Abstract
Naive pluripotent stem cells have the highest developmental potential but their in vivo existence in the blastocyst is transient. Here we report a blastocyst motif substrate for the in vitro reversion of mouse and human pluripotent stem cells to a naive state. The substrate features randomly varied microstructures, which we call motifs, mimicking the geometry of the blastocyst. Motifs representing mouse-blastocyst-scaled curvature ranging between 15 and 62 mm−1 were the most efficient in promoting reversion to naivety, as determined by time-resolved correlative analysis. In these substrates, apical constriction enhances E-cadherin/RAC1 signalling and activates the mechanosensitive nuclear transducer YAP, promoting the histone modification of pluripotency genes. This results in enhanced levels of pluripotency transcription factor NANOG, which persist even after cells are removed from the substrate. Pluripotent stem cells cultured in blastocyst motif substrates display a higher development potential in generating embryoid bodies and teratomas. These findings shed light on naivety-promoting substrate design and their large-scale implementation.
Similar content being viewed by others
Main
In nature, naive and primed states of pluripotent stem cells emerge sequentially in pre- and post-implantation embryos1,2. These states are distinct in morphology, clonogenic capacity, transcriptome and epigenetic signatures3. Naive pluripotent stem cells (nPSCs) have shown robust proliferation and enhanced directed differentiation4, especially towards extra-embryonic lineages5,6, compared with their primed counterparts. Therefore, generating and stabilizing nPSCs in vitro is of great importance for a comprehensive modelling of early embryo and foetal anomalies.
Current strategies of primed–naive reversion rely on small-molecule or genetic approaches7,8, which are tedious, cost intensive and subject to safety concerns. This warrants the search for robust, scalable, rapid and safe approaches. Structural cues are powerful morphogens in embryo development9. At the epiblasts (Epi)–trophectoderm (TE) interface in preimplantation blastocysts, nPSCs show compacted apical cell–cell contacts due to the cellular tension of the surrounding TE10,11, which presents an axially symmetric concave geometry12 characterized by its height, width and curvature.
Here we hypothesize that a blastocyst-deduced topographical microenvironment on a cell culture substrate can revert cells to naivety by physical means. We anticipate that the cell constraint, mediated by the certain curvature range within the dimensional space of the blastocyst, could elicit reversion. We collectively designate this parameter as the blastocyst-scaled curvature range (BSCR). When the microtopographical units on a substrate encompass the BSCR, they are referred to as BSCR+ motifs. We intend to address the question, whether the reversion requires for BSCR in a radial-symmetric manner, as provided by the Epi–TE interface, or in only a limited number of directions. However, given the impracticality of the side-by-side integration of a multitude of BSCR motifs in a closed three-dimensional architecture like a blastocyst, a structured, that is, open 2.5-dimensional polymeric substrate should be engaged.
To test our hypothesis, the blastocyst motif substrate (BMS) was designed and fabricated featuring thousands of randomly varied motifs, covering not only biological size and shape heterogeneity but also following our reductionist paradigm. Electrical discharge machining was utilized to create the microstructure on metal mould surfaces. The resulting surfaces were then inverted into the targeted BMSs by the injection moulding of polystyrene (Supplementary Table 1 lists the surface parameters). Our hypothesis was validated by employing spatiotemporal analysis, which correlates geometrical information with cellular behaviour. The BSCR—not necessarily radial symmetric—was able to elicit the in situ reversion of primed pluripotent stem cells (PSCs) to a sustainable naive state. The PSCs on BSCR+ motifs presented apical constriction and enhanced E-cad/RAC1 signalling, with weakened cell–substrate interaction. The propagation of physical cues from the BSCR interface to the nuclei through a series of force-dependent elements led to histone modification (H3K27me3 and H3K4me3) at the promoter regions of Nanog and Zic2 and at a distal enhancer region of Pou5f1. Topographical cues abstracted from nature’s blueprint offer inherent stability and robustness, which can be easily integrated into the PSC substrate for large-scale implementation in biomanufacturing processes.
Bioinspired design and characterization of BMS
We analysed the architecture of blastocysts (embryonic day, E4.25–E4.50)10 and determined the dimensional limits of nPSCs in Epi (height, 21 ± 9 µm; width, 59 ± 10 µm), as well as the curvature κ of the Epi–TE interface (BSCR), ranging between 15 and 62 mm−1 (Fig. 1a and Supplementary Fig. 1).
To grasp both in silico and real-world 2.5-dimensional BMS topographies with respect to the blastocyst curvature properties, we calculated and mapped the spatial information of the BMS surface based on the voxel datasets13 from micro-computed tomography (micro-CT) (Supplementary Fig. 2a,b). Noteworthily, we developed a scaled three-point approach (Fig. 1b,c and Supplementary Fig. 3) to measure the asymmetrical curvatures in 18 discrete directions around each surface point on the substrate. A given point was set to a radial distance of 30 µm, that is, half the ~60 µm Epi width, to match the blastocyst scale (Fig. 1a and Supplementary Fig. 1d).
The morphological features of BMS, that is, convex, flat and concave, were then distinguished based on the mean value of the curvatures13 〈κ〉 in 18 directions. The individual curvature value κ was used to define the BSCR+ area, in which the given point has at least one curvature within the BSCR (Fig. 1d and Supplementary Fig. 2c–e). The representative image of a BMS shows about 8% of the BSCR+ points. The maximum BSCR count lies between 4 and 5. The high proportion of BSCR count at 18 directions indicated the relatively higher proportion of bowl-like structures on the substrates. The BSCR count and morphological features correlate in an intricate way, with the higher number of BSCR located on concave motifs (Fig. 1d).
Establishment of such topographical maps enabled the identification and investigation of specific surface features, including the aforementioned motifs14. Correlating these maps with microscopy images showing nPSC distribution (Supplementary Fig. 4), we were able to analyse the in situ effect of geometrical cues on PSC reversion (Fig. 1e).
BSCR reverts PSCs from primed to naive state
To obtain primed cells, PhiC31 and iPS-MEF-Ng-492B-4 mouse PSCs were chemically adapted for at least three passages to the primed-like state by using 1i medium containing GSK3β inhibitor and AXIN stabilizer (Supplementary Fig. 5), followed by seeding on laminin-coated BMS (Supplementary Fig. 6). Compared with the plain substrate (laminin-coated polystyrene without microstructure), PSCs on BMS exhibited the upregulated naivety genes (Extended Data Fig. 1a), with NANOG, STELLA, OCT4 and SSEA1 proteins at similar levels as the naive control (TCP, 2i/L) (Fig. 2a). For the SSEA4– (a negative marker of mouse PSC) population, the percentage of SSEA1+ (a naive marker of mouse PSC) cells was higher on the BMS (94 ± 1%) than the plain substrate (89 ± 1%) (Extended Data Fig. 1b,c). Contrarily, lineage-specific gene expression was decreased on BMS (Extended Data Fig. 1d). ZIC2, which drives naive–primed transition15, was downregulated (Fig. 2a).
To correlate the in situ effect of detailed structural cues on PSC naivety, we sought to discriminate those nPSCs (NANOGhi and STELLAhi) using a fluorescence threshold based on the mean fluorescence intensity (MFI) on a plain control (Supplementary Fig. 7) and overlaying those signals with the motif and BSCR maps (Fig. 2b and Supplementary Fig. 4). The number of nPSCs was remarkably higher in concave motifs, than in convex and flat on days 1 and 3 (Fig. 2b and Extended Data Fig. 1e,f). Comparison of concave areas with and without BSCR revealed that BSCR+ motif, rather than concave itself, played the pivotal role to revert the naivety of PSCs (Fig. 2b). These results were supported by analysing NANOGhi and STELLAhi cells on different motifs with varying BSCR counts. On days 1 and 3, a significantly higher percentage of nPSCs was found on BSCR+ compared with BSCR– motif (Fig. 2c,d), and the naivety of nPSCs was correlated with the BSCR counts (Fig. 2e,f and Extended Data Fig. 1g). In particular, even the motifs with BSCR in only one direction (BSCR 1) was sufficient to increase the naivety (Extended Data Fig. 1h,i), indicating that BSCR is the key naive-pluripotency-associated geometrical parameter.
