You are on page 1of 7

Available online at www.sciencedirect.

com

ScienceDirect

Bioengineered pluripotent stem cell models:


new approaches to explore early human embryo
development
Agnes M Resto Irizarry1, Sajedeh Nasr Esfahani1 and
Jianping Fu1,2,3

Human development is a complex process in which place across multiple spatial and temporal scales, with
environmental signals and factors encoded by the genome processes at the subcellular level affecting the behavior of
interact to engender cell fate changes and self-organization individual cells and leading to complex emergent multi-
that drive the progressive formation of the human body. Herein, cellular phenomena. The difficulty of studying human
we discuss engineered biomimetic platforms with controllable embryo development in vivo has led researchers to turn to
environments that are being used to develop human pluripotent the use of animal models. Monkey embryos have been
stem cell (hPSC)-based embryo models (or embryoids) that used to gain insights into primate development [1–4].
recapitulate a wide range of early human embryonic However, working with monkey embryos is expensive,
developmental events. Coupled with genome editing tools, time-consuming, and may raise its own ethical concerns
single-cell analysis, and computational models, they can be [5]. Research with the mouse is more ubiquitous, and
used to parse the spatiotemporal dynamics that lead to most of our current knowledge of mammalian develop-
differentiation, patterning, and growth in early human ment is obtained from the mouse embryo. Importantly,
development. Furthermore, we discuss ongoing efforts in great strides have been made in recent years with the
human extraembryonic lineage derivation and what can be creation of in vitro embryo models (or embryoids) using
learned from mouse embryoid models that have used both mouse stem cells [6,7,8,9,10]. However, we’ve come
embryonic and extraembryonic stem cells. Finally, we discuss to discover that there are key differences between the
promising bioengineering tools for the generation of more mouse and the human even at the early days of the
controllable systems and the need for validation of findings embryo development [11–14]. Another approach to study
from hPSC-based embryoid models. human development has been the culture of human
embryos obtained from in vitro fertilization [15–17]. This
Addresses approach has yielded insights into the autonomy and self-
1
Department of Mechanical Engineering, University of Michigan, Ann
organizing properties of human embryo development
Arbor, MI 48109, USA
2
even in the absence of the maternal environment. How-
Department of Biomedical Engineering, University of Michigan, Ann
ever, there is limited accessibility to human embryos, and
Arbor, MI 48109, USA
3
Department of Cell and Developmental Biology, University of Michigan studies of human embryos are further limited by the small
Medical School, Ann Arbor, MI 48109, USA window of research afforded by 14-day rule [18,19]. The
limitations of the aforementioned approaches have led
Corresponding author: Fu, Jianping (jpfu@umich.edu) researchers to take a bottom-up approach and use human
stem cells, particularly human pluripotent stem cells
Current Opinion in Biotechnology 2020, 66:52–58 (hPSCs), to construct synthetic models of human devel-
This review comes from a themed issue on Tissue, cell and pathway
opment (or human embryoid systems) [20,21].
engineering
Edited by Li Tang, Peng Xu and Haoran Zhang
Stem cell behavior is governed by both endogenous,
genome-encoded mechanisms, and exogenous signals
For a complete overview see the Issue and the Editorial
provided by the surrounding extracellular matrix
Available online 13th July 2020 (ECM) and cell–cell interactions. In order to construct
https://doi.org/10.1016/j.copbio.2020.06.005 embryoid models, researchers must provide stem cells
0958-1669/ã 2020 Elsevier Ltd. All rights reserved. with an environment that recapitulates the necessary
exogenous signals in the in vivo setting. Bioengineering
tools can be used to create biomimetic niches in which
parameters such as adhesive surface area, dimensionality,
and mechanical and chemical properties of the environ-
ment can be dynamically modulated to guide the pro-
Introduction gressive development of embryoid models to recapitulate
As one of the key areas in modern biology, human different aspects of embryonic development [22,23]. For
development is a complex process that involves cell example, microcontact printing is a commonly used
differentiation, self-organization, and growth. It takes tool to control adhesive surface area in order to obtain

