Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2025 Mar 7.
Published in final edited form as: Cell Stem Cell. 2024 Feb 20;31(3):312–333. doi: 10.1016/j.stem.2024.01.009

Hallmarks of Totipotent and Pluripotent Stem Cell States

Peng Du 1,2,*, Jun Wu 3,4,5,*
PMCID: PMC10939785  NIHMSID: NIHMS1963678  PMID: 38382531

Abstract

Though totipotency and pluripotency are transient during early embryogenesis, they establish the foundation for the development of all mammals. Studying these in vivo has been challenging due to limited access and ethical constraints, particularly in humans. Recent progress has led to diverse culture adaptations of epiblast cells in vitro in the form of totipotent and pluripotent stem cells, which not only deepen our understanding of embryonic development but also serve as invaluable resources for animal reproduction and regenerative medicine. This review delves into the hallmarks of totipotent and pluripotent stem cells, shedding light on their key molecular and functional features.

Short summary for the e-table of contents:

In this review, Peng Du and Jun Wu present a detailed overview of the growth conditions, molecular characteristics, and functional attributes of totipotent and pluripotent stem cells generated in vitro. Additionally, they discuss the limitations, challenges, emerging concepts, and future prospects associated with stem cells generated from early embryos.

1. Introduction

Mammalian early embryonic development is characterized by a high degree of self-organization and autonomy, crucial for the formation of a complete organism. This process begins with fertilization and progresses to gastrulation, involving a sequence of cell-fate decisions and symmetry breaking events. Post-fertilization, the embryo undergoes multiple cleavage divisions, evolving into a pre-implantation blastocyst. The blastocyst comprises the epiblast, primitive endoderm, and trophectoderm, three foundational tissues of a developing conceptus1. Following blastocyst implantation, substantial tissue reorganization and lineage development occur. Notably, epiblast cells undergo epithelialization and influenced by extraembryonic tissues, form the anterior-posterior axis. This leads to gastrulation, a process marked by complex cellular activities and the formation of the three primary germ layers: ectoderm, mesoderm, and endoderm2, which are instrumental in the development of various tissues and organs.

Pre-gastrulation development encompasses the stages of embryonic totipotency and pluripotency. In a natural embryo, totipotency is a trait of the blastomeres, whereas pluripotency is characteristic of the epiblast cells. A single blastomere, present from the two-cell stage up to the four-cell stage (though this can vary among species), possesses the remarkable ability to generate all embryonic and extraembryonic tissues and develop into a complete organism if isolated. On the other hand, epiblast cells, emerging in later stages of pre-implantation development, are pluripotent. They can differentiate into almost all cell types within the organism, except for extraembryonic tissues such as the placenta. Totipotency and pluripotency can be recapitulated in vitro in dynamic states of totipotent3 and pluripotent stem cells4 under specific conditions. The ability of these stem cells to maintain aspects of totipotency and pluripotency outside the natural embryonic context underscores their significance as powerful tools in basic and translational research.

Despite the potential, however, notable differences exist between in vivo and in vitro conditions. In vivo, cells are tightly regulated by the embryonic microenvironment, existing in a state of continuous development and actively transitioning through various stages. In contrast, cultured stem cells are typically maintained in nutrient-rich environments designed to promote proliferation and prevent differentiation, settings that are quite different from the natural embryonic environment. Despite ongoing efforts to develop conditions that more closely mimic the in vivo embryonic niche and support more authentic totipotent and pluripotent cell states in vitro, iťs important to recognize that significant differences in transcriptional, epigenetic, and metabolic characteristics exist between embryonic cells in their natural state and those cultured in laboratory settings.

Embryonic cells are characterized by their remarkable plasticity, allowing them to transdifferentiate or dedifferentiate under certain in vitro conditions into cell types not typically observed in vivo. This plasticity, which should be distinguished from intrinsic cell potency (a range of cell types a progenitor or stem cell can develop into), reflects how a cell reacts to environmental stimuli or experimental conditions, revealing adaptability that goes beyond its inherent potency. For instance, epiblast cells in embryos and their in vitro counterparts, pluripotent stem cells (PSCs), can generate extraembryonic trophoblast cells under certain in vitro conditions. This capability, usually absent in developing embryos, indicates that external factors can markedly influence the behavior and and developmental path of a pluripotent cell. The plasticity of embryonic cells, together with their spatiotemporal characteristics, contributes to the array of totipotent and pluripotent stem cell states that are maintained in vitro under various culture conditions, which should be carefully evaluated and differentiated from stem cell potency.