To validate the in situ primed–naive reversion, rather than other mechanisms such as cell selection and migration, we investigated the cell–substrate interaction from the initial cell attachment. After seeding, the cells exhibited a random distribution on different motifs of the BMS, with similar density, NANOG expression and cell size 9 h post-cell seeding (Fig. 3a–d). The BSCR motif did not affect the necrosis of PSCs. The apoptosis level of PSCs in the BSCR+ area remained similar to BSCR– on day 0, but increased to a significantly higher level on day 3 (Extended Data Fig. 2a,b). This might be attributed to the higher sensitivity to cellular stress in naive PSCs than their primed counterparts16,17, as a result of elevated mitochondrial priming18. In addition, the high cell confluence in the BSCR+ area, resulting from the enhanced proliferation of nPSCs, could trigger apoptotic cell death19,20.
To evaluate the reversion kinetics, we utilized time-lapse imaging to track live cells using a PSC line with NANOG reporter–GFP21. The gradually increased GFP signals was observed in the BSCR+ area as early as 21 h after cell seeding (Fig. 3e and Extended Data Fig. 2c). By analysing the migration of single cells, we further confirmed the in situ reversion within the BSCR+ motif. From day 0 to day 1, the migration distance of the cells on BMS was limited to 10 µm. From day 1 to day 3, rare migration was observed from BSCR– to BSCR+ (Fig. 3f). These results excluded the possibility that nPSCs in BSCR+ are from BSCR– as a result of remote migration. In particular, more nPSCs were found in the BSCR– area at later time points (Fig. 3e), which is consistent with the results that a similar percentage of nPSCs were found in different motifs on day 4 (Fig. 2c,d and Extended Data Fig. 1e,f). The analysis of the trajectory video revealed that nPSCs in the BSCR– area are mainly from the BSCR+ area, with high confluence and subsequent expansion/migration (Fig. 3f, Extended Data Fig. 2d and Supplementary Videos 1 and 2). The impact of motifs on PSC naivety reversion and proliferation was further examined with time-resolved imaging analysis (Extended Data Figs. 3 and 4). Consistent with the reported results22,23,24, the naive cells (2i/L-PSCs) proliferated faster than the primed counterparts (1i-PSCs) and the naive/primed mixed PSCs, showing the highest increasing rate of cell density on all the tested motifs. BSCR+ motif could revert primed cells to a more naive state. For the 1i-PSCs and mixed PSCs, which contains the primed cells, a significant increase in GFPhi cell percentage was observed 21 h after cell seeding in the BSCR+ area, compared with the BSCR– and plain substrates. However, the percentage of Ki67+ cells remained at a similar level at/before these time points. This result indicated that the increased number of naive cells in the BSCR+ area at the early stage is mainly attributed to the reversion of—rather than the proliferation of—naive cells.
Next, to verify the effect of BSCR on naivety reversion, we created substrates with uniform microbowls (radial symmetry) and microgrooves (axial symmetry) (Extended Data Fig. 5 and Supplementary Tables 2 and 3). Consistent with BMS, reversion correlates with the BSCR counts, regardless of the microstructure shape. The microbowls without BSCR, despite their similarity in shape with a blastocyst, demonstrated a lower reversion capacity than the microgrooves with BSCR (Extended Data Fig. 6a,b). The uniform microstructures with higher BSCR counts induced higher levels of naivety (NANOG-GFP, SSEA1, TBX3 and STELLA) (Extended Data Fig. 6). When PSCs were cultured on substrates with microbowls with and without BSCR, they showed similar proliferation profile from days 1 to 3 (Extended Data Fig. 6f–h), but the higher percentage of GFPhi PSCs could be found as early as day 1 on microbowl-containing BSCR (Extended Data Fig. 6b). This observation is in line with that on BMS, suggesting that the increase in naive cells in the BSCR+ area in the early stage is not from proliferation but is a result of naivety reversion.
BSCR triggers mechanosensation and histone modification
PSCs inside the BSCR+ area presented the morphological characteristics of naive cells, with compacted colony and decreased cell interspace along the direction of BSCR (Supplementary Figs. 1f and 7a,b). The mere increase in the cell density on a plain substrate did not increase the NANOG level (Extended Data Fig. 7c), suggesting the indispensable role of BSCR curvature on naivety reversion.
It is known that the preservation of PSC naivety can be strengthened by cytoskeleton-bound cell–cell junctions25,26,27. Here cell clusters in the BSCR+ area displayed strong apical F-actin and E-cad compared with the BSCR– area (Fig. 4a,b). At the population level, E-cad and its stabilizer RAC1 (refs. 28,29,30) were upregulated on BMS (Fig. 4c,d). The inhibition of E-cad and RAC1 significantly decreased BSCR-mediated naivety reversion on both BMS (Fig. 4f–i) and substrates with uniformed microbowls (Extended Data Fig. 6c). Furthermore, the nPSCs rapidly lost their naivety on E-cad and RAC1 inhibition, suggesting the essential role of E-cad/RAC1 signalling in the preservation of naivety (Fig. 4j). We further investigated the intracellular components related to mechanical force propagation. A strong upregulation of pMLC2 and a higher cell contractility were observed in BMS–PSCs, which was abolished on E-cad neutralization (Extended Data Fig. 7d–g), suggesting the intermediary role of cell–cell adhesion in force transmission. PSCs on BMS exhibited a weakened cell–substrate interaction, with about 20% decrease in active FAK (phosphorylated/total FAK ratio) (Fig. 4e), which is in agreement with the previous report that concave curvature weakened the cell–substrate interaction31. The naive markers NANOG, STELLA and primed marker ZIC2 in BMS–PSCs were not altered following FAK inhibition (Fig. 4f). These results suggested that the naivety of BMS–PSCs indispensably relied on cell–cell adhesion and E-cad/RAC1 signalling, but not on FAK-mediated cell–substrate adhesion.
These findings led us to further examine the reversion of human PSCs, which might require different intracellular signalling compared with mouse PSCs2,3. Similarly, BSCR+ motifs efficiently reverted human PSCs to a naive state (Extended Data Fig. 8a), with enhanced naive markers CD7 and NANOG and reduced prime markers CD24, CD57 and CD90 (refs. 32,33,34) (Extended Data Fig. 8b). Cells were more compacted on BMS than on the plain substrate, showing smaller cell size (Extended Data Fig. 8c) and strongly activated E-cad (Extended Data Fig. 8d). The level of NANOG was significantly reduced on the inhibition of E-cad, RAC1 and YAP, but not affected by FAK inhibition (Extended Data Fig. 8e). These results might indicate the similarity in key components for curvature sensing and naivety reversion between mouse and human PSCs, highlighting the feasibility of BMS for applications in human PSCs.
E-cad/RAC1 signalling can further mediate the activation of YAP35,36,37, which regulates stem cell fate and pluripotency as a mechanotransducer38,39,40. Compared with the plain group, the total YAP level was unaltered, whereas inactive phosphorylated YAP was significantly reduced in BMS–PSCs (Fig. 5a). PCR array analysis showed that YAP downstream target genes were upregulated, whereas upstream suppressor genes were downregulated on BMS (Extended Data Fig. 9a). In particular, phosphorylated AmotL1—an inhibitor of YAP—decreased in BMS–PSCs (Extended Data Fig. 9b). Given the central role of YAP in cell cycle progression40,41, we analysed the cell cycles and found a naivety-featured rapid G1/S transition, a higher proliferation rate and clonogenic potential in BMS–PSCs (Extended Data Fig. 9c–e). The inhibition of RAC1 reduced the YAP activity on BMS (Fig. 5a) and resulted in decreased cell proliferation (Extended Data Fig. 9d), suggesting the involvement of E-cad/RAC1 signalling in YAP activation. The role of YAP on PSC naivety was further validated via the inhibition of YAP, which significantly decreased NANOG and STELLA but upregulated ZIC2 (Fig. 5b and Extended Data Fig. 6c).
