RESULTS
Time course of gastruloid development To study gastruloid development, we generated gastruloids as described by Beccari et al.11 (see STAR Methods) and performed scRNA-seq time course experiments with sampling from 0 to 120 h (Figure 1A). To identify gastruloid cell states, we clustered single-cell transcriptomes globally (Figure S1A) and from individual time points (Figure S1B) and used cluster alignment tool (CAT)22 to compare the clusters with annotated cell types from a published in vivo dataset21,23 (see STAR Methods). For most gastruloid clusters, the analysis resulted in single or strong matches to a particular embryonic cell type (Tables S1, S2, and S3). Based on the results from this analysis and marker gene expression, we annotated the cells generating a comprehensive atlas of gastruloid development (Figures 1B and S1C; Tables S4 and S5). Cells originated as naive pluripotent cells and were exiting this state during the first 24 h (Figure 1C). At 36 h, the cells resided in a broad epiblast state until 48 h when they received Wnt activation. During this activation, between 60 and 72 h, most of the cells started differentiating via a primitive streak-like state. At later time points, between 84 and 120 h, most of the cells fully committed to the three germ layers (Figure 1D). As expected, the gastruloids had an underrepresentation of anterior structures and rostral neuronal fates8,10 with a clear population of neuro-mesodermal progenitors (NMPs). We also saw the emergence of the definitive endoderm lineage, which further differentiated into the gut. Mesoderm was the most diverse lineage including cells with pre-somitic mesoderm (PSM), somite, and paraxial mesodermal identity. We report high similarities between gastruloid cell types and their respective in vivo counterparts (Figure 1E). Surprisingly, during Wnt activation, some cells (cluster 4 in Figure 1B; see temporal dynamics in Figures 1C and 1D) reverted to a population we term ectopic pluripotency (EP), as it displayed strong similarities with naive ES cells and expressed pluripotency markers such as Sox2, Esrrb, and Zfp42 (Figures 1E and S1C).24–26 To systematically compare gastruloids with in vivo embryonic development, we integrated and co-embedded gastruloid and embryonic cells23 (Figures 2A, 2B, S2A, and S2B). The gastruloid cell types from time points after Wnt activation (>72 h) mostly coclustered with their in vivo counterparts (Figure S2C). In contrast with these strong similarities, the cell types from earlier time points did not co-cluster as prominently, likely because the time points sampled in the reference dataset (E.6.5–E8.5) were not equivalent to early gastruloid time points. To further characterize our epiblast population, we compared it with an in vivo dataset that identified anterior, transition, and posterior states in early post-implantation epiblast and captured the acquisition of primitive streak propensity from E5.25 to E6.5 (Figures 2C and S2E).27 Post-implantation epiblast cells formed a continuum with amajor axis of cellular state variability (see t-distributed stochastic neighbor embedding, t-SNE 2) corresponding to the AP axis (respective markers: Fgf4, Trh, and Wnt3) (Figures 2C and S2D). Gene signatures for the three epiblast states (Table S6) allowed us to generate temporal expression maps (Figures 2D and S2G) and showed that epiblast cells in gastruloids change from an anterior-like epiblast state at 36 h (pre-Wnt pulse) to a mixed transitioning and posterior-like state onWnt activation (56–60 h). At the same time, we report the emergence of the EP at 60 h. This population was very similar to naive pluripotent cells and a population of cells found in a published dataset (primordial germ cell [PGC]-like in van den Brink et al.15) (Figure 2E). Dynamic analysis of the EP (Figure S2F; Table S6) (see STAR Methods) revealed early, intermediate, and late gene expression modules with epiblast markers, such as Dnmt3b and the transitioning epiblast marker Trh gradually decreasing over time (Figure 2F). Conversely, the expression of pluripotency genes like Zfp42 increases gradually over time. In later time points (>84 h), a subset of EP starts to upregulate the PGC marker Dppa3, (PGCs marker). Interestingly, the CAT analysis shows that early EP had only a few matches, whereas the later EP matched to numerous distinct and mature cell types (Figure S1B). This indicates that EP started homogeneously and then acquired more heterogeneity over time, likely due to increasing complexity in the tissue context. At 120 h, one match of the EP was PGC, suggesting some similarities to in vivo PGCs (Figure S2H). However, we did not find EP co-clustering with in vivo PGCs (Figure 2A) which suggested that a subset of late EP might have had the potential to acquire but did not fully commit to a PGC identity at the assessed time points. Overall, we report a good resemblance between gastruloids with their in vivo counterparts. Nonetheless, we observed two phenomena, which were aberrant from in vivo gastrulation, namely the emergence of a mixed transitioning and posterior epiblast state and the existence of an EP population. To further characterize the EP population during Wnt activation, we performed multiome sequencing (scRNA-seq + scATAC-seq) on gastruloids sampled at 48 and 52 h. Although the (legend on next page) 870 Cell Stem Cell 30, 867–884, June 1, 2023 cells were in an epiblast state, the promoter regions of naive pluripotency genes such as Klf2 and Klf4 as well as those of primitive streak genes like T were accessible. However, the expression of these genes was not detected (Figures 2G, 2H, S2I, and S2J). Gene activity scores for single cells based on promoter and gene body accessibilities (see STAR Methods) revealed gene accessibility for naive, EP, and primitive streak signatures (Figures 2I and S2K). Interestingly, although there was no significant difference in the gene activity for the naive signature, we detected an increase in gene activity at 52 h for the EP and primitive streak-like signatures. Unsupervised clustering using both multiome-modalities (see STAR Methods) revealed several clusters at 52 h (e.g., cluster 6), (Figures 2J and S2L). Interestingly, the percentage of cells at 52 h in cluster 6 was 7.5%, which was similar to the fraction of cells annotated as EP at 60 h (5.6%) in the scRNA-seq data. Cell fate bias analysis toward the EP and primitive streak-like populations showed that cluster 6 had a higher fate bias toward EP comparedwith the primitive streak-like population (Figure 2K). These data suggest that on Wnt activation, there is a differential response to Wnt in epiblast cells, which drives a binary fate response: EP and primitive streak-like. Spatial cell-type organization during gastruloid development To study their spatial organization, we established an automated handling procedure and a pipeline for high-throughput culture, compound and genomic perturbations, immunofluorescence staining, sample clearing, and high-content imaging of tens of thousands of gastruloids (Figure 3A). This approach and some aspects described here28 allowed us to increase the elongation efficiency from the previously reported 70%8 to 100% (Figure S3A). Gastruloid images were then automatically segmented and processed with a custom workflow extracting features at multiple levels as illustrated in Figure 3B (see STAR Methods). We established that radial symmetry breaking, axial elongation, aswell as themajority of cell types are formedwithin the first 96 h (Figure 1D). We therefore performed time course experiments starting from 24 to 96 h with fixation intervals of 12 