Current Opinion in Biotechnology 2020, 66:52–58 www.sciencedirect.com


Bioengineered models of human embryo development Resto Irizarry, Nasr Esfahani and Fu 53

two-dimensional (2D) cell colonies of specific shapes and (Figure 1). Human embryoids aiming to recapitulate
sizes. The technique consists of coating a micropatterned the peri-implantation development of the human embryo
substrate or the stamp with adhesive ECM proteins would ideally include stem cell derivatives associated
before stamping it onto a planar 2D surface where cells with all of the three cell lineages, either from co-culture
will be seeded. Microcontact printing has allowed for the or from using human stem cells with the potency to give
study of how biomechanical mechanisms involving cell rise to both extraembryonic and embryonic lineages.
density and shape affect stem cell fate [24] as well as for Significant recent effort has been devoted to the devel-
the study of signaling dynamics within stem cell colonies opment of the human hypoblast stem cells from naı̈ve
[25–28]. Basement membrane matrixes and synthetic hPSCs [32,33]. While progress has been made to derive
hydrogels are often used to create three-dimensional human trophoblast stem cells from human blastocysts and
(3D) environments containing adhesive molecules. Stiff- placenta [34], many researchers have been using BMP4
ness of these 3D scaffolds can be tuned by changing the treatments to coax primed hPSCs to differentiate into
gel concentration or the crosslinker density in synthetic trophoblast-like cells [35]. Recent studies have also
hydrogels. This allows for assay optimization to obtain reported human expanded potential stem cells (hEPSCs)
desired behaviors of stem cells. Furthermore, it allows for [36–38] that seem to be able to give rise to both embry-
studies of how the mechanical properties of the environ- onic and extraembryonic lineages. However, the poten-
ment affect hPSC differentiation and self-organization cies of primed hPSCs and hEPSCs to give rise to extra-
[29]. These 3D scaffolds can be incorporated into a embryonic lineages are still under debate. A recent study
variety of bioengineering platforms that allow for the by Guo et al. [12] has shed doubt on the validity of these
control of initial cluster size of stem cells, including methods. They show that both primed hPSCs and
microwells and microfluidic devices. The current state hEPSCs have lost the capacity to differentiate into tro-
of microfabrication technologies is such that stem cell phoblast-like cells and that BMP4 treatment causes dif-
culture platforms can be made arbitrarily complex with ferentiation of both cell types into amniotic cells as shown
the use of microfluidic devices capable of creating by Shao et al. [29]. They further show that naı̈ve hPSCs
dynamic chemical gradients and the fabrication of sub- [39–41] are able to give rise to trophoblast-like cells but
strates with different topographies. Importantly, even require an unpredicted dedifferentiation process to the
simple bioengineering platforms that provide a homoge- earlier inner cell mass (ICM) state [12,42]. While it is clear
neous 2D or 3D environment are able to engender the that we still have gaps in our knowledge regarding the
formation of complex human embryoid structures from potencies of different human stem cell types, researchers
hPSCs [25,30,31]. have been developing hPSC-based embryoid models that
yield complex multicellular structures that recapitulate
The pre-implantation human embryo or the human blas- different aspects of the human embryonic development.
tocyst contains two extraembryonic lineages (trophoblast In this review, we highlight current developments in
and hypoblast) and one embryonic lineage (epiblast) hPSC-based models of human embryogenesis and discuss

Figure 1

16-cell morula Blastocyst Implantation Trophoblast


(E3) (E5) (E6-7)
Hypoblast
Blastomere Epiblast
Amnion
Pre-PS EPI
Zona pellucida Primordial germ
Maternal tissue
cell

Pro-amniotic cavity Amniotic sac Primitive streak formation

Epiblast cyst formation Amniotic sac formation Onset of gastrulation


(E7-12) (E9-12) (E15-17)
Current Opinion in Biotechnology

Schematic showing development of the human embryo in the first two weeks of pregnancy.

