mechanisms of human embryo development: from cell fate to … · development have so far been...

14
REVIEW Mechanisms of human embryo development: from cell fate to tissue shape and back Marta N. Shahbazi* ABSTRACT Gene regulatory networks and tissue morphogenetic events drive the emergence of shape and function: the pillars of embryo development. Although model systems offer a window into the molecular biology of cell fate and tissue shape, mechanistic studies of our own development have so far been technically and ethically challenging. However, recent technical developments provide the tools to describe, manipulate and mimic human embryos in a dish, thus opening a new avenue to exploring human development. Here, I discuss the evidence that supports a role for the crosstalk between cell fate and tissue shape during early human embryogenesis. This is a critical developmental period, when the body plan is laid out and many pregnancies fail. Dissecting the basic mechanisms that coordinate cell fate and tissue shape will generate an integrated understanding of early embryogenesis and new strategies for therapeutic intervention in early pregnancy loss. KEY WORDS: Human embryo, Morphogenesis, Cell fate, Embryonic stem cells, Aneuploidy, Polarisation Introduction Developmental biologists are faced with a question of form and function. Starting from a relatively simple spore, seed or egg, multiple cell types arise that perform specific functions. These different cells organise into diverse configurations, leading to the emergence of tissues of a defined form. Development requires mechanical changes in cell and tissue shape, which drive morphogenesis, and gene expression changes, which regulate cell fate decisions and tissue patterning. These two developmental processes are not independent, as changes in cell fate can modify the architecture of tissues, and vice versa (Chan et al., 2017). Mechanochemical feedback at the molecular, cellular and tissue levels coordinates this crosstalk and instructs tissue self-organisation (see Glossary, Box 1) (Hannezo and Heisenberg, 2019). Molecular motors generate mechanical forces that are transmitted across tissues via the cytoskeleton and cell-cell adhesion proteins (Heisenberg and Bellaiche, 2013). In the embryo, this leads to changes in cell shape and position, to large tissue-scale deformations, and consequently to morphogenesis. Cells sense mechanical cues a process termed mechanosensation via extracellular matrix (ECM) adhesion proteins (Schwartz, 2010), cell-cell adhesion proteins (Benham-Pyle et al., 2015), stretch- activated ion channels (Coste et al., 2010), mechanosensitive transcription factors (Dupont et al., 2011) or directly by the nucleus (Kirby and Lammerding, 2018). Once sensed, mechanical cues are transduced into biochemical signals a process known as mechanotransduction (Chan et al., 2017). The conversion of mechanical cues into biochemical signals leads to changes in gene expression and protein activity that control cell behaviour, cell fate specification and tissue patterning. Current consensus focuses on two main ideas to explain the emergence of tissue patterns in response to morphogen (see Glossary, Box 1) signals. In the positional informationmodel (Wolpert, 1969), the concentration of a morphogen serves as a coordinate of the position of a cell within a tissue. Cells respond to a specific morphogen concentration making a specific fate choice. Therefore, an existing asymmetry is transformed into a pattern of cell fates. On the contrary, in the reaction-diffusion model (Turing, 1952), pattern formation is a self-organising phenomenon. The basic idea behind this model is the presence of a short-range activator that induces its own expression and the expression of a long-range inhibitor. A spontaneous increase in the local concentration of the activator (a symmetry-breaking event) will induce a further increase in the levels of the activator and expression of the inhibitor. However, as the inhibitor has a higher diffusion rate, this will lead to a peak of activation surrounded by a valley of repression. These two mechanisms, positional information and reaction-diffusion, co-exist during development to generate patterns (Green and Sharpe, 2015). The use of model organisms allows us to explore the mechanisms that orchestrate the crosstalk between cell fate and tissue shape. For example, cytoskeletal forces lead to the acquisition of cell polarity and the differential segregation of cell fate determinants in C. elegans (Goehring et al., 2011), an emerging lumen traps fibroblast growth factor (FGF) molecules, which lead to differentiation in the lateral line primordium of zebrafish (Durdu et al., 2014), and tissue deformations control midgut differentiation in Drosophila embryos (Desprat et al., 2008). As human beings, our own development remains the most significant to study, yet is still mysterious due to technical and ethical limitations (Box 2). In this Review, I draw upon knowledge gained from studies in model organisms, embryonic stem cell research and human embryology to propose mechanistic models of three critical developmental events: compaction and polarisation at the cleavage stage; embryonic epithelialisation at the time of implantation; and pluripotent cell differentiation at gastrulation (Fig. 1). The emerging picture supports a role for the crosstalk between tissue shape and cell fate as a determinant of human embryogenesis. Compaction and polarisation: the first morphogenetic transformation Compaction At the 8- to 16-cell stage, human embryos undergo the process of compaction; blastomeres (see Glossary, Box 1) adhere tightly to MRC Laboratory of Molecular Biology, Francis Crick Avenue, Cambridge Biomedical Campus, Cambridge CB2 0QH, UK. *Author for correspondence ([email protected]) M.N.S., 0000-0002-1599-5747 This is an Open Access article distributed under the terms of the Creative Commons Attribution License (http://creativecommons.org/licenses/by/4.0), which permits unrestricted use, distribution and reproduction in any medium provided that the original work is properly attributed. 1 © 2020. Published by The Company of Biologists Ltd | Development (2020) 147, dev190629. doi:10.1242/dev.190629 DEVELOPMENT

Upload: others

Post on 18-Jul-2020

1 views

Category:

Documents


0 download

TRANSCRIPT

Page 1: Mechanisms of human embryo development: from cell fate to … · development have so far been technically and ethically challenging. However, recent technical developments provide

REVIEW

Mechanisms of human embryo development: from cell fateto tissue shape and backMarta N. Shahbazi*

ABSTRACTGene regulatory networks and tissue morphogenetic events drive theemergence of shape and function: the pillars of embryo development.Although model systems offer a window into the molecular biology ofcell fate and tissue shape, mechanistic studies of our owndevelopment have so far been technically and ethically challenging.However, recent technical developments provide the tools todescribe, manipulate and mimic human embryos in a dish, thusopening a new avenue to exploring human development. Here, Idiscuss the evidence that supports a role for the crosstalk betweencell fate and tissue shape during early human embryogenesis. This isa critical developmental period, when the body plan is laid out andmany pregnancies fail. Dissecting the basic mechanisms thatcoordinate cell fate and tissue shape will generate an integratedunderstanding of early embryogenesis and new strategies fortherapeutic intervention in early pregnancy loss.

KEY WORDS: Human embryo, Morphogenesis, Cell fate, Embryonicstem cells, Aneuploidy, Polarisation

IntroductionDevelopmental biologists are faced with a question of form andfunction. Starting from a relatively simple spore, seed or egg,multiple cell types arise that perform specific functions. Thesedifferent cells organise into diverse configurations, leading to theemergence of tissues of a defined form. Development requiresmechanical changes in cell and tissue shape, which drivemorphogenesis, and gene expression changes, which regulate cellfate decisions and tissue patterning. These two developmentalprocesses are not independent, as changes in cell fate can modify thearchitecture of tissues, and vice versa (Chan et al., 2017).Mechanochemical feedback at the molecular, cellular and tissuelevels coordinates this crosstalk and instructs tissue self-organisation(see Glossary, Box 1) (Hannezo and Heisenberg, 2019).Molecular motors generate mechanical forces that are transmitted

across tissues via the cytoskeleton and cell-cell adhesion proteins(Heisenberg and Bellaiche, 2013). In the embryo, this leads tochanges in cell shape and position, to large tissue-scaledeformations, and consequently to morphogenesis. Cells sensemechanical cues – a process termed mechanosensation – viaextracellular matrix (ECM) adhesion proteins (Schwartz, 2010),cell-cell adhesion proteins (Benham-Pyle et al., 2015), stretch-

activated ion channels (Coste et al., 2010), mechanosensitivetranscription factors (Dupont et al., 2011) or directly by the nucleus(Kirby and Lammerding, 2018). Once sensed, mechanical cuesare transduced into biochemical signals – a process known asmechanotransduction (Chan et al., 2017). The conversion ofmechanical cues into biochemical signals leads to changes ingene expression and protein activity that control cell behaviour, cellfate specification and tissue patterning.

Current consensus focuses on two main ideas to explain theemergence of tissue patterns in response tomorphogen (see Glossary,Box 1) signals. In the ‘positional information’ model (Wolpert,1969), the concentration of a morphogen serves as a coordinate of theposition of a cell within a tissue. Cells respond to a specificmorphogen concentration making a specific fate choice. Therefore,an existing asymmetry is transformed into a pattern of cell fates. Onthe contrary, in the reaction-diffusion model (Turing, 1952), patternformation is a self-organising phenomenon. The basic idea behindthis model is the presence of a short-range activator that induces itsown expression and the expression of a long-range inhibitor. Aspontaneous increase in the local concentration of the activator (asymmetry-breaking event) will induce a further increase in the levelsof the activator and expression of the inhibitor. However, as theinhibitor has a higher diffusion rate, this will lead to a peak ofactivation surrounded by a valley of repression. These twomechanisms, positional information and reaction-diffusion, co-existduring development to generate patterns (Green and Sharpe, 2015).

The use of model organisms allows us to explore the mechanismsthat orchestrate the crosstalk between cell fate and tissue shape. Forexample, cytoskeletal forces lead to the acquisition of cell polarityand the differential segregation of cell fate determinants inC. elegans (Goehring et al., 2011), an emerging lumen trapsfibroblast growth factor (FGF) molecules, which lead todifferentiation in the lateral line primordium of zebrafish (Durduet al., 2014), and tissue deformations control midgut differentiationinDrosophila embryos (Desprat et al., 2008). As human beings, ourown development remains the most significant to study, yet is stillmysterious due to technical and ethical limitations (Box 2). In thisReview, I draw upon knowledge gained from studies in modelorganisms, embryonic stem cell research and human embryology topropose mechanistic models of three critical developmental events:compaction and polarisation at the cleavage stage; embryonicepithelialisation at the time of implantation; and pluripotent celldifferentiation at gastrulation (Fig. 1). The emerging picturesupports a role for the crosstalk between tissue shape and cell fateas a determinant of human embryogenesis.

Compaction and polarisation: the first morphogenetictransformationCompactionAt the 8- to 16-cell stage, human embryos undergo the process ofcompaction; blastomeres (see Glossary, Box 1) adhere tightly to

MRC Laboratory of Molecular Biology, Francis Crick Avenue, CambridgeBiomedical Campus, Cambridge CB2 0QH, UK.

*Author for correspondence ([email protected])

M.N.S., 0000-0002-1599-5747

This is an Open Access article distributed under the terms of the Creative Commons AttributionLicense (http://creativecommons.org/licenses/by/4.0), which permits unrestricted use,distribution and reproduction in any medium provided that the original work is properly attributed.

1

© 2020. Published by The Company of Biologists Ltd | Development (2020) 147, dev190629. doi:10.1242/dev.190629

DEVELO

PM

ENT

Page 2: Mechanisms of human embryo development: from cell fate to … · development have so far been technically and ethically challenging. However, recent technical developments provide

each other, maximising their cell-cell contact area, which leads theembryo to acquire a compressed morphology. This process isconserved in all mammalian species studied so far, but the timingvaries. The precise timing of the onset and completion ofcompaction is important for the successful development of bothmouse and human embryos (Mizobe et al., 2017; Kim et al., 2017;Skiadas et al., 2006). Human embryos in which compaction is eitherinitiated prematurely or delayed have a lower potential to formblastocysts (see Glossary, Box 1). Premature compaction isassociated with cytokinetic failure and multi-nuclear blastomeres(Iwata et al., 2014), whereas delayed compaction is associated witha reduced number of inner cell mass (ICM) cells (see Glossary,Box 1) (Ivec et al., 2011).

The mechanisms that drive compaction in human embryosremain unknown and, instead, have mainly been explored usingmouse embryos. The key players are the actomyosin cytoskeletonand the cell adhesion protein E-cadherin. The actomyosincytoskeleton undergoes pulsed contractions that generate forceand increase membrane tension. This happens specifically at thecell-free surface of the embryo, as E-cadherin keeps contractilitylow at cell-cell adhesion sites (Maitre et al., 2015). An alternativemodel proposes that E-cadherin-containing filopodia are the majordrivers of compaction. By binding to and promoting elongation of

Box 1. GlossaryAmnion. An extra-embryonic membrane that protects thedeveloping foetus from mechanical damage and the maternalimmune response.Amniotes. A clade of vertebrates in which the embryo develops whilebeing protected by extra-embryonic membranes, including the amnion. Itencompasses reptiles, birds and mammals.Amniotic cavity. Fluid-filled sac located between the amnion and theembryo, the function of which is to protect the embryo from mechanicaldamage and the maternal immune system.Blastocoel. Fluid-filled cavity present in blastocyst stage embryos.Blastocyst. An embryo composed of an outer trophectoderm layer, aninner cell mass and an internal blastocoel cavity.Blastomeres. Cells of the early embryo, formed by cleavage of thezygote.Cytotrophoblast. Trophectoderm stem cell compartment of the humanembryo. It plays a key role during blastocyst implantation and it generatesdifferentiated trophectoderm cells such as syncytiotrophoblast andextravillous trophoblast.Ectoderm. Outermost primary germ layer that gives rise to the nervoussystem, the epidermis and its appendages.Embryonic genome activation. The process whereby zygotictranscription is initiated and takes over the maternally stored mRNAsand proteins. In human embryos, it takes place at the four- to eight-cellstage transition.Embryoid body. Three dimensional aggregates of pluripotent stem cellscommonly used to study differentiation.Endoderm. Innermost primary germ layer that gives rise to theepithelial cells of the gastrointestinal, respiratory, endocrine andurinary systems.Epiblast. The pluripotent embryonic tissue that gives rise to germlineand soma, as well as the extra-embryonic amnion.Epithelial-to-mesenchymal transition. Cellular program that leads tothe loss of epithelial features (i.e. polarised organisation and cell-celladhesion) and the acquisition of a mesenchymal phenotype. It plays akey role during gastrulation.Extra-embryonic ectoderm. Trophectoderm-derived tissue that formsduring early post-implantation in mouse embryos.Inner cell mass. A group of cells located inside the blastocyst that givesrise to the epiblast and the hypoblast.Morphogen. A signalling molecule that determines cell fates in aconcentration-dependent manner.Mesoderm. Primary germ layer positioned in between the ectoderm andthe endoderm. It gives rise to muscle, bone, connective tissue, bloodvessels, red and white blood cells, and the mesenchyme of variousorgans such as skin, kidney or gonads.Primary yolk sac. Extra-embryonic membrane formed by hypoblastcells around 7 to 8 days post-fertilisation. By E12-15, it gives rise to asecondary yolk sac, the primary functions of which are hematopoiesisand nutrient transport.Primitive endoderm (mouse) or hypoblast (human). An extra-embryonic tissue characteristic of pre-implantation blastocysts thatgives rise to the yolk sac.Primitive streak. Transient structure that appears in the posteriorepiblast and marks the start of gastrulation.Primordial germ cells. Specialised cells, precursors of the gametes,that are specified in the epiblast of mammalian embryos during earlypost-implantation development.Pro-amniotic cavity. Luminal cavity that is formed in the mouse epiblastat E5.0. The fusion between the epiblast and extra-embryonic ectodermcavities leads to the formation of the amniotic cavity by E5.75 in mouseembryos.Self-organisation. Spontaneous generation of ordered structures thatresults from the interaction between elements that have no previouspattern.Sialomucins. Glycosylated proteins of the mucin family that contain theacidic sugar sialic acid.Trophectoderm. An extra-embryonic tissue present in pre-implantationembryos that gives rise to the placenta.