Further, we performed chromatin immunoprecipitation (ChIP) polymerase chain reaction (PCR) to gain a better understanding of YAP in the epigenetic regulation of pluripotency genes40,42. BMS–PSCs showed significantly decreased H3K27me3 but increased H3K4me3 occupancy on ‘naive loci’ including Nanog promoter and Pou5f1 distal enhancer. There were increased H3K27me3 on the promoter region of Zic2 in BMS–PSCs (Fig. 5c). YAP inhibition significantly decreased the level of H3K4me3 on Nanog promoter and Pou5f1 distal enhancer, as well as the level of H3K27me3 on Zic2 promoter (Fig. 5c,d), suggesting that YAP activation in BMS–PSCs played a key role in the histone modification of pluripotency genes. These data suggest a mechanistic relationship between BSCR, enhanced E-cad/RAC1 signalling and YAP activation, which consequently increased permissive versus repressive histone patterns in line with a gain of naivety (Fig. 5e).
BSCR stabilizes PSC naivety
To determine how long the mechanical memory and naivety can persist after the removal of BMS, nPSCs were harvested from BMS and reseeded or injected to demonstrate the sustainable effect of BSCR regulation (Fig. 6a). The upregulated E-cad expression and YAP activity of BMS–PSCs remained for at least 15 days after reseeding (Fig. 6b–d). The enhancement in NANOG by BSCR was preserved for at least 10 days (Fig. 6e). The newly formed colonies from BMS–PSCs were more compact, with a higher proliferation rate than those from the plain substrate (Extended Data Fig. 10a,b). Larger embryoid bodies (EBs) were generated from cells preconditioned on BMS (Fig. 6f,g), retaining a higher E-cad level and YAP activity, as well as an accelerated cell cycle progression (Fig. 6h and Extended Data Fig. 10c–e). To exclude the potential influence of EB size on YAP43, we generated EBs with controlled dimensions, and a higher YAP activity was still retained in EBs derived from BMS–PSCs (Fig. 6i). Teratoma formation was performed to determine the in vivo proliferation and pluripotency of PSCs. Although cells from both groups were able to form teratomas containing three germ layers, BMS–PSCs were able to develop into teratomas with larger volume (Fig. 6j,k), suggesting their higher development potential/capacity acquired from BMS40,44.
Outlook
In nature, although naive PSCs only transiently exist in the blastocyst, the topographical features of a blastocyst (for example, Epi–TE curvature) may still provide unlimited inspiration. Here learning from nature, we explored the functionality of topographical structures derived from blastocysts on the reversion of PSCs back to a naive state. We established a method to define specific surface topographical maps, and analysed in situ the complexed microstructures on PSC reversion by correlating with time-resolved microscopy images. We concluded that the curvature constraint in a single direction enabled the successful cell reversion through the activation of cell–cell cohesion, intracellular signalling and epigenetic modulation, without necessarily recapitulating the relative radial symmetry of the blastocyst.
We demonstrated that the reverted naivety of PSCs was sustained for at least 10 days after the removal of BMS. The BMS–PSCs processed the higher development potential in generating EBs and teratomas, compared with cells on non-structured substrates.
The BSCR reported here is inspired and simplified from nature, but goes beyond nature, which provides the design criteria for preparing a functional substrate with the purpose to achieve high sustainability in stem cell applications. Importantly, our results indicated the possibility of the BSCR for reverting human PSCs to a more naive state. Although the underlying mechanism of human PSCs should be further explored, the results highlighted the importance of substrate design in the reversion of human PSC naivety and profound applications such as organoid generation, drug screening, disease models and personalized medicine.
Methods
Ethical compliance
The animal studies were performed by Experimental Pharmacology and Oncology (EPO), Berlin-Buch, under the approval of the Landesamt für Gesundheit und Soziales Berlin, which reviews animal husbandry based on the German Animal Welfare Act (no. H 0023/09) and in compliance with the EU guideline ‘European convention for the protection of vertebrate animals used for experimental and other scientific purposes (ETS 123)’.
Determination of mouse blastocyst geometry
The confocal cross views of the preimplantation blastocysts (E4.25–E4.50) from the literature45,46,47,48,49,50,51,52,53,54,55,56 were imported to ImageJ software with the Bio-Formats Importer plugin (National Institutes of Health). The width of the nPSC layer was measured by detecting the line segments between the two endpoints of Epi in contact with polar TE (Supplementary Fig. 1). The distance from the midpoint of the width line to the Epi–TE interface was defined as a height of the outer layer of Epi. The radii of the arcs, curvatures, arc lengths and cell densities of the outer layer of Epi were calculated based on the following formulas: arc radius = width2/ (8 × height) + height/2; curvature = 1/arc radius; arc length = 2 × radius × arcsin (width/(2 × radius)); cell density = cell number/arc length. The polar TE layer provides a relatively well-defined curvature of 15 to 62 mm–1 (31 ± 8 mm–1, means ± standard deviation (s.d.)), which we refer to as the BSCR.
Defining motifs on cell culture substrates
The scaled three-point approach (Supplementary Methods) was used to measure the curvature of a given point on the BMS in 18 directions. The curvature value of a single direction, κ, and the mean curvature values of 18 directions, 〈κ〉, were used to define the motif regions on the BMS. Here we define a point as BSCR+ if at least one curvature κ for that point is within the BSCR (15 < κ < 62 mm–1). Next, we distinguished regions that have a positive mean curvature 〈κ〉 value (concave) and negative 〈κ〉 value (convex). Between both concave and convex regions, we defined a flat region, with some tolerance for positive and negative average curvature values. The regions were defined in detail as follows: convex (〈κ〉 < −2.5 mm−1), flat (–2.5 < 〈κ〉 < 2.5 mm–1), concave (〈κ〉 > 2.5 mm–1).
Cell culture
The non-viral integrating mouse PSC line (PhiC31, BioCat; reprogrammed from C57BL/6 mouse embryonic fibroblasts (MEFs) with a plasmid encoding OCT4, SOX2, KLF4 and c-Myc) was used for assessing the reversion to naivety. The mouse cell line with NANOG reporter–GFP (iPS-MEF-Ng-492B-4 cells, CiRA, Kyoto University) was used for live-cell tracking. The initial culture of both mouse PSC lines was performed using mitomycin-C-treated feeder MEFs (BioCat) and 0.2% gelatin-coated culture plate, in feeder-dependent PSC culture medium (KnockOut DMEM basal medium containing 15.0% KnockOut Serum Replacement, 1.0% MEM Non-Essential Amino Acids, 1.0% GlutaMAX-I, 0.1% 2-mercaptoethanol and 10 ng ml–1 mouse LIF; Life Technologies). Undifferentiated PSCs were isolated using magnetically activated cell sorting technology with Feeder Removal MicroBeads and Pluripotent Stem Cell Isolation Kit (Miltenyi Biotec); PSCs were then maintained in ESGRO-2i/L medium (containing GSK3β and MEK 1/2 inhibitors and 0.5 × 106 U ml–1 mouse LIF, Merck Chemicals), and cultured on 9 µg ml–1 Cultrex Mouse Laminin (Bio-Techne)-coated six-well tissue culture plates. The PSCs were chemically adapted for at least three passages to the primed-like state by using 1i medium containing GSK3β inhibitor (3 µM CHIR99021, Stemgent) and AXIN stabilizer (2 µM XAV-939, Stemgent)3. The 2i/L-PSCs and 1i-PSCs maintained in the tissue culture plates were used as positive and negative controls for naive cells, respectively (Supplementary Fig. 4a). The culture media were changed daily and all the PSCs under the aforementioned culture conditions were used for experiments before reaching passage 50. All the PSCs applied for the experiments detected negative for mycoplasma using MycoFluor Mycoplasma Detection Kit (Thermo Fisher Scientific). The morphology of the PSCs under different culture conditions was monitored; compared with the 2i/L-PSCs, the 1i-PSCs presented primed-like characteristics, including the elongated cell shape and flattened colonies (Supplementary Fig. 4b), as well as decreased levels of NANOG reporter–GFP (the MFI of GFP on plain substrates was set as the threshold) and NANOG proteins (Supplementary Fig. 4c,d).