www.sciencedirect.com Current Opinion in Biotechnology 2020, 66:52–58


54 Tissue, cell and pathway engineering

how these embryoid models are anticipated to increase in 3D biomimetic platform with a soft gel bed made with the
complexity over the next few years. basement membrane matrix GeltrexTM and a 3D overlay
made with a low concentration of GeltrexTM diluted in
Embryoid models using human pluripotent culture medium (Figure 2c). hPSCs plated as single cells
stem cells cluster and undergo lumenogenesis to form pluripotent
During implantation of the human blastocyst, the epiblast cysts enclosing a central lumen, mimicking the formation
polarizes and undergoes lumenogenesis to form the pro- of the pro-amniotic cavity in the epiblast of the peri-
amniotic cavity (Figure 1). The lumenal epiblast struc- implantation human embryo. Importantly, a subset of
ture subsequently undergoes symmetry breaking, leading these cysts undergo symmetry breaking and yield the
to the formation of the bipolar amniotic sac, with squa- post-implantation amniotic sac embryoid (PASE). This
mous amniotic cells at the uterine-proximal pole and the asymmetric cyst consists of a central lumen, enclosed by
epiblast comprising the embryonic disc at the hypoblast- amniotic cells at one pole and epiblast-like cells at the
proximal pole (Figure 1). Soon after the formation of the opposite pole (Figure 2c), resembling the post-implanta-
amniotic sac, gastrulation is initiated with the formation tion human amniotic sac structure (Figure 1). The PASE
of the primitive streak at the prospective posterior end of was further able to recapitulate key events of gastrulation
the epiblast, resulting in the formation of the three germ including basal lamina breakdown, EMT, mesoderm
layers and establishment of the body axes. Researchers induction, and cell dissemination. BMP-SMAD signaling
have successfully leveraged the developmental potential was found to be important for amniotic differentiation of
and self-organizing property of hPSCs to develop embry- hPSCs [30]. However, the low yield of the asymmetric
oid models that help break down these developmental PASE structure and the lack of controllability of its
stages and enable mechanistic studies [25,30,31]. symmetry breaking and embryonic-extraembryonic axis
formation makes in depth mechanistic studies difficult.
Simple and controllable human embryoid systems can be More recently, Simunovic et al. [31] developed a 3D
used to recapitulate a variety of developmental events. hydrogel environment in which to study BMP4-induced
Combined with single-cell analysis and mathematical symmetry breaking events in lumenal pluripotent cysts
modeling, they can help us understand how processes formed by primed hPSCs (Figure 2b). By varying BMP4
at the subcellular level have a direct effect on tissue-level concentration, Simunovic et al. were able to identify a
emergent behaviors. Warmflash et al. [25] used micro- condition that resulted in spontaneous patterning with
contact printing to create the first human embryoid model SOX2+ and BRA+ cells occupying two opposite poles of
in which primed hPSCs are constrained to 2D circular the cyst (Figure 2b). Progressive development of this 3D
adhesive islands (Figure 2a). Uniform supplementation of human embryoid model further displayed the formation
BMP4 into culture medium resulted in ring-shaped gene of a primitive streak-like structure, cell dissemination,
expression patterns, with CDX2+ extraembryonic cells on and mesoderm/endoderm marker expression.
the colony edge followed by mesoendoderm and ending
with ectoderm in the colony center, mimicking certain Mechanistic studies using embryoid models would bene-
aspects of germ layer patterning during the human gas- fit from platforms that allow for precise control of system
trulation. Varying pattern size, an edge sensing mecha- parameters and for a clear understanding of how the cells
nism was discovered to be responsible for patterning in and the structures they form are oriented with respect to
the 2D human gastrulation model. The use of knockout their surroundings. Zheng et al. [43] recently developed
lines and computational modeling revealed that this edge a microfluidic platform that provides a highly controllable
sensing mechanism involved gradual cell polarization and 3D biomimetic environment for the PASE development
diffusion of the BMP4 inhibitor NOGGIN [28]. Further (Figure 2d). This microfluidic device contains three par-
work in the 2D human gastrulation model revealed that allel channels, with an induction channel and a cell
WNT and ACTIVIN-NODAL signaling act downstream loading channel separated by a central gel channel. Addi-
of BMP4 to control primitive streak formation, timing of tion of GeltrexTM to the gel channel leads to the forma-
epithelial-mesenchymal transition (EMT), and pattern- tion of pockets in which hPSCs will settle and aggregate.
ing of mesoderm and endoderm (Figure 2a) [26]. A 2D The size of the initial cell clusters can be controlled by
partial differential equation (PDE) model was generated modulating initial cell seeding density. Both the induc-
to show that their system dynamics could be described by tion and cell loading channels can be used for exogenous
waves resulting from bistability [27]. chemical stimulations. Notably, exposing clusters of
primed hPSCs to BMP4 through the induction channel
While 2D embryoid models are more amenable to live leads to the formation of a posteriorized PASE or poster-
imaging and quantitative measurements and perturba- iorized embryonic-like sac (P-ELS) (Figure 2d), which
tions, spatial constraints of hPSCs on 2D lead to a lack of begins with patterning of the lumenal pluripotent hPSC
3D cellular architecture and morphogenetic events that cysts where the cells facing BMP4 differentiate into
are needed to model the 3D human embryo develop- amnion-like cells (AMLCs) and the cells at the opposite
ment. To address this issue, Shao et al. [30] engineered a pole show markers suggesting a pre-primitive streak

Current Opinion in Biotechnology 2020, 66:52–58 www.sciencedirect.com


Bioengineered models of human embryo development Resto Irizarry, Nasr Esfahani and Fu 55