h and stained for Bra, monitoring tail bud and mesodermal induction,8,10 and for Sox2, expressed in naive pluripotent cells, epiblast, NMPs, and neural progenitors in vivo.29–31 The choice Figure 2. In vivo comparison and characterization of epiblast and plur (A) UMAP of co-embedded gastruloid and embryonic cells highlighting embryon (B) UMAP of co-embedded gastruloid and embryonic cells highlighting gastruloid primitive streak/definitive endoderm. (C) t-SNE map of single-cell transcriptomes from Cheng et al.27 highlighting thre (D) Temporal gene expression maps of anterior, transition, and posterior gen expression. (E) Scatter plot and inferred linear regression comparing ectopic and naive plurip expression of individual genes in ectopic and naive pluripotency populations or (F) Temporal gene expression maps of Dppa3, Zfp42, Dnmt3b, of EP cells. y axis: (G) Coverage plot of chromatin accessibility for Klf2 and 1,000 bp upstream re expression of Klf2 in the same cells. Top: averaged frequency of DNA fragments region for single cells. Lower: arrows indicate transcriptional direction. Bottom: p (H) Coverage plot of chromatin accessibility for T and 1,000 bp upstream region (I) Boxplots: aggregated gene activity scores for the EP and primitive streak-like (J) UMAP of the scATAC-seq data-modality from the multiome highlighting clust (K) Boxplots of fate bias analysis at 52 h for multiome clusters toward EP and pr of Sox2 allowed us to follow the cell type annotations 1–4 and 16 and is continuously expressed throughout gastruloid development. Making use of the extracted features, we further created ‘‘meta-gastruloids’’ showing Sox2 and Bra expression patterns in an average gastruloid representation (Figure 3C). Gastruloids from ESCs grown in S/L exhibit Bra expression before 48 h,10,21 although starting from cells in the S/L/2i medium, Bra protein expression started only at 60 h and displayed a salt-and-pepper yet peripheral pattern. Interestingly, the initially uniform Sox2 staining developed into a heterogeneous pattern at 36 h, forming a gradient with high levels in the gastruloid core. The Chir pulse converted this gradient into a binary pattern with only former Sox2-low regions expressing Bra and a persistent Sox2-positive core population of Bra-negative cells in the center. Spatial variability in Sox2 expression therefore preceded the induction of Bra expression. Remarkably, the Sox2positive and Bra-positive populations exhibited an increasing bias toward opposing poles starting at 72 h,marking the initiation of radial symmetry breaking and axial organization, culminating in the translocation of the Sox2-positive cell population from the core to the anterior tip and the establishment of a primary body axis. We then used a panel of antibodies to profile the expression of 21 cell types, adhesion, and signaling activity markers (see STAR Methods). In each case, we co-stained one marker from the panel with DAPI (40,6-diamidino-2-phenylindole) and Sox2 as a fiducial marker. Gastruloids do not show a perfectly synchronous developmental progression (Figures 3D and S3B; see STAR Methods). To gain developmental resolution, we inferred an image-based gastruloid trajectory.32,33 This pseudotime trajectory enabled us to correlate expression patterns along gastruloid formation (Figures 3E, S3C, and S3D) at the whole gastruloid (Figure S3E) as well as at the segment and inside/outside level (Figures 3F, S3F, and S3G). Patterning maps aligned to the trajectory showed a robust formation of the AP axis, as evidenced by the progressive anterior and posterior localization of polarized markers (Figures 3F and 3G). Of note, the anterior localization of Sox2-positive cells was not marking a rostral neural identity, as we saw segment-level colocalization with pluripotency markers (e.g., Oct4). As shown by scRNA-seq, the only population that expressed a pluripotency signature at 96 h was the EP population. Thus, we used anterior Sox2-positive cells to mark the EP ipotency states ic cell types from Pijuan-Sala et al.23 cell types. PS, primitive streak; dif., differentiation; Ant. PS/Def. endo., anterior e embryonic epiblast states and expression maps of Fgf4, Trh, and Wnt3. e signatures for embryonic and gastruloid epiblast cells. y axis: normalized otency populations (left) and PGC-like15 populations (right). Scatter plot: mean PGC-like populations. normalized expression. Self-organizing map (SOM) modules (see Figure S2F). gion from transcription start site (TSS) at 48 and 52 h. Right: multiome RNA within the genomic region. Middle: frequency of fragments within the genomic eak coordinates within genome region. from the TSS containing the promoter. state. ers. Time point: 52 h. imitive steak-like populations. Cell Stem Cell 30, 867–884, June 1, 2023 871 (A) Scheme illustrating automatized handling workflow. Right: representative image of gastruloids fixed at 120 h. Maximum intensity projection (MIP) of z stack: DAPI and antibody stainings for Sox2 and Bra. Scale bars, 150 mm. (legend continued on next page) 872 Cell Stem Cell 30, 867–884, June 1, 2023 state in post-Wnt pulse gastruloids. From here onward, we refer to the EP population also as the ‘‘gastruloid core.’’ Other markers such as the mesodermal and epithelial-tomesenchymal transition (EMT) regulator Eomes34 and Hes1 formed an anteriorly biased band pattern but did not fully reach the anterior pole. Although Hes1 suggested Notch activity near the Sox2 core, we saw that Wnt and Nodal activity (b-catenin and pSmad2 antibodies, respectively) was posteriorly polarized. We observed N-cadherin expression at the posterior, consistent with mesoderm specification, whereas E-cadherin was globally expressed. This segment-level co-expression suggested incomplete EMT at 96 h. We also observed the expression of transcription factors suggesting the emergence of the endodermal (Foxa2) and neural lineage (Sox1). Remarkably, the ECMcomponent Fibronectin 1 (Fn1) showed similar behaviors as the Sox2 core (Figures 3H and 3I). Fn1 already defined a domain at the core 24 h post-seeding and continuously overlapped with Sox2 expression throughout morphogenesis. Molecular regulators and regime-dependent phenotypic differences Our analysis revealed a three-step process of symmetry breaking: (1) establishment of cellular variability in Sox2 levels as a possible consequence of differentiation progression, (2) a binary response to Wnt activation, and the formation of two cell populations whose organization ultimately culminates in (3) radial symmetry breaking and elongation. To systematically identify molecular regulators of each step, we designed an image-based compound screen (Figure 4A). The screening library consisted of 84 compounds (Cpd) (Figure S4A; Table S7) selected from a pre-screening of 200 small molecules (Figure S4B). The library was annotated with 68 unique primary targets. Compound treatment was performed in three separate regimes: from 32 to 72 h (‘‘variable differentiation’’), 48 to 72 h (‘‘Bra induction’’), or 72 to 96 h (‘‘axial elongation’’). In all treatment regimes, gastruloids were fixed at 120 h and stained for markers of mesoderm (Bra),35 neuroectoderm (Sox1),36 epiblast or endoderm (E-cadherin),12 and DAPI. 40 gastruloids per condition and regime were then imaged, analyzed, and quality controlled (Figure S4C) and used to generate a multivariate feature set on the whole gastruloid and a segment level of 9,000 gastruloids. To generate a phenotypic landscape of gastruloid development, we grouped gastruloids by phenotypic similarity by sepa- (B) Scheme illustrating extracted features and super pixel analysis. (C) Representative