Human PSC line (BIHi001-A) was a gift from the Stem Cell Core Facility, Berlin Institute of Health. This exogene-integration-free cell line was generated using Sendai virus vectors. Detailed information is available in the hPSCreg database (https://hpscreg.eu/cell-line/BIHi001-A). Human PSCs were maintained on Geltrex (Thermo Fisher Scientific)-coated TCP under feeder-free conditions using Essential 8 medium (Thermo Fisher Scientific). The human PSCs were adapted for ten passages to a naive-like stage using RseT medium (STEMCELL Technologies) in the presence of irradiated CF1 MEFs (Thermo Fisher Scientific) under hypoxic condition (5% O2). The RseT–human PSCs were set as a positive control for human PSC analysis.
Validation of motif function on PSC naivety reversion
The bright-field microscopy images of the BMS were first recognized to be precisely overlaid onto the micro-CT scanning images. The defined BSCR+ area and the concave, flat and convex motifs based on the micro-CT scanning and curvature calculation were then mapped and overlaid with the microscopy images to evaluate the effect of geometrical motifs on cells. In brief, the maximum-intensity-projection confocal laser scanning microscopy (CLSM) images of cells and the motif maps in the same region were imported into Photoshop software (CS6, Adobe Systems), and the motif maps were used as a mask to study the cellular behaviour and parameters on defined areas, including cell size, density, distribution, migration and naive marker level.
Live-cell tracking
NANOG reporter–GFP integrated in iPS-MEF-Ng-492B-4 cells (CiRA, Kyoto University) was detected by an argon channel (488 nm excitation) to track the living-mouse PSCs21. To monitor naivety reversion and cell migration, the real-time fluorescence images of PSCs and bright-field images of the BMS surface in the Z stack were acquired using CLSM (Carl Zeiss). PSC expansion on BMS in a single colony were recorded at 20 min intervals using a time-lapse fluorescent microscope IX81 with xCellence/cell^R software (Olympus).
For image analysis, the maximum intensity projection was first performed for all the Z-stack CLSM images. The GFP intensity of cells was analysed using ImageJ software (National Institutes of Health). The maximum GFP intensity of the cells on plain substrates was set as a threshold. On BMS, the cells with higher intensity than the threshold were defined as GFPhi naive PSCs. MountainsMap (Digital Surf, Besancon) and Spyder (Python 3.8, Anaconda) were then used to analyse the effect of motifs on GFPhi cell occupancy and naivety level in a time-resolved manner.
E-cad, RAC1, FAK and YAP activity and inhibition
To examine the distribution and expression of E-cad and F-actin with respect to motifs, cells were fixed, permeabilized and blocked using Image-iT Fixation/Permeabilization Kit (Life Technologies). The anti-E-cad-PE antibody (rabbit monoclonal; 1:10; New England Biolabs) was used for labelling E-cad. F-actin was detected using ActinRed 555 ReadyProbes Reagent (Life Technologies). The CLSM images of the samples were then overlaid with the motif maps following the method shown in Supplementary Fig. 4.
To quantify the E-cad level, 1 × 106 cells were freshly harvested from the substrates at the indicated time points and then immediately processed for anti-E-cad-PE antibody (rabbit monoclonal; 1:50; New England Biolabs) staining in the dark at room temperature for 30 min. The labelled cells were measured by flow cytometry (MACSQuant, Miltenyi Biotec) and analysed using FlowJo software (version 10.4, Tree Star Inc.).
The concentration of phosphorylated FAK and total FAK in the cell extract were measured using the pFAK (pY397)/tFAK enzyme-linked immunosorbent assay (ELISA) kits (Thermo Fisher Scientific). The same amount of total protein determined using a BCA protein assay kit (Thermo Fisher Scientific) was applied for ELISA.
For the quantification of YAP and AMOTL1 phosphorylation levels in PSCs, day-3 cells growing on different substrates were harvested, fixed with 4% paraformaldehyde and permeabilized with prechilled 90% methanol for 30 min on ice, then stained with anti-YAP-Alexa Fluor 647 and anti-phospho-YAP (S127) (rabbit monoclonal; 1:50; New England Biolabs) or anti-phospho-AMOTL1 (S262) antibodies (rabbit polyclonal; 1:100; Covalab Biotechnology) for 45 min. The cells were then stained with secondary anti-rabbit IgG (H + L)-Alexa Fluor 647 and anti-rabbit IgG (H + L) Alexa Fluor 488 antibodies (goat polyclonal; 1:500; New England Biolabs) for 30 min, followed by flow cytometry analysis.
Inhibition experiments were performed by treating the cells using the RAC1 activity inhibitor NSC 23766 (50 µM; Bio-Techne), the selective FAK inhibitor PF-573228 (10 µM; Sigma-Aldrich) and the YAP activation inhibitor Verteporfin (20 nM; Sigma-Aldrich). For E-cad neutralization, cells were treated with E-cad antibody (25 µg ml–1; Merck) for 24 h before harvesting.
ChIP PCR
ChIP was performed using the SimpleChIP Plus Enzymatic Chromatin IP Kit (New England Biolabs) following the given instruction. ChIP employed anti-H3, anti-H3K27me3 and anti-H3K4me3 antibodies (rabbit monoclonal; 1:50; New England Biolabs) with magnetic beads. After washes, the samples were eluted and treated with RNase and proteinase K overnight. The released DNA was used for real-time PCR quantification. Enrichment was calculated relative to the input DNA data and expressed as per cent: input = 10% × 2(CT 10% input sample – CT ChIP sample) and then normalized to the total H3 enrichment level. Normal rabbit IgG (1:50; New England Biolabs) was used as an isotype control. The oligonucleotide sequences were Nanog promoter forward primer 5′-GGAGAATAGGGGGTGGGTAG and reverse 5′-CAGCCTTCCCACAGAAAGAG. Zic2 promoter forward primer 5′- TGGACTCTTCTCCTCCTCCA and reverse 5′-GGTGGAAATACTGGCGACTG. Pou5f1 distal enhancer forward primer 5′- GGCTGCAGGCATACTTGAAC and reverse 5′-AGGGCAGAGCTATCATGCAC.
Stability of naivety of BMS–PSCs
PSCs were cultured on plain substrate and BMS for 5 days and then harvested and reseeded to evaluate the stability of naivety. On laminin-coated TCP, the cells were seeded at a density of 2 × 104 cm–2 and cultured in a monolayer for four passages, followed by anti-E-cad-PE (rabbit monoclonal; 1:10; New England Biolabs), anti-YAP-Alexa Fluor 488 (rabbit monoclonal; D8H1X; 1:100; New England Biolabs) and anti-NANOG-APC (recombinant human monoclonal; 1:11; Miltenyi Biotec) staining. The samples were then examined with a CLSM (LSM780, Carl Zeiss) with Zen 2012 software (Carl Zeiss) and flow cytometry.