Figure 2

(a) (b)
Micropatterned culture 48 h SOX2/COX17/BRA Plate and top with medium BRA SOX2

hPSC

(c) (d)
OCT4TFAP2A
Day 1 Day 5 HOECHST / WGA

Current Opinion in Biotechnology

(a, left) hPSCs in micropatterned culture [25]. Scale bar, 500 mm. (a, right) hPSCs grown in 1000 mm micropatterns, stimulated with WNT3A,
WNT3A + SB or WNT3A + activin and stained after 48 hour for germ layer markers [27]. (b, left) Protocol used by Ref. [31] for creation of 3D
environment. (b, right) Cyst with asymmetric expression of SOX2 (yellow) and BRA (green) resulting from BMP4 treatment. (c, right) Schematic of
the biomimetic system used by [30] showing the formation of asymmetric cysts from hPSC. (c, left) Day five asymmetric cyst, stained for TFAP2A
(green), OCT4 (red), and WGA (purple). HOECHST (blue) counterstains nuclei. Scale bar, 50 mm. (d, top) Schematic of microfluidic device designed
by [43], showing three parallel channels partitioned by trapezoid-shaped supporting posts. (d, bottom) P-ELS at 36 h stained for TFAP2A (green)
and T (purple) (left) and NANOG (red) and SOX17 (purple) (right). TFAP2C + SOX17+ hPGCLCs are marked by color-coded arrowheads to indicate
spatial localizations (blue, amniotic ectoderm-like compartment; yellow, amniotic ectoderm– epiblast junction; white, PrePS-EPI-like compartment).

epiblast (PrePS-EPI) phenotype. NANOG + TFAP2C (XEN) cells [44], which represent the stem cell popula-
+ SOX17+ human primordial germ cell-like cells tions of the extraembryonic ectoderm and the primitive
(hPGCLCs) are evident and scattered throughout the endoderm in the mouse embryo, researchers have created
posteriorized PASE structure. Progressive development mouse embryoid models that incorporate these extraem-
of this structure showed gastrulation-like events, with bryonic cell lineages. Rivron et al. [8] co-cultured mouse
EMT, mesoderm induction, and cell dissemination from ESCs (mESCs) with mTSCs in non-adherent hydrogel
the PrePS-EPI compartment. The microfluidic PASE microwells to induce the cells to self-organize into struc-
model has shown superior controllability and reproduc- tures mimicking the mouse blastocyst (blastoid;
ibility for recapitulating consecutive hallmarks of the Figure 3a). Harrison et al. [6] co-cultured mESCs and
early human post-implantation development. mTSCs in a 3D Matrigel culture to generate a mouse
embryoid model with mESC-derived and mTSC-derived
Mouse embryoid models incorporating compartments (coined ETS embryoid model) to recapit-
extraembryonic stem cells ulate certain events of the post-implantation mouse
The 3D hPSC-based embryoid models lack extraembry- development, including mesoderm and PGC induction.
onic lineages associated with the trophoblast and the Adding mouse XEN cells to the ETS embryoid using an
hypoblast, which may be critical for the proper organiza- inverted pyramid microwell plate resulted in the ETX
tion and continuous development of these human embry- embryoid, showing adjoining mTSC-derived and mESC-
oid models. Given the availability of mouse trophoblast derived compartments surrounded by XEN cells [9].
stem cells (mTSCs) and extra-embryonic endoderm Importantly, the ETX embryoid showed an improved

www.sciencedirect.com Current Opinion in Biotechnology 2020, 66:52–58


56 Tissue, cell and pathway engineering

Figure 3

(a) DAPI PDGFRa TS:eGFP OCT4

EPS-blastoid after
72h in IVC
Extended Trophoblast
Pluripotent Stem Cells
Stem (EPS)
Cells

(b)

ESCs/microwell
TSCs/microwell

Current Opinion in Biotechnology

(a, left) Platform used by Ref. [45] for generating EPS-blastoids. (a, right) EPS-blastoid at 72 hour showing an implantation-like structure ost-
implantation-like structure with TS cell and EPS cell compartments. (b, left) Non-adherent hydrogel microwell arrays used by Ref. [8] for co-
culture of mPSCs and TS cells. (b, right) Immunofluorescent staining of blastoids showing expression of NANOG (green, left), OCT4 (green,
middle), and CDX2 (green, right). Scale bars, 50 mm.