images at indicated time points (middle z plane of a z stack sho superpixel intensities of Sox2. Kernel density plots: distribution of intensities alon center (bottom). Co-expression hexbin plots: expression of Sox2 and Bra. Kern numbers (n) are indicated. (D) UMAP plots of n = 2,862 gastruloids color-coded by time points. Bottom left (E) Inferred pseudotime (top left) and scheme of pseudotime ordering, trajectory (F) Heatmaps depicting distribution of stainings from the anterior (left) to the poster gastruloids. Anterior bias (light green), posterior bias (light red), and unbiased ma (G) Representative images at 96 h. (MIPs of z stack showing DAPI and antibody Sox2). Scale bars, 100 mm. (H) Top: representative images at indicated pseudotime and sampling time poin 100 mm. Bottom: heatmaps depicting distribution of Fn1 and Sox2 staining from (I) Top: line plots of mean staining intensity for Sox2 and Fn1 (n = 82). Blue bars: in from anterior (top) to posterior (bottom) along pseudotime. Bottom: heatmap de rately clustering37,38 whole gastruloid and segment features (Figures S4D and S4E). Each gastruloid was thus unambiguously assigned to a whole gastruloid and to an AP-pattern class. At the whole gastruloid level, we observed nine phenotypes ranging over four major groups (Figure 4B): wild-type phenotype (classes 1–2) to which the majority (97.6%) of control gastruloids was assigned; Sox1-enriched (classes 3–5) with an increased or exclusive expression of the neuroectoderm marker, indicating failure to produce primitive streak-derived cell lineages; E-cadherin-enriched, exhibiting increased expression of the epithelial marker either together with Sox1 (class 6) or Bra (class 7) expression; and Bra-enriched (classes 8 and 9) with an increased or exclusive expression of Bra (Figure 4B). At the segment level, we detected 10 pattern classes, among them, classes observed in control conditions (classes I–IV, ‘‘wildtype’’ classes) and those that occurred mostly under perturbation (classes VI–X, ‘‘perturbed’’ classes) (Figure S4E). The latter included gastruloids with an increased polarized expression of Bra (class VII) or Sox1 (classes VIII and IX), localization of Bra to the center (class VI), or expression of the two markers on opposing poles (class X). We then highlighted gastruloids from each treatment regime separately (Figure 4C) and inferred a network of functional annotations and color-coded the nodes by the most frequently assigned phenotype for each treatment regime (see STAR Methods). Each phenotypic class was detected in all three temporal regimes. However, their ratios differed significantly, especially between the earliest regime and the latter two. Although perturbation during the establishment of ‘‘variable differentiation’’ (32–72 h) favored classes 8 (light pink) and 6 (purple), the abundance of Sox1-enriched phenotypes (classes 3–5, shades of green) increased when perturbing in the latter two regimes (Figure S4F). Regulatory modules of gastruloid development To systematically uncover regulatory modules, we combined the abundance of the whole gastruloid and segment classes in the three regimes into a phenotypic signature (57-feature vector for each compound, Figures S5A–S5C). This revealed 4 regulatory modules that were divided into categories grouping compounds with similar phenotypic effects over time (Figure 5A). We selected hits by significance and robustness (see STAR Methods and Figures S5D and S5E) for a final hitlist of 38 compounds (Table S7). These were predominantly assigned to modules A–C that included gastruloids with delayed development wing DAPI, Bra, and Sox2. Scale bars, 100 mm. Hexbin plots: mean normalized g the x and y axes; dist. to center, normalized distance of superpixel to object el density plots: distribution of intensities along x and y axes (right). Sample : scheme illustrates increasing heterogeneity in later time points. inference, and molecular progression. ior (right) pole along pseudotime (progressing from bottom to top). n, number of rkers (light yellow). stainings for Oct4, Cdx2, Eomes, Hes1, Foxa2, Sox1, E-Cad and N-Cad, and ts. Middle z plane: DAPI and antibody stainings for Fn1 and Sox2. Scale bars, anterior (left) to posterior (right). dividual gastruloids shown in (H). Middle: heatmaps depicting Fn1 distribution picting Sox2 and Fn1 in/out ratio measured on the middle z plane. Cell Stem Cell 30, 867–884, June 1, 2023 873 (A) Scheme of experimental outline, image processing, and analysis of the screen. (B) Left: representative maximum intensity projection images of whole gastruloid phenotypes. Stainings: DAPI, Sox1, Brachyury, and E-cadherin, Scale bars, 100 mm. Right: UMAP plot color-coded by whole gastruloid class. Data points: individual gastruloids, n = 8,740. Heatmap: mean values of indicated features for each class, Z score normalized. (C) UMAP plots and pie charts color-coded by whole gastruloid classes from indicated treatment regimens (left to right: 32–72 h, 48–72 h, and 72–96 h). (module A), increased Bra expression (module B), or increased Sox1 expression (module C) (see Figures S5E and S5F). Module A contained compounds that produced gastruloids with minor phenotypes indicated by high correlations to DMSO controls. Indeed, the perturbation of targets in module A1 such as Akt1, Igf1r, or Pik3ca resulted in gastruloids with a slight increase in Bra expression (class 8) that exhibited a delay but not a full developmental failure, as gastruloids at 120 h resembledwild-type gastruloids at an earlier time point (96 h). Inhibition of Ccr5, Mapk14, or Prkcb during or after the Chir pulse (module A2) produced gastruloids of class 3 (light green) with both Sox1 and Bra-positive domains. Module B contained compounds that had an increased Bra expression with either elongated (module B1, class 7, and dusky pink) or almost spherical morphology (module B2, class 9, and red). Unexpectedly, inhibition of Ctnnb1 (b-catenin) and Porcn (porcupine O-acyltransferase), members of the Wnt signaling pathway, produced spherical, Bra-increased gastruloids (class 9) when treated before or during the Chir pulse and elongated, Sox1/Bra double-positive gastruloids (class 3) when treated later. To understand the counterintuitive emergence of Bra-positive phenotypes, we performed follow-up experiments for Wnt pathway-related hits including additional compounds (IWP2 and XAV939) (Figures 5B, 5C, S5G, and S5H). Gastruloids 874 Cell Stem Cell 30, 867–884, June 1, 2023 were treated from 48 to 72 h or from 72 to 96 h and fixed every 24 h after treatment up to 144 h and stained for Sox1, Sox2, and Bra. Wnt-agonistic treatments (Gsk3b inhibitors) caused an increase in Bra at early time points but resulted in mild phenotypes with only limited effect by 120 and 144 h. This suggests that Wnt overexposure does not cause an increase in Bra expression at the expense of Sox1 at later time points but rather that cell ratios found in late gastruloids are not strongly dependent on the dose of Wnt activation during the Chir pulse. Inhibitors of b-catenin (Cpd54), tankyrase (XAV939), and porcupine (IWP2, Cpd58, and Cpd63), on the other hand, had very drastic effects. All compounds except Cpd54 caused AP axis failure. When treated between 48 and 72 h, gastruloids had a strongly reduced Bra expression (72 h), which resulted in increased levels of Bra, Sox1, and Sox2 at 120 and 144 h. The delayed expression of Bra after the Chir pulse suggested that endogenousWnt activity is sufficient to cause Bra induction. Thus, endogenously secreted Wnt ligands and their gradients play an important role in primary axial elongation.10 Here, we detected an expanded