Box 2. Historical perspective of human embryodevelopmentThe birth of human embryology as a scientific discipline is intimatelylinked to the creation of human embryo collections (Yamada et al., 2015;Gasser et al., 2014). The pioneering work of Franklin Mall led to thecreation of the Carnegie collection in 1887, which harbours more than10,000 human embryo specimens, and established the basic stagingcriteria for the developmental classification of human embryos (Keibeland Mall, 1912). Other collections were later created, such as the Kyotocollection, which today holds ∼44,000 specimens (Nishimura et al.,1968). Much of our current textbook knowledge of human development isderived from the early descriptive studies of these samples.The development of in vitro fertilisation (IVF) of human eggs initiated a

revolution in human embryo and stem cell research and humanreproduction (Edwards et al., 1969; Rock and Menkin, 1944; Shettles,1955). This initial milestone was followed by the development ofconditions to culture fertilised human eggs in vitro for up to 5-6 days(Edwards et al., 1970; Steptoe et al., 1971), and ultimately led to the birthof the first IVF baby in 1978, thanks to the tireless efforts of RobertEdwards, Patrick Steptoe and Jean Purdy. Since then, the field of humanembryology has flourished. IVF has allowed scientists to describe thedynamics of key morphogenetic processes during early humandevelopment, such as cleavage, compaction and blastulation (Wonget al., 2010; Marcos et al., 2015; Iwata et al., 2014); to characterise celllineage specification events by studying the transcriptional andepigenetic profiles of all the cells present in a developing humanembryo (Niakan and Eggan, 2013; Petropoulos et al., 2016; Braudeet al., 1988; Zhu et al., 2018); to identify genetic and chromosomalabnormalities that compromise human embryo development (Munneet al., 2009; Vanneste et al., 2009); and, perhaps more importantly, toestablish human embryonic stem cell lines (Thomson et al., 1998), whichon their own have revolutionised our approach to studying humandevelopment and devising regenerative therapies. However, untilrecently, gene function could not be studied in the context of humanembryos. The recent generation of OCT4 knockout human embryosrepresents a turning point in the field (Fogarty et al., 2017). This studyhighlighted differences in gene function between mouse and humans,and established a gold standard for functional studies in humanembryos. Thus, human embryology is becoming an experimentalscience; I argue that, in the years to come, we will witness a surge inthe number of mechanistic studies exploring our own development.

2

REVIEW Development (2020) 147, dev190629. doi:10.1242/dev.190629

DEVELO

PM

ENT

Page 3: Mechanisms of human embryo development: from cell fate to … · development have so far been technically and ethically challenging. However, recent technical developments provide

the apical surface of neighbouring blastomeres, they trigger the cellshape changes that take place during compaction in a non-cell-autonomous manner (Fierro-Gonzalez et al., 2013). Whether thesemechanisms are active in human embryos remains unknown. Interms of timing, studies in mouse embryos suggest that the onset ofcompaction could be controlled by the decrease in the nuclear/cytoplasmic ratio that takes place during the early cleavage divisionsand/or the dilution of a potential inhibitor of compaction present inthe zygote (Kojima et al., 2014). These are interesting hypothesesthat require further experimental validation.

PolarisationConcomitant with the process of compaction, cells acquireapicobasal polarity. In mouse embryos, the basolateral domainconcentrates cell adhesion proteins, such as E-cadherin, whereas theapical domain is enriched in apical polarity proteins, such as thepartitioning defective (Par) complex and cytoskeletal components(Leung et al., 2016). Recent studies in mice have revealed that

protein kinase C (PKC) activation at the eight-cell stage triggersactomyosin polarisation via RhoA (Zhu et al., 2017). In addition,transcription is required for Par complex polarisation. Thetranscription factors Tfap2c and Tead4 are expressed afterembryonic genome activation (EGA, see Glossary, Box 1) andinduce the expression of several regulators of the actin cytoskeleton,which become progressively upregulated from the two- to eight-cellstage. This leads to a remodelling of the actin network that isnecessary for the clustering of apical polarity proteins, andculminates with the formation of the mature apical domain. As aresult, the combined artificial activation of RhoA and expression ofTfap2c and Tead4 is sufficient to induce premature embryopolarisation and to advance morphogenesis (Zhu et al., 2020).

Studies in human embryos revealed the presence of apicalmicrovilli and basolateral E-cadherin by the time embryos compact(Alikani, 2005; Nikas et al., 1996), but the exact sequence of eventsand the mechanisms leading to human embryo polarisation remainto be explored. Given that EGA precedes polarisation, it is tempting

Embryonic genome activation

Compaction and polarisation

Inside cell generation

Pre-implantation lineage

specification

Naive pluripotency

exit

Epiblast epithelialization

Pro-amniotic cavity

formation

Pluripotency loss and lineage commitment

Epithelial-to-mesenchymal

transition

BlastocoelAmniotic

cavityPrimary yolk sac Primitive streak Yolk sac

Amnion

Placenta

Human developmental time

0 1.5 2 2.5 3 4 5.5 6.5 9.5

Amnion

Mouse developmental time

Fate

Shap

e

Embryonic genome activation

Compaction and polarisation

Pre-implantation lineage

specification

Inside cell generation

Naive pluripotency

exit

Epiblast epithelialisation

Amniotic cavity formation

Amnion specification

Pluripotency loss and lineage commitment

Epithelial-to-mesenchymal

transition

Fate

Shap

e

Implantation

Implantation Pro-amniotic cavity

0 1 2 3 4 6 10 14 80

Fig. 1. Overview of human and mouse embryo development. Upon fertilisation, mouse and human embryos undergo a series of cleavage divisions. Theembryonic genome becomes activated by the two-cell stage in mouse embryos and at the four/eight-cell stage transition in human embryos. It is followed bycompaction and polarisation, which occur at the eight-cell stage in mouse embryos, and between the eight- to 16-cell stage in human embryos. Formation of ahollow cavity, the blastocoel, denotes the formation of the blastocyst, which, by embryonic day E4 (mouse) and E6 (human), is composed of three main tissues:epiblast, hypoblast and trophectoderm. Upon implantation (E5 in mouse and E7 in human), embryos undergo a global morphological transformation. Theembryonic epiblast loses its naïve pluripotent character, becomes epithelial and forms the pro-amniotic cavity (mouse), which spans both the epiblast and thetrophectoderm-derived extra-embryonic ectoderm, and the amniotic cavity (human). A key difference between mouse and human embryos relates to theformation of the amnion.Whereas in mouse embryos amnion formation takes place during gastrulation, in human embryos a subset of epiblast cells differentiatesto form the squamous amniotic epithelium during early post-implantation (E10). As a result, the human epiblast acquires a disc shape and the mouse epiblast acylindrical morphology. On embryonic days 6.5 (mouse) and 14 (human), gastrulation is initiated in the posterior epiblast. Cells undergo an epithelial-to-mesenchymal transition, lose their pluripotent character and commit to one of the germ layers. Epiblast-derived tissues are shown in pink, hypoblast-derivedtissues are shown in blue and trophectoderm-derived tissues are shown in green.

3

REVIEW Development (2020) 147, dev190629. doi:10.1242/dev.190629

DEVELO

PM

ENT

Page 4: Mechanisms of human embryo development: from cell fate to … · development have so far been technically and ethically challenging. However, recent technical developments provide

to speculate that, as in mouse embryos, EGA controls the timing ofhuman embryo polarisation via as yet unknown transcription factors(Zhu et al., 2020) (Fig. 2).

The first cell fate decisionPolarisation is fundamental for the divergence of embryonic andextra-embryonic lineages in mouse embryos. In support of thisnotion, loss-of-function studies demonstrate that Par complexcomponents are needed for the acquisition of an extra-embryonictrophectoderm (TE) (see Glossary, Box 1) fate (Plusa et al., 2005;Alarcon, 2010). Cells that inherit the apical domain differentiatetowards TE, whereas cells that do not inherit the apical domain andare internalised become the ICM, and hence the precursors of theembryonic epiblast (see Glossary, Box 1) and extra-embryonicprimitive endoderm (PE) (Korotkevich et al., 2017) (see Glossary,Box 1). Inner cell allocation is mainly regulated by apicalconstriction, heterogeneities in cortical tension and, to a lesserextent, by the orientation of cell division (Samarage et al., 2015;Anani et al., 2014). This leads to the first spatial segregation of cellsin the mouse embryo: outer polarised cells and inner apolar cells.Therefore, two morphogenetic cues, polarity and position, regulatethe first cell fate decision in mouse embryos.Mechanistically, the transcription factor Yap is responsible for

the ICM-TE bifurcation in mouse embryos (Sasaki, 2015). Inpolarised cells, Yap translocates to the nucleus and binds thetranscription factor Tead4, which leads to the expression of TEgenes such as Cdx2 (Rayon et al., 2014). In turn, Cdx2 inhibits theexpression of the ICM-specific pluripotency markers Oct4 (Niwaet al., 2005) and Nanog (Strumpf et al., 2005). By the 32-cell stage,Cdx2+ cells are irreversibly committed to the TE fate (Posfai et al.,2017; Nichols and Gardner, 1984; Gardner, 1983). Yap localisationis regulated by several upstream factors. In apolar cells, the Hippopathway component angiomotin activates the Hippo kinase Lats,which phosphorylates Yap, leading to its cytoplasmic sequestration.In polarised cells, angiomotin is recruited to the apical domain andbecomes inactivated, allowing Yap to translocate to the nucleus(Leung and Zernicka-Goetz, 2013; Hirate et al., 2013). Differences

in actomyosin contractility and tension between polar and apolarcells also regulate Yap localisation (Maitre et al., 2016).

Our knowledge of lineage specification in human embryosremains rudimentary. Single-cell sequencing analyses havegenerated a spatiotemporal catalogue of gene expression and haverevealed a number of transcriptional differences between mouse andhuman embryos (Niakan and Eggan, 2013; Blakeley et al., 2015;Petropoulos et al., 2016; Yan et al., 2013). At the compacted morulastage (embryonic day E4), the transcription of TE genes such asGATA3 and PDGFA is initiated, although cells still express ICMgenes (Petropoulos et al., 2016). It is not until the early blastocyststage (embryonic day E5) when lineage-specific markers start tobecome mutually exclusive. Aggregation experiments have shownthat E5 TE cells still retain the ability to form ICM cells (De Paepeet al., 2013), and, conversely, isolated human ICMs can alsogenerate TE cells (Guo et al., 2020). These results indicate that, evenif the ICM-TE lineage segregation takes places at the blastocyststage (Petropoulos et al., 2016), cells are not yet fully committed.

Analysis of protein expression and localisation has alsouncovered interesting differences between mouse and humanembryos. YAP localises to the nucleus both in TE and epiblastcells in human blastocysts (Qin et al., 2016), but becomes restrictedto the TE at the late blastocyst stage (Noli et al., 2015), in agreementwith a delayed ICM-TE segregation in human embryos whencompared with mouse. Supporting a potential role for YAP duringTE specification, loss of YAP prevents human embryonic stem cell(ESC) differentiation to the TE fate (Mischler et al., 2019; Guoet al., 2020). Surprisingly, CDX2, a master TE regulator in mouseembryos, is not expressed until the late blastocyst stage in humanembryos (Niakan and Eggan, 2013); its target genes in the mouse,Eomes and Elf5, are not expressed in pre-implantation human TE(Blakeley et al., 2015); and loss of OCT4 in human embryos leads toa downregulation of CDX2 expression (Fogarty et al., 2017),indicating that, in contrast to mouse embryos, OCT4 and CDX2 donot mutually repress each other in human embryos (Niwa et al.,2005; Ralston et al., 2010). Globally, these findings raise thepossibility that different transcription factors control the

Cell fate:gene expression changes

Embryonic genome activation

RNA

Time

Transcription factors

Morphogenesis:tissue shape changes

Polarisation

Apical polarity proteinsEmbryonic

genes

Nuclear YAPTrophectoderm

specification

Transcriptionfactors

Four-cell stageEight-cell stage

Maternal factors

Cytoskeletal proteins

Cytoskeletalproteins

Blastocyst

?

?

Fig. 2. Proposed cell fate-tissue shape crosstalk during human pre-implantation development. The onset of apicobasal polarisation at the eight-cellstage may be controlled by the expression of embryonic genes, potentially encoding transcription factors. As recently shown in mouse embryos, embryonicgenome activation induces the expression of regulators of the actin cytoskeleton. This leads to a remodelling of the actin network that is necessary for theclustering of apical polarity proteins, and culminates in the formation of the mature apical domain. In turn, the acquisition of apicobasal polarisation maylead to nuclear YAP localisation, as seen in the mouse, and expression of trophectoderm-specific transcription factors. This model remains to befunctionally validated in human embryos. Question marks denote molecular connections that have not been validated in human embryos.