efficiency and was able to recapitulate axial mesoderm Further, the aforementioned mouse embryoid models
and definitive endoderm specification in the mouse still suffer from low yields, which hinders their use for
embryo. in depth mechanistic studies. Regardless of current pit-
falls, the success seen with mouse embryoid systems has
Researchers have begun to use mouse EPSCs (mEPSCs) set a precedent for future development of human embry-
to generate mouse embryoid models. The first attempt by oid models. It is clear that the development of human
Li et al. [10] showed that mEPSCs cultured in inverted embryoid models that more closely resemble the in vivo
pyramid arrays could spontaneously give rise to a blastoid system will require the integration of extraembryonic
structure capable of implanting in utero. In a more recent lineages. However, there is still work to be done for
work, Sozen et al. [45] co-cultured mEPSCs with mTSCs the generation of human extraembryonic stem cell
in inverted pyramid arrays (Figure 3b) to obtain a blastoid lineages. It also remains to be determined whether
structure, in which a mTSC cyst enclosing the blastocoel- hEPSCs could be used to create human blastoid struc-
like cavity had an internal mEPSC compartment. The tures, like their mouse counterpart, the mEPSCs.
cavity was then flanked by mEPSC-derived primitive
endoderm-like cells. Importantly, this blastoid was able Outlook and challenges
to develop further and form an elongated, cylindrical Future work with human embryoid models will involve
structure with epiblast and extraembryonic ectoderm-like the application of a wider range of bioengineering tools
compartments covered by a visceral endoderm-like cell for the creation of environments that better capture
layer (Figure 3b). signaling mechanisms seen in the in vivo embryo niche.
A very promising approach is the use of microfluidic
The range of successive developmental events that these devices, which has been used already to control cell
mouse embryoid models are able to recapitulate high- cluster size and apply asymmetric chemical stimulations
lights the importance of stem cell potency and crosstalk [43]. Microfluidic devices could further be used to
between embryonic and extraembryonic stem cells to create morphogen gradients reminiscent of those seen
promote the proper organization and continuous devel- in the in vivo environment. Another promising bioengi-
opment of embryoid models. However, the development neering tool to increase controllability of embryoid mod-
of extraembryonic mouse lineages is still an ongoing els is optogenetics [47]. Researchers have already been
effort. The potential of mEPSCs to yield extraembryonic able to use optogenetics for control of cell differentiation
lineages has been challenged by a recent study [46]. [48]. Optogenetics could facilitate gain-of-function and

Current Opinion in Biotechnology 2020, 66:52–58 www.sciencedirect.com


Bioengineered models of human embryo development Resto Irizarry, Nasr Esfahani and Fu 57

