expanding mTOR physiology. Rev Reprod 2: 121–7. signaling. Cell Res 17: 666–81. ment of epithelial polarity in the post- 15. Bedzhov I, Liszewska E, Kanzler B, 32. Bi L, Okabe I, Bernard DJ, Nussbaum RL. implantation EPI [55, 56]. A final Stemmler MP. 2012. Igf1r signaling is 2002. Early embryonic lethality in mice question remains: what is the exact indispensable for preimplantation develop- deficient in the p110beta catalytic subunit ment and is activated via a novel function of of PI 3-kinase. Mamm Genome 13: 169–72. signalling that consolidates signals E-cadherin. PLoS Genet 8: e1002609. 33. Zhou XA, Takatoh J, Wang F. 2011. The from the BM and central epithelial 16. Bedzhov I, Stemmler MP. 2015. Applying mammalian class 3 PI3K (PIK3C3) is required factors such as Cdc42 to establish the Proximity Ligation Assay (PLA) to mouse for early embryogenesis and cell proliferation. apical-basal polarity in the embryonic preimplantation embryos for identifying pro- PLoS One 6. tein-protein interactions in situ. Methods Mol 34. Hakem R, de la Pompa JL, Mak TW. 1998. lineage, and how this process is devel- Biol 1233: 57–64. Developmental studies of Brca1 and Brca2 opmentally regulated remains to be 17. Hardy K, Spanos S. 2002. Growth factor knock-out mice. J Mammary Gland Biol determined. expression and function in the human and Neoplasia 3: 431–5.

Bioessays 37: 372–378, ß 2015 The Authors. Bioessays published by WILEY Periodicals, Inc. 377 I. Bedzhov and M. Zernicka-Goetz Insights & Perspectives.....

35. Liu CY, Flesken-Nikitin A, Li S, Zeng Y,etal. cell polarization and tissue organization. Dev 55. Chen F, Ma L, Parrini MC, Mao X,etal. 1996. Inactivation of the mouse Brca1 gene Cell 4: 613–24. 2000. Cdc42 is required for PIP2-induced leads to failure in the morphogenesis of the 46. Niakan KK, Ji H, Maehr R, Vokes SA,etal. actin polymerization and early develop- egg cylinder in early postimplantation devel- 2010. Sox17 promotes differentiation in ment but not for cell viability. Curr Biol 10: opment. Genes Dev 10: 1835–43. mouse embryonic stem cells by directly 758–65. 36. Suzuki A, delaPompa JL, Hakem R, Elia A, regulating extraembryonic gene expression 56. Wu XW, Li SH, Chrostek-Grashoff A, et al. 1997. Brca2 is required for embryonic and indirectly antagonizing self-renewal. Czuchra A,etal. 2007. Cdc42 is crucial for cellular proliferation in the mouse. Genes Dev Genes Dev 24: 312–26. the establishment of epithelial polarity during 11: 1242–52. 47. Smyth N, Vatansever HS, Murray P, Meyer early mammalian development. Dev Dynam 37. Jones SN, Roe AE, Donehower LA, Bradley M,etal. 1999. Absence of basement 236: 2767–78. A. 1995. Rescue of embryonic lethality in membranes after targeting the LAMC1 gene 57. Lobascio AM, Klinger FG, Scaldaferri ML, Mdm2-deficient mice by absence of p53. results in embryonic lethality due to failure of Farini D,etal. 2007. Analysis of programmed Nature 378: 206–8. endoderm differentiation. J Cell Biol 144: cell death in mouse fetal oocytes. Reproduc-