4

REVIEW Development (2020) 147, dev190629. doi:10.1242/dev.190629

DEVELO

PM

ENT

Page 5: Mechanisms of human embryo development: from cell fate to … · development have so far been technically and ethically challenging. However, recent technical developments provide

specification of human TE (Fig. 2). GATA3 is an interestingcandidate, as its downregulation in primate embryos inhibits TEdifferentiation (Krendl et al., 2017) and because it is a downstreamtarget of TEAD4/YAP (Home et al., 2012) that inhibits OCT4expression (Krendl et al., 2017) in human ESCs differentiated to TE.Future studies are needed to determine whether YAP and GATA3play a role in TE specification in human embryos. This is especiallyimportant as protocols to differentiate human ESCs into TE may notreflect the in vivo specification route. Differentiation of humanESCs upon addition of BMP4 has been widely used to obtain TE-like cells (Amita et al., 2013; Xu et al., 2002). However, variousreports suggest that cells acquire an extra-embryonic and embryonicmesoderm and/or extra-embryonic amniotic fate rather than a TEfate (Guo et al., 2020; Bernardo et al., 2011). Although newdifferentiation protocols are being developed (Guo et al., 2020;Mischler et al., 2019; Horii et al., 2019; Dong et al., 2020), these donot reflect the 3D organisation of the embryo, and the potentialcontribution of embryo polarisation and cell position to TEspecification cannot be explored. Therefore, the molecular playersinvolved in human TE specification, and whether, as in the mouse,polarisation controls the first lineage segregation in human embryosremain to be determined.

The implanting embryo: a global transformationThe second cell fate decisionHuman and mouse embryos reach the blastocyst stage at embryonicdays E5 and E3.5, respectively. Blastocyst formation involves aprocess of cavitation to form the blastocoel (see Glossary, Box 1). Inmouse embryos, blastocoel formation is driven by the accumulationof pressurised fluid between cells. This fluid moves from the outsidemedium into the intercellular space as a consequence of an osmoticgradient, leading to the formation of microlumens that coarsen toform a single lumen (Dumortier et al., 2019). In turn, luminalpressure leads to increased cortical tension in the TE and tightjunction maturation to allow lumen expansion (Chan et al., 2019).This is a perfect example of how mechanical cues, in this casehydrostatic pressure, affect differentiation (Chan and Hiiragi, 2020).Once the embryo reaches the blastocyst stage, it is ready to

implant in the maternal uterus. At this stage, mouse and humanembryos undergo a second cell fate specification event: the ICMsegregates into pluripotent epiblast and extra-embryonic hypoblast(see Glossary, Box 1). Whereas in mouse embryos the specificationof TE cells precedes the segregation of ICM cells into epiblast andPE (the mouse equivalent of the human hypoblast), the timing ofepiblast-hypoblast segregation in human embryos is still underdebate. Although some studies report a concomitant specification ofthe three lineages, epiblast, hypoblast and TE, in human embryos(Petropoulos et al., 2016), others have identified an early ICM statethat precedes the epiblast-hypoblast split (Stirparo et al., 2018).Upon specification, human hypoblast cells become apicobasallypolarised and express apical proteins such as the sialomucin (seeGlossary, Box 1) protein podocalyxin (Shahbazi et al., 2017). Inmouse embryos, polarisation is required for PE maintenance.Inhibition of atypical protein kinase C (aPKC), a component of thePar complex, or β1-integrin, a cell-ECM adhesion protein, leads todefects in epithelialisation and PE maturation (Saiz et al., 2013; Liuet al., 2009). Studies using human ESCs suggest that aPKC mayalso control the epiblast-hypoblast lineage segregation. Inhibition ofaPKC is needed to maintain naïve pre-implantation-like pluripotenthuman ESCs (Takashima et al., 2014). These cells are competent todifferentiate into extra-embryonic endoderm cells, and this has beenproposed to be dependent on aPKC signalling (Linneberg-

Agerholm et al., 2019), although this hypothesis remains to beformally validated.

A recent study in mouse embryos revealed an unexpected role forthe blastocoel during epiblast-PE specification (Ryan et al., 2019).The authors propose that lumen expansion is needed for correctlineage segregation (Ryan et al., 2019). Interestingly, in isolatedICMs, epiblast and PE progenitors segregate, although with anincreased proportion of PE to epiblast cells (Wigger et al., 2017).This result suggests that, although the second cell fate decision cantake place in the absence of the blastocoel, it may be required to fine-tune the relative numbers of epiblast and PE progenitors.

Human embryo culture beyond implantationOur knowledge of human embryo development at and beyondimplantation (E7) is very scarce. It has been mainly limited to theanalysis of human embryo specimens from the Carnegie collection(Box 2), which have revealed that implanting blastocysts undergoglobal morphological remodelling to form a disc-shaped embryo(Hertig et al., 1956). Conventional human embryo culture methodsonly allow embryo growth and survival up to E6-7 (late blastocyststage). Co-cultures with endometrial cells have traditionally beenused to explore the crosstalk between the embryo and the uterus(Weimar et al., 2013; Lindenberg et al., 1985), but whether theembryo undergoes proper morphogenesis in this setting is unclear.Recently, human embryos have been cultured up to day 13 in vitro,thus permitting insight into the major morphologicaltransformations of post-implantation human development – TEdifferentiation (West et al., 2019), amniotic cavitation (see Glossary,Box 1) and primary yolk sac (see Glossary, Box 1) formation – inthe absence of interactions with maternal tissues (Deglincerti et al.,2016; Shahbazi et al., 2016). This culture method has been takenforward by the addition of a 3D matrix, which allows amnion (seeGlossary, Box 1) specification and further development up to thegastrula stage (Xiang et al., 2019). These systems open the door tostudy human embryo development beyond implantation; e.g. theyhave been used to generate a single-cell transcriptome andmethylome map of post-implantation human embryos (Zhouet al., 2019; Xiang et al., 2019). But given the lack of an in vivoreference, comparisons with primate embryos are important forvalidation (Nakamura et al., 2016; Boroviak and Nichols, 2017; Maet al., 2019; Niu et al., 2019).

Epiblast epithelialisation and amniotic cavitationThe first transformation that epiblast cells undergo is the formationof an epithelial tissue that lines the amniotic cavity. This process isevolutionarily conserved in all amniotes (see Glossary, Box 1) andis a fundamental prerequisite for gastrulation, lineage specificationand developmental progression (Sheng, 2015). Mutations thatimpair pro-amniotic cavity (see Glossary, Box 1) formation lead todevelopmental arrest in mouse embryos and failed differentiation inembryoid bodies (see Glossary, Box 1) (Sakai et al., 2003; Lianget al., 2005; Smyth et al., 1999).

In mouse and human embryos, epiblast epithelialisation takesplace during implantation (Shahbazi and Zernicka-Goetz, 2018). Inresponse to ECM proteins secreted by the extra-embryonic tissues(Li et al., 2003), epiblast cells become polarised and form a rosette-like structure that undergoes lumenogenesis to form the amnioticcavity (Bedzhov and Zernicka-Goetz, 2014; Luckett, 1975). In themouse, epiblast polarisation is triggered by β1-integrin signallingand requires phosphatase and tensin homologue (Pten) and integrin-linked kinase (Ilk) activity (Meng et al., 2017; Sakai et al., 2003),whereas lumenogenesis is triggered by exocytosis of apical vesicles

5

REVIEW Development (2020) 147, dev190629. doi:10.1242/dev.190629

DEVELO

PM

ENT

Page 6: Mechanisms of human embryo development: from cell fate to … · development have so far been technically and ethically challenging. However, recent technical developments provide

and membrane separation (Shahbazi et al., 2017), a process known ashollowing (Bryant and Mostov, 2008). In the human context, furthermechanistic insight has been gained through the use of 3D ESCcultures, which recapitulate the processes of epithelialisation andcavity formation (Shahbazi et al., 2016; Taniguchi et al., 2015).Whereas actin polymerisation promotes cavity formation, actomyosincontractility and the activity of Rho-associated protein kinase (ROCK)prevent lumenogenesis (Taniguchi et al., 2015). Accordingly, theaddition of a ROCK inhibitor to human and monkey embryo culturepromotes amniotic cavity formation (Xiang et al., 2019; Niu et al.,2019). The positive effect of ROCK inhibition during lumenogenesishas been described in other models of hollowing (Bryant et al., 2014),suggesting it is evolutionarily conserved.Once the incipient amniotic cavity is formed, mouse and human

embryos acquire different morphologies (Fig. 1). In the mouseembryo, the TE-derived extra-embryonic ectoderm also undergoes acavitation event. The subsequent fusion of the epiblast and extra-embryonic ectoderm cavities leads to the formation of the pro-amniotic cavity, which spans both tissues (Christodoulou et al.,2018). In contrast, the human TE gives rise to the cytotrophoblast(see Glossary, Box 1). Epiblast cells in contact with thecytotrophoblast differentiate to form an extra-embryonic tissue,the amnion, whereas epiblast cells opposite the cytotrophoblastremain pluripotent (Shahbazi and Zernicka-Goetz, 2018). As aresult, whereas mouse embryos acquire a cylindrical shape, humanembryos display a disc-shaped morphology.

Pluripotency and epiblast shapeEpiblast cells at the blastocyst stage display naïve pluripotency,characterised by global hypomethylation, expression of transcriptionfactors promoting the naïve state and two active X-chromosomes infemale cells (Nichols and Smith, 2012; Theunissen et al., 2016). Thisdevelopmental ‘blank state’ is lost as embryos implant: genes thatpromote the naïve state are downregulated, whereas post-implantationfactors become upregulated, methylation levels increase and one ofthe X chromosomes is inactivated in female cells (Nakamura et al.,2016; Xiang et al., 2019; Kalkan and Smith, 2014). Importantly, exitfrom the naïve pluripotent state happens concomitantly with themorphological transformation of the epiblast into an epithelial tissuethat lines the amniotic cavity, but whether these two events aremechanistically linked was unknown until recently. Using 3Dcultures of mouse and human ESCs, and mouse and humanembryo cultures, it has been shown that amniotic cavity formationis transcriptionally controlled by a pluripotent state transition(Shahbazi et al., 2017). Naïve pluripotent cells polarise in responseto the underlying ECM, but apical vesicle exocytosis is compromised.Consequently, failure to dismantle the naïve state leads to impairedamniotic cavitation and epiblast epithelialisation both in mouse andhuman embryos (Shahbazi et al., 2017). In the mouse epiblast, thetranscription factors Oct4 and Otx2 induce the expression ofpodocalyxin, which participates in lumen opening by promotingelectrostatic membrane repulsion of apposing membranes duringvesicle exocytosis. This transcriptional regulation may not beconserved in human embryos, as OTX2 is weakly expressed in thehuman epiblast (Xiang et al., 2019), andOCT4 has different functionsin mouse and human blastocysts (Fogarty et al., 2017). Therefore, thetranscription factors that control amniotic cavity formation in humanembryos remain to be determined. Notwithstanding the specificmechanism, this is a very clear example of how a cell identity changeis necessary for a change in tissue shape.The fate-shape relationship is bidirectional, as changes in cell

shape and mechanics also control pluripotent cell identity.

Experiments in human ESCs revealed that epithelialisation leadsto decreased pluripotent gene expression via integrin β1 activation(Hamidi et al., 2020), and therefore epiblast epithelialisation couldfacilitate pluripotency exit in vivo. In mouse ESCs, a decrease inmembrane tension is necessary for naïve pluripotency exit (Bergertet al., 2019), as it allows the activation of FGF signalling, a trigger ofnaïve exit, via endocytosis of FGF receptors (De Belly et al., 2019).In human ESCs FGF signalling inhibition is implied as a keyrequirement for naïve human ESCs, and its activation is associatedwith naïve pluripotency exit (Di Stefano et al., 2018). Thus, it istempting to speculate that loss of the naïve state in human embryoscould also be regulated by changes in membrane tension leading toFGF receptor endocytosis (Fig. 3). Future studies are needed todetermine whether similar changes in membrane tension andsignalling are observed beyond 2D cultures, using systems thatrecapitulate both epiblast gene expression and shape. Cell-cell andcell-matrix interactions, as well as the mechanical properties of theECM, are major regulators of pluripotent stem cell identity in vitro(Ranga et al., 2014; Przybyla et al., 2016; Pieters and Van Roy, 2014;Hamidi et al., 2020). Upon naïve pluripotency exit, cells becomeresponsive to mechanical stimuli. In particular, cell stretching leads toan increase in the intracellular levels of calcium, which activate stressand metabolic pathways (Verstreken et al., 2019).

Altogether, these studies provide a glimpse of the complexinterplay between transcriptional responses, mechanics, geometryand signalling in pluripotent cells. However, functional studies in thehuman embryo are needed to determine whether these mechanismsare conserved from ESCs to embryos and from mice to humans.

Amnion differentiationA key event in the development of implanting human embryos is thesplit of epiblast cells into the pluripotent epiblast disc and thedifferentiated extra-embryonic amniotic epithelium. Our knowledgeof the mechanisms that govern this event is very rudimentary, as inmost animal models amnion formation takes place at gastrulation(Dobreva et al., 2010). Much of what we know is derived fromstudies in monkey embryos (Hill, 1932). Upon implantation,epiblast cells in contact with the cytotrophoblast adopt asquamous epithelial morphology and downregulate pluripotencyfactors such as NANOG and SOX2, whereas epiblast cells incontact with the hypoblast form a columnar epithelium and remainpluripotent (Luckett, 1975; Sasaki et al., 2016). This fate split couldbe regulated by a gradient of BMP and/or WNT activity, as themonkey cytotrophoblast and amnion are a source ofWNT and BMPligands, whereas the hypoblast secretes WNT and BMP inhibitors(Sasaki et al., 2016). In agreement with this notion, stimulation ofhuman ESCs with BMP leads to the formation of amnion-like cells(Guo et al., 2020; Minn et al., 2020), BMP signalling is required foramnion specification in mouse embryos (Dobreva et al., 2018) andculture of human ESCs in a soft 3D matrix generates amnion-likesquamous spheroids in a BMP-dependent manner (Shao et al.,2016). Mechanical cues may cooperate with BMP signalling, asplating human ESCs on a hard surface prevents amniogenesis (Shaoet al., 2016). Amnion formation is also regulated by cell density;high cell densities preserve pluripotency and inhibit amniogenesis,whereas low cell densities promote amnion specification (Shaoet al., 2017). However, the exact mechanism of amnionspecification awaits further investigation.