loss-of-function assays to understand how exogenous and References and recommended reading
endogenous signaling interact to bring about system level Papers of particular interest, published within the period of review,
have been highlighted as:
behaviors. Furthermore, local control over differentiation
using optogenetics could facilitate creation of local sig-  of special interest
 of outstanding interest
naling centers to understand the role of paracrine signal-
ing in pattern formation and tissue growth. Precise local 1. Ma H, Zhai J, Wan H, Jiang X, Wang X, Wang L, Xiang Y, He X,
control over the environment and cell fates will not only Zhao ZA, Zhao B et al.: In vitro culture of cynomolgus monkey
embryos beyond early gastrulation. Science (80-) 2019, 366.
increase reproducibility, but also facilitate the develop-
ment of computational models for mechanistic studies. 2. Niu Y, Sun N, Li C, Lei Y, Huang Z, Wu J, Si C, Dai X, Liu C, Wei J
et al.: Dissecting primate early post-implantation development
Advances in control of stem cell fate will not only lead to using long-term in vitro embryo culture. Science (80-) 2019,
more robust and controllable in vitro embryo models but 366:eaaw5754.
could also further our ability to generate stem cell-derived 3. Sasaki K, Nakamura T, Okamoto I, Yabuta Y, Iwatani C,
functional cell lineages and tissues for cell replacement Tsuchiya H, Seita Y, Nakamura S, Shiraki N, Takakuwa T et al.: The
Germ cell fate of cynomolgus monkeys is specified in the
therapies and tissue engineering applications. nascent amnion. Dev Cell 2016, 39:169-185.
4. Nakamura T, Okamoto I, Sasaki K, Yabuta Y, Iwatani C,
The development of in vitro models of human develop- Tsuchiya H, Seita Y, Nakamura S, Yamamoto T, Saitou M: A
ment could lead to a better understanding of problems developmental coordinate of pluripotency among mice,
monkeys and humans. Nature 2016, 537:57-62.
like infertility and dysmorphogenesis. However, embry-
oid models by themselves can’t be used to draw definitive 5. Sawai T, Hatta T, Fujita M: Japan significantly relaxes its
human-animal chimeric embryo research regulations. Cell
conclusions about the mechanisms at work during devel- Stem Cell 2019, 24:513-514.
opment without including some sort of validation. Mor- 6. Harrison SE, Sozen B, Christodoulou N, Kyprianou C, Zernicka-
gani et al. [49] have started efforts to use the mouse Goetz M: Assembly of embryonic and extraembryonic stem
cells to mimic embryogenesis in vitro. Science (80-) 2017, 356.
embryo to validate patterning mechanisms seen in the
2D mouse gastrulation model [25–28]. An exciting area 7. Beccari L, Moris N, Girgin M, Turner DA, Baillie-Johnson P,
 Cossy A-C, Lutolf MP, Duboule D, Arias AM: Multi-axial self-
where significant progress has been achieved recently is organization properties of mouse embryonic stem cells into
prolonged in vitro culture of monkey embryos. Research- gastruloids. Nature 2018, 562:272-276
This work shows that mESCs cultured as aggregates can be induced to
ers have been able to culture monkey embryos in vitro for undergo gastrulation-like events and form gastruloids with a pattern of
longer than ever before [1,2]. The possibility of using neural, mesodermal, and endodermal derivatives. The self-organizing
monkey models to understand human development and capability of these gastruloids makes them a promising model to study
early axial patterning events of mammalian embryogenesis.
help validate findings from human embryoid models is
8. Rivron NC, Frias-Aldeguer J, Vrij EJ, Boisset JC, Korving J, Vivié J,
exciting, but nevertheless, there are still differences  Truckenmüller RK, Van Oudenaarden A, Van Blitterswijk CA,
between monkey and human development. As we keep Geijsen N: Blastocyst-like structures generated solely from
stem cells. Nature 2018, 557:106-111
seeking understanding of early human embryogenesis Using a co-culture system, this work shows that mESCs and mTSCs can
with the development of human embryoid models and self-organize into a mouse blastocyst-like structure, which can implant
alternative animal models, the need for validation will into the mouse uterus. Signals from mESCs regulate mTSC development.
continue to grow. Researchers have been discussing for 9. Sozen B, Amadei G, Cox A, Wang R, Na E, Czukiewska S, Chappell L,
Voet T, Michel G, Jing N et al.: Self-assembly of embryonic and two
years whether technical advances warrant a revision of the extra-embryonic stem cell types into gastrulating embryo-like
14-day rule [50,51], and some hope that the new devel- structures. Nat Cell Biol 2018, 20:979-989.
opments with human embryoid research and monkey 10. Li R, Zhong C, Yu Y, Liu H, Sakurai M, Yu L, Min Z, Shi L, Wei Y,
embryo culture will push this policy to extend [52].  Takahashi Y et al.: Generation of blastocyst-like structures
from mouse embryonic and adult cell cultures. Cell 2019,
Whether or not the 14-day rule changes, our understand- 179:687-702.e18
ing of human stem cell potency is still developing, and This work shows the use of mEPSCs to generate blastocyst-like structures
that contain all three blastocyst cell lineages and are able to implant in utero.
more complex human embryoid models are still to be
made, since human TSCs and hypoblast stem cells just 11. Rossant J, Tam PPL: New insights into early human
development: lessons for stem cell derivation and
become available. For now, there is still much to learn differentiation. Cell Stem Cell 2017, 20:18-28.
within the 14-day limit.
12. Guo G, Stirparo GG, Strawbridge S, Yang J, Clarke J, Li MA,
Myers S, Ozel BN, Nichols J, Smith A: Trophectoderm potency is
retained exclusively in human Naı̈ve cells. bioRxiv 2020 http://
Conflict of interest statement dx.doi.org/10.1101/2020.02.04.933812.
Nothing declared. 13. Blakeley P, Fogarty NME, Valle I, Wamaitha SE, Hu TX, Elder K,
Snell P, Christie L, Robson P, Niakan KK et al.: Erratum to
defining the three cell lineages of the human blastocyst by
single-cell RNA-seq (Development, (2015) 142, 3151-3165).
Acknowledgements Development 2015, 142:3613.
14. Molè MA, Weberling A, Zernicka-Goetz M: Comparative analysis
This work is supported by the University of Michigan Mechanical
of human and mouse development: from zygote to pre-
Engineering Faculty Support Fund, the Michigan-Cambridge Research gastrulation. Curr Top Dev Biol 2020, 136:113-138.
Initiative, and the University of Michigan Mcubed Fund. A.M.R.I. is
partially supported by the National Science Foundation Graduate Research 15. Deglincerti A, Etoc F, Guerra MC, Martyn I, Metzger J, Ruzo A,
Fellowship under grant no. DGE 1256260. Simunovic M, Yoney A, Brivanlou AH, Siggia E et al.: Self-