This review seeks to provide a comprehensive summary and detailed comparison of the dynamic states of totipotent and pluripotent stem cells identified thus far. We will focus on outlining the molecular and functional hallmarks used to define these states (Figure 1) and discuss how they correspond to their in vivo counterparts.

Figure 1. Hallmarks of totipotent and pluripotent stem cells.

Figure 1.

A list of key molecular and functional hallmarks used to define and/or distinguish different totipotent and pluripotent stem cell states.

2. Dynamic Pluripotent Stem Cell States

In vitro cultured PSCs exhibit various pluripotency states, each with different levels of similarity in global gene expression, chromatin states, and developmental potential compared to in vivo epiblasts. Numerous culture methods have been developed to stabilize naïve, intermediate/formative, and primed PSCs in both mice and humans (Figure 2). It should be noted that PSC states are not as clearly defined in humans as in mice, with considerable variations observed across protocols used by different laboratories. Given the limited variety of PSCs reported from species beyond rodents and primates, we will only briefly touch on these.

Figure 2. Growth conditions for mouse and human pluripotent stem cells.

Figure 2.

Culture components, such as growth factors, cytokines, and/or chemicals, included in each growth condition for mouse and human PSCs are indicated using blue color boxes. The related signaling pathways are noted on the left.

2.1. Naïve Pluripotent Stem Cell States

Naïve pluripotency is defined by an unfettered developmental capacity to differentiate into all somatic and germline lineages. Mouse embryonic stem cells (mESCs) are considered the benchmark for naïve pluripotency because they are functionally equivalent to the naïve epiblast cells in the inner cell mass (ICM) of an embryonic day 4.5 (E4.5) blastocyst5. mESCs express core pluripotency transcription factors (TFs) and can generate all adult lineages in a chimera6 and a whole mouse through tetraploid complementation7. mESCs were first derived from the inbred 129 strain and cultured on mouse embryonic fibroblasts (MEFs) with media containing serum to maintain pluripotency8,9. Advances have led to feeder-free and/or serum-free cultures that require leukemia inhibitory factor (LIF)10,11, bone morphogenetic proteins (BMPs)12, or the use of inhibitors that block differentiation and promote self-renewal13,14. Two most widely used growth conditions for mESCs include serum with LIF (designated as S/L herein) and a “2i/L” culture using a combination of MEK inhibition (PD0325901, or PD03) and GSK3 inhibition (CHIR99021, or CHIR) with LIF13. S/L-cultured mESCs exhibit variable expression of pluripotency-related TFs1517, suggesting they are in a metastable state18. S/L culture cannot support derivation of mESCs from non-permissive strains such as CBA, MF1 and nonobese diabetic (NOD) mice. 2i/L-cultured mESCs, in contrast, show reduced heterogeneity in pluripotency TF expression17 and can be derived from a variety of mouse strains1922. Interestingly, several substates, reflecting a developmental continuum of naive pluripotency, emerge in response to different combinations of extrinsic 2i/L and S/L signaling cues23.

Despite the success, 2i/L culture is not without its limitations. Extended cultivation of mESCs in 2i/L leads to permanent changes in their genetic and epigenetic makeup, which detrimentally impacts their developmental capabilities24,25. These deficiencies are in part due to prolonged MEK inhibition that induces alterations in DNA methyltransferase expression24. In supporting this, mESCs deficient in Erk1 and Erk2 couldn’t be maintained beyond four passages and exhibited slowed growth, G1 arrest, increased apoptosis, and genomic instability26. To address these limitations, several alternative conditions have been developed: 1) Tuning down the concentration of PD0325; 2) Replacing the MEK inhibitor with a Src inhibitor24,25,27,28. 3) Supplementation of vitamin C, which acts as a cofactor for Fe(ii) 2-oxoglutarate dioxygenase enzymes like the Tet family, has been shown to stimulate Tet-dependent DNA demethylation, fostering a DNA methylation landscape in mESCs that more closely resembles that of the ICM29. 4) Blocking certain protein kinase C (PKC) isoforms, especially PKCζ, preserves the pluripotency of mESCs without needing to activate STAT3 or block MEK/GSK3 signaling30. 5) Inhibiting mediator kinase CDK8/19 (CDK8/19i) could recapitulate naïve pluripotency in mESCs without affecting FGF/MEK signaling or global DNA methylation31. 6) Development of conditions for PSCs with extended or expanded potential32,33, which initially were thought to be totipotent-like but later studies revealed they are more similar to naïve mESCs34,35.