Gastrulation: from pluripotency loss to lineage specificationThe day 14 rule limits human embryo research to the first 14 days ofdevelopment (Pera, 2017) and, thus, to the period prior to the

6

REVIEW Development (2020) 147, dev190629. doi:10.1242/dev.190629

DEVELO

PM

ENT

Page 7: Mechanisms of human embryo development: from cell fate to … · development have so far been technically and ethically challenging. However, recent technical developments provide

formation of the primitive streak (see Glossary, Box 1).Consequently, gastrulation cannot be studied in human embryos.Given that it is ‘not birth, marriage or death, but gastrulation whichis truly the most important time in your life’ (Wolpert and Vicente,2015), it is not surprising that multiple groups are putting a greateffort into recreating human embryogenesis using stem cells(Shahbazi et al., 2019; Simunovic and Brivanlou, 2017). Duringgastrulation, cells undergo concomitant changes in shape and fate.At the site where the primitive streak forms, epiblast cells undergoepithelial-to-mesenchymal transition (EMT) (see Glossary, Box 1),lose their pluripotent features and generate mesoderm (see Glossary,Box 1) and endoderm (see Glossary, Box 1) (Tam and Behringer,1997). Epiblast progenitors that do not ingress through the streakbecome ectoderm (see Glossary, Box 1). The onset of gastrulationis regulated by the signalling crosstalk between embryonic andextra-embryonic cells, mediated by the concerted action of BMP,WNT and activin A/Nodal signals and their inhibitors (Tam andLoebel, 2007). These signalling interactions may be furthermodulated by mechanical cues and the physical structure of thetissue. Modelling the human embryo using stem cells disentanglesthis complexity by modulating single parameters at a time (Siggiaand Warmflash, 2018).

Self-organisation of human ESCsIn human ESC cultures, cell density and colony shape and size canbe precisely controlled using micropattern technology (Théry andPiel, 2009; Bauwens et al., 2008; Nazareth et al., 2013). Theseinitial studies led to the conclusion that colony size and compositionaffect human ESC identity in the absence of exogenous cues(Peerani et al., 2007).Subsequent experiments applied micropattern technology to the

study of tissue patterning. When human ESCs are cultured inmicropatterns of 0.5 mm, they reach a cell density equivalent to that

observed in the pre-gastrulating human epiblast (Etoc et al., 2016).Addition of BMP4 triggers pluripotency exit and lineagecommitment. Despite the homogenous presentation of themorphogen, germ layers become radially organised, with TE andamnion on the outside, followed by endoderm, mesoderm andectoderm towards the centre of the colony (Warmflash et al., 2014;Minn et al., 2020). Mechanistic studies revealed that patterning wasa consequence of the diffusion of the BMP4 inhibitor noggin(NOG) and the epithelial architecture of the colony (Etoc et al.,2016). As a result of the epithelial nature of human ESCs, BMPreceptors become basally localised and therefore inaccessible to theapically presented BMP4. However, cells in the border of the colonyhave a less pronounced polarised phenotype, which allows ligand-receptor interactions and, hence, differentiation of the outermostcells. Negative feedback by NOG, which is directly induced byBMP4, is also required to explain the fate patterning (Etoc et al.,2016). Interestingly, a recent report has shown that BMP receptorsare basolaterally localised in the early post-implantation mouseepiblast, and this is required for the formation of a robust BMPsignalling gradient (Zhang et al., 2019), indicating tissue geometrycontrols pluripotent cell differentiation both in mice and humans(Fig. 4).

An alternative mechanistic explanation for the emergence ofpatterning upon BMP4 stimulation has been proposed by Tewaryet al. (2017). A reaction-diffusionmechanism is initially responsiblefor the generation of a self-organised BMP4 gradient. Thesubsequent interpretation of that gradient and the patterned fateacquisition is consistent with the positional information model. Insupport of this idea, decreasing the concentration of BMP4 issufficient to preserve fate patterning in micropatterns of smaller size(0.2-0.3 mm). This represents a potential mechanism to adapt tochanges in embryo size, and hence maintain developmentalrobustness (Tewary et al., 2017). However, a recent report has

Cell fate:gene expression changes

Naive pluripotency exit

RNA

Time

Increased FGF activity?

Morphogenesis:tissue shape changes

Exocytosis

Lumenogenesis

Post-implantation factors

Endocytosis

Membrane tension

Transcription factors

Blastocyst Blastocyst

Epiblast disc

Exocytosis and luminal proteins

Naive genes

Fig. 3. Proposed cell fate-tissue shape crosstalk during human implantation development. Upon implantation, epiblast cells exit from the naïvepluripotent state and initiate the expression of post-implantation factors. Studies in mouse ESCs have shown that this is controlled by a decrease in membranetension, which promotes endocytosis and, as a consequence, increased fibroblast growth factor (FGF) signalling activity, which is required for naïvepluripotency exit. Therefore, membrane tension affects the pluripotent state of a cell. Whether this pathway is active in human embryos remains to beexplored. Upon naïve pluripotency exit, genes involved in exocytosis become expressed and initiate the process of lumenogenesis to form the amnioticcavity. Exocytic vesicles provide apical membrane and luminal proteins, which are necessary to build a lumen de novo. The transcription factors involvedin this process remain to be identified. Question mark denotes a molecular event that has not beem validated in human embryos.

7

REVIEW Development (2020) 147, dev190629. doi:10.1242/dev.190629

DEVELO

PM

ENT

Page 8: Mechanisms of human embryo development: from cell fate to … · development have so far been technically and ethically challenging. However, recent technical developments provide

proposed that BMP4 does not act as a classical morphogen. In verysmall colonies (one to eight cells), the response to BMP4 is binary:cells adopt either a pluripotent or a TE fate (Nemashkalo et al.,2017). The appearance of mesendodermal fates in a primitivestreak-like region additionally requires WNT and Nodal activation(Siggia and Warmflash, 2018).Self-organised patterns also emerge upon stimulation with WNT

(Martyn et al., 2018), which leads to the appearance of an outermesendoderm ring while inner cells remain pluripotent. Formationof this fate pattern requires E-cadherin cell-cell adhesions, which aremore prominent in the colony centre, and dampen WNT activity bysequestering β-catenin. In addition, the WNT inhibitor DKK1 isexpressed in response to WNT and controls the size of themesendoderm ring (Martyn et al., 2019). In agreement with thesemicropattern studies, a group of cells in the primitive streak ofmouse and rabbit embryos expresses both activators and inhibitorsof the BMP and WNT signalling pathways (Peng et al., 2016;Idkowiak et al., 2004), indicating there are epiblast-intrinsicmechanisms that regulate the position of the primitive streak.These examples demonstrate how simplified 2D models of the

embryo can help us dissect the chemical control of tissue patterning;but mechanical cues also play a role in patterning. For example,mechanical strains during gastrulation lead to mesodermspecification via induction of the transcription factor brachyury,and this mechanism may be conserved from flies to humans (Brunetet al., 2013). Although the 2D self-organisation of human ESCsdescribed so far does not recapitulate the mechanical features of thedeveloping embryo, recent technological advances provide anopportunity to explore the contribution of mechanics (Vianello andLutolf, 2019). The use of hydrogels led to the observation thatmatrix stiffness, degradability and composition influenceproliferation, tissue structure and differentiation in the neurallineage (Ranga et al., 2016; Sun et al., 2014). Similarly, mesodermand endoderm differentiation are enhanced in soft substrates butinhibited in stiffer ones (Przybyla et al., 2016; Chen et al., 2020).Combining micropattern technology with embryo-like compliant

hydrogels revealed that gastrulation-like domains emerge in areas ofhigh tension due to the release of β-catenin from cell-cell adhesionsites and its nuclear translocation, both in mouse and human ESCs(Muncie et al., 2020; Blin et al., 2018). To dissect the role ofmechanics during development, future experimental strategies needto: be based on the use of robust, reproducible and relevant stem cellmodels of the embryo (Huch et al., 2017; Van Den Brink et al.,2020); allow mapping of intrinsic stresses and application ofextrinsic forces in a precise manner (Vianello and Lutolf, 2019); anduse appropriate readouts to assess changes in patterning andmorphogenesis.

Embryonic-extra-embryonic crosstalkWe can add an extra layer of complexity to self-organising modelsof human ESCs by investigating how extra-embryonic cellsmodulate human embryonic stem cell fate and shape atgastrulation. This is particularly important given the differentorganisation of extra-embryonic tissues in mouse and humanembryos. Microfluidics has been recently used to model theepiblast-amnion fate split (Zheng et al., 2019). This led to thecharacterisation of the amnion as a crucial signalling centre.Amniotic epithelial-like cells express high levels of BMP4 andinduce gastrulation-like events in a WNT-dependent manner inpluripotent human ESCs and specification of primordial germ cell-like cells (see Glossary, Box 1) (Zheng et al., 2019). Therefore,whereas in mouse embryos the extra-embryonic ectoderm is the keydriver of gastrulation (Tam and Behringer, 1997), in humanembryos this role may be played by the amnion. Conversely, thefunction of the hypoblast as an inhibitor of BMP andWNT signallingseems to be conserved in mouse, monkey and humans (Xiang et al.,2019; Stower and Srinivas, 2014; Sasaki et al., 2016). Hypoblast cellssecrete BMP and WNT inhibitors in a polarised manner, andtherefore the combined action of TE/amnion and hypoblast leads tothe generation of a gradient of BMP and WNT activity across theepiblast. Future studies are needed to dissect the specificcontributions of the cytotrophoblast, hypoblast and amnion in

Cell fate:gene expression changes

Polarized mesoderm marker expression

RNA

A P

Morphogenesis:tissue shape changes

Mesenchymal shape

EMT transcription factors

Decreased cell-cell adhesion

Brachyury

Unpolarised BMP receptors

Increased BMP and WNT signalling

Mesendoderm specification

Epiblast disc

Gastrula

Epiblast disc

Fig. 4. Proposed cell fate-tissue shape crosstalk during human post-implantation development. BMP and WNT signals secreted by the amnion and thetrophectoderm lead to expression of mesoderm markers, such as the transcription factor brachyury, in the posterior epiblast. Brachyury induces expression ofepithelial-to-mesenchymal transition (EMT) transcription factors, which cause a decrease in the levels of E-cadherin. As a consequence, cell-cell adhesionsweaken and cells lose their epithelial morphology and their polarised organisation, such as the polarised localisation of BMP receptors. The unpolarised BMPreceptors are able to interact with BMP ligands, increasing BMP signalling activity and inducing mesendoderm specification.

8

REVIEW Development (2020) 147, dev190629. doi:10.1242/dev.190629

DEVELO

PM

ENT

Page 9: Mechanisms of human embryo development: from cell fate to … · development have so far been technically and ethically challenging. However, recent technical developments provide

patterning the human epiblast. This could be achieved by combiningstudies on in vitro cultured human embryos (Xiang et al., 2019) within vivo and in vitro cultured monkey embryos (Nakamura et al., 2016;Niu et al., 2019; Ma et al., 2019) and the development of new stemcell models of the human embryo comprising both embryonic andextra-embryonic stem cells, as previously used in the mouse(Harrison et al., 2017; Sozen et al., 2018; Rivron et al., 2018).

When things go wrong: aneuploidy and pregnancy lossUnderstanding the basic mechanisms of human development isfundamental to tackling an important clinical problem: theremarkable inefficiency of human reproduction. It is estimatedthat only 30-40% of conceptions progress to live birth (Larsen et al.,2013). Development may fail at any time between fertilisation andterm, but the early stages are especially crucial. Epidemiologicalstudies estimate that 40-50% of embryos are lost before implantation(Wilcox et al., 2020). Once the embryo implants, the ongoingpregnancy can be detected based on the increased levels of humanchorionic gonadotropin (hCG) (Wilcox et al., 1999). Based on hCGserum levels, 20-30% of implanted embryos fail to progress beyondweek 6 (Wilcox et al., 1988). Owing to the lack of ultrasoundverification, these cases are termed biochemical losses (Larsen et al.,2013;Macklon et al., 2002). The reasons behind this high rate of earlypregnancy loss remain poorly understood. Multiple potentialmechanisms have been involved, including both alterations inembryo development and maternal causes, but further research isneeded to understand their contribution.The observation that 50-75% of spontaneous abortions present

karyotypic abnormalities and morphological malformations(Philipp et al., 2003; Boué et al., 1975) has resulted in a focus onthe genetic causes of pregnancy loss. While the incidence of meioticaneuploidy in most species is remarkably low, 10-30% of fertilisedhuman eggs are aneuploid due to female meiotic errors (Hassoldand Hunt, 2001; Nagaoka et al., 2012). These chromosomesegregation errors are more prevalent in young women and inwomen of advanced age, resulting from whole-chromosomenondisjunction and premature sister chromatid separation,respectively (Gruhn et al., 2019). Why, when and how embryosharbouring specific aneuploidies fail during embryogenesis remainsunknown. All single chromosome aneuploidies can potentially formblastocysts, but they do so at a lower rate and with a higher incidenceof morphological abnormalities. Aneuploid embryos are morelikely to arrest during EGA, to be developmentally delayed and topresent a low-quality TE and/or ICM (i.e. few cells and cellularfragmentation indicative of cell death) (Minasi et al., 2016; Fragouliet al., 2013). Moreover, despite forming morphologically normalblastocysts, aneuploid embryos display transcriptional andepigenetic alternations that may compromise their subsequentdevelopment (McCallie et al., 2016; Licciardi et al., 2018; Starostiket al., 2020). Autosomal monosomies typically lead to biochemicallosses, whereas autosomal trisomies are associated with silentmiscarriage (presence of extra-embryonic membranes but lack ofembryonic structures) and first-trimester miscarriage (Ljunger et al.,2005; Martinez et al., 2010; Ferro et al., 2003; Stephenson et al.,2002). In addition, alterations in the number of chromosomes can bea consequence of mitotic alterations during the first three cleavagedivisions, which lead to the formation of mosaic human embryos(Popovic et al., 2019; Starostik et al., 2020; Mantikou et al., 2012).Given the inaccessibility of human embryos at these stages, severalfundamental questions remain to be addressed. How do specificalterations in the number of chromosomes impact onmorphogenesisor cell fate specification? Is the response to aneuploidy different in

embryonic and extra-embryonic cells? What is the fate of aneuploidcells in mosaic embryos? Healthy babies can potentially be bornfrom mosaic aneuploid human embryos (Greco et al., 2015),suggesting aneuploid cells may be selectively eliminated duringdevelopment. Could this selective elimination be a consequence ofthe impaired response of aneuploid cells to physical and/orbiochemical cues? And given that 25-50% of spontaneousmiscarriages do not show chromosomal abnormalities, what is thereason behind their failed development? Could alterations in the cellfate-tissue shape crosstalk be responsible for some of these earlypregnancy losses? Addressing these questions and understandingthe mechanisms that lead to pregnancy loss is a first step towards thefuture development of therapeutic interventions.