Sox1- and Sox2-positive territory during Ctnnb1 inhibition and formation of rosette-like structures in XAV939 treatment (Figures S5G–S5I). Module C mainly contained Sox1-enriched gastruloids. Lack of Bra expression implied the absence of inductive signals and (legend on next page) Cell Stem Cell 30, 867–884, June 1, 2023 875 failure of mesodermal differentiation, which consequently skewed development toward neural differentiation.39 This suggests that calcium, hedgehog, mitogen-activated protein kinase (MAPK), and TGF-b signaling control induction and/or maturation of mesodermal cell types. Although perturbation of Tgfbr1, Src, Gli1, and Met (module C1) produced Sox1-enriched gastruloids (classes 3–5) irrespective of the treatment regime, the inhibition of Map2k1, Fgfr1, Braf, or Acvr resulted in the phenotypic class 6 when perturbed before the Chir pulse. Intriguingly, class 6 gastruloids exhibited themost severe defect with a nearly complete absence of the mesoderm. The phenotypic signature of MAPK/fibroblast growth factor (FGF) signaling inhibition underlined its importance throughout gastruloid development: although its inhibition at later time points impeded mesoderm and favors neural induction, at earlier time points, it reduced differentiation in general. This suggests that a perturbation during the variable differentiation states (32–72 h) is incompatible with subsequent development. To better understand the role of MAPK/FGF signaling in pluripotency exit, we analyzed the core in screening hits related to class 6 and FGF-mediated MAPK signaling. After treating gastruloids between 32 and 72 h and fixing at 48, 72, and 120 h, we stained for Bra, Sox2, Fn1, and the pluripotency marker Dppa4 (Figures 5D and 5E). Inhibition of FGF receptors as well as their downstream target MAP2K1 resulted in an expansion of the Sox2 and Dppa4-positive cells as well as an increase in Fn1 at the expense of Bra expression and axial elongation. Inhibition of MAPK/FGF signaling in combination with Wnt activation maintained naive pluripotency40 and under the inhibition of FGF-mediated MAPK signaling, pluripotency is maintained for longer preventing gastruloids to enter a state that is competent for Bra induction on Wnt activation. Cell-state heterogeneity in early gastruloids and characterization of the gastruloid core We then addressed five aspects of the EP core population: the pluripotency state, the timing of differentiation competency, the role of Fn1, gastruloid size dependence, and the reproducibility between multiple cell lines. To verify the presence of a pluripotent subpopulation, we performed clonogenicity assays (Figures 6A and 6B), which select for naive pluripotent stem cells in the N2B27/2i medium.41,42 When dissociated into single cells, gastruloids showed a colony forming unit of 3.5% at 48 h with an increase after Wnt pulse. Cloning efficiency of ESCs is usually below 50%.41 Given that EP cells made up for 5.6% of all sequenced cells at 72 h, the efficiency exceeded this clonogenicity score. The colony formation was Chir-dependent since the lack of Wnt activation caused a decrease <1.5%. The EPs maintained naive pluripotency, since PGCs and embryonic germ cells cannot be maintained in N2B27/2i in the absence of leukemia inhibitory factor (LIF).43 The same assay was performed on a miR-290-mCherry/miR-302-eGFP reporter line.10 We sorted cells and assessed different pluripotency states with miR-290-mCherry expressed in E3.5–E6.5 embryos and miR-302-eGFP expressed from E5.5 to E8.0.44 In early gastruloids (48–72 h), we found cells corresponding to pre-implantation (mCherry+/GFP ) and early post-implantation epiblast (mCherry+/GFP+). Displaying a continuum, cells shifted toward a double-positive post-implantation epiblast state. By 72 h, 9.3% of the sorted cells remained mCherry single positive (Figure S6A). As expected, colony-forming efficiency was dependent on the pluripotency state, and we saw an enrichment for that state after the Wnt pulse. To assess the effects of different levels of differentiation competency, we performed Wnt pulses at different time points (Figures 6C and 6Di–6Diii). An early pulse showed an increase in Sox2 expression and a delayed Bra expression onset. At 144 h post-seeding, the prematurely pulsed gastruloids also had multiple Bra+ foci instead of a single tail bud. We also performed bulk RNA-sequencing of prematurely pulsed gastruloids (24 h) and assessedgenesignaturesobtained fromscRNA-seq (Figures6Eand S6B). We saw that gastruloids pulsed at 24 h showed an upregulation of genes associatedwith EP, naive, and exiting pluripotency signatures. Interestingly, gastruloids that received an early pulse showed a downregulation of the epiblast signature at 48 h suggesting that cells in early gastruloids did not maintain an epiblast identity longer but responded with an EP signature. Late primitive streakandanteriorprimitivestreak/definitiveendodermsignatures showed lower expression levels at 72 h when pulsed prematurely. Genes associatedwith PSMorNMPswere only expressed in 96 h gastruloids when pulsed at 48 h. To understand the importance of Sox2 levels when gastruloids receiveWnt activation, we successfully performed siRNA knockdown (KD) experiments (Figure S6C), adding siRNA during aggregation. Sox2 KD had a minor effect on pluripotency markers Oct4 and Dppa4, however, caused an increased expression of Nanog at 48 h, which can act as an early primitive streak marker.45 Sox2 KD also resulted in an increase of Bra expression by 72 h and failed axial elongation by 120 h suggesting that increased levels of differentiation in early gastruloids before Wnt activation lead to failure of efficient axial elongation, which was in line with previous studies.21,28 Another plausible explanation for the failure of axial elongation is the depletion of Sox2-dependent lineages such as NMP cells, which are important for in vivo axial elongation.46 Fn1 colocalized with the Sox2-positive core and was expressed by naive pluripotent and core cells (Figures S2D and (legend on next page) Cell Stem Cell 30, 867–884, June 1, 2023 877 S6E).47 To understand if Fn1 plays a functional role in core maintenance, we treated gastruloids with RGD-peptide (minimal integrin binding motif48 preventing cellular attachment to Fn1 and thus inhibiting downstream signaling49–51). RGD treatment increased Sox2 expression and decreased Bra expression (Figure S6F). Inhibition of focal adhesion kinase (FAK) caused an analogous but stronger effect (Figure S6G) with complete failure of axial elongation. This suggests that deposited Fn1 may keep the level and extent of pluripotency within certain boundaries. A crucial aspect of gastruloid development8 that could have an effect on the core is the initial seeding cell number. Accordingly, seeding gastruloids with different cell numbers (150, 300, and 500) resulted in a size-dependent relative expansion of the core as well as the Fn1 expression (Figures 6F and 6H). Gastruloids generated from 150 cells showed impaired Fn1 and Sox2 core formation, whereas gastruloids generated from 500 cells showed an expanded core. In gastruloids generated from 500 cells, the peripheral cells also showed reduced Bra expression at 72 h. Fn1 secretion is stimulated under hypoxia, which could also explain the cell number-dependent increase of Fn1 expression.52 Finally, we explored if the core is a unique feature of gastruloids generated from the Sox1-GFP::Brachyury-mCherry (SBR) cells and their parental line CGR8.53 We tested additional cell lines (Figures 6I–6K) using a BramCh/Sox2Ve reporter line,14 E14, and a Mesp1 reporter line.54 Although SBR and BramCh/ Sox2Ve