www.sciencedirect.com Current Opinion in Biotechnology 2020, 66:52–58


58 Tissue, cell and pathway engineering

organization of human embryonic stem cells on 34. Okae H, Toh H, Sato T, Hiura H, Takahashi S, Shirane K,
micropatterns. Nat Protoc 2016, 11:2223-2232. Kabayama Y, Suyama M, Sasaki H, Arima T: Derivation of human
trophoblast stem cells. Cell Stem Cell 2018, 22:50-63.e6.
16. Shahbazi MN, Jedrusik A, Vuoristo S, Recher G, Hupalowska A,
Bolton V, Fogarty NME, Campbell A, Devito LG, Ilic D et al.: Self- 35. Gamage TK, Chamley LW, James JL: Stem cell insights into
organization of the human embryo in the absence of maternal human trophoblast lineage differentiation. Hum Reprod Update
tissues. Nat Cell Biol 2016, 18:700-708. 2016, 23:77-103 http://dx.doi.org/10.1093/humupd/dmw026.
17. Xiang L, Yin Y, Zheng Y, Ma Y, Li Y, Zhao Z, Guo J, Ai Z, Niu Y, 36. Yang Y, Liu B, Xu J, Wang J, Wu J, Shi C, Xu Y, Dong J, Wang C,
Duan K et al.: A developmental landscape of 3D-cultured Lai W et al.: Derivation of pluripotent stem cells with in vivo
human pre-gastrulation embryos. Nature 2019 http://dx.doi.org/ embryonic and extraembryonic potency. Cell 2017, 169:243-
10.1038/s41586-019-1875-y. 257.e25.

18. Hyun I, Wilkerson A, Johnston J: Embryology policy: revisit the 37. Yang J, Ryan DJ, Lan G, Zou X, Liu P: In vitro establishment of
14-day rule. Nature 2016, 533:169-171. expanded-potential stem cells from mouse pre-implantation
embryos or embryonic stem cells. Nat Protoc 2019, 14:350-378.
19. Rivron N, Pera M: Debate ethics of embryo models from stem
cells. Nature 2018, 564:183-185. 38. Gao X, Nowak-Imialek M, Chen X, Chen D, Herrmann D, Ruan D,
Chen ACH, Eckersley-Maslin MA, Ahmad S, Lee YL et al.:
20. Rossant J, Tam PPL: Exploring early human embryo Establishment of porcine and human expanded potential stem
development. Science (80-) 2018, 360:1075-1076. cells. Nat Cell Biol 2019, 21:687-699.
21. Shahbazi MN, Siggia ED, Zernicka-Goetz M: Self-organization of 39. Guo G, Von Meyenn F, Santos F, Chen Y, Reik W, Bertone P,
stem cells into embryos: a window on early mammalian Smith A, Nichols J: Naive pluripotent stem cells derived directly
development. Science (80-) 2019, 364:948-951. from isolated cells of the human inner cell mass. Stem Cell Rep
2016, 6:437-446.
22. Shao Y, Fu J: Integrated micro/nanoengineered functional
biomaterials for cell mechanics and mechanobiology: a 40. Guo G, von Meyenn F, Rostovskaya M, Clarke J, Dietmann S,
materials perspective. Adv Mater 2014, 26:1494-1533. Baker D, Sahakyan A, Myers S, Bertone P, Reik W et al.: Erratum:
correction: Epigenetic resetting of human pluripotency
23. Sun Y, Chen CS, Fu J: Forcing stem cells to behave: a (doi:10.1242/dev.146811) (Development (Cambridge, England)
biophysical perspective of the cellular microenvironment. (2017) 144 15 (2748-2763) PII: dev166397). Development 2018, 145.
Annu Rev Biophys 2012, 41:519-542.
41. Pastor WA, Liu W, Chen D, Ho J, Kim R, Hunt TJ, Lukianchikov A,
24. Xue X, Sun Y, Resto-Irizarry AM, Yuan Y, Aw Yong KM, Zheng Y, Liu X, Polo JM, Jacobsen SE et al.: TFAP2C regulates
Weng S, Shao Y, Chai Y, Studer L et al.: Mechanics-guided transcription in human naive pluripotency by opening
embryonic patterning of neuroectoderm tissue from human enhancers. Nat Cell Biol 2018, 20:553-564.
pluripotent stem cells. Nat Mater 2018, 17:633-641.
42. Rostovskaya M, Stirparo GG, Smith A: Capacitation of human
25. Warmflash A, Sorre B, Etoc F, Siggia ED, Brivanlou AH: A method naı̈ve pluripotent stem cells for multi-lineage differentiation.
to recapitulate early embryonic spatial patterning in human Development 2019, 146:1-15.
embryonic stem cells. Nat Methods 2014, 11:847-854.
43. Zheng Y, Xue X, Shao Y, Wang S, Esfahani SN, Li Z, Muncie JM,
26. Martyn I, Kanno TY, Ruzo A, Siggia ED, Brivanlou AH: Self-  Lakins JN, Weaver VM, Gumucio DL et al.: Controlled modelling
organization of a human organizer by combined Wnt and of human epiblast and amnion development using stem cells.
Nodal signaling. Nature 2018, 558:132-135. Nature 2019, 573:421-425
This work shows the use of a highly controllable microfluidic platform for
27. Martyn I, Brivanlou AH, Siggia ED: A wave of WNT signaling the formation of either posteriorized or anteriorized amniotic sac models.
balanced by secreted inhibitors controls primitive streak These microfluidic amniotic sac models can capture key human embryo
formation in micropattern colonies of human embryonic stem developmental events including symmetry breaking of the epiblast cyst,
cells. Development 2019, 146. appearance of hPGCLCs, EMT, and mesoderm induction.
28. Etoc F, Metzger J, Ruzo A, Kirst C, Yoney A, Ozair MZ, 44. Kunath T, Arnaud D, Uy GD, Okamoto I, Chureau C, Yamanaka Y,
Brivanlou AH, Siggia ED: A balance between secreted inhibitors Heard E, Gardner RL, Avner P, Rossant J: Imprinted X-
and edge sensing controls gastruloid self-organization. Dev inactivation in extra-embryonic endoderm cell lines from
Cell 2016, 39:302-315. mouse blastocysts. Development 2005, 132:1649-1661.
29. Shao Y, Taniguchi K, Gurdziel K, Townshend RF, Xue X, Yong KMA, 45. Sozen B, Cox AL, De Jonghe J, Bao M, Hollfelder F, Glover DM,
Sang J, Spence JR, Gumucio DL, Fu J: Self-organized Zernicka-Goetz M: Self-organization of mouse stem cells into
amniogenesis by human pluripotent stem cells in a biomimetic an extended potential blastoid. Dev Cell 2019, 51:698-712.e8.
implantation-like niche. Nat Mater 2017, 16:419-427.
46. Posfai E, Schell JP, Janiszewski A, Rovic I, Murray A, Bradshaw B,
30. Shao Y, Taniguchi K, Townshend RF, Miki T, Gumucio DL, Fu J: A Pardon T, Bakkali MEl, Talon I, De Geest N et al.: Defining
pluripotent stem cell-based model for post-implantation totipotency using criteria of increasing stringency. bioRxiv
human amniotic sac development. Nat Commun 2017, 8:1-15. 2020 http://dx.doi.org/10.1101/2020.03.02.972893.