ConclusionsDespite remarkable advances in the field of human development, untilvery recently human embryo research was mainly descriptive and, asWilhelm Roux already realised by the end of the 19th century, tocomprehend the mechanisms of development, we must interfere withit. Today, we are ideally suited to tackle this challenge and decipherhow form and function emerge during human embryogenesis. Recentevidence indicates that the crosstalk between cell fate and tissue shapeplays an important role during key human developmental events,including compaction and polarisation in pre-implantation embryos,embryonic epithelialisation at the time of implantation and germlineage commitment at gastrulation. We are just beginning tounderstand how cell fate specification events modulate tissueorganisation and, vice versa, how changes in tissue shape modulatecell identity and tissue patterning, leading to the emergence of self-organisation. A number of crucial questions still remain to beaddressed. What are the mechanisms that remodel the implantinghuman embryo? How do physical and chemical cues coordinatemorphogenesis and patterning? What signals trigger cell fatespecification during gastrulation? And how are all these eventsaffected when chromosomal alterations are present? To address thesequestions, we now possess an adequate set of experimental tools:genome editing has already been successfully used to study genefunction in human embryos (Fogarty et al., 2017); novel imagingtechniques allow us to map cellular behaviours with unprecedentedresolution (McDole et al., 2018); new culture methods have extendedthe window of human development amenable to investigation(Shahbazi et al., 2016; Deglincerti et al., 2016; Xiang et al., 2019);and human ESC-embryo chimeras could be potentially used as a goldstandard to study cell potency (De Los Angeles et al., 2015;Alexandrova et al., 2016; Tachibana et al., 2012; Chen et al., 2015),although stringent criteria to assess lineage contribution and ethicalconsiderations need to be applied (Posfai et al., 2020). In addition,human ESCs can mimic different aspects of embryo development invitro (Shahbazi et al., 2019). These stem cell models of the embryo areallowing researchers to deconstruct the complexity of humandevelopment and to decipher the basic principles of self-organisation. Last and importantly, the overall increase in assistedreproductive technology cycles over the years, together with theimprovement in embryo selection procedures, leads to the generationof vast numbers of surplus human embryos (Kushnir et al., 2017),which represent an invaluable resource for research. Using this newset of tools, we can start to unravel the mysteries of our owndevelopment.

AcknowledgementsI am grateful to Eric Siggia, Meng Zhu, Bailey Weatherbee, Kirsty Mackinlay andMichele Petruzzelli for critical reading of the manuscript.

9

REVIEW Development (2020) 147, dev190629. doi:10.1242/dev.190629

DEVELO

PM

ENT

Page 10: Mechanisms of human embryo development: from cell fate to … · development have so far been technically and ethically challenging. However, recent technical developments provide

Competing interestsThe authors declare no competing or financial interests.

FundingWork in M.N.S.’s group is funded by the Medical Research Council (MC_UP_1201/24) and the European Molecular Biology Organization. Deposited in PMC forimmediate release.

ReferencesAlarcon, V. B. (2010). Cell polarity regulator PARD6B is essential for trophectodermformation in the preimplantation mouse embryo. Biol Reprod 83, 347-358. doi:10.1095/biolreprod.110.084400

Alexandrova, S., Kalkan, T., Humphreys, P., Riddell, A., Scognamiglio, R.,Trumpp, A. and Nichols, J. (2016). Selection and dynamics of embryonic stemcell integration into early mouse embryos. Development 143, 24-34. doi:10.1242/dev.124602

Alikani, M. (2005). Epithelial cadherin distribution in abnormal human pre-implantation embryos. Hum Reprod 20, 3369-3375. doi:10.1093/humrep/dei242

Amita, M., Adachi, K., Alexenko, A. P., Sinha, S., Schust, D. J., Schulz, L. C.,Roberts, R. M. and Ezashi, T. (2013). Complete and unidirectional conversion ofhuman embryonic stem cells to trophoblast by BMP4. Proc Natl Acad Sci U S A110, E1212-E1221. doi:10.1073/pnas.1303094110

Anani, S., Bhat, S., Honma-Yamanaka, N., Krawchuk, D. and Yamanaka, Y.(2014). Initiation of Hippo signaling is linked to polarity rather than to cell position inthe pre-implantation mouse embryo. Development 141, 2813-2824. doi:10.1242/dev.107276

Bauwens, C. L., Peerani, R., Niebruegge, S., Woodhouse, K. A., Kumacheva, E.,Husain, M. and Zandstra, P. W. (2008). Control of human embryonic stem cellcolony and aggregate size heterogeneity influences differentiation trajectories.Stem Cells 26, 2300-2310. doi:10.1634/stemcells.2008-0183

Bedzhov, I. and Zernicka-Goetz, M. (2014). Self-organizing properties of mousepluripotent cells initiate morphogenesis upon implantation. Cell 156, 1032-1044.doi:10.1016/j.cell.2014.01.023

Benham-Pyle, B. W., Pruitt, B. L. and Nelson, W. J. (2015). Cell adhesion.Mechanical strain induces E-cadherin-dependent Yap1 and beta-cateninactivation to drive cell cycle entry. Science 348, 1024-1027.

Bergert, M. L., Lembo, S., Milovanovic, D., Bormel, M., Neveu, P. and Diz-Mun oz, A. (2019). Cell surface mechanics gate stem cell differentiation. bioRxiv.

Bernardo, A. S., Faial, T., Gardner, L., Niakan, K. K., Ortmann, D., Senner, C. E.,Callery, E. M., Trotter, M. W., Hemberger, M., Smith, J. C. et al. (2011).Brachyury and CDX2 mediate BMP-induced differentiation of human and mousepluripotent stem cells into embryonic and extraembryonic lineages.Cell Stem Cell9, 144-155. doi:10.1016/j.stem.2011.06.015

Blakeley, P., Fogarty, N. M., Del Valle, I., Wamaitha, S. E., Hu, T. X., Elder, K.,Snell, P., Christie, L., Robson, P. and Niakan, K. K. (2015). Defining the threecell lineages of the human blastocyst by single-cell RNA-seq. Development 142,3151-3165. doi:10.1242/dev.123547

Blin, G., Wisniewski, D., Picart, C., Thery, M., Puceat, M. and Lowell, S. (2018).Geometrical confinement controls the asymmetric patterning of brachyury incultures of pluripotent cells. Development, 145.

Boroviak, T. and Nichols, J. (2017). Primate embryogenesis predicts the hallmarksof human naive pluripotency. Development 144, 175-186. doi:10.1242/dev.145177

Boue, J., Bou, A. and Lazar, P. (1975). Retrospective and prospectiveepidemiological studies of 1500 karyotyped spontaneous human abortions.Teratology 12, 11-26. doi:10.1002/tera.1420120103

Braude, P., Bolton, V. and Moore, S. (1988). Human gene expression first occursbetween the four- and eight-cell stages of preimplantation development. Nature332, 459-461. doi:10.1038/332459a0

Brunet, T., Bouclet, A., Ahmadi, P., Mitrossilis, D., Driquez, B., Brunet, A.-C.,Henry, L., Serman, F., Bealle, G., Menager, C. et al. (2013). Evolutionaryconservation of early mesoderm specification by mechanotransduction inBilateria. Nat Commun 4, 2821. doi:10.1038/ncomms3821

Bryant, D. M. and Mostov, K. E. (2008). From cells to organs: building polarizedtissue. Nat Rev Mol Cell Biol 9, 887-901. doi:10.1038/nrm2523

Bryant, D. M., Roignot, J., Datta, A., Overeem, A. W., Kim, M., Yu, W., Peng, X.,Eastburn, D. J., Ewald, A. J., Werb, Z. et al. (2014). A molecular switch for theorientation of epithelial cell polarization. Dev Cell 31, 171-187. doi:10.1016/j.devcel.2014.08.027

Chan, C. J. and Hiiragi, T. (2020). Integration of luminal pressure and signalling intissue self-organization. Development, 147.

Chan, C. J., Heisenberg, C. P. and Hiiragi, T. (2017). Coordination ofMorphogenesis and Cell-Fate Specification in Development. Curr Biol 27,R1024-R1035. doi:10.1016/j.cub.2017.07.010

Chan, C. J., Costanzo, M., Ruiz-Herrero, T., Monke, G., Petrie, R. J., Bergert, M.,Diz-Munoz, A., Mahadevan, L. and Hiiragi, T. (2019). Hydraulic control ofmammalian embryo size and cell fate. Nature 571, 112-116. doi:10.1038/s41586-019-1309-x

Chen, Y., Niu, Y., Li, Y., Ai, Z., Kang, Y., Shi, H., Xiang, Z., Yang, Z., Tan, T., Si, W.et al. (2015). Generation of Cynomolgus Monkey Chimeric Fetuses usingEmbryonic Stem Cells. Cell Stem Cell 17, 116-124. doi:10.1016/j.stem.2015.06.004

Chen, Y.-F., Li, Y.-S. J., Chou, C.-H., Chiew, M. Y., Huang, H.-D., Ho, J. H.-C.,Chien, S. and Lee, O. K. (2020). Control of matrix stiffness promotes endodermlineage specification by regulating SMAD2/3 via lncRNA LINC00458. Sci. Adv., 6.

Christodoulou, N., Kyprianou, C., Weberling, A., Wang, R., Cui, G., Peng, G.,Jing, N. and Zernicka-Goetz, M. (2018). Sequential formation and resolution ofmultiple rosettes drive embryo remodelling after implantation. Nat Cell Biol 20,1278-1289. doi:10.1038/s41556-018-0211-3

Coste, B., Mathur, J., Schmidt, M., Earley, T. J., Ranade, S., Petrus, M. J., Dubin,A. E. and Patapoutian, A. (2010). Piezo1 and Piezo2 are essential componentsof distinct mechanically activated cation channels. Science 330, 55-60. doi:10.1126/science.1193270

De Belly, H., Jones, P. H., Paluch, E. K. and Chalut, K. J. (2019). Membranetension mediated mechanotransduction drives fate choice in embryonic stemcells. bioRxiv.

De Los Angeles, A., Ferrari, F., Xi, R., Fujiwara, Y., Benvenisty, N., Deng, H.,Hochedlinger, K., Jaenisch, R., Lee, S., Leitch, H. G. et al. (2015). Hallmarks ofpluripotency. Nature 525, 469-478. doi:10.1038/nature15515

De Paepe, C., Cauffman, G., Verloes, A., Sterckx, J., Devroey, P., Tournaye, H.,Liebaers, I. and Van De Velde, H. (2013). Human trophectoderm cells are not yetcommitted. Hum Reprod 28, 740-749. doi:10.1093/humrep/des432

Deglincerti, A., Croft, G. F., Pietila, L. N., Zernicka-Goetz, M., Siggia, E. D. andBrivanlou, A. H. (2016). Self-organization of the in vitro attached human embryo.Nature 533, 251-254. doi:10.1038/nature17948

Desprat, N., Supatto, W., Pouille, P. A., Beaurepaire, E. and Farge, E. (2008).Tissue deformation modulates twist expression to determine anterior midgutdifferentiation in Drosophila embryos.Dev Cell 15, 470-477. doi:10.1016/j.devcel.2008.07.009

Di Stefano, B., Ueda, M., Sabri, S., Brumbaugh, J., Huebner, A. J., Sahakyan,A., Clement, K., Clowers, K. J., Erickson, A. R., Shioda, K. et al. (2018).Reduced MEK inhibition preserves genomic stability in naive human embryonicstem cells. Nat Methods 15, 732-740. doi:10.1038/s41592-018-0104-1

Dobreva, M. P., Pereira, P. N., Deprest, J. and Zwijsen, A. (2010). On the origin ofamniotic stem cells: of mice andmen. Int J Dev Biol 54, 761-777. doi:10.1387/ijdb.092935md

Dobreva, M. P., Abon Escalona, V., Lawson, K. A., Sanchez, M. N., Ponomarev,L. C., Pereira, P. N. G., Stryjewska, A., Criem, N., Huylebroeck, D., Chuva DeSousa Lopes, S. M. et al. (2018). Amniotic ectoderm expansion in mouse occursvia distinct modes and requires SMAD5-mediated signalling. Development, 145.

Dong, C., Beltcheva, M., Gontarz, P., Zhang, B., Popli, P., Fischer, L. A., Khan,S. A., Park, K. M., Yoon, E. J., Xing, X. et al. (2020). Derivation of trophoblaststem cells from naive human pluripotent stem cells. Elife, 9.