showed a tight clustering of Sox2 cells, E14 and Mesp1-GFP gastruloids did not have the same level of organization. We also stained for Fn1 (Figure S6H), which was only expressed in SBR and BramCh/Sox2Ve gastruloids. In all cell lines, we observed variability of Sox2 expression and a differential Wnt response, with some cells maintaining high levels of Sox2 after the Wnt pulse. The separation between Sox2- and Bra-positive cells did not happen as efficiently in E14 andMesp1-GFP gastruloids. At 96 h, E14 andMesp1-GFP gastruloids had a larger NMP population indicated by the co-expression of Sox2 and Bra.55 At 120 h only SBR, CGR8 (not shown) and BramCh/Sox2Ve gastruloids have anteriorly localized Sox2 pluripotent cells. It is possible that there are convergent mechanisms of gastruloid for- mation56 that might also be dependent on cell line-specific differences.57 The window of competency to differentiation in gastruloids is dependent onmultiple factors such as pluripotency state, aggregate size, and time of Wnt activation, as well as cell line-specific aspects. Careful assessment of these aspects is necessary to generate developmentally meaningful cell types in gastruloids. Dual WNT modulation causes anterior structures We then hypothesized that the core population and surrounding cells might be an opportunity to reach a better representation of anterior embryonic identities in gastruloids. To successfully form anterior parts of the in vivo gastrula, respective cells are situated in a region that is shielded fromWnt andNodal activation by local inhibitors (Dkk1, Lefty1, and Cer1) secreted by the anterior visceral endoderm (AVE).58 Screening hits that could restrict caudalizing gradients and phenocopy the effect of the AVE are thus compounds targeting the Wnt pathway and the TGF-b superfamily (especially Nodal signaling). For the TGF-b superfamily inhibition, we performed treatments (Cpd56 [ALK2i], Cpd66 [TGFRi], Cpd74 [TGFRi], and SB43 [ALK4,5,7i]), with different concentrations, in 24 h treatment windows starting at 48, 72, or 96 h, fixed at 144 h and stained for Sox1, Sox2, and Otx2, which marks rostral neurectoderm.59–61 Otx2 is also associated with the foregut and anterior foregut and thus also marks anterior endodermal derivatives.62,63 TGFb superfamily inhibitors caused a strong increase in Sox1 and Sox2 levels with a drastic expansion of neural lineages, although having only a minor effect on elongation (Figures 7A and S7A– S7D). All conditions, however, did not show an increased Otx2 expression suggesting the absence of anterior neural and endodermal structures. For the Wnt pathway, we utilized the porcupine inhibitors (IWP2, Cpd58, and Cpd63) as they affect Wnt secretion and the formation of endogenous AP-gradients. We also used a Ctnnb1 inhibitor (Cpd54) as it previously caused an increase in Sox1 and Sox2 while maintaining the AP axis. Each Wnt inhibitor showed a dose-dependent upregulation of the three markers (Sox1, Sox2, and Otx2), with a striking anterior localization of Cell Stem Cell 30, 867–884, June 1, 2023 879 Otx2 (Figures 7B and S7E–S7H). In themost promising condition (2.5 mM Cpd63 administered at 48 h), gastruloids displayed neuronal maturation, as indicated by laterally and anterior Tuj1 expression and long cell protrusions (Figures 7C and 7D). We also observed Pax6- and Nestin-positive cells localized between the AP poles with a potential spinal cord identity (Figure 7E).64,65 In some cases, anterior Tuj1-positive cells were in proximity to a bi-layered Otx2 and Sox17 double-positive ring (Figure 7F). The morphology of this structure is reminiscent of endoderm compartments in 168-h gastruloids previously annotated as anterior foregut.12 Our results are also in line with previous conclusions that gastruloids develop endodermal progenitors that do not transition through EMT or a Bra-positive state.
INCLUSION AND DIVERSITY
One or more of the authors of this paper self-identifies as an underrepresented ethnic minority in their field of research or within their geographical location. One or more of the authors of this paper self-identifies as a gender minority in their field of research. One or more of the authors of this paper self-identifies as a member of the LGBTQIA+ community. We support inclusive, diverse, and equitable conduct of research. Received: February 22, 2023 Revised: April 16, 2023 Accepted: April 25, 2023 Published: May 19, 2023 REFERENCES 1. Rivron, N.C., Frias-Aldeguer, J., Vrij, E.J., Boisset, J.-C., Korving, J., Vivié, J., Truckenm€uller, R.K., van Oudenaarden, A., van Blitterswijk, C.A., and Geijsen, N. (2018). Blastocyst-like structures generated solely from stem cells. Nature 557, 106–111. https://doi.org/10.1038/s41586018-0051-0. 2. Yu, L., Wei, Y., Duan, J., Schmitz, D.A., Sakurai, M., Wang, L., Wang, K., Zhao, S., Hon, G.C., and Wu, J. (2021). Blastocyst-like structures generated from human pluripotent stem cells. Nature 591, 620–626. https:// doi.org/10.1038/s41586-021-03356-y. 3. 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., and Fu, J. (2019). Controlled modelling of human epiblast and amnion development using stem cells. Nature 573, 421–425. https://doi.org/10.1038/s41586-0191535-2. 4. Warmflash, A., Sorre, B., Etoc, F., Siggia, E.D., and Brivanlou, A.H. (2014). Amethod to recapitulate early embryonic spatial patterning in human embryonic stem cells. Nat. Methods 11, 847–854. https://doi.org/ 10.1038/nmeth.3016. 5. Sozen, B., Amadei, G., Cox, A., Wang, R., Na, E., Czukiewska, S., Chappell, L., Voet, T., Michel, G., Jing, N., et al. (2018). Self-assembly of embryonic and two extra- embryonic stem cell types into gastrulating embryo-like structures. Nat. Cell Biol. 20, 979–989. https://doi.org/10. 1038/s41556-018-0147-7. 6. Sozen, B., Cox, A.L., De Jonghe, J., Bao, M., Hollfelder, F., Glover, D.M., and Zernicka-Goetz, M. (2019). Self-organization of mouse stem cells into an extended potential blastoid. Dev. Cell 51, 698–712.e8. https:// doi.org/10.1016/j.devcel.2019.11.014. 7. 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 amniotic sac development. Nat. Commun. 8, 208. https://doi.org/ 10.1038/s41467-017-00236-w. 8. van den Brink, S.C., Baillie-Johnson, P., Balayo, T., Hadjantonakis, A.K., Nowotschin, S., Turner, D.A., and Martinez Arias, A. (2014). Symmetry breaking, germ layer specification and axial organisation in aggregates of mouse embryonic stem cells. Development 141, 4231–4242. https:// doi.org/10.1242/dev.113001. 9. Moris, N., Anlas, K., van den Brink, S.C., Alemany, A., der, J. S.x.F., Ghimire, S., Balayo, T., van Oudenaarden, A., and Arias, A.M. (2020). An in vitro model of early anteroposterior organization during human development. Nature 582, 410–415. https://doi.org/10.1038/s41586020-2383-9. 10. Turner, D.A., Girgin, M., Alonso-Crisostomo, L., Trivedi, V., Baillie- Johnson, P., Glodowski, C.R., Hayward, P.C., Collignon, J., Gustavsen, C., Serup, P., et al. (2017). Anteroposterior polarity and elongation in the absence of extra-embryonic tissues and of spatially localised signalling in gastruloids: mammalian embryonic organoids. Development 144, 3894–3906. https://doi.org/10.1242/dev.150391. 11. Beccari, L., Moris, N., Girgin, M., Turner, D.A., Baillie-Johnson, P., Cossy, A.C., Lutolf, M.P., Duboule, D., and Arias, A.M. (2018). Multi-axial self-organization properties of mouse embryonic stem cells into gastruloids. Nature 562, 272–276. https://doi.org/10.1038/s41586-018-0578-0. 12. Vianello, S., Rosa, V.S., and Lutolf, M.P. (2021). In vitro endoderm emer- gence and self-organisation in the absence of extraembryonic tissues and embryonic architecture. Preprint at bioRxiv. https://doi.org/10. 