EBs were obtained by reseeding the cells (2 × 104 cm–2) on uncoated TCP, and cultured in complete KnockOut Serum Replacement EB medium (Thermo Fisher Scientific). To form EBs with uniform dimensions, a 96-well V-bottom plate (Sigma-Aldrich) was used. The generated EBs were fixed, permeabilized and incubated with anti-YAP (rabbit monoclonal; D8H1X; 1:100; New England Biolabs) overnight at 4 °C. The secondary anti-rabbit IgG (H + L)-Alexa Fluor 647 or anti-rabbit IgG (H + L)-Alexa Fluor 488 antibodies (goat polyclonal; 1:500; New England Biolabs) were used for labelling. After 1 h of culture at room temperature, the fluorescence was detected and imaged by LSM780 (Carl Zeiss) with Zen 2012 and Zen 2.3 software (Carl Zeiss). The YAP phosphorylation level was examined using western blot (Supplementary Methods).
The teratoma formation experiment was performed by EPO and carried out in accordance with the German Animal Protection Law and approved by the local responsible authorities. EPO complies with the EU guideline ‘European convention for the protection of vertebrate animals used for experimental and other scientific purposes (ETS 123)’. Animals were handled according to the ‘Regulation on the protection of experimental scientific purposes or other purposes used animals’. Compliance with the above rules and regulations is monitored by the Landesamt fuer Gesundheit und Soziales, which is the responsible regulatory authority monitoring the animal husbandry based on the German Animal Welfare Act. Approval (code H 0023/09) was given after careful inspection of the site including bedding, feeding and water, ventilation, temperature and humidity, and cleaning and hygiene concepts. The day-5 PSCs derived from plain substrates and BMSs were subcutaneously injected into eight-week-old male NSG immunodeficient mice (1 × 106 PSCs for each mouse). The teratoma diameter measurements were performed twice weekly until the end day of the study when teratoma reached 1.5 cm3. Teratoma volume was calculated using the following formula: volume = width2 × length/2. The dissected teratomas were cryoconserved or formalin fixed for haematoxylin and eosin staining.
Statistical analysis
Statistical analysis for biological evaluation was performed using Prism 8.0 (GraphPad) software. Comparisons between two experimental groups or the same group with and without inhibitors were analysed using two-tailed unpaired sample Student’s t-tests. Before performing the Student’s t-test, the homogeneity of variance for the two groups was verified by an F test. The differences among three or more independent groups were analysed using one- or two-way analysis of variance (ANOVA) followed by Bonferroni’s multiple comparisons test. In particular, the two-way ANOVA was applied to analyse the individual effects of motifs and BSCR counts on regulating cell distribution, proliferation and naive markers, as well as their overall effects over time. The sample sizes for each experimental group and the statistical test used to determine significances among groups are reported in the figure legends. Briefly, for the following in vitro and in vivo studies, statistical analyses were applied to biologically independent samples (separate batches/plates of cells, cell lysates and immunodeficient mice) across plain substrate and BMS: quantification of naive-pluripotency-associated markers, E-cad, FAK, YAP, histone H3 modification at naive gene loci, cell death, migration, cell cycle sub-phases, cell expansion and teratoma formation. For image-based analysis with ImageJ software (version 2.0.0-rc-65/1.52b, National Institutes of Health), the numbers of images, cell nuclei and colonies were indicated in the corresponding figure legends. In all the cases, P values of less than 0.05 were considered statistically significant and the exact P values are indicated in the figures. Unless indicated otherwise, the quantification data are presented as means ± s.d.
Reporting summary
Further information on research design is available in the Nature Portfolio Reporting Summary linked to this article.
Data availability
All data supporting the findings of this study are available within the Article and its Supplementary Information. Source data are available via Figshare at https://doi.org/10.6084/m9.figshare.25827706 as well as are provided with this Article.
Code availability
The custom code written for this paper uses Python 3.8 and the accompanying packages numpy, numpy-stl, matplotlib, scipy and pandas were used to analyse the different substrates mentioned in the paper. The code scripts are available via Zenodo at https://doi.org/10.5281/zenodo.10732261 (ref. 57).
References
Nichols, J. & Smith, A. I. Naive and primed pluripotent states. Cell Stem Cell 4, 487–492 (2009).
Fu, J., Warmflash, A. & Lutolf, M. P. Stem-cell-based embryo models for fundamental research and translation. Nat. Mater. 20, 132–144 (2021).
Davidson, K. C., Mason, E. A. & Pera, M. F. The pluripotent state in mouse and human. Development 142, 3090–3099 (2015).
Weinberger, L., Ayyash, M., Novershtern, N. & Hanna, J. H. Dynamic stem cell states: naive to primed pluripotency in rodents and humans. Nat. Rev. Mol. Cell Biol. 17, 155–169 (2016).
Dong, C. et al. Derivation of trophoblast stem cells from naive human pluripotent stem cells. eLife 9, e52504 (2020).
Dong, C., Fischer, L. A. & Theunissen, T. W. Recent insights into the naive state of human pluripotency and its applications. Exp. Cell. Res. 385, 111645 (2019).
Gafni, O. et al. Derivation of novel human ground state naive pluripotent stem cells. Nature 504, 282–286 (2013).
Takashima, Y. et al. Resetting transcription factor control circuitry toward ground-state pluripotency in human. Cell 158, 1254–1269 (2014).
Shao, Y. et al. Self-organized amniogenesis by human pluripotent stem cells in a biomimetic implantation-like niche. Nat. Mater. 16, 419–425 (2017).
Boroviak, T. & Nichols, J. The birth of embryonic pluripotency. Phil. Trans. R. Soc. B 369, 20130541 (2014).
Biggins, J. S., Royer, C., Watanabe, T. & Srinivas, S. Towards understanding the roles of position and geometry on cell fate decisions during preimplantation development. Semin. Cell Dev. Biol. 47–48, 74–79 (2015).
Gardner, R. L. The early blastocyst is bilaterally symmetrical and its axis of symmetry is aligned with the animal-vegetal axis of the zygote in the mouse. Development 124, 289–301 (1997).
Schamberger, B. et al. Curvature in biological systems: its quantification, emergence, and implications across the scales. Adv. Mater. 35, e2206110 (2023).
Bäckemo, J., Heuchel, M., Reinthaler, M., Kratz, K. & Lendlein, A. Predictive topography impact model for Electrical Discharge Machining (EDM) of metal surfaces. MRS Adv. 5, 621–632 (2019).
Matsuda, K. et al. ChIP-seq analysis of genomic binding regions of five major transcription factors highlights a central role for ZIC2 in the mouse epiblast stem cell gene regulatory network. Development 144, 1948–1958 (2017).
Kim, E. & Hyun, S. H. Apoptosis in porcine pluripotent cells: from ICM to iPSCs. Int. J. Mol. Sci. 17, 1533 (2016).
Bahat, A. & Gross, A. Mitochondrial plasticity in cell fate regulation. J. Biol. Chem. 294, 13852–13863 (2019).
Liu, J. C. et al. High mitochondrial priming sensitizes hESCs to DNA-damage-induced apoptosis. Cell Stem Cell 13, 483–491 (2013).
Pavel, M. et al. Contact inhibition controls cell survival and proliferation via YAP/TAZ-autophagy axis. Nat. Commun. 9, 2961 (2018).
Vicar, T., Raudenska, M., Gumulec, J. & Balvan, J. The quantitative-phase dynamics of apoptosis and lytic cell death. Sci. Rep. 10, 1566 (2020).
Okita, K., Nakagawa, M., Hong, H. J., Ichisaka, T. & Yamanaka, S. Generation of mouse induced pluripotent stem cells without viral vectors. Science 322, 949–953 (2008).
Chen, C. et al. Translational and post-translational control of human naive versus primed pluripotency. iScience 25, 103645 (2022).
Coronado, D. et al. A short G1 phase is an intrinsic determinant of naive embryonic stem cell pluripotency. Stem Cell Res 10, 118–131 (2013).
Hu, Z. et al. Generation of naivetropic induced pluripotent stem cells from Parkinson’s disease patients for high-efficiency genetic manipulation and disease modeling. Stem Cells Dev. 24, 2591–2604 (2015).