Dumortier, J. G., Le Verge-Serandour, M., Tortorelli, A. F., Mielke, A., De Plater,L., Turlier, H. andMaitre, J. L. (2019). Hydraulic fracturing and active coarseningposition the lumen of the mouse blastocyst. Science 365, 465-468. doi:10.1126/science.aaw7709

Dupont, S., Morsut, L., Aragona, M., Enzo, E., Giulitti, S., Cordenonsi, M.,Zanconato, F., Le Digabel, J., Forcato, M., Bicciato, S. et al. (2011). Role ofYAP/TAZ in mechanotransduction. Nature 474, 179-183. doi:10.1038/nature10137

Durdu, S., Iskar, M., Revenu, C., Schieber, N., Kunze, A., Bork, P., Schwab, Y.and Gilmour, D. (2014). Luminal signalling links cell communication to tissuearchitecture during organogenesis. Nature 515, 120-124. doi:10.1038/nature13852

Edwards, R. G., Bavister, B. D. and Steptoe, P. C. (1969). Early stages offertilization in vitro of human oocytes matured in vitro. Nature 221, 632-635.doi:10.1038/221632a0

Edwards, R. G., Steptoe, P. C. and Purdy, J. M. (1970). Fertilization and cleavagein vitro of preovulator human oocytes. Nature 227, 1307-1309. doi:10.1038/2271307a0

Etoc, F., Metzger, J., Ruzo, A., Kirst, C., Yoney, A., Ozair, M. Z., Brivanlou, A. H.and Siggia, E. D. (2016). A Balance between Secreted Inhibitors and EdgeSensing Controls Gastruloid Self-Organization. Dev Cell 39, 302-315. doi:10.1016/j.devcel.2016.09.016

Ferro, J., Martinez, M. C., Lara, C., Pellicer, A., Remohi, J. and Serra, V. (2003).Improved accuracy of hysteroembryoscopic biopsies for karyotyping early missedabortions. Fertil Steril 80, 1260-1264. doi:10.1016/S0015-0282(03)02195-2

Fierro-Gonzalez, J. C., White, M. D., Silva, J. C. andPlachta, N. (2013). Cadherin-dependent filopodia control preimplantation embryo compaction. Nat Cell Biol 15,1424-1433. doi:10.1038/ncb2875

Fogarty, N. M. E., Mccarthy, A., Snijders, K. E., Powell, B. E., Kubikova, N.,Blakeley, P., Lea, R., Elder, K., Wamaitha, S. E., Kim, D. et al. (2017). Genomeediting reveals a role for OCT4 in human embryogenesis. Nature 550, 67-73.doi:10.1038/nature24033

Fragouli, E., Alfarawati, S., Spath, K., Jaroudi, S., Sarasa, J., Enciso, M. andWells, D. (2013). The origin and impact of embryonic aneuploidy.HumGenet 132,1001-1013. doi:10.1007/s00439-013-1309-0

10

REVIEW Development (2020) 147, dev190629. doi:10.1242/dev.190629

DEVELO

PM

ENT

Page 11: Mechanisms of human embryo development: from cell fate to … · development have so far been technically and ethically challenging. However, recent technical developments provide

Gardner, R. L. (1983). Origin and differentiation of extraembryonic tissues in themouse. Int Rev Exp Pathol 24, 63-133.

Gasser, R. F., Cork, R. J., Stillwell, B. J. and Mcwilliams, D. T. (2014). Rebirth ofhuman embryology. Dev Dyn 243, 621-628. doi:10.1002/dvdy.24110

Goehring, N. W., Trong, P. K., Bois, J. S., Chowdhury, D., Nicola, E. M., Hyman,A. A. and Grill, S. W. (2011). Polarization of PAR proteins by advective triggeringof a pattern-forming system. Science 334, 1137-1141. doi:10.1126/science.1208619

Greco, E., Minasi, M. G. and Fiorentino, F. (2015). Healthy Babies afterIntrauterine Transfer of Mosaic Aneuploid Blastocysts. N Engl J Med 373,2089-2090. doi:10.1056/NEJMc1500421

Green, J. B. and Sharpe, J. (2015). Positional information and reaction-diffusion:two big ideas in developmental biology combine. Development 142, 1203-1211.doi:10.1242/dev.114991

Gruhn, J. R., Zielinska, A. P., Shukla, V., Blanshard, R., Capalbo, A.,Cimadomo, D., Nikiforov, D., Chan, A. C., Newnham, L. J., Vogel, I. et al.(2019). Chromosome errors in human eggs shape natural fertility overreproductive life span. Science 365, 1466-1469. doi:10.1126/science.aav7321

Guo, G., Stirparo, G. G., Strawbridge, S., Yang, J., Clarke, J., Li, M. A., Myers, S.,Ozel, B. N., Nichols, J. and Smith, A. (2020). Trophectoderm potency is retainedexclusively in human naive cells. bioRxiv.

Hamidi, S., Nakaya, Y., Nagai, H., Alev, C., Kasukawa, T., Chhabra, S., Lee, R.,Niwa, H., Warmflash, A., Shibata, T. et al. (2020). Mesenchymal-epithelialtransition regulates initiation of pluripotency exit before gastrulation.Development, 147.

Hannezo, E. and Heisenberg, C.-P. (2019). Mechanochemical Feedback Loops inDevelopment and Disease. Cell 178, 12-25. doi:10.1016/j.cell.2019.05.052

Harrison, S. E., Sozen, B., Christodoulou, N., Kyprianou, C. and Zernicka-Goetz, M. (2017). Assembly of embryonic and extraembryonic stem cells to mimicembryogenesis in vitro. Science, 356.

Hassold, T. and Hunt, P. (2001). To err (meiotically) is human: the genesis ofhuman aneuploidy. Nat Rev Genet 2, 280-291. doi:10.1038/35066065

Heisenberg, C. P. and Bellaiche, Y. (2013). Forces in tissue morphogenesis andpatterning. Cell 153, 948-962. doi:10.1016/j.cell.2013.05.008

Hertig, A. T., Rock, J. and Adams, E. C. (1956). A description of 34 human ovawithin the first 17 days of development. Am J Anat 98, 435-493. doi:10.1002/aja.1000980306

Hill, J. P. (1932). The developmental history of the primates. Philos Trans R SocLond B Biol Sci 221, 45-178. doi:10.1098/rstb.1932.0002

Hirate, Y., Hirahara, S., Inoue, K., Suzuki, A., Alarcon, V. B., Akimoto, K., Hirai,T., Hara, T., Adachi, M., Chida, K. et al. (2013). Polarity-dependent distribution ofangiomotin localizes Hippo signaling in preimplantation embryos. Curr Biol 23,1181-1194. doi:10.1016/j.cub.2013.05.014

Home, P., Saha, B., Ray, S., Dutta, D., Gunewardena, S., Yoo, B., Pal, A., Vivian,J. L., Larson, M., Petroff, M. et al. (2012). Altered subcellular localization oftranscription factor TEAD4 regulates first mammalian cell lineage commitment.Proc Natl Acad Sci U S A 109, 7362-7367. doi:10.1073/pnas.1201595109

Horii, M., Bui, T., Touma, O., Cho, H. Y. and Parast, M. M. (2019). An ImprovedTwo-Step Protocol for Trophoblast Differentiation of Human Pluripotent StemCells. Curr Protoc Stem Cell Biol 50, e96. doi:10.1002/cpsc.96

Huch, M., Knoblich, J. A., Lutolf, M. P. and Martinez-Arias, A. (2017). The hopeand the hype of organoid research. Development 144, 938-941. doi:10.1242/dev.150201

Idkowiak, J., Weisheit, G., Plitzner, J. and Viebahn, C. (2004). Hypoblast controlsmesoderm generation and axial patterning in the gastrulating rabbit embryo. DevGenes Evol 214, 591-605. doi:10.1007/s00427-004-0436-y

Ivec, M., Kovacic, B. and Vlaisavljevic, V. (2011). Prediction of human blastocystdevelopment from morulas with delayed and/or incomplete compaction. Fertil.Steril., 96, 1473-1478e2.

Iwata, K., Yumoto, K., Sugishima, M., Mizoguchi, C., Kai, Y., Iba, Y. and Mio, Y.(2014). Analysis of compaction initiation in human embryos by using time-lapsecinematography. J Assist Reprod Genet 31, 421-426. doi:10.1007/s10815-014-0195-2

Kalkan, T. and Smith, A. (2014). Mapping the route from naive pluripotency tolineage specification. Philos. Trans. R. Soc. Lond. B Biol. Sci., 369.

Keibel, F. and Mall, F. P. (1912). Manual of Human Embryology. JB LippincottCompany.

Kim, J., Kim, S. H. and Jun, J. H. (2017). Prediction of blastocyst development andimplantation potential in utero based on the third cleavage and compaction timesin mouse pre-implantation embryos. J Reprod Dev 63, 117-125. doi:10.1262/jrd.2016-129

Kirby, T. J. and Lammerding, J. (2018). Emerging views of the nucleus as a cellularmechanosensor. Nat Cell Biol 20, 373-381. doi:10.1038/s41556-018-0038-y

Kojima, Y., Tam, O. H. and Tam, P. P. (2014). Timing of developmental events inthe early mouse embryo. Semin Cell Dev Biol 34, 65-75. doi:10.1016/j.semcdb.2014.06.010

Korotkevich, E., Niwayama, R., Courtois, A., Friese, S., Berger, N., Buchholz, F.and Hiiragi, T. (2017). The Apical Domain Is Required and Sufficient for the FirstLineage Segregation in the Mouse Embryo. Dev. Cell, 40, 235-247e7.

Krendl, C., Shaposhnikov, D., Rishko, V., Ori, C., Ziegenhain, C., Sass, S.,Simon, L., Muller, N. S., Straub, T., Brooks, K. E. et al. (2017). GATA2/3-TFAP2A/C transcription factor network couples human pluripotent stem celldifferentiation to trophectoderm with repression of pluripotency. Proc Natl AcadSci U S A 114, E9579-E9588. doi:10.1073/pnas.1708341114

Kushnir, V. A., Barad, D. H., Albertini, D. F., Darmon, S. K. and Gleicher, N.(2017). Systematic review of worldwide trends in assisted reproductive technology2004-2013. Reprod Biol Endocrinol 15, 6. doi:10.1186/s12958-016-0225-2

Larsen, E. C., Christiansen, O. B., Kolte, A. M. and Macklon, N. (2013). Newinsights into mechanisms behind miscarriage. BMC Med 11, 154. doi:10.1186/1741-7015-11-154

Leung, C. Y. and Zernicka-Goetz, M. (2013). Angiomotin prevents pluripotentlineage differentiation in mouse embryos via Hippo pathway-dependent and-independent mechanisms. Nat Commun 4, 2251. doi:10.1038/ncomms3251

Leung, C. Y., Zhu, M. and Zernicka-Goetz, M. (2016). Polarity in Cell-FateAcquisition in the Early Mouse Embryo. Curr Top Dev Biol 120, 203-234. doi:10.1016/bs.ctdb.2016.04.008

Li, S., Edgar, D., Fassler, R., Wadsworth, W. and Yurchenco, P. D. (2003). Therole of laminin in embryonic cell polarization and tissue organization. Dev Cell 4,613-624. doi:10.1016/S1534-5807(03)00128-X

Liang, X., Zhou, Q., Li, X., Sun, Y., Lu, M., Dalton, N., Ross, Jr., J. and Chen, J.(2005). PINCH1 plays an essential role in early murine embryonic developmentbut is dispensable in ventricular cardiomyocytes. Mol Cell Biol 25, 3056-3062.doi:10.1128/MCB.25.8.3056-3062.2005

Licciardi, F., Lhakhang, T., Kramer, Y. G., Zhang, Y., Heguy, A. and Tsirigos, A.(2018). Human blastocysts of normal and abnormal karyotypes display distincttranscriptome profiles. Sci Rep 8, 14906. doi:10.1038/s41598-018-33279-0

Lindenberg, S., Nielsen, M. H. and Lenz, S. (1985). In vitro studies of humanblastocyst implantation. Ann N Y Acad Sci 442, 368-374. doi:10.1111/j.1749-6632.1985.tb37541.x

Linneberg-Agerholm, M., Wong, Y. F., Romero Herrera, J. A., Monteiro, R. S.,Anderson, K. G. V. and Brickman, J. M. (2019). Naive human pluripotent stemcells respond toWnt, Nodal and LIF signalling to produce expandable naive extra-embryonic endoderm. Development, 146.

Liu, J., He, X., Corbett, S. A., Lowry, S. F., Graham, A. M., Fassler, R. and Li, S.(2009). Integrins are required for the differentiation of visceral endoderm. JCell Sci122, 233-242. doi:10.1242/jcs.037663

Ljunger, E., Cnattingius, S., Lundin, C. and Anneren, G. (2005). Chromosomalanomalies in first-trimester miscarriages. Acta Obstet Gynecol Scand 84,1103-1107. doi:10.1111/j.0001-6349.2005.00882.x

Luckett, W. P. (1975). The development of primordial and definitive amnioticcavities in early Rhesus monkey and human embryos. Am J Anat 144, 149-167.doi:10.1002/aja.1001440204

Ma, H., Zhai, J., Wan, H., Jiang, X., Wang, X., Wang, L., Xiang, Y., He, X., Zhao,Z. A., Zhao, B. et al. (2019). In vitro culture of cynomolgus monkey embryosbeyond early gastrulation. Science, 366.

Macklon, N. S., Geraedts, J. P. and Fauser, B. C. (2002). Conception to ongoingpregnancy: the ‘black box’ of early pregnancy loss. Hum Reprod Update 8,333-343. doi:10.1093/humupd/8.4.333

Maitre, J. L., Niwayama, R., Turlier, H., Nedelec, F. and Hiiragi, T. (2015).Pulsatile cell-autonomous contractility drives compaction in the mouse embryo.Nat Cell Biol 17, 849-855. doi:10.1038/ncb3185

Maitre, J. L., Turlier, H., Illukkumbura, R., Eismann, B., Niwayama, R., Nedelec,F. and Hiiragi, T. (2016). Asymmetric division of contractile domains couples cellpositioning and fate specification. Nature 536, 344-348. doi:10.1038/nature18958

Mantikou, E., Wong, K. M., Repping, S. and Mastenbroek, S. (2012). Molecularorigin of mitotic aneuploidies in preimplantation embryos. Biochim Biophys Acta1822, 1921-1930. doi:10.1016/j.bbadis.2012.06.013

Marcos, J., Perez-Albala, S., Mifsud, A., Molla, M., Landeras, J. and Meseguer,M. (2015). Collapse of blastocysts is strongly related to lower implantationsuccess: a time-lapse study. Hum Reprod 30, 2501-2508. doi:10.1093/humrep/dev216

Martinez, M. C., Mendez, C., Ferro, J., Nicolas, M., Serra, V. and Landeras, J.(2010). Cytogenetic analysis of early nonviable pregnancies after assistedreproduction treatment. Fertil Steril 93, 289-292. doi:10.1016/j.fertnstert.2009.07.989

Martyn, I., Kanno, T. Y., Ruzo, A., Siggia, E. D. and Brivanlou, A. H. (2018). Self-organization of a human organizer by combinedWnt and Nodal signalling.Nature558, 132-135. doi:10.1038/s41586-018-0150-y

Martyn, I., Brivanlou, A. H. and Siggia, E. D. (2019). A wave of WNT signalingbalanced by secreted inhibitors controls primitive streak formation in micropatterncolonies of human embryonic stem cells. Development, 146.