mESCs have set the standard for identifying and characterizing naïve pluripotency states in other species. 2i/L culture also enabled the derivation of germline-competent rat ESCs (rESCs)36,37, which facilitated the generation of genetically modified rats by homologous recombination3840. 2i/L causes rESCs to express differentiation-related genes, such as Cdx2 and T, hinting at an overly strong inhibition of GSK341,42. Indeed, lowering GSK3 inhibition (titrated 2i/L, t2iL) in rESCs suppresses differentiation and promotes self-renew41,42. These observations underscore the need for carefully balanced GSK3 inhibition to maintain stable naïve rESCs. Nevertheless, even in a t2iL condition rESCs can generate fertile offspring via tetraploid complementation only at very early passages43. Interestingly, inhibition of PKC signaling (PKCi) also promotes self-renewal of rESCs without compromising their developmental potency44. Similar to mice, PKCi culture appears to be more effective than 2i/L to repress differentiation in rESCs44. However, it is still unclear if PKCi-cultured rESCs are competent in tetraploid complementation assays.

The generation of naïve-like human PSCs (hPSCs) was achieved by overexpression of OCT4, SOX2, NANOG, and LIN28 in fibroblasts, and culturing in a medium containing human LIF and inhibitors specifically targeting MEK (PD03), GSK3 (CHIR), and ALK5 (A83–01)45. Other pioneering studies in generating mESC-like hPSCs also relied on the ectopic expression of known pluripotency-related TFs in fibroblasts or in conventional hPSCs4652, which paved the way for developing transgene-independent conditions to stabilize human naïve pluripotency in vitro. The initial cultures supporting transgene-independent naïve-like hPSCs were established by testing combined actions of different factors known to regulate naïve pluripotency or employing unbiased screens, which include NHSM53, t2iLGö50, 5i/L/A49, 2iF54, 3iL55, 2i/LIF/FGF2/Ascorbic Acid/Forskolin56, LCDM32, and hEPSCM57. Subsequent refinements were made, leading to the evolution of t2iLGö to PXGL58; NHSM to HENSM59; 5i/L/A to m5i/L/A/F60 (reduced MEK inhibition) or AXGY61. More recent progress has introduced alternative conditions, including 2iLI62, CDK8/19i31, 4CL63, AIC-N64 and prEpiSC65 cultures. The unexpected variety of growth conditions for naïve hPSCs could be partially attributed to the greater developmental plasticity of the human naïve epiblast that retains the ability to feed into extraembryonic trophoblast lineages well past the early pre-implantation stages66. Several naïve-like hPSC cultures, some modified, have been used on non-human primates (NHPs). These include STAT3 overexpression and culture systems such as t2iLGö, PXGL, NHSMV, E-NHSM, 5i/L/A, 4CL, and 4i/L/b6770. When applied to NHP PSCs they result in varied colony morphology, variable expression levels of naïve TFs, and assorted X-chromosome inactivation (XCI) states in female cells.

2.2. Primed Pluripotent Stem Cell States

During the post-implantation phase, the epiblast is subjected to inductive cues from extraembryonic tissues, effectively ‘priming’ them for differentiation. Rodent epiblast stem cells (EpiSCs) are derived from post-implantation epiblast and represent primed pluripotency. EpiSCs are typically maintained in media containing FGF2 and Activin-A (F/A) that supports their developmentally more advanced epiblast state71,72, which bears transcriptional and functional similarity to anterior late-gastrula primitive streak cells73,74.

Under F/A conditions, EpiSCs self-renew into substates reflecting different post-implantation epiblast stages75,76. This heterogeneity likely originates from dynamic epiblast cell populations that exist during the peri-gastrulation stage and are influenced by WNT/β-catenin signaling 77. Inhibiting WNT signaling, either genetically or chemically (with XAV939 and IWR-1), alongside F/A, stabilizes a more uniform primed pluripotent state and enhances EpiSCs’ clonal expansion and derivation efficiency78. Notably, EpiSCs cultured with FGF2 and IWR-1, known as region-selective EpiSCs (rsEpiSCs), show transcriptional similarity and selectively engraft to the posterior epiblast79. This characteristic indicates that different primed conditions impart specific regional traits to EpiSCs4.

Conventional hPSCs, for example cultured in mTeSR1 medium80 and categorized as 'primed' PSCs, share many characteristics with mouse EpiSCs (mEpiSCs) but have distinct molecular and functional traits. For instance, PRDM14 is essential for maintaining hPSCs, but not mEpiSCs81. Different from mEpiSCs, conventional hPSCs don’t exhibit increased FGF5 or N-CADHERIN expression but maintain the expression of E-CADHERIN and REX153,81. Furthermore, they can efficiently differentiate into both primordial germ cell-like cells (PGC-LCs) and amniotic epithelium, unlike mEpiSCs8284. Similar to mEpiSCs, inhibiting WNT signaling in hPSCs reduces heterogeneity and spontaneous differentiation, enhancing quality and clonal efficiency79,85.