1101/2020.06.07.138883. 13. Rossi, G., Broguiere, N., Miyamoto, M., Boni, A., Guiet, R., Girgin, M., Kelly, R.G., Kwon, C., and Lutolf, M.P. (2020). Capturing cardiogenesis in gastruloids. Stem Cells 28, 230–240.e6. https://doi.org/10.1016/j. stem.2020.10.013. 14. Veenvliet, J.V., Bolondi, A., Kretzmer, H., Haut, L., Scholze-Wittler, M., Schifferl, D., Koch, F., Guignard, L., Kumar, A.S., Pustet, M., et al. (2020). Mouse embryonic stem cells self-organize into trunk-like Cell Stem Cell 30, 867–884, June 1, 2023 881 structures with neural tube and somites. Science 370, eaba4937– eaba4939. https://doi.org/10.1126/science.aba4937. 15. van den Brink, S.C., Alemany, A., van Batenburg, V., Moris, N., Blotenburg, M., Vivié, J.V., Baillie-Johnson, P., Nichols, J., Sonnen, K.F., Arias, A.M., and van Oudenaarden, A. (2020). Single-cell and spatial transcriptomics reveal somitogenesis in gastruloids. Nature 582, 405–409. https://doi.org/10.1038/s41586-020-2024-3. 16. Girgin, M.U., Broguiere, N., Hoehnel, S., Brandenberg, N., Mercier, B., Arias, A.M., and Lutolf, M.P. (2021). Bioengineered embryoids mimic post-implantation development in vitro. Nat. Commun. 12, 5140. https://doi.org/10.1038/s41467-021-25237-8. 17. Girgin, M.U., Broguiere, N., Mattolini, L., and Lutolf, M.P. (2021). Gastruloids generated without exogenous Wnt activation develop anterior neural tissues. Stem Cell Rep. 16, 1143–1155. https://doi.org/10. 1016/j.stemcr.2021.03.017. 18. Xu, P.-F., Borges, R.M., Fillatre, J., De Oliveira-Melo, M., Cheng, T., Thisse, B., and Thisse, C. (2021). Construction of a mammalian embryo model from stem cells organized by a morphogen signalling centre. Nat. Commun. 12, 3277. https://doi.org/10.1038/s41467-021-23653-4. 19. Bérenger-Currias, N.M., Mircea, M., Adegeest, E., Van Den Berg, P.R., Feliksik, M., Hochane, M., Idema, T., Tans, S.J., and Semrau, S. (2022). A gastruloid model of the interaction between embryonic and extra-embryonic cell types. J. Tissue Eng. 13, 20417314221103042. https://doi.org/10.1177/20417314221103042. 20. Wehmeyer, A.E., Sch€ule, K.M., Conrad, A., Schröder, C.M., Probst, S., and Arnold, S.J. (2022). Chimeric 3D gastruloids – a versatile tool for studies of mammalian peri-gastrulation development. Development 149, dev200812. https://doi.org/10.1242/dev.200812. 21. Anlas, K., Gritti, N., Oriola, D., Arató, K., Nakaki, F., Le Lim, J., Sharpe, J., and Trivedi, V. (2021). Dynamics of anteroposterior axis establishment in amammalian embryo-like system. Preprint at bioRxiv. https://doi.org/10. 1101/2021.02.24.432766. 22. Rothová, M.M., Nielsen, A.V., Proks, M., Wong, Y.F., Riveiro, A.R., Linneberg-Agerholm, M., David, E., Amit, I., Trusina, A., and Brickman, J.M. (2022). Identification of the central intermediate in the extra-embryonic to embryonic endoderm transition through single-cell transcriptomics. Nat. Cell Biol. 24, 833–844. https://doi.org/10.1038/s41556-02200923-x. 23. Pijuan-Sala, B., Griffiths, J.A., Guibentif, C., Hiscock, T.W., Jawaid, W., Calero-Nieto, F.J., Mulas, C., Ibarra-Soria, X., Tyser, R.C.V., Ho, D.L.L., et al. (2019). A single-cell molecular map of mouse gastrulation and early organogenesis. Nature 566, 490–495. https://doi.org/10.1038/s41586019-0933-9. 24. Masui, S., Nakatake, Y., Toyooka, Y., Shimosato, D., Yagi, R., Takahashi, K., Okochi, H., Okuda, A., Matoba, R., Sharov, A.A., et al. (2007). Pluripotency governed by Sox2 via regulation of Oct3/4 expression in mouse embryonic stem cells. Nat. Cell Biol. 9, 625–635. https://doi. org/10.1038/ncb1589. 25. Festuccia, N., Osorno, R., Halbritter, F., Karwacki-Neisius, V., Navarro, P., Colby, D., Wong, F., Yates, A., Tomlinson, S.R., and Chambers, I. (2012). Esrrb is a direct Nanog target gene that can substitute for Nanog function in pluripotent cells. Stem Cells 11, 477–490. https:// doi.org/10.1016/j.stem.2012.08.002. 26. Shi,W.,Wang,H.,Pan,G.,Geng,Y.,Guo,Y., andPei,D. (2006).Regulation of the pluripotencymarker Rex-1 by Nanog and Sox2. J. Biol. Chem. 281, 23319–23325. https://doi.org/10.1074/jbc.M601811200. 27. Cheng, S., Pei, Y., He, L., Peng, G., Reinius, B., Tam, P.P.L., Jing, N., and Deng, Q. (2019). Single-cell RNA-seq reveals cellular heterogeneity of pluripotency transition and X chromosome dynamics during early mouse development. Cell Rep. 26, 2593–2607.e3. https://doi.org/10.1016/j.celrep.2019.02.031. 28. Cermola, F., D’Aniello, C., Tatè, R., De Cesare, D., Martinez-Arias, A., Minchiotti, G., and Patriarca, E.J. (2021). Gastruloid development competence discriminates different states of pluripotency. Stem Cell Rep. 16, 354–369. https://doi.org/10.1016/j.stemcr.2020.12.013. 882 Cell Stem Cell 30, 867–884, June 1, 2023 29. Koch, F., Scholze, M., Wittler, L., Schifferl, D., Sudheer, S., Grote, P., Timmermann, B., Macura, K., and Herrmann, B.G. (2017). Antagonistic activities of Sox2 and brachyury control the fate choice of neuro-mesodermal progenitors. Dev. Cell 42, 514–526.e7. https://doi.org/10.1016/ j.devcel.2017.07.021. 30. Bergsland, M., Ramsköld, D., Zaouter, C., Klum, S., Sandberg, R., and Muhr, J. (2011). Sequentially acting Sox transcription factors in neural lineage development. Genes Dev. 25, 2453–2464. https://doi.org/10. 1101/gad.176008.111. 31. Avilion, A.A., Nicolis, S.K., Pevny, L.H., Perez, L., Vivian, N., and Lovell- Badge, R. (2002). Multipotent cell lineages in early mouse development depend on SOX2 function. Genes Amp Dev. 17, 126–140. 32. Setty, M., Kiseliovas, V., Levine, J., Gayoso, A., Mazutis, L., and Pe’er, D. (2019). Characterization of cell fate probabilities in single-cell data with Palantir. Nat. Biotechnol. 37, 451–460. https://doi.org/10.1038/s41587019-0068-4. 33. Serra, D., Mayr, U., Boni, A., Lukonin, I., Rempfler, M., Challet Meylan, L.C., Stadler, M.B., Strnad, P., Papasaikas, P., Vischi, D., et al. (2019). Self-organization and symmetry breaking in intestinal organoid development. Nature 569, 66–72. https://doi.org/10.1038/s41586-019-1146-y. 34. Arnold, S.J., Hofmann, U.K., Bikoff, E.K., and Robertson, E.J. (2008). Pivotal roles for eomesodermin during axis formation, epithelium-tomesenchyme transition and endoderm specification in the mouse. Development 135, 501–511. https://doi.org/10.1242/dev.014357. 35. Herrmann, B.G. (1991). Expression pattern of the brachyury gene in whole mount Twis/Twis mutant embryos. Development 113, 913–917. 36. Pevny, L.H., Sockanathan, S., Placzek, M., and Lovell-Badge, R. (1998). A role for Sox1 in neural determination. Development 125, 1967–1978. 37. Levine, J.H., Simonds, E.F., Bendall, S.C., Davis, K.L., Amir, E.-a.D., Tadmor, M.D., Litvin, O., Fienberg, H.G., Jager, A., Zunder, E.R., et al. (2015). Data-driven phenotypic dissection of AML reveals progenitorlike cells that correlate with prognosis. Cell 162, 184–197. https://doi. org/10.1016/j.cell.2015.05.047. 38. Lukonin, I., Serra, D., Challet Meylan, L.C., Volkmann, K., Baaten, J., Zhao, R., Meeusen, S., Colman, K., Maurer, F., Stadler, M.B., et al. (2020). Phenotypic landscape of intestinal organoid regeneration. Nature 586, 275–280. https://doi.org/10.1038/s41586-020-2776-9. 39. Hemmati-Brivanlou, A., and Melton, D. (1997). Vertebrate embryonic cells will become nerve cells unless told otherwise. Cell 88, 13–17. https://doi.org/10.1016/S0092-8674(00)81853-X. 40. Ying, Q.L., Wray, J., Nichols, J., Batlle-Morera, L., Doble, B., Woodgett, J., Cohen, P., and Smith, A. (2008). The ground state of embryonic stem cell self-renewal. Nature 453, 519–523. https://doi.org/10.1038/ nature06968. 41. Wray, J., Kalkan, T., and Smith, A.G. (2010). The ground state of plurip- otency. Biochem. Soc. Trans. 38, 1027–1032. https://doi.org/10.1042/ BST0381027. 