31. Simunovic M, Metzger JJ, Etoc F, Yoney A, Ruzo A, Martyn I, 47. Deisseroth K: Optogenetics. Nat Methods 2011, 8:26-29.
 Croft G, You DS, Brivanlou AH, Siggia ED: A 3D model of a human
epiblast reveals BMP4-driven symmetry breaking. Nat Cell Biol 48. Polstein LR, Juhas M, Hanna G, Bursac N, Gersbach CA: An
2019, 21:900-910 engineered optogenetic switch for spatiotemporal control of
This work presents a hPSC-based epiblast model to study BMP4-driven gene expression, cell differentiation, and tissue
symmetry breaking events. Using a 3D hydrogel environment and uniform morphogenesis. ACS Synth Biol 2017, 6:2003-2013.
BMP4 treatment, pluripotent hPSC cysts exhibit asymmetric expression 49. Morgani SM, Metzger JJ, Nichols J, Siggia ED, Hadjantonakis AK:
of SOX2 and BRA, followed by morphogenetic events mimicking primitive Micropattern differentiation of mouse pluripotent stem cells
streak formation and mesoderm dissemination. recapitulates embryo regionalized cell fate patterning. eLife
32. Anderson KGV, Hamilton WB, Roske FV, Azad A, Knudsen TE, 2018, 7:1-35.
Canham MA, Forrester LM, Brickman JM: Insulin fine-tunes self- 50. Hurlbut JB, Hyun I, Levine AD, Lovell-Badge R, Lunshof JE,
renewal pathways governing naive pluripotency and extra- Matthews KRW, Mills P, Murdoch A, Pera MF, Scott CT et al.:
embryonic endoderm. Nat Cell Biol 2017, 19:1164-1177. Erratum: revisiting the warnock rule. Nat Biotechnol 2017,
35:1211.
33. Linneberg-Agerholm M, Wong YF, Romero Herrera JA,
Monteiro RS, Anderson KGV, Brickman JM: Naı̈ve human 51. Pera MF: Human embryo research and the 14-day rule. Dev
pluripotent stem cells respond to Wnt, Nodal and LIF 2017, 144:1923-1925.
signalling to produce expandable naı̈ve extra-embryonic
endoderm. Development 2019, 146:dev180620. 52. Cyranoski BD: in the Lab for Longer. 2019, 575:17–18.

Current Opinion in Biotechnology 2020, 66:52–58 www.sciencedirect.com

You might also like