Primed PSC cultures show more consistency across species compared to naïve conditions. F/A conditions have successfully derived primed PSCs from a wide range of species, including endangered species like drills and northern white rhinoceros86, as well as bats87. WNT inhibition conditions are also broadly effective in various species, including mice, humans, rats88,89, cattle9092, sheep92,93, pigs92, rabbits94 and NHPs79,95. WNT-inhibited PSCs exhibit species-specific differences in colony morphology93, clonogenicity79, and responsiveness to PGC-LCs induction89,94,95. These differences may stem from inherent species-specific traits, variations in FGF and Nodal/Activin signaling levels, or the WNT inhibitors used.

2.3. Intermediate and Formative Pluripotent Stem Cell States

Early post-implantation epiblast cells undergo substantial morphological, molecular, and metabolic changes, leading to gastrulation. This period involves shifting growth factor dependencies for epiblast self-renewal. Studies have identified various intermediate mouse pluripotency states between the naïve and primed epiblast cells35,75,85,96107. A notable intermediate state is “formative pluripotency”, found in E5-E6 epiblasts, characterized by unique molecular traits and the ability to contribute to germline chimeras and respond to PGC induction by BMP signaling108.

The interplay of FGF, TGF-β/Nodal/Activin, and WNT signaling is key in defining the temporal identity of epiblast cells cultured in vitro. For maintaining mESCs in their ground state, active WNT signaling is essential, whereas FGF/ERK signaling is inhibited to block differentiation, and TGF signaling is dispensable. During early post-implantation development, epiblast cells transition from the apolar ICM to the organized rosette structures of the E5 epiblast109. The combination of LIF with WNT and FGF/ERK inhibitors has enabled the derivation of rosette-like stem cells (RSCs) from both blastocysts and mESCs101, mimicking the E5 epiblast’s cellular architecture. RSCs express markers of both naïve and formative pluripotency (e.g. OTX2) and their chromatin landscape resembles the primed state, positioning them between naïve and primed pluripotency101.

TGF and FGF signaling activation is crucial for advancing pluripotency beyond the naïve state110,111. The combination of Activin-A with BMP4, CHIR, and LIF (in ABCL medium) stabilizes advanced pluripotent stem cells (ASCs), which are developmentally more advanced than the ground state and characterized by a stable hypermethylated epigenome and preserved genomic imprints98. Treating mESCs with FGF2 and Activin-A for 2 days produces transient formative epiblast-like cells (EpiLCs), responsive to PGC-LC induction and chimera competence, similar to E5.75 epiblasts104. Another study discovered a transient intermediate cell type during EpiLC differentiation, representing a poised state of pluripotency between the naïve and formative states99. These cells, losing the expression of naïve genes and not yet expressing formative markers, are marked by the activation of various miRNAs mediated by ISY1, resembling peri-implantation epiblasts around E4.5-E5.0. However, these cells require further characterization and have not been stably cultured yet.

Activating WNT signaling by CHIR balances the differentiation effects induced by Activin-A and FGF2, maintaining transient EpiLCs as metastable epiblast-like stem cells (EpiLSCs) in culture. EpiLSCs have bivalent energy metabolism, distinctive transcriptomic and chromatin accessibility profiles, and the competence for PGC-LC induction105. Stable and uniform populations of formative-like FTW-ESCs35 or Intermediate Pluripotent Stem Cells (INTPSCs)100 can be directly derived from blastocysts, converted from mESCs, or reprogrammed from MEFs when cultured on feeders with FGF2, Activin-A, and CHIR. FTW-ESCs show lower levels of naïve markers than mESCs and moderate expression of formative markers. Their gene expression profile closely resembles the E5.0-E5.5 epiblast, and they are responsive to PGC-LC induction and competent for germline chimera formation following blastocyst injection35.

Subpopulations of F/A-EpiSCs and EpiSCs cultured under different conditions also reveal intermediate states between naïve and primed pluripotency. F/A-EpiSCs carrying the Oct4-GFP transgene contain both GFP-positive and GFP-negative populations. GFP-positive cells exist transiently in early passages and contribute to blastocyst chimeras, suggesting a pre-gastrulation epiblast state75. Germline transmission, however, is not observed with these cells. A modified EpiSC medium with FGF4 stabilizes the Oct4-GFP-positive population, maintaining their chimeric potential106. IWPs, a porcupine inhibitor, stabilizes EpiSCs in a pre-gastrulation state (IWP2-EpiSCs) when combined with F/A, leading to increased efficiency in reverting to naïve mESCs and chimera formation85. The effects differ from EpiSCs cultured with XAV or IWR-1, possibly due to varying endogenous WNT signaling levels. Interestingly, combining a WNT activator (CHIR) and a WNT inhibitor (XAV939 or IWR1, but not IWP2) stabilizes EpiSCs at a stage closer to mESCs (CX/CR1-EpiSCs)107. This combination’s mechanism involves containing β-CATENIN in the cytoplasm, separate from its role in regulating transcription.