42. Betschinger, J., Nichols, J., Dietmann, S., Corrin, P.D., Paddison, P.J., and Smith, A. (2013). Exit frompluripotency is gated by intracellular redistribution of the bHLH transcription factor Tfe3. Cell 153, 335–347. https:// doi.org/10.1016/j.cell.2013.03.012. 43. Leitch, H.G., Nichols, J., Humphreys, P., Mulas, C., Martello, G., Lee, C., Jones, K., Surani, M.A., and Smith, A. (2013). Rebuilding pluripotency from primordial germ cells. Stem Cell Rep. 1, 66–78. https://doi.org/10. 1016/j.stemcr.2013.03.004. 44. Parchem, R.J., Ye, J., Judson, R.L., LaRussa, M.F., Krishnakumar, R., Blelloch, A., Oldham, M.C., and Blelloch, R. (2014). Two miRNA clusters reveal alternative paths in late-stage reprogramming. Cell Stem Cell 14, 617–631. https://doi.org/10.1016/j.stem.2014.01.021. 45. Hart, A.H., Hartley, L., Ibrahim, M., and Robb, L. (2004). Identification, cloning and expression analysis of the pluripotency promoting Nanog genes in mouse and human. Dev. Dyn. 230, 187–198. https://doi.org/ 10.1002/dvdy.20034. 46. Sambasivan, R., and Steventon, B. (2020). Neuromesodermal progeni- tors: A basis for robust axial patterning in development and evolution. Front. Cell Dev. Biol. 8, 607516. https://doi.org/10.3389/fcell.2020. 607516. 47. Hunt, G.C., Singh, P., and Schwarzbauer, J.E. (2012). Endogenous pro- duction of fibronectin is required for self-renewal of cultured mouse embryonic stem cells. Exp. Cell Res. 318, 1820–1831. https://doi.org/10. 1016/j.yexcr.2012.06.009. 48. Kapp, T.G., Rechenmacher, F., Neubauer, S., Maltsev, O.V., Cavalcanti- Adam, E.A., Zarka, R., Reuning, U., Notni, J.,Wester, H.-J., Mas-Moruno, C., et al. (2017). A comprehensive evaluation of the activity and selectivity profile of ligands for RGD-binding integrins. Sci. Rep. 7, 39805. https:// doi.org/10.1038/srep39805. 49. Ruoslahti, E., and Pierschbacher, M.D. (1987). New perspectives in cell adhesion: RGD and integrins. Science 238, 491–497. https://doi.org/ 10.1126/science.2821619. 50. Desrochers, L.M., Bordeleau, F., Reinhart-King, C.A., Cerione, R.A., and Antonyak, M.A. (2016). Microvesicles provide a mechanism for intercellular communication by embryonic stem cells during embryo implantation. Nat. Commun. 7, 11958. https://doi.org/10.1038/ncomms11958. 51. Hur, Y.H., Feng, S., Wilson, K.F., Cerione, R.A., and Antonyak, M.A. (2020). Embryonic stem-cell-derived extracellular vesicles maintain ESC stemness by activating FAK. Dev. Cell 56, 277–291.e6. https://doi. org/10.1016/j.devcel.2020.11.017. 52. Lee, S.H., Lee, Y.J., and Han, H.J. (2011). Role of hypoxia-induced fibro- nectin-integrin b1 expression in embryonic stem cell proliferation and migration: involvement of PI3K/Akt and FAK. J. Cell. Physiol. 226, 484–493. https://doi.org/10.1002/jcp.22358. 53. Deluz, C., Friman, E.T., Strebinger, D., Benke, A., Raccaud, M., Callegari, A., Leleu, M., Manley, S., and Suter, D.M. (2016). A role for mitotic bookmarking of SOX2 in pluripotency and differentiation. Genes Dev. 30, 2538–2550. https://doi.org/10.1101/gad.289256.116. 54. Bondue, A., T€annler, S., Chiapparo, G., Chabab, S., Ramialison, M., Paulissen, C., Beck, B., Harvey, R., and Blanpain, C. (2011). Defining the earliest step of cardiovascular progenitor specification during embryonic stem cell differentiation. J. Cell Biol. 192, 751–765. https://doi.org/ 10.1083/jcb.201007063. 55. Wymeersch, F.J., Huang, Y., Blin, G., Cambray, N., Wilkie, R., Wong, F.C., and Wilson, V. (2016). Position-dependent plasticity of distinct progenitor types in the primitive streak. eLife 5, e10042. https://doi.org/10. 7554/eLife.10042. 56. Rosen, L.U., Stapel, L.C., Argelaguet, R., Barker, C.G., Yang, A., Reik, W., and Marioni, J.C. (2022). Inter-gastruloid heterogeneity revealed by single cell transcriptomics time course: implications for organoid based perturbation studies. Preprint at bioRxiv. https://doi.org/10.1101/2022. 09.27.509783. 57. Ortmann, D., Brown, S., Czechanski, A., Aydin, S., Muraro, D., Huang, Y., Tomaz, R.A., Osnato, A., Canu, G., Wesley, B.T., et al. (2020). Naive pluripotent stem cells exhibit phenotypic variability that is driven by genetic variation. Stem Cells 27, 470–481.e6. https://doi.org/10.1016/j. stem.2020.07.019. 58. Stower, M.J., and Srinivas, S. (2014). Heading forwards: anterior visceral endoderm migration in patterning the mouse embryo. Philos. Trans. R. Soc. Lond. B Biol. Sci. 369, 20130546. https://doi.org/10.1098/rstb. 2013.0546. 59. Ang, S.L., Jin, O., Rhinn, M., Daigle, N., Stevenson, L., and Rossant, J. (1996). A targetedmouseOtx2mutation leads to severe defects in gastrulation and formation of axial mesoderm and to deletion of rostral brain. Development 122, 243–252. https://doi.org/10.1242/dev.122.1.243. 60. Acampora, D., Mazan, S., Lallemand, Y., Avantaggiato, V., Maury, M., Simeone, A., and Brûlet, P. (1995). Forebrain and midbrain regions are deleted in Otx2 / mutants due to a defective anterior neuroectoderm specification during gastrulation. Development 121, 3279–3290. https://doi.org/10.1242/dev.121.10.3279. 61. Matsuo, I., Kuratani, S., Kimura, C., Takeda, N., and Aizawa, S. (1995). Mouse Otx2 functions in the formation and patterning of rostral head. Genes Dev. 9, 2646–2658. https://doi.org/10.1101/gad.9.21.2646. 62. Vianello, S.D., and Lutolf, M. (2020). In vitro endoderm emergence and self-organisation in the absence of extraembryonic tissues and embryonic architecture. Preprint at bioRxiv. https://doi.org/10.1101/2020.06. 07.138883. 63. Nowotschin, S., Setty, M., Kuo, Y.-Y., Liu, V., Garg, V., Sharma, R., Simon, C.S., Saiz, N., Gardner, R., Boutet, S.C., et al. (2019). The emergent landscape of the mouse gut endoderm at single-cell resolution. Nature 569, 361–367. https://doi.org/10.1038/s41586-019-1127-1. 64. Osumi, N., Shinohara, H., Numayama-Tsuruta, K., and Maekawa, M. (2008). Concise review: Pax6 transcription factor contributes to both embryonic and adult neurogenesis as amultifunctional regulator. StemCells 26, 1663–1672. https://doi.org/10.1634/stemcells.2007-0884. 65. Bel-Vialar, S., Medevielle, F., and Pituello, F. (2007). The on/off of Pax6 controls the tempo of neuronal differentiation in the developing spinal cord. Dev. Biol. 305, 659–673. https://doi.org/10.1016/j.ydbio.2007. 02.012. 66. Chhabra, S., Liu, L., Goh, R., Kong, X., and Warmflash, A. (2019). Dissecting the dynamics of signaling events in the BMP, WNT, and NODAL cascade during self-organized fate patterning in human gastruloids. PLoS Biol. 17, e3000498. https://doi.org/10.1371/journal.pbio. 3000498. 67. 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 edge sensing controls gastruloid self-organization. Dev. Cell 39, 302–315. https://doi.org/10.1016/j.devcel.2016.09.016. 68. Liu, L., Nemashkalo, A., Rezende, L., Jung, J.Y., Chhabra, S., Guerra, M.C., Heemskerk, I., and Warmflash, A. (2022). Nodal is a short-range morphogen with activity that spreads through a relay mechanism in human gastruloids. Nat. Commun. 13, 497. https://doi.org/10.1038/ s41467-022-28149-3. 69. Kramer, B.A., Sarabia del Castillo, J., and Pelkmans, L. (2022). Multimodal perception links cellular state to decision-making in single cells. Science 377, 642–648. https://doi.org/10.1126/science.abf4062. 70. Xavier da Silveira Dos Santos, A., and Liberali, P. (2018). From single cells to tissue self-organization. FEBS Journal 18. 483–419. https://doi.org/10. 1111/febs.14694. 71. Kinney, B.A., Al Anber, A., Row, R.H., Tseng, Y.-J., Weidmann, M.D., Knaut, H., and Martin, B.L. (2020). Sox2 and canonical Wnt signaling interact to activate a developmental checkpoint coordinating morphogenesis with mesoderm fate acquisition. Cell Rep. 33, 108311. https:// doi.org/10.1016/j.celrep.2020.108311. 