Zenker, J. et al. Expanding actin rings zipper the mouse embryo for blastocyst formation. Cell 173, 776–791e717 (2018).
Bergert, M. et al. Cell surface mechanics gate embryonic stem cell differentiation. Cell Stem Cell 28, 209–216e204 (2021).
Sharaireh, A. M., Fitzpatrick, L. M., Ward, C. M., McKay, T. R. & Unwin, R. D. Epithelial cadherin regulates transition between the naive and primed pluripotent states in mouse embryonic stem cells. Stem Cells 38, 1292–1306 (2020).
Nakagawa, M., Fukata, M., Yamaga, M., Itoh, N. & Kaibuchi, K. Recruitment and activation of Rac1 by the formation of E-cadherin-mediated cell-cell adhesion sites. J. Cell Sci. 114, 1829–1838 (2001).
Liu, W. F., Nelson, C. M., Pirone, D. M. & Chen, C. S. E-cadherin engagement stimulates proliferation via Rac1. J. Cell Biol. 173, 431–441 (2006).
Shibata, K. et al. Rac1 regulates myosin II phosphorylation through regulation of myosin light chain phosphatase. J. Cell. Physiol. 230, 1352–1364 (2015).
Broaders, K. E., Cerchiari, A. E. & Gartner, Z. J. Coupling between apical tension and basal adhesion allow epithelia to collectively sense and respond to substrate topography over long distances. Integr. Biol. 7, 1611–1621 (2015).
Shakiba, N. et al. CD24 tracks divergent pluripotent states in mouse and human cells. Nat. Commun. 6, 7329 (2015).
Collier, A. J. et al. Comprehensive cell surface protein profiling identifies specific markers of human naive and primed pluripotent states. Cell Stem Cell 20, 874–890e877 (2017).
Trusler, O., Huang, Z., Goodwin, J. & Laslett, A. L. Cell surface markers for the identification and study of human naive pluripotent stem cells. Stem Cell Res 26, 36–43 (2018).
Sabra, H. et al. β1 integrin-dependent Rac/group I PAK signaling mediates YAP activation of Yes-associated protein 1 (YAP1) via NF2/merlin. J. Biol. Chem. 292, 19179–19197 (2017).
Aragona, M. et al. A mechanical checkpoint controls multicellular growth through YAP/TAZ regulation by actin-processing factors. Cell 154, 1047–1059 (2013).
Benham-Pyle, B. W., Pruitt, B. L. & Nelson, W. J. Mechanical strain induces E-cadherin–dependent Yap1 and β-catenin activation to drive cell cycle entry. Science 348, 1024–1027 (2015).
Yang, C., Tibbitt, M. W., Basta, L. & Anseth, K. S. Mechanical memory and dosing influence stem cell fate. Nat. Mater. 13, 645–652 (2014).
Dupont, S. et al. Role of YAP/TAZ in mechanotransduction. Nature 474, 179–183 (2011).
Lian, I. et al. The role of YAP transcription coactivator in regulating stem cell self-renewal and differentiation. Gene Dev. 24, 1106–1118 (2010).
Aksoy, I. et al. Apoptosis, G1 phase stall, and premature differentiation account for low chimeric competence of human and rhesus monkey naive pluripotent stem cells. Stem Cell Rep. 16, 56–74 (2021).
Oh, H. et al. Yorkie promotes transcription by recruiting a histone methyltransferase complex. Cell Rep. 8, 448–458 (2014).
Zhao, B., Li, L., Lei, Q. & Guan, K. L. The Hippo-YAP pathway in organ size control and tumorigenesis: an updated version. Genes Dev. 24, 862–874 (2010).
Chan, Y. S. et al. Induction of a human pluripotent state with distinct regulatory circuitry that resembles preimplantation epiblast. Cell Stem Cell 13, 663–675 (2013).
Acampora, D. et al. Loss of the Otx2-binding site in the nanog promoter affects the integrity of embryonic stem cell subtypes and specification of inner cell mass-derived epiblast. Cell Rep. 15, 2651–2664 (2016).
Chen, Y. H. & Yu, J. Ectopic expression of Fgf3 leads to aberrant lineage segregation in the mouse parthenote preimplantation embryos. Dev. Dyn. 241, 1651–1664 (2012).
Dietrich, J. E. et al. Venus trap in the mouse embryo reveals distinct molecular dynamics underlying specification of first embryonic lineages. EMBO Rep. 16, 1005–1021 (2015).
Grabarek, J. B. et al. Differential plasticity of epiblast and primitive endoderm precursors within the ICM of the early mouse embryo. Development 139, 129–139 (2012).
Kang, M. J., Piliszek, A., Artus, J. & Hadjantonakis, A. K. FGF4 is required for lineage restriction and salt-and-pepper distribution of primitive endoderm factors but not their initial expression in the mouse. Development 140, 267–279 (2013).
Kim, S. et al. PRMT5 protects genomic integrity during global DNA demethylation in primordial germ cells and preimplantation embryos. Mol. Cell 56, 564–579 (2014).
Papanayotou, C. et al. A novel nodal enhancer dependent on pluripotency factors and Smad2/3 signaling conditions a regulatory switch during epiblast maturation. PLoS Biol. 12, e1001890 (2014).
Plusa, B., Piliszek, A., Frankenberg, S., Artus, J. & Hadjantonakis, A. K. Distinct sequential cell behaviours direct primitive endoderm formation in the mouse blastocyst. Development 135, 3081–3091 (2008).
Smith, E. R. et al. Nuclear envelope structural proteins facilitate nuclear shape changes accompanying embryonic differentiation and fidelity of gene expression. BMC Cell Biol. 18, 8 (2017).
Strumpf, D. et al. Cdx2 is required for correct cell fate specification and differentiation of trophectoderm in the mouse blastocyst. Development 132, 2093–2102 (2005).
Thomas, F. C. et al. Contribution of JAM-1 to epithelial differentiation and tight-junction biogenesis in the mouse preimplantation embryo. J. Cell Sci. 117, 5599–5608 (2004).
Wicklow, E. et al. HIPPO pathway members restrict SOX2 to the inner cell mass where it promotes ICM fates in the mouse blastocyst. PLoS Genet. 10, e1004618 (2014).
Bäckemo, J. D. V. & Heuchel, M. Three-point curvature analysis. Zenodo https://doi.org/10.5281/zenodo.10732261 (2024).
Acknowledgements
We acknowledge R. Bendisch and M. Rettschlag for the fabrication of sterilized BMS, Y. Pieper and M. Keller for technical support, M. Gossen for critical reading and editing our manuscript and I. Ullah and R. Machatschek for critically reading our manuscript. This work was financially supported by the Helmholtz Association of German Research Centers through program-oriented funding (X.X., Weiwei Wang, Y.L., J.B., M.H., Y.N., I.I., K.K., A.L. and N.M.); I2B Funds ‘high-resolution imaging and computational analysis to study the dynamics of stem cell-biomaterial interaction’ (received by X.X., Weiwei Wang, I.I. and N.M.); Helmholtz Imaging Project ‘AIOrganoid’ (received by X.X., Weiwei Wang, Y.N. and N.M.); Helmholtz Cross Programme Initiative ‘Technology and Medicine—Adaptive Systems’ (received by N.M.); Helmholtz Graduate School for Macromolecular Bioscience (MacroBio, grant no. VH-GS-503, received by N.M.); Helmholtz Virtual Institute, Multifunctional Biomaterials for Medicine (grant no. VH-VI-423, received by A.L. and N.M.); and the Federal Ministry of Education and Research, Germany, for funding through the Program Health Research (grant no. 13GW0098 and 13GW0099, received by N.M.). Wei Wang is grateful for support from the China Scholarship Council (no. 202206170040).
Funding
Open access funding provided by Helmholtz-Zentrum hereon GmbH (4216).