Two studies have generated formative PSCs resembling E6 epiblast, using modified EpiSC culture conditions with XAV, Activin-A and FGF2. The first study, using low Activin-A stimulation, XAV, and a retinoic acid receptor inverse agonist (BMS 493) - the AloXR condition – derived so-called formative stem (FS) cells. These cells show low naïve marker expression, high formative marker expression (Fgf5, Otx2, Oct6), XCI, direct PGC-LC responsiveness, and chimera formation102. The second study focused on post-implantation epiblast progression, culturing mESCs and E5.5–E6.5 epiblasts in 3D Matrigel with Activin-A, FGF2, and XAV. This culture condition led to epiblast-like embryoids (epiblastoids) forming rosettes and lumens, and stable formative cell lines (fPSCs)103. These findings, along with FTW-ESCs35 and EpiLSCs105, suggest that the formative state spans a continuum between naïve and primed states. Future research is needed to determine whether other intermediate mouse PSCs also fit within this formative pluripotency spectrum.

Studies on cultured hPSCs also propose intermediate states of pluripotent epiblast cells35,102,112115. Reassessing hPSCs under conventional conditions revealed cells at stages earlier than primed pluripotency. One study identified a subset of NCAD+ cells at the periphery of hPSC colonies with high self-renewal capacity and a tendency towards the extraembryonic endoderm lineage112. This finding suggests these NCAD+ cells represent a stage before primed pluripotency, as naïve hPSCs can differentiate into extraembryonic endoderm cells116, but primed hPSCs usually don’t. Another study found that lipid deprivation induces a stable intermediate pluripotency state in conventional hPSCs, characterized by high self-renewal, expression of naïve markers, and a bivalent metabolic profile113. Additionally, a distinct subpopulation at the periphery of hPSC colonies, similar to NCAD+ cells, showed potential for germline and somatic differentiation, a shorter G1 cell cycle, and a unique open chromatin profile, without lineage priming114. More recently, hPSCs cultured on laminin-111 in a defined primed condition displayed features similar to the early post-implantation epiblast115.

Comparative transcriptomic studies show that hPSCs cultured under various naïve conditions align more with early post-implantation (E8–E10) than pre-implantation (E6) human epiblasts, indicating they are in intermediate pluripotency states35. Additional culture conditions supporting hPSCs between naïve and primed states have been reported. Two mouse formative PSC conditions, FTW35 and AloXR102, have been adapted for human cells. AloXR-hPSCs, not expressing naïve markers, resemble early post-implantation monkey epiblasts and share some traits with primed hPSCs102. These cells rapidly differentiate into neural, endodermal, and mesodermal lineages, but their PGC-LC differentiation competence is untested102. FTW-hPSCs exhibit gene expression profiles between naïve and primed states, resembling E8 human epiblasts35, and show potential for PGC specification and interspecies chimera formation35,117. Given the transient nature of the naïve state in human and NHP blastocysts118,119, compared to the stable gene expression of early post-implantation primate epiblasts120, capturing the intermediate and primed states in primates may be more feasible than capturing the naïve state.

3. Hallmarks of Pluripotent Stem Cell States

The characteristics of naïve and primed PSCs were initially identified by contrasting these two states against each other121. Yet, the uniqueness of these defining features of naïve and primed pluripotency has been challenged by the identification and characterization of PSCs in intermediate stages. Additionally, variations between species call for different benchmarks to define pluripotency across organisms. Moreover, the ethical restrictions on using hPSCs in germline chimera and tetraploid complementation assays necessitate alternative methods to functionally define pluripotency in human research. This review provides a summary of the distinguishing features, or hallmarks, of various PSC states (Figure 3). Readers seeking a more in-depth understanding are encouraged to consult comprehensive recent reviews4,96,122129. As the variety of PSCs from different species continues to grow, the characteristics used to differentiate them will need frequent updates. In this review, we include only those hallmarks that are currently applicable to the majority of mouse and human PSCs identified to date. The criteria for defining PSC states are generally classified into two main groups: molecular and functional hallmarks (Figure 3).