72. Gouti, M., Delile, J., Stamataki, D., Wymeersch, F.J., Huang, Y., Kleinjung, J., Wilson, V., and Briscoe, J. (2017). A gene regulatory network balances neural and mesoderm specification during vertebrate trunk development. Dev. Cell 41, 243–261.e7. https://doi.org/10.1016/j. devcel.2017.04.002. 73. Blassberg, R., Patel, H., Watson, T., Gouti, M., Metzis, V., Delás, M.J., and Briscoe, J. (2020). Sox2 levels configure the WNT response of epiblast progenitors responsible for vertebrate body formation 17. 3165–3145. https://doi.org/10.1101/2020.12.29.424684. 74. Posfai, E., Schell, J.P., Janiszewski, A., Rovic, I., Murray, A., Bradshaw, B., Yamakawa, T., Pardon, T., El Bakkali, M., Talon, I., et al. (2021). Evaluating totipotency using criteria of increasing stringency. Nat. Cell Biol. 23, 49–60. https://doi.org/10.1038/s41556-020-00609-2. 75. Yanagida, A., Spindlow, D., Nichols, J., Dattani, A., Smith, A., and Guo, G. (2021). Naive stem cell blastocyst model captures human embryo lineage segregation. Cell Stem Cell 28, 1016–1022.e4. https://doi.org/10. 1016/j.stem.2021.04.031. 76. Kagawa, H., Javali, A., Khoei, H.H., Sommer, T.M., Sestini, G., Novatchkova, M., Scholte op Reimer, Y., Castel, G., Bruneau, A., Maenhoudt, N., et al. (2022). Human blastoids model blastocyst Cell Stem Cell 30, 867–884, June 1, 2023 883 development and implantation. Nature 601, 600–605. https://doi.org/10. 1038/s41586-021-04267-8. 77. Tarazi, S., Aguilera-Castrejon, A., Joubran, C., Ghanem, N., Ashouokhi, S., Roncato, F., Wildschutz, E., Haddad, M., Oldak, B., Gomez-Cesar, E., et al. (2022). Post-gastrulation synthetic embryos generated ex utero from mouse naive ESCs. Cell 185, 3290–3306.e25. https://doi.org/10. 1016/j.cell.2022.07.028. 78. Amadei, G., Handford, C.E., Qiu, C., De Jonghe, J., Greenfeld, H., Tran, M., Martin, B.K., Chen, D.Y., Aguilera-Castrejon, A., Hanna, J.H., et al. (2022). Embryo model completes gastrulation to neurulation and organogenesis. Nature 610, 143–153. https://doi.org/10.1038/s41586-02205246-3. 79. Merle, M., Friedman, L., Chureau, C., and Gregor, T. (2023). Precise and scalable self-organization in mammalian pseudo-embryos. Preprint at arXiv. https://doi.org/10.48550/arXiv.2303.17522. 80. Ghimire, S., Van der Jeught, M., Neupane, J., Roost, M.S., Anckaert, J., Popovic, M., Van Nieuwerburgh, F., Mestdagh, P., Vandesompele, J., Deforce, D., et al. (2018). Comparative analysis of naive, primed and ground state pluripotency in mouse embryonic stem cells originating from the same genetic background. Sci. Rep. 8, 5884. https://doi.org/ 10.1038/s41598-018-24051-5. 81. Baillie-Johnson, P., van den Brink, S.C., Balayo, T., Turner, D.A., and Martinez Arias, A. (2015). Generation of aggregates of mouse embryonic stem cells that show symmetry breaking, polarization and emergent collective behaviour in vitro. J. Vis. Exp. https://doi.org/10.3791/53252. 82. Hama, H., Hioki, H., Namiki, K., Hoshida, T., Kurokawa, H., Ishidate, F., Kaneko, T., Akagi, T., Saito, T., Saido, T., et al. (2015). ScaleS: an optical clearing palette for biological imaging. Nat. Neurosci. 18, 1518–1529. https://doi.org/10.1038/nn.4107. 83. Zhu, X., Huang, L., Zheng, Y., Song, Y., Xu, Q., Wang, J., Si, K., Duan, S., and Gong, W. (2019). Ultrafast optical clearing method for three-dimensional imaging with cellular resolution. Proc. Natl. Acad. Sci. USA 116, 11480–11489. https://doi.org/10.1073/pnas.1819583116. 84. Ortiz, R., deMedeiros, G., Peters, A.H.F.M., Liberali, P., andRempfler, M. (2020). RDCNet: Instance Segmentation with a Minimalist Recurrent Residual Network. Held in Cham (Springer International Publishing), pp. 434–443. 85. Zappia, L., and Oshlack, A. (2018). Clustering trees: a visualization for evaluating clusterings at multiple resolutions. GigaScience 7, 1–9. https://doi.org/10.1093/gigascience/giy083. 86. Lawrence, M., Huber, W., Pagès, H., Aboyoun, P., Carlson, M., Gentleman, R., Morgan, M.T., and Carey, V.J. (2013). Software for computing and annotating genomic ranges. PLoS Comp. Biol. 9, e1003118. https://doi.org/10.1371/journal.pcbi.1003118. 87. Patro, R., Duggal, G., Love, M.I., Irizarry, R.A., and Kingsford, C. (2017). Salmon provides fast and bias-aware quantification of transcript expression. Nat. Methods 14, 417–419. https://doi.org/10.1038/nmeth.4197. 88. Srivastava, A., Malik, L., Smith, T., Sudbery, I., and Patro, R. (2019). Alevin efficiently estimates accurate gene abundances from dscRNAseq data. Genome Biol. 20, 65. https://doi.org/10.1186/s13059-0191670-y. 884 Cell Stem Cell 30, 867–884, June 1, 2023 89. Love, M.I., Soneson, C., Hickey, P.F., Johnson, L.K., Pierce, N.T., Shepherd, L., Morgan, M., and Patro, R. (2020). Tximeta: reference sequence checksums for provenance identification in RNA-seq. PLoS Comp. Biol. 16, e1007664. https://doi.org/10.1371/journal.pcbi.1007664. 90. McCarthy, D.J., Campbell, K.R., Lun, A.T., and Wills, Q.F. (2017). Scater: pre-processing, quality control, normalization and visualization of singlecell RNA-seq data in R. Bioinformatics 33, 1179–1186. https://doi.org/10. 1093/bioinformatics/btw777. 91. Hafemeister, C., and Satija, R. (2019). Normalization and variance stabi- lization of single-cell RNA-seq data using regularized negative binomial regression. Genome Biol. 20, 296. https://doi.org/10.1186/s13059-0191874-1. 92. Butler, A., Hoffman, P., Smibert, P., Papalexi, E., and Satija, R. (2018). Integrating single-cell transcriptomic data across different conditions, technologies, and species. Nat. Biotechnol. 36, 411–420. https://doi. org/10.1038/nbt.4096. 93. Stuart, T., Butler, A., Hoffman, P., Hafemeister, C., Papalexi, E., Mauck, W.M., Hao, Y., Stoeckius, M., Smibert, P., and Satija, R. (2019). Comprehensive integration of single-cell data. Cell 177, 1888– 1902.e21. https://doi.org/10.1016/j.cell.2019.05.031. 94. Wolf, F.A., Angerer, P., and Theis, F.J. (2018). SCANPY: large-scale sin- gle-cell gene expression data analysis. Genome Biol. 19, 15. https://doi. org/10.1186/s13059-017-1382-0. 95. Herman, J.S., Sagar, and Gr€un, D. (2018). FateID infers cell fate bias in multipotent progenitors from single-cell RNA-seq data. Nat. Methods 15, 379–386. https://doi.org/10.1038/nmeth.4662. 96. Anders, S., and Huber, W. (2010). Differential expression analysis for sequence count data. Genome Biol. 11, R106. https://doi.org/10.1186/ gb-2010-11-10-r106. 97. Cusanovich, D.A., Daza, R., Adey, A., Pliner, H.A., Christiansen, L., Gunderson, K.L., Steemers, F.J., Trapnell, C., and Shendure, J. (2015). Multiplex single-cell profiling of chromatin accessibility by combinatorial cellular indexing. Science 348, 910–914. https://doi.org/10.1126/science.aab1601. 98. Martin, M. (2011). Cutadapt removes adapter sequences from high- throughput sequencing reads. EMBnet.journal 17, 10. https://doi.org/ 10.14806/ej.17.1.200. 99. Dobin, A., Davis, C.A., Schlesinger, F., Drenkow, J., Zaleski, C., Jha, S., Batut, P., Chaisson, M., and Gingeras, T.R. (2013). STAR: ultrafast universal RNA-seq aligner. Bioinformatics 29, 15–21. https://doi.org/10. 1093/bioinformatics/bts635. 100. Trapnell, C., Williams, B.A., Pertea, G., Mortazavi, A., Kwan, G., Van Baren, M.J., Salzberg, S.L., Wold, B.J., and Pachter, L. (2010). Transcript assembly and quantification by RNA-Seq reveals unannotated transcripts and isoform switching during cell differentiation. Nat. Biotechnol. 28, 511–515. https://doi.org/10.1038/nbt.1621. 101. Roberts, A., Pimentel, H., Trapnell, C., and Pachter, L. (2011). Identification of novel transcripts in annotated genomes using RNASeq. Bioinformatics 27, 2325–2329. https://doi.org/10.1093/bioinformatics/btr355. STAR+METHODS