Mccallie, B. R., Parks, J. C., Patton, A. L., Griffin, D. K., Schoolcraft, W. B. andKatz-Jaffe, M. G. (2016). Hypomethylation and Genetic Instability in MonosomyBlastocysts May Contribute to Decreased Implantation Potential. PLoS One 11,e0159507. doi:10.1371/journal.pone.0159507

Mcdole, K., Guignard, L., Amat, F., Berger, A., Malandain, G., Royer, L. A.,Turaga, S. C., Branson, K. and Keller, P. J. (2018). In Toto Imaging andReconstruction of Post-Implantation Mouse Development at the Single-Cell Level.Cell, 175, 859-876e33.

11

REVIEW Development (2020) 147, dev190629. doi:10.1242/dev.190629

DEVELO

PM

ENT

Page 12: Mechanisms of human embryo development: from cell fate to … · development have so far been technically and ethically challenging. However, recent technical developments provide

Meng, Y., Cai, K. Q., Moore, R., Tao, W., Tse, J. D., Smith, E. R. and Xu, X.-X.(2017). Pten facilitates epiblast epithelial polarization and proamniotic lumenformation in early mouse embryos. Dev Dyn 246, 517-530. doi:10.1002/dvdy.24503

Minasi, M. G., Colasante, A., Riccio, T., Ruberti, A., Casciani, V., Scarselli, F.,Spinella, F., Fiorentino, F., Varricchio, M. T. and Greco, E. (2016). Correlationbetween aneuploidy, standard morphology evaluation and morphokineticdevelopment in 1730 biopsied blastocysts: a consecutive case series study.Hum Reprod 31, 2245-2254. doi:10.1093/humrep/dew183

Minn, K. T., Fu, Y. C., He, S., George, S. C., Anastasio, M. A., Morris, S. A. andSolnica-Krezel, L. (2020). High-resolution transcriptional and morphogeneticprofiling of cells from micropatterned human embryonic stem cell gastruloidcultures. bioRxiv.

Mischler, A., Karakis, V., Mahinthakumar, J., Carberry, C., San Miguel, A.,Rager, J., Fry, R. and Rao, B. M. (2019). Two distinct trophectoderm lineagestem cells from human pluripotent stem cells. bioRxiv.

Mizobe, Y., Ezono, Y., Tokunaga, M., Oya, N., Iwakiri, R., Yoshida, N., Sato, Y.,Onoue, N. and Miyoshi, K. (2017). Selection of human blastocysts with a highimplantation potential based on timely compaction. J Assist Reprod Genet 34,991-997. doi:10.1007/s10815-017-0962-y

Muncie, J. M., Ayad, N. M. E., Lakins, J. N. and Weaver, V. M. (2020). Mechanicsregulate human embryonic stem cell self-organization to specify mesoderm.bioRxiv.

Munne, S., Tomkin, G. and Cohen, J. (2009). Selection of embryos by morphologyis less effective than by a combination of aneuploidy testing and morphologyobservations. Fertil Steril 91, 943-945. doi:10.1016/j.fertnstert.2007.06.082

Nagaoka, S. I., Hassold, T. J. and Hunt, P. A. (2012). Human aneuploidy:mechanisms and new insights into an age-old problem. Nat Rev Genet 13,493-504. doi:10.1038/nrg3245

Nakamura, T., Okamoto, I., Sasaki, K., Yabuta, Y., Iwatani, C., Tsuchiya, H.,Seita, Y., Nakamura, S., Yamamoto, T. and Saitou, M. (2016). A developmentalcoordinate of pluripotency among mice, monkeys and humans. Nature 537,57-62. doi:10.1038/nature19096

Nazareth, E. J., Ostblom, J. E., Lucker, P. B., Shukla, S., Alvarez, M. M., Oh,S. K., Yin, T. and Zandstra, P. W. (2013). High-throughput fingerprinting ofhuman pluripotent stem cell fate responses and lineage bias. Nat Methods 10,1225-1231. doi:10.1038/nmeth.2684

Nemashkalo, A., Ruzo, A., Heemskerk, I. and Warmflash, A. (2017). Morphogenand community effects determine cell fates in response to BMP4 signaling inhuman embryonic stem cells. Development 144, 3042-3053. doi:10.1242/dev.153239

Niakan, K. K. and Eggan, K. (2013). Analysis of human embryos from zygote toblastocyst reveals distinct gene expression patterns relative to the mouse. DevBiol 375, 54-64. doi:10.1016/j.ydbio.2012.12.008

Nichols, J. and Gardner, R. L. (1984). Heterogeneous differentiation of externalcells in individual isolated early mouse inner cell masses in culture. J Embryol ExpMorphol 80, 225-240.

Nichols, J. and Smith, A. (2012). Pluripotency in the embryo and in culture. ColdSpring Harb Perspect Biol 4, a008128. doi:10.1101/cshperspect.a008128

Nikas, G., Ao, A., Winston, R. M. and Handyside, A. H. (1996). Compaction andsurface polarity in the human embryo in vitro. Biol Reprod 55, 32-37. doi:10.1095/biolreprod55.1.32

Nishimura, H., Takano, K., Tanimura, T. and Yasuda, M. (1968). Normal andabnormal development of human embryos: first report of the analysis of 1213intact embryos. Teratology 1, 281-290. doi:10.1002/tera.1420010306

Niu, Y., Sun, N., Li, C., Lei, Y., Huang, Z., Wu, J., Si, C., Dai, X., Liu, C., Wei, J.et al. (2019). Dissecting primate early post-implantation development using long-term in vitro embryo culture. Science, 366.

Niwa, H., Toyooka, Y., Shimosato, D., Strumpf, D., Takahashi, K., Yagi, R. andRossant, J. (2005). Interaction between Oct3/4 and Cdx2 determinestrophectoderm differentiation. Cell 123, 917-929. doi:10.1016/j.cell.2005.08.040

Noli, L., Capalbo, A., Ogilvie, C., Khalaf, Y. and Ilic, D. (2015). Discordant Growthof Monozygotic Twins Starts at the Blastocyst Stage: A Case Study. Stem CellReports 5, 946-953. doi:10.1016/j.stemcr.2015.10.006

Peerani, R., Rao, B. M., Bauwens, C., Yin, T.,Wood, G. A., Nagy, A., Kumacheva,E. and Zandstra, P. W. (2007). Niche-mediated control of human embryonic stemcell self-renewal and differentiation.Embo J 26, 4744-4755. doi:10.1038/sj.emboj.7601896

Peng, G., Suo, S., Chen, J., Chen,W., Liu, C., Yu, F., Wang, R., Chen, S., Sun, N.,Cui, G. et al. (2016). Spatial Transcriptome for the Molecular Annotation ofLineage Fates and Cell Identity in Mid-gastrula Mouse Embryo. Dev Cell 36,681-697. doi:10.1016/j.devcel.2016.02.020

Pera, M. F. (2017). Human embryo research and the 14-day rule. Development 144,1923-1925. doi:10.1242/dev.151191

Petropoulos, S., Edsgard, D., Reinius, B., Deng, Q., Panula, S. P., Codeluppi, S.,Plaza Reyes, A., Linnarsson, S., Sandberg, R. and Lanner, F. (2016). Single-Cell RNA-Seq Reveals Lineage and X Chromosome Dynamics in HumanPreimplantation Embryos. Cell, 165, 1012-1026.

Philipp, T., Philipp, K., Reiner, A., Beer, F. and Kalousek, D. K. (2003).Embryoscopic and cytogenetic analysis of 233 missed abortions: factors involved

in the pathogenesis of developmental defects of early failed pregnancies. HumReprod 18, 1724-1732. doi:10.1093/humrep/deg309

Pieters, T. and Van Roy, F. (2014). Role of cell-cell adhesion complexes inembryonic stem cell biology. J Cell Sci 127, 2603-2613. doi:10.1242/jcs.146720

Plusa, B., Frankenberg, S., Chalmers, A., Hadjantonakis, A. K., Moore, C. A.,Papalopulu, N., Papaioannou, V. E., Glover, D. M. and Zernicka-Goetz, M.(2005). Downregulation of Par3 and aPKC function directs cells towards the Icm inthe preimplantation mouse embryo. J Cell Sci 118, 505-515. doi:10.1242/jcs.01666

Popovic, M., Dhaenens, L., Taelman, J., Dheedene, A., Bialecka, M., De Sutter,P., Chuva De Sousa Lopes, S. M., Menten, B. and Heindryckx, B. (2019).Extended in vitro culture of human embryos demonstrates the complex nature ofdiagnosing chromosomal mosaicism from a single trophectoderm biopsy. Hum.Reprod.

Posfai, E., Petropoulos, S., De Barros, F. R. O., Schell, J. P., Jurisica, I.,Sandberg, R., Lanner, F. and Rossant, J. (2017). Position- and Hippo signaling-dependent plasticity during lineage segregation in the earlymouse embryo.Elife, 6.

Posfai, E., Schell, J. P., Janiszewski, A., Rovic, I., Murray, A., Bradshaw, B.,Pardon, T., El Bakkali, M., Talon, I., De Geest, N. et al. (2020). Definingtotipotency using criteria of increasing stringency. bioRxiv.

Przybyla, L., Lakins, J. N. and Weaver, V. M. (2016). Tissue MechanicsOrchestrate Wnt-Dependent Human Embryonic Stem Cell Differentiation. CellStem Cell 19, 462-475. doi:10.1016/j.stem.2016.06.018

Qin, H., Hejna, M., Liu, Y., Percharde, M., Wossidlo, M., Blouin, L., Durruthy-Durruthy, J., Wong, P., Qi, Z., Yu, J. et al. (2016). YAP Induces Human NaivePluripotency. Cell Rep 14, 2301-2312. doi:10.1016/j.celrep.2016.02.036

Ralston, A., Cox, B. J., Nishioka, N., Sasaki, H., Chea, E., Rugg-Gunn, P., Guo,G., Robson, P., Draper, J. S. and Rossant, J. (2010). Gata3 regulatestrophoblast development downstream of Tead4 and in parallel to Cdx2.Development 137, 395-403. doi:10.1242/dev.038828

Ranga, A., Gobaa, S., Okawa, Y., Mosiewicz, K., Negro, A. and Lutolf, M. P.(2014). 3D niche microarrays for systems-level analyses of cell fate. Nat Commun5, 4324. doi:10.1038/ncomms5324

Ranga, A., Girgin, M., Meinhardt, A., Eberle, D., Caiazzo, M., Tanaka, E. M. andLutolf, M. P. (2016). Neural tube morphogenesis in synthetic 3Dmicroenvironments. Proc Natl Acad Sci U S A 113, E6831-E6839. doi:10.1073/pnas.1603529113

Rayon, T., Menchero, S., Nieto, A., Xenopoulos, P., Crespo, M., Cockburn, K.,Canon, S., Sasaki, H., Hadjantonakis, A. K., De La Pompa, J. L. et al. (2014).Notch and hippo converge on Cdx2 to specify the trophectoderm lineage in themouse blastocyst. Dev Cell 30, 410-422. doi:10.1016/j.devcel.2014.06.019

Rivron, N. C., Frias-Aldeguer, J., Vrij, E.-J., Boisset, J. C., Korving, J., Vivie, J.,Truckenmuller, R. K., Van Oudenaarden, A., Van Blitterswijk, C. A. andGeijsen, N. (2018). Blastocyst-like structures generated solely from stem cells.Nature 557, 106-111. doi:10.1038/s41586-018-0051-0

Rock, J. and Menkin, M. F. (1944). In Vitro Fertilization and Cleavage of HumanOvarian Eggs. Science 100, 105-107. doi:10.1126/science.100.2588.105

Ryan, A. Q., Chan, C. J., Graner, F. and Hiiragi, T. (2019). Lumen ExpansionFacilitates Epiblast-Primitive Endoderm Fate Specification during MouseBlastocyst Formation. Dev. Cell, 51, 684-697e4.

Saiz, N., Grabarek, J. B., Sabherwal, N., Papalopulu, N. and Plusa, B. (2013).Atypical protein kinase C couples cell sorting with primitive endoderm maturationin the mouse blastocyst. Development 140, 4311-4322. doi:10.1242/dev.093922

Sakai, T., Li, S., Docheva, D., Grashoff, C., Sakai, K., Kostka, G., Braun, A.,Pfeifer, A., Yurchenco, P. D. and Fassler, R. (2003). Integrin-linked kinase (ILK)is required for polarizing the epiblast, cell adhesion, and controlling actinaccumulation. Genes Dev 17, 926-940. doi:10.1101/gad.255603

Samarage, C. R., White, M. D., Alvarez, Y. D., Fierro-Gonzalez, J. C., Henon, Y.,Jesudason, E. C., Bissiere, S., Fouras, A. and Plachta, N. (2015). CorticalTension Allocates the First Inner Cells of the Mammalian Embryo. Dev Cell 34,435-447. doi:10.1016/j.devcel.2015.07.004

Sasaki, H. (2015). Position- and polarity-dependent Hippo signaling regulates cellfates in preimplantation mouse embryos. Semin Cell Dev Biol 47-48, 80-87.doi:10.1016/j.semcdb.2015.05.003

Sasaki, K., Nakamura, T., Okamoto, I., Yabuta, Y., Iwatani, C., Tsuchiya, H.,Seita, Y., Nakamura, S., Shiraki, N., Takakuwa, T. et al. (2016). The Germ CellFate of Cynomolgus Monkeys Is Specified in the Nascent Amnion. Dev Cell 39,169-185. doi:10.1016/j.devcel.2016.09.007

Schwartz, M. A. (2010). Integrins and extracellular matrix in mechanotransduction.Cold Spring Harb Perspect Biol 2, a005066. doi:10.1101/cshperspect.a005066

Shahbazi, M. N. and Zernicka-Goetz, M. (2018). Deconstructing andreconstructing the mouse and human early embryo. Nat Cell Biol 20, 878-887.doi:10.1038/s41556-018-0144-x

Shahbazi, M. N., Jedrusik, A., Vuoristo, S., Recher, G., Hupalowska, A., Bolton,V., Fogarty, N. M., Campbell, A., Devito, L. G., Ilic, D. et al. (2016). Self-organization of the human embryo in the absence of maternal tissues. Nat CellBiol 18, 700-708. doi:10.1038/ncb3347

Shahbazi, M. N., Scialdone, A., Skorupska, N., Weberling, A., Recher, G., Zhu,M., Jedrusik, A., Devito, L. G., Noli, L., Macaulay, I. C. et al. (2017). Pluripotent

12

REVIEW Development (2020) 147, dev190629. doi:10.1242/dev.190629

DEVELO

PM

ENT

Page 13: Mechanisms of human embryo development: from cell fate to … · development have so far been technically and ethically challenging. However, recent technical developments provide

state transitions coordinate morphogenesis in mouse and human embryos.Nature 552, 239-243. doi:10.1038/nature24675

Shahbazi, M. N., Siggia, E. D. and Zernicka-Goetz, M. (2019). Self-organization ofstem cells into embryos: A window on early mammalian development. Science364, 948-951. doi:10.1126/science.aax0164

Shao, Y., Taniguchi, K., Gurdziel, K., Townshend, R. F., Xue, X., Yong, K. M.,Sang, J., Spence, J. R., Gumucio, D. L. and Fu, J. (2016). Self-organizedamniogenesis by human pluripotent stem cells in a biomimetic implantation-likeniche. Nat. Mater.