Figure 3. Molecular and functional hallmarks of dynamic pluripotent stem cells.

Figure 3.

Left, A summary that highlights the selected molecular and functional characteristics useful for distinguishing mouse and human PSCs in various states of pluripotency. Right, the hallmarks each type of PSC meets are highlighted with color-coded boxes.

3.1. Molecular Hallmarks

Signaling dependency.

Most naïve PSC cultures use the 2i/L framework or its variants, highlighting the importance of LIF/STAT3 and WNT signaling, and inhibiting FGF/ERK signaling, to maintain naïve pluripotency in vitro. Recent methods have employed tankyrase inhibitors to support human naïve pluripotency58,59,61. FGF2 is also included in various naïve hPSC cultures47,49,5356, suggesting FGF signaling might positively influence naïve hPSCs through non-ERK pathways, though the exact mechanisms are unclear. In several species, including mice, rats, bovines, marmosets, and humans, strong NANOG expression in preimplantation blastocysts occurs without FGF/ERK signaling, suggesting ERK independence as a universal feature of naïve pluripotency130133. Nonetheless, 2i/L lone is not enough to maintain naïve pluripotency in species beyond mice and rats, necessitating modulation of other signaling pathways. Moreover, the reliance on specific signaling pathways for naïve pluripotency varies across species; for instance, TGF signaling is dispensible for mESCs but is needed for naïve human pluripotency134,135.

Stabilizing primed pluripotency in vitro hinges on activating FGF and TGF pathways, a common requirement across species. Adding WNT inhibitors can result in a 'ground state' of primed pluripotency with uniform pluripotent gene expression. However, intermediate/formative pluripotency, which includes several temporal states, has different signaling dependencies. Various methods are used to maintain formative PSCs, with some activating WNT signaling (as in FTW-PSCs and EpiLSCs) and others inhibiting it (as in AloXR-PSCs and fPSCs), in combination with F/A.

Expression of pluripotency state-specific marker genes.

Various genes help distinguish naïve (e.g. Klf2, Klf4, Tfcp2l1), intermediate/formative (e.g. Otx2, Sox3, Oct6), and primed (e.g. Fgf5, Lef1, Nodal) PSCs5,35,102. However, care should be taken in defining pluripotency states based on gene expressions due to inherent heterogeneity. In addition, significant species-specific differences exist. Certain mouse naïve markers, such as Klf2, Nrob1, Bmp4, and Esrrb, are not expressed in human or marmoset ICMs. Conversely, the human preimplantation epiblast expresses unique genes, such as KLF17, and members of the TGF signaling pathway (e.g., NODAL, GDF3, and LEFTY1131,135), which are absent in mice. Furthermore, transcriptome profiling reveals continuous expression of NANOG and PRDM14 in post-implantation epiblasts until gastrulation in NHPs120.

Transcriptomic similarity to in vivo epiblasts.

Comparing transcripotomes of cultured PSCs with those of in vivo epiblasts at various developmental stages, particularly at the single cell level, can provide a more accurate assessment of their pluripotency states. Recent advances in transcriptomic analysis of mammalian embryos before, during, and after implantation have yielded a wealth of data crucial for such comparisons5,120,131,135141. Ideally, the expression patterns of genes in naïve, intermediate/formative, and primed PSCs should mirror those in pre-implantation, pre-gastrulation, and peri-gastrulation epiblasts, respectively. However, it is important to account for differences between in vivo and in vitro conditions, which can cause variations in gene expression.

Global DNA methylation status.

Mouse and human preimplantation epiblasts feature widespread DNA hypomethylation142146, a key trait of naïve pluripotency, except in imprinted regions. 2i/L-mESCs exhibit low DNA methylation17,147, likely attributable to JMJD2C-dependent TET1 activation and DNMT3A/B protein degradation mediated by PRDM14/G9a148, a feature not maintained under the S/L condition. Naïve hPSCs grown in 5i/L/A49 and t2iLGö/PXGL50,149 conditions show DNA hypomethylation similar to the human ICM but lose genomic imprinting150152. Naïve hPSC cultures with slightly higher DNA methylation levels (~42%) than the human ICM but lower than primed hPSCs (~75%) show improved genomic stability and imprint preservation5961,63. These hPSCs are in more advanced pluripotency states than 5i/L/A and t2iLGö/PXGL cells, reminiscent of mouse ASCs (~70% CpG methylation). This level is higher than 2i/L-mESCs (~20%) but lower than EpiSCs (~90%)98. Unlike 2i/L-mESCs, where imprints are erased98,153, most imprints are retained in ASCs98. The methylation status of most intermediate hPSCs is unreported, but those cultured on laminin-111 and in lipid-deprived conditions show methylation levels similar to or slightly lower than primed hPSCs113,115.