Author information
Authors and Affiliations
Contributions
X.X., Weiwei Wang, N.M. and A.L. designed the experiments. X.X. and Weiwei Wang performed the in vitro PSC experiments, immunostaining, fluorescence-activated cell sorting and western blotting; generated ChIP PCR; and analysed the gene expression array and in vivo teratoma formation data. J.B., K.K. and A.L. designed the BMS. X.X., Weiwei Wang, Y.L. and N.M. designed and fabricated the microbowl and microgroove substrates. J.B., M.H. and K.K. performed the experimental surface characterization and metrological analysis of BMS and electrical-discharge-machining-finished steel cylinders. X.X., Weiwei Wang, Y.L., J.B., M.H., Wei Wang, Y.N., I.I., K.K., N.M. and A.L. analysed the data and wrote the manuscript. N.M. and A.L. supervised the project. All authors edited and approved the final manuscript.
Corresponding authors
Ethics declarations
Competing interests
A.L. and K.K. are inventors of a patent (WO 2011/054601 (A1)), related to cell culture inserts. X.X., Weiwei Wang and N.M. are inventors on a provisional patent application (EP 24160389.3 and 24161228.2) covering the biophysical methods for naivety reversion. The remaining authors declare no competing interests.
Peer review
Peer review information
Nature Materials thanks Milica Radisic and the other, anonymous, reviewer(s) for their contribution to the peer review of this work.
Additional information
Publisher’s note Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Extended data
Extended Data Fig. 1 Enhanced naivety of PSCs on BMS.
a. Real-time PCR array analysis of naive and primed pluripotent gene expression in PSCs on different substrates. b. Representative fluorescence microscopic image of SSEA1 (green) and nuclei (blue) of PSCs on Plain and BMS substrate (Images represent the results from 6 independent experiments; Scale bar, 100 μm). c. Quantitative analysis of the percentage of SSEA1+/SSEA4– cell population in day-5 PSCs on different substrates (n = 6 biologically independent experiments; Data presented as means ± SD.; Statistical significance was calculated via two-tailed Student’s t test). d. Heat map of real-time PCR array screening of germ layer differentiation associated genes in day-5 PSCs. Percentage of NANOGhi (e) and STELLAhi (f) PSC covered area compared to the whole area of corresponding motifs (n = 4; Statistical significance was calculated via two-way ANOVA with Bonferroni’s multiple comparisons test). g. Analysis of intensity of NANOG in cells located in BSCR+ areas with different counts. h. Comparison of NANOG intensity of PSCs located in Concave+ and Concave- motifs with one out of 18 directions fitting BSCR (BSCR 1) and without BSCR (BSCR 0). i. Quantification of the proportion of NANOGhi cells located at BSCR 0 and BSCR 1 areas in and outside Concave motifs (n = 4 biologically independent experiments; Data presented as means ± SD.; Statistical significance was calculated via two-tailed Student’s t test).
Extended Data Fig. 2 Primed-to-naive reversion of PSCs on BMS.
Representative images and quantitative analysis of PSC necrosis (a) and apoptosis (b) in BSCR+ and BSCR- areas at day 0 (9 h post cell seeding; n = 8 images for necrosis and n = 10 images for apoptosis) and day 3 (n = 10 images for necrosis and n = 5 images for apoptosis; Data presented as means ± SD.; Statistical significance was calculated via two-tailed Student’s t test; N.S. statistically non-significance). c. Detection of NANOG-GFPhi nPSCs on BSCR+ area of BMS from 12 h to 39 h post cell seeding. d. Expansion of NANOG-GFPhi nPSCs from BSCR+ to BSCR- areas. The outlines of BSCR+ areas were illustrated with yellow dash lines. Images in a-d represent the results from 3 independent experiments. Scale bar, 100 μm.
Extended Data Fig. 3 PSC naivety and proliferation activity on different substrates.
Representative fluorescence images of Ki67+ proliferating cells, NANOG-GFPhi naive PSCs in BSCR+ and BSCR- areas of BMS, and on Plain substrate (Images represent the results from 3 independent experiments. Scale bar, 100 μm). The outlines of BSCR+ areas were illustrated with yellow dash lines. 1i-PSCs, Mixed PSCs (Naive:Primed = 1:4 in number) were maintained under primed PSC culture condition in 1i medium. 2i/L-PSCs used as positive control were cultured under naive PSC culture condition in 2i/L medium.
Extended Data Fig. 4 Quantification of PSC naivety and proliferation activity on different substrates.
The NANOG-GFPhi proportion, cell density and percentage of Ki67+ proliferating cells in BSCR + , BSCR- areas of BMS and on Plain substrate within 48 h post cell seeding were quantified based on the fluorescence images from 5 biologically independent experiments (For GFPhi cell analysis over time, 1i-PSCs group: nBSCR+=35, nBSCR-=26, nPlain = 25; Mixed PSCs group: nBSCR+=27, nBSCR-=26, nPlain = 26; 2i/L-PSCs group: n = 25. For cell density analysis, 1i-PSCs group: nBSCR+=44, nBSCR-=32, nPlain = 30; Mixed PSCs group: nBSCR+=33, nBSCR-=33, nPlain = 32; 2i/L-PSCs group: n = 30. For Ki67+ cell analysis, 1i-PSCs group: nBSCR+=35, nBSCR-=26, nPlain = 25; Mixed PSCs group: nBSCR+=27, nBSCR-=27, nPlain = 26; 2i/L-PSCs group: n = 25. Data presented as means ± SD.; Statistical significance was calculated via two-way ANOVA with Bonferroni’s multiple comparisons test, for effect of BSCR; *BSCR+ vs. Plain, #BSCR+ vs. BSCR-).
Extended Data Fig. 5 Topography of microbowl and microgroove substrates.
a. Height map reconstruction of substrates based on optical profilometry analysis. b. Representative scanning electron microscopic images of microstructured substrates. Confocal microscopic images of tilted-view (c) and side-view (d) of laminin-coated microbowls and microgrooves (Images represent the results from 3 independent experiments; Scale bar, 200 μm).
Extended Data Fig. 6 Primed-to-naive reversion and expansion of PSCs on substrates with uniform motifs.
a. Representative confocal microscopic images of NANOG-GFP PSCs on microbowls and microgrooves with different BSCR counts at day 3 after cell seeding (Fluorescence images represent the results from 3 independent experiments; Scale bar, 100 μm). b. Quantification of the area fraction of GFPhi in GFP+ cells and MFI of GFP in PSCs growing on microbowls and microgrooves with different BSCR counts for 3 days (n = 5 biologically independent experiments; Data presented as means ± SD.; Statistical significance was calculated via two-way ANOVA with Bonferroni’s multiple comparisons test, for effects of BSCR and time). c. the area fraction of GFPhi in GFP+ cells and MFI of GFP in day-3 PSCs on BSCR+ microbowls with and without E-cad, RAC1 and YAP inhibition (n = 5 biologically independent experiments; Data presented as means ± SD.; Statistical significance was calculated via one-way ANOVA with Bonferroni’s multiple comparisons). d. Quantification of fraction and MFI of SSEA1+ PSCs on BSCR+ and BSCR- microbowls (n = 5 biologically independent experiments; Data presented as means ± SD.; Statistical significance was calculated via one-way ANOVA with Bonferroni’s multiple comparisons). e. Representative confocal images showing the alteration of TBX3 and STELLA in PSCs on microbowls with different BSCR counts (Images represent the results from 3 independent experiments; Scale bar, 50 μm). Cell cycle progression patterns of day-1 (f) and day-3 (g) PSCs on microbowls with different BSCR counts (n = 3 biologically independent experiments; Data presented as means ± SD.; Statistical significance was calculated via two-way ANOVA with Bonferroni’s multiple comparisons test). h. Expansion curves of PSCs on microbowls with different BSCR counts over time (n = 3 biologically independent experiments; Data presented as means ± SD.; Statistical significance was calculated via two-way ANOVA with Bonferroni’s multiple comparisons test, for effects of BSCR and time).