Shao, Y., Taniguchi, K., Townshend, R. F., Miki, T., Gumucio, D. L. and Fu, J.(2017). A pluripotent stem cell-based model for post-implantation human amnioticsac development. Nat Commun 8, 208. doi:10.1038/s41467-017-00236-w

Sheng, G. (2015). Epiblast morphogenesis before gastrulation.Dev Biol 401, 17-24.doi:10.1016/j.ydbio.2014.10.003

Shettles, L. B. (1955). Amorula stage of human ovum developed in vitro. Fertil Steril6, 287-289. doi:10.1016/S0015-0282(16)32040-4

Siggia, E. D. and Warmflash, A. (2018). Modeling Mammalian Gastrulation WithEmbryonic Stem Cells. Curr Top Dev Biol 129, 1-23. doi:10.1016/bs.ctdb.2018.03.001

Simunovic, M. andBrivanlou, A. H. (2017). Embryoids, organoids and gastruloids:new approaches to understanding embryogenesis. Development 144, 976-985.doi:10.1242/dev.143529

Skiadas, C. C., Jackson, K. V. and Racowsky, C. (2006). Early compaction on day3 may be associated with increased implantation potential. Fertil Steril 86,1386-1391. doi:10.1016/j.fertnstert.2006.03.051

Smyth, N., Vatansever, H. S., Murray, P., Meyer, M., Frie, C., Paulsson, M. andEdgar, D. (1999). Absence of basement membranes after targeting the LAMC1gene results in embryonic lethality due to failure of endoderm differentiation. J CellBiol 144, 151-160. doi:10.1083/jcb.144.1.151

Sozen, B., Amadei, G., Cox, A., Wang, R., Na, E., Czukiewska, S., Chappell, L.,Voet, T., Michel, G., Jing, N. et al. (2018). Gastrulating complete embryo-likestructures reconstituted from stem cells in vitro. Nat. Cell Biol.

Starostik, M. R., Sosina, O. A. andMccoy, R. (2020). Single-cell analysis of humanembryos reveals diverse patterns of aneuploidy and mosaicism. bioRxiv.

Stephenson, M. D., Awartani, K. A. and Robinson, W. P. (2002). Cytogeneticanalysis of miscarriages from couples with recurrent miscarriage: a case-controlstudy. Hum Reprod 17, 446-451. doi:10.1093/humrep/17.2.446

Steptoe, P. C., Edwards, R. G. and Purdy, J. M. (1971). Human blastocysts grownin culture. Nature 229, 132-133. doi:10.1038/229132a0

Stirparo, G. G., Boroviak, T., Guo, G., Nichols, J., Smith, A. and Bertone, P.(2018). Integrated analysis of single-cell embryo data yields a unifiedtranscriptome signature for the human pre-implantation epiblast. Development,145.

Stower, M. J. and Srinivas, S. (2014). Heading forwards: anterior visceralendodermmigration in patterning the mouse embryo.Philos. Trans. R. Soc. Lond.B Biol. Sci., 369.

Strumpf, D., Mao, C. A., Yamanaka, Y., Ralston, A., Chawengsaksophak, K.,Beck, F. and Rossant, J. (2005). Cdx2 is required for correct cell fatespecification and differentiation of trophectoderm in the mouse blastocyst.Development 132, 2093-2102. doi:10.1242/dev.01801

Sun, Y., Yong, K. M., Villa-Diaz, L. G., Zhang, X., Chen,W., Philson, R.,Weng, S.,Xu, H., Krebsbach, P. H. and Fu, J. (2014). Hippo/YAP-mediated rigidity-dependent motor neuron differentiation of human pluripotent stem cells.Nat Mater13, 599-604. doi:10.1038/nmat3945

Tachibana, M., Sparman, M., Ramsey, C., Ma, H., Lee, H. S., Penedo, M. C. andMitalipov, S. (2012). Generation of chimeric rhesus monkeys. Cell 148, 285-295.doi:10.1016/j.cell.2011.12.007

Takashima, Y., Guo, G., Loos, R., Nichols, J., Ficz, G., Krueger, F., Oxley, D.,Santos, F., Clarke, J., Mansfield, W. et al. (2014). Resetting Transcription FactorControl Circuitry toward Ground-State Pluripotency in Human. Cell 158,1254-1269. doi:10.1016/j.cell.2014.08.029

Tam, P. P. and Behringer, R. R. (1997). Mouse gastrulation: the formation of amammalian body plan.Mech Dev 68, 3-25. doi:10.1016/S0925-4773(97)00123-8

Tam, P. P. and Loebel, D. A. (2007). Gene function in mouse embryogenesis: getset for gastrulation. Nat Rev Genet 8, 368-381. doi:10.1038/nrg2084

Taniguchi, K., Shao, Y., Townshend, R. F., Tsai, Y. H., Delong, C. J., Lopez,S. A., Gayen, S., Freddo, A. M., Chue, D. J., Thomas, D. J. et al. (2015). LumenFormation Is an Intrinsic Property of Isolated Human Pluripotent Stem Cells. StemCell Reports 5, 954-962. doi:10.1016/j.stemcr.2015.10.015

Tewary, M., Ostblom, J., Prochazka, L., Zulueta-Coarasa, T., Shakiba, N.,Fernandez-Gonzalez, R. and Zandstra, P. W. (2017). A stepwise model ofReaction-Diffusion and Positional-Information governs self-organized humanperi-gastrulation-like patterning. Development.

Thery, M. and Piel, M. (2009). Adhesive micropatterns for cells: a microcontactprinting protocol. Cold Spring Harb Protoc, doi:10.1101/pdb.prot5255

Theunissen, T. W., Friedli, M., He, Y., Planet, E., O’neil, R. C., Markoulaki, S.,Pontis, J.,Wang, H., Iouranova, A., Imbeault, M. et al. (2016). Molecular Criteriafor Defining the Naive Human Pluripotent State. Cell Stem Cell 19, 502-515.doi:10.1016/j.stem.2016.06.011

Thomson, J. A., Itskovitz-Eldor, J., Shapiro, S. S., Waknitz, M. A., Swiergiel,J. J., Marshall, V. S. and Jones, J. M. (1998). Embryonic stem cell lines derivedfrom human blastocysts. Science 282, 1145-1147. doi:10.1126/science.282.5391.1145

Turing, A. M. (1952). The chemical basis of morphogenesis. Philos Trans R SocLond B Biol Sci 237, 37-72. doi:10.1098/rstb.1952.0012

VanDenBrink, S. C., Alemany, A., VanBatenburg, V., Moris, N., Blotenburg, M.,Vivie, J., Baillie-Johnson, P., Nichols, J., Sonnen, K. F., Martinez Arias, A.et al. (2020). Single-cell and spatial transcriptomics reveal somitogenesis ingastruloids. Nature.

Vanneste, E., Voet, T., Le Caignec, C., Ampe, M., Konings, P., Melotte, C.,Debrock, S., Amyere, M., Vikkula, M., Schuit, F. et al. (2009). Chromosomeinstability is common in human cleavage-stage embryos. Nat Med 15, 577-583.doi:10.1038/nm.1924

Verstreken, C. M., Labouesse, C., Agley, C. C. and Chalut, K. J. (2019).Embryonic stem cells becomemechanoresponsive upon exit from ground state ofpluripotency. Open Biol 9, 180203. doi:10.1098/rsob.180203

Vianello, S. and Lutolf, M. P. (2019). Understanding the Mechanobiology of EarlyMammalian Development through Bioengineered Models. Dev Cell 48, 751-763.doi:10.1016/j.devcel.2019.02.024

Warmflash, A., Sorre, B., Etoc, F., Siggia, E. D. and Brivanlou, A. H. (2014). Amethod to recapitulate early embryonic spatial patterning in human embryonicstem cells. Nat Methods 11, 847-854. doi:10.1038/nmeth.3016

Weimar, C. H., Post Uiterweer, E. D., Teklenburg, G., Heijnen, C. J. andMacklon, N. S. (2013). In-vitro model systems for the study of human embryo-endometrium interactions. Reprod Biomed Online 27, 461-476. doi:10.1016/j.rbmo.2013.08.002

West, R. C., Ming, H., Logsdon, D. M., Sun, J., Rajput, S. K., Kile, R. A.,Schoolcraft, W. B., Roberts, R. M., Krisher, R. L., Jiang, Z. et al. (2019).Dynamics of trophoblast differentiation in peri-implantation-stage humanembryos. Proc Natl Acad Sci U S A 116, 22635-22644. doi:10.1073/pnas.1911362116

Wigger, M., Kisielewska, K., Filimonow, K., Plusa, B., Maleszewski, M. andSuwinska, A. (2017). Plasticity of the inner cell mass in mouse blastocyst isrestricted by the activity of FGF/MAPK pathway. Sci Rep 7, 15136. doi:10.1038/s41598-017-15427-0

Wilcox, A. J., Weinberg, C. R., O’connor, J. F., Baird, D. D., Schlatterer, J. P.,Canfield, R. E., Armstrong, E. G. and Nisula, B. C. (1988). Incidence of earlyloss of pregnancy. N Engl J Med 319, 189-194. doi:10.1056/NEJM198807283190401

Wilcox, A. J., Baird, D. D. and Weinberg, C. R. (1999). Time of implantation of theconceptus and loss of pregnancy. N Engl J Med 340, 1796-1799. doi:10.1056/NEJM199906103402304

Wilcox, A. J., Harmon, Q., Doody, K., Wolf, D. P. and Adashi, E. Y. (2020).Preimplantation loss of fertilized human ova: estimating the unobservable. Hum.Reprod.

Wolpert, L. (1969). Positional information and the spatial pattern of cellulardifferentiation. J Theor Biol 25, 1-47. doi:10.1016/S0022-5193(69)80016-0

Wolpert, L. and Vicente, C. (2015). An interview with Lewis Wolpert. Development142, 2547-2548. doi:10.1242/dev.127373

Wong, C. C., Loewke, K. E., Bossert, N. L., Behr, B., De Jonge, C. J., Baer, T. M.and Reijo Pera, R. A. (2010). Non-invasive imaging of human embryos beforeembryonic genome activation predicts development to the blastocyst stage. NatBiotechnol 28, 1115-1121. doi:10.1038/nbt.1686

Xiang, L., Yin, Y., Zheng, Y., Ma, Y., Li, Y., Zhao, Z., Guo, J., Ai, Z., Niu, Y., Duan,K. et al. (2019). A developmental landscape of 3D-cultured human pre-gastrulation embryos. Nature.

Xu, R. H., Chen, X., Li, D. S., Li, R., Addicks, G. C., Glennon, C., Zwaka, T. P. andThomson, J. A. (2002). BMP4 initiates human embryonic stem cell differentiationto trophoblast. Nat Biotechnol 20, 1261-1264. doi:10.1038/nbt761

Yamada, S., Hill, M. and Takakuwa, T. (2015). Human embryology. In NewDiscoveries in Embryology (ed. B. Wu), pp. 97-124. IntechOpen.

Yan, L., Yang, M., Guo, H., Yang, L., Wu, J., Li, R., Liu, P., Lian, Y., Zheng, X.,Yan, J. et al. (2013). Single-cell RNA-Seq profiling of human preimplantationembryos and embryonic stem cells. Nat Struct Mol Biol 20, 1131-1139. doi:10.1038/nsmb.2660

Zhang, Z., Zwick, S., Loew, E., Grimley, J. S. and Ramanathan, S. (2019). Mouseembryo geometry drives formation of robust signaling gradients through receptorlocalization. Nat. Commun. 10, 4516. doi:10.1038/s41467-019-12533-7

Zheng, Y., Xue, X., Shao, Y., Wang, S., Esfahani, S. N., Li, Z., Muncie, J. M.,Lakins, J. N., Weaver, V. M., Gumucio, D. L. et al. (2019). Controlled modellingof human epiblast and amnion development using stem cells. Nature 573,421-425. doi:10.1038/s41586-019-1535-2

Zhou, F., Wang, R., Yuan, P., Ren, Y., Mao, Y., Li, R., Lian, Y., Li, J., Wen, L., Yan,L. et al. (2019). Reconstituting the transcriptome and Dna methylome landscapesof human implantation. Nature 572, 660-664. doi:10.1038/s41586-019-1500-0

Zhu, M., Leung, C. Y., Shahbazi, M. N. and Zernicka-Goetz, M. (2017).Actomyosin polarisation through PLC-PKC triggers symmetry breaking of themouse embryo. Nat Commun 8, 921. doi:10.1038/s41467-017-00977-8

13

REVIEW Development (2020) 147, dev190629. doi:10.1242/dev.190629

DEVELO

PM

ENT

Page 14: Mechanisms of human embryo development: from cell fate to … · development have so far been technically and ethically challenging. However, recent technical developments provide

Zhu, P., Guo, H., Ren, Y., Hou, Y., Dong, J., Li, R., Lian, Y., Fan, X., Hu, B., Gao, Y.et al. (2018). Single-cell DNA methylome sequencing of human preimplantationembryos. Nat Genet 50, 12-19. doi:10.1038/s41588-017-0007-6

Zhu, M., Wang, P., Handford, C. E., Na, J. and Zernicka-Goetz, M. (2020).Transcriptional control of apical protein clustering drives de novo cell polarityestablishment in the early mouse embryo. bioRxiv.

14

REVIEW Development (2020) 147, dev190629. doi:10.1242/dev.190629

DEVELO

PM

ENT