From E4.0 to E6.5, mouse epiblasts experience a genome-wide increase in DNA methylation, coinciding with upregulation of de novo methyltransferase 3 (DNMT3) enzymes154. In mammals, three types of DNMT3s have been identified: DNMT3a and DNMT3b are catalytically active, whereas DNMT3L acts as a cofactor. Dnmt3b expression starts at E3.5, followed by Dnmt3a at E4.5, with Dnmt3l expressed briefly between E4.5 and E6.5141. Human embryos show a similar increase in median DNA methylation from 26.1% on day 6 to 60.0% by day 10141.

DNA methylation patterns in various in vitro PSCs largely reflect in vivo patterns. 2iL-mESCs show global hypomethylation similar to ICM cells, whereas S/L-mESCs have higher DNA methylation, resembling the early post-implantation epiblast153,155. Interestingly, the intermediate pluripotent state regulated by ISY1/miRNAs, despite having a similar transcriptomic signature to S/L-mESCs, has a hypomethylated genome as in 2iL-mESCs99. ASCs, further along in development, exhibit high methylation levels across various genomic regions98. Contrasting with 2iL-mESCs, which exhibit lower expression of DNA methyltransferases17,147,153,155, RSCs express Dnmt3b and Dnmt3l, showing elevated 5mC levels intermediate between naïve and primed cells101. Globally, fPSCs exhibit high DNA methylation similar to EpiLCs and E6.5 epiblast cells103. EpiSCs and rsEpiSCs, at the far end of the pluripotency spectrum, have a hypermethylated epigenome (~87%)79. rsEpiSCs show hypermethylation in regions linked to regionalization and anterior/posterior pattern specification79. Notably, the global level of 5-methylcytosine in EpiSCs is similar to S/L-mESCs153,156. However, EpiSCs show a distinct bias towards hypermethylation at promoter regions, especially those with high CpG island promoters. This DNA methylation signature is unique to EpiSCs and differs from their in vivo counterparts, the E6.5 and E7 gastrulating epiblasts, indicating potential culture artifacts157.

X chromosome inactivation status in female cells.

XCI is a crucial developmental process in female mammals to balance the dosage of X-chromosomal genes between the sexes. It occurs dynamically during mammalian development and has two forms: imprinted and random. Imprinted XCI, happening at the preimplantation stage in mice, involves selective silencing of the paternal X chromosome at the 4-cell stage122,158, with the cis-acting, non-coding RNA Xist (X-inactive specific transcript) playing a key role158. Imprinted XCI begins to be erased in the ICM of early blastocysts, leading to a temporary reactivation of both X chromosomes. Subsequently, random XCI begins in the post-implantation epiblast136,158. A study tracking Xist expression during peri-implantation development found that Xist erasure starts around E3.75 and completes by E4.5 in mice. Notably, Xist re-expression was observed as early as E4.75, indicating the early initiation of random XCI in these cells159. This study also uncovered that a notable proportion of epiblast cells displayed biallelic Xist expression at E5.25 (13.9%) and E5.5 (9.3%)159. Interestingly, similar expression patterns were noted in male epiblast cells at comparable stages160. These findings suggest substantial epigenomic changes during this phase of developmental transition. By E6.5, nearly all cells exhibited a single Xist cloud, indicating that random XCI is likely completed by this stage159.

Pre-implantation human embryos exhibit a unique XCI pattern compared to mice, lacking paternal XIST imprints and showing random XCI in both embryonic and extraembryonic tissues136,158,161. Notably, ~85% of cells in day 6 and 7 female blastocysts display bi-allelic XIST coating, indicating a distinct mechanism of XCI in humans compared to mice136,158. Interestingly, despite XIST accumulation, bi-allelic expression of X-linked genes is evident at the chromosome-wide level throughout pre-implantation development, starting as early as E3136,158. These studies establish that human embryos are in a pre-XCI state before implantation (XaXIST+XaXIST+). Information on human random XCI timing is limited due to restricted access to peri-implantation embryos. XCI involves the accumulation of histone 3 lysine 27 trimethylation (H3K27me3) marks on the condensed X-chromosome. In a study modeling human implantation, day 5 human blastocysts were cultured for an additional three days on decidualized endometrial stromal cells. Intriguingly, at the end of this co-culture, H3K27me3 foci were absent in epiblast cells, suggesting that random XCI did not initiate at this stage162. Advances in extending human blastocyst culture beyond implantation stages163,164 have provided insights into XCI during early post-implantation. Studies examining allele-specific expression of X-linked genes from days 6 to 12 in these cultured embryos suggest that random XCI begins at peri-implantation stages without being complete by day 12141.