Extended Data Fig. 7 PSC compaction on BMS.
a. Representative top- and side-view of confocal microscopic images and quantitative analysis showing spatial organization of PSC colonies in crater and channel like areas on BMS. Cell nuclei were stained with DAPI (Data acquired from 9 biologically independent experiments; BSCR+ Crater: n = 9, BSCR+ Channel: n = 90, BSCR- Channel: n = 36; Data presented as means ± SD.; Statistical significance was calculated via one-way ANOVA with Bonferroni’s multiple comparisons test). b. Imaging analysis of cell density, top-view colony area, and flow cytometry analysis of relative cell size (Cell density: ncolony=15 and 20 for Plain and BMS substrate from 10 biologically independent samples; Colony area: n = 8 and 22, ncolony=53 and 91 for Plain and BMS substrate from 10 biologically independent samples, respectively; Cell size: n = 3 biologically independent samples; Data presented as means ± SD.; Statistical significance was calculated via two-tailed Student’s t test; N.S., non-significance). c. The flow cytometry analysis of NANOG of day-5 PSCs on Plain substrates with and without high cell seeding density (5×104 /cm2). TCP, 2i/L control was set as 1 (n = 3 biologically independent experiments; Data presented as means ± SD.; Statistical significance was calculated via one-way ANOVA with Bonferroni’s multiple comparisons test; N.S., non-significance). Representative confocal images and (d) the quantification of pMLC2 level (e) in PSC colonies at BSCR+ and BSCR- areas (Scale bar, 100 μm; ncolony = 19 from 3 biologically independent experiments; Data presented as means ± SD.; Statistical significance was calculated via two-tailed Student’s t test). f. Comparison of pMLC2 levels of cells using flow cytometry (n = 3 biologically independent experiments; Data presented as means ± SD.; Statistical significance was calculated via two-tailed Student’s t test). g. Contractility assay based on the PSCs/collagen gel size measurement. The cells were harvested from Plain and BMS with/without E-cad inhibition (n = 3 biologically independent experiments; Data presented as means ± SD.; Statistical significance was calculated via two-tailed Student’s t test).
Extended Data Fig. 8 Primed-to-naive reversion of human PSCs on BMS.
a. Representative bright field (BF) image and laser scanning microscopic images of human PSCs on BMS at day 5. The nuclei (blue) were stained with Hoechst 33342 to visualize the whole population, and the NANOGhi (green) and STELLAhi (red) cells were filtered out to show the naive population (Images represent the results from 3 independent experiments; Scale bars, 100 μm). b. Flow cytometry analysis of naive markers (CD7, NANOG) and primed markers (CD24, CD57, CD90) of day-8 human PSCs adapted in RSeT medium on TCP (Pos ctrl), on Plain and BMS substrates in IPS-brew medium (n = 3 biologically independent experiments; Data presented as means ± SD.; Statistical significance was calculated via one-way ANOVA with Bonferroni’s multiple comparisons test). c. Flow cytometry analysis of relative cell size (based on FSC-A value) of human PSCs cultured on Plain and BMS surfaces (n = 6 biologically independent experiments; Data presented as means ± SD.; Statistical significance was calculated via two-tailed Student’s t test). d. Quantitative analysis of E-cad+ fractions and E-cad expression levels of cells under different culture conditions (n = 3 biologically independent experiments; Data presented as means ± SD.; Statistical significance was calculated via one-way ANOVA with Bonferroni’s multiple comparisons test). e. The level of NANOG in day-8 human PSCs on BMS with and without E-cad, RAC1 and FAK inhibition. The level of untreated PSCs was set as 1 (n = 3 biologically independent experiments; Data presented as means ± SD.; Statistical significance was calculated via one-way ANOVA with Bonferroni’s multiple comparisons).
Extended Data Fig. 9 YAP activation and cell proliferation in response to culture on a BMS substrate.
a. Real-time PCR array analysis of YAP signaling suppressor and target genes in day-3 PSCs from Plain and BMS substrates. b. The representative flow cytometry dot plots showing the phosphorylated level of AmotL1 (S262) in PSCs in the indicated conditions. c. Percentage of cells in the phases of cell cycle on Plain and BMS substrates (n = 3 biologically independent experiments; Data presented as means ± SD.; Statistical significance was calculated via two-way ANOVA with Bonferroni’s multiple comparisons test). d. Growth curve of PSCs on Plain and BMS substrates, with and without RAC1 inhibition (n = 9 biologically independent experiments; Data presented as means ± SD.; Statistical significance was calculated via two-way ANOVA with Bonferroni’s multiple comparisons test; N.S, non-significance). e. Alkaline phosphatase staining of PSCs on Plain and BMS at indicated time points. Image represents the results from 3 biologically independent experiments. Scale bar, 5 mm.
Extended Data Fig. 10 BMS stabilizes naive pluripotency of PSCs.
a. Morphology of reseeded PSCs pre-conditioned on different substrates. Scale bar, 50 μm. b. Proliferation curves of reseeded PSCs. n = 4 biologically independent experiments; Data presented as means ± SD.; Statistical significance was calculated via two-way ANOVA for main effect of substrate. Western blot analysis of E-cad (c), pYAP (at S127) and tYAP levels (d) in day-3 EBs formed by reseeding PSCs pre-conditioned on Plain (Plain-Re) and BMS (BMS-Re) substrates. GAPDH was used as a loading control. e. Cell cycle progression patterns of day-3 EBs (n = 3 biologically independent experiments; Data presented as means ± SD.; Statistical significance was calculated via two-way ANOVA with Bonferroni’s multiple comparisons test).
Supplementary information
Supplementary Information
Supplementary Figs. 1–8, Tables 1–3, captions for Videos 1 and 2, Methods and references.
Supplementary Video 1
Migration of whole population of PSCs on BMS from day 0 (12 h) to day 1 (25 h). The iPS-MEF-Ng-492B-4 mouse PSC line with NANOG reporter–GFP was used for live-cell tracking. Images were recorded within 60 min intervals. The outlines of BSCR+ areas are illustrated with dashed yellow lines. The video was exported and played at 5 fps. Scale bar, 100 μm.
Supplementary Video 2
Migration of NANOG-GFPhi naive PSCs on BMS from day 1 (36.0 h) to day 3 (73.5 h). The iPS-MEF-Ng-492B-4 mouse PSC line with NANOG reporter–GFP was used for live naive PSCs tracking. GFPhi naive PSCs were discriminated using a fluorescence threshold based on the plain control. Images were recorded within 90 min intervals. The outlines of BSCR+ areas are illustrated with dashed yellow lines. The video was exported and played at 5 fps. Scale bar, 100 μm.
Source data
Source Data Fig. 1
Statistical source data.
Source Data Fig. 2
Statistical source data.
Source Data Fig. 3
Statistical source data.
Source Data Fig. 4
Statistical source data.
Source Data Fig. 5
Statistical source data.
Source Data Fig. 6
Statistical source data.
Source Data Extended Data Fig. 1
Statistical source data.
Source Data Extended Data Fig. 2
Statistical source data.
Source Data Extended Data Fig. 4
Statistical source data.
Source Data Extended Data Fig. 6
Statistical source data.
Source Data Extended Data Fig. 7
Statistical source data.
Source Data Extended Data Fig. 8
Statistical source data.
Source Data Extended Data Fig. 9
Statistical source data.
Source Data Extended Data Fig. 10
Statistical source data.
Uncropped Blots for Extended Data Fig. 10
Unprocessed western blots.
Rights and permissions
Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article’s Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article’s Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by/4.0/.
About this article
Cite this article
Xu, X., Wang, W., Liu, Y. et al. Substrates mimicking the blastocyst geometry revert pluripotent stem cell to naivety. Nat. Mater. (2024). https://doi.org/10.1038/s41563-024-01971-4
Received:
Accepted:
Published:
DOI: https://doi.org/10.1038/s41563-024-01971-4