Think again 38. Montes deOca, Luna R, Wagner DS, 151–60. tion 134: 241–52. Lozano G. 1995. Rescue of early embryonic 48. Bedzhov I, Leung CY, Bialecka M, Zer- 58. De Felici M, Lobascio AM, Klinger FG. lethality in mdm2-deficient mice by deletion of nicka-Goetz M. 2014. In vitro culture of 2008. Cell death in fetal oocytes: many p53. Nature 378: 203–6. mouse blastocysts beyond the implantation players for multiple pathways. Autophagy 4: 39. Sah VP, Attardi LD, Mulligan GJ, Williams stages. Nat Protoc 9: 2732–9. 240–2. BO,etal. 1995. A subset of p53-deficient 49. Fassler R, Meyer M. 1995. Consequences of 59. Fabian D, Cikos S, Rehak P, Koppel J. 2014. embryos exhibit exencephaly. Nat Genet 10: lack of beta 1 integrin gene expression in Do embryonic polar bodies commit suicide? 175–80. mice. Genes Dev 9: 1896–08. Zygote 22: 10–7. 40. Arnold SJ, Robertson EJ. 2009. Making a 50. Stephens LE, Sutherland AE, Klimanskaya 60. Ajduk A, Ilozue T, Windsor S. Yu Y,etal. commitment: cell lineage allocation and axis IV, Andrieux A,etal. 1995. Deletion of beta 1 2011. Rhythmic actomyosin-driven contrac- patterning in the early mouse embryo. Nat integrins in mice results in inner cell mass tions induced by sperm entry predict mam- Rev Mol Cell Biol 10: 91–103. failure and peri-implantation lethality. Genes malian embryo viability. Nat Commun 2: 41. Bedzhov I, Graham SJ, Leung CY, Zernicka- Dev 9: 1883–95. 417. Goetz M. 2014. Developmental plasticity, cell 51. Li S, Bordoy R, Stanchi F, Moser M,etal. 61. Tarkowski AK. 1959. Experiments on the fate specification and morphogenesis in the 2005. PINCH1 regulates cell-matrix and cell- development of isolated blastomeres of early mouse embryo. Philos Trans R Soc Lond cell adhesions, cell polarity and cell survival mouse eggs. Nature 184: 1286–7. BBiolSci369: pii:20130538. during the peri-implantation stage. J Cell Sci 62. Rossant J. 1976. Postimplantation develop- 42. Takaoka K, Hamada H. 2012. Cell fate 118: 2913–21. ment of blastomeres isolated from 4- and 8- decisions and axis determination in the early 52. Liang XQ, Zhou Q, Li XD, Sun YF,etal. 2005. cell mouse eggs. J Embryol Exp Morphol 36: mouse embryo. Development 139: 3–14. PINCH1 plays an essential role in early murine 283–90. 43. Wang H, Dey SK. 2006. Roadmap to embryo embryonic development but is dispensable in 63. Morris SA, Guo Y, Zernicka-Goetz M. 2012. implantation: clues from mouse models. Nat ventricular cardiomyocytes. Mol Cell Biol 25: Developmental plasticity is bound by pluri- Rev Genet 7: 185–99. 3056–62. potency and the Fgf and Wnt signaling 44. Martin-Belmonte F, Yu W, Rodriguez-Fra- 53. Sakai T, Li SH, Docheva D, Grashoff C,etal. pathways. Cell Rep 2: 756–65. ticelli AE, Ewald AJ,etal. 2008. Cell-polarity 2003. Integrin-linked kinase (ILK) is required 64. Piotrowska-Nitsche K, Zernicka-Goetz M. dynamics controls the mechanism of lumen for polarizing the epiblast, cell adhesion, and 2005. Spatial arrangement of individual 4-cell formation in epithelial morphogenesis. Curr controlling actin accumulation. Genes Dev 17: stage blastomeres and the order in which they Biol 18: 507–13. 926–40. are generated correlate with blastocyst pat- 45. Li S, Edgar D, Fassler R, Wadsworth W, 54. Etienne-Manneville S. 2004. Cdc42–the tern in the mouse embryo. Mech Develop 122: et al. 2003. The role of laminin in embryonic centre of polarity. J Cell Sci 117: 1291–300. 487–500.

378 Bioessays 37: 372–378, ß 2015 The Authors. Bioessays published by WILEY Periodicals, Inc.