In female mouse PSCs, XCI status is another key indicator of their pluripotency state and resemblance to in vivo development stages. Female mESCs have two active X chromosomes (XaXa), mirroring pre-implantation epiblast cells. Intermediate PSCs transition from this XaXa status to having one active and one inactive chromosome (XaXi). Both RSCs and FTW-ESCs retain the XaXa status, consistent with E5.0-E5.5 epiblast stages35,101. In contrast, primed F/A-EpiSCs and rsEpiSCs79, intermediate CX/CR1-EpiSCs107, IWP2-EpiSCs85, fPSCs103 and FS cells102 exhibit XaXi status, corresponding to later epiblast stages (E6.5-E7.75).

XCI in female hPSCs is more complex than in mice. Initially, conventional hPSCs were thought to have two active X chromosomes without XIST expression (XaXIST−XaXIST−)165. Later studies revealed significant variations in XCI status across different hPSC lines and even within subcultures of the same line166170. Additionally, factors like oxygen concentration during derivation and culture can influence the XCI status in primed hPSCs171. Primed hPSCs show XCI instability, often losing XCI characteristics over multiple passages, unlike differentiated cells. This instability is characterized by the loss of XIST expression and partial reactivation of the inactive X chromosome172,173. Similarly, naïve hPSCs exhibit a varying XCI status under different culture conditions, with some (5i/L/A49,151 and t2iLGö50) aligning with a pre-XCI state indicated by biallelic expression of X-linked genes and XIST accumulation on active X chromosomes174,175. However, the extent to which these cells mirror in vivo pre-implantation epiblasts is debatable, as most show monoallelic XIST expression174,175. There is also a lack of consensus regarding the XCI marker H3K27me3. Some studies reported an enrichment of H3K27me3 on active X chromosomes that are coated with XIST174. Others did not observe this accumulation175, aligning instead with findings in embryos158. Moreover, during differentiation, naïve hPSCs undergo de novo XCI, but this process differs from the XCI observed in embryos as it follows a non-random pattern151,174. In another study insufficient inhibition of autocrine FGF2 led to varied X chromosome status in naïve hPSCs. The authors successfully isolated a uniform population of naïve hPSCs with an XaXIST+XaXIST+ status, which is closely related to the XCI status of human pre-implantation epiblast. In this study, it was also observed that when these cells transitioned from a naïve to primed state, random XCI ensued, mimicking the natural embryonic process176.

Metabolic features.

PSC states are closely linked to specific metabolic profiles. Although a high glycolytic flux is essential for maintaining pluripotency177181, its role varies across different pluripotency states182. Primed PSCs rely almost entirely on glycolysis. In contrast, naïve PSCs are characterized by a bivalent metabolism, leaning more towards oxidative metabolism49,50,183,184. Intermediate and formative PSCs show mixed metabolic states. EpiLSCs and FTW-ESCs display an intermediate level of maximal mitochondrial activity between mESCs and mEpiSCs. They rely on oxidative phosphorylation during basal respiration but shift to glycolysis during maximal respiration35,105. FS cells, similar to mEpiSCs, predominantly use glycolysis for energy production105. These observations mirror the metabolic preference of the early embryos, which initially depend on pyruvate from ovarian follicle cells for oxidative metabolism. As development progresses, increased glucose availability and transporter expression lead to a rise in glycolytic activity182,185,186. Interestingly, primed PSCs have low mitochondrial activity but elongated mitochondria with defined cristae, in contrast to the spherical but active mitochondria in naïve PSCs50,54,183,187,188. Formative EpiLSCs show an intermediate mitochondrial morphology with a mix of spherical and elongated structures105.

Lipids are essential in early mammalian development, acting as energy sources, signaling molecules, and membrane components189. Prior to implantation, lipids come from the oocyte's endogenous reserves and are also synthesized de novo190. Lipid availability influences acetyl-coenzyme A and α-ketoglutarate levels, impacting ERK signaling and inducing an intermediate pluripotency state with heightened de novo lipogenesis113,191. This intermediate state thrives in lipid-free culture conditions, promoting active lipid biosynthesis and adding lipids to the medium disrupts this state. Supporting this observation, numerous genes highly expressed in fPSCs are involved in cholesterol/sterol biosynthetic and lipid metabolic processes, underlining the significance of lipid metabolism in maintaining intermediate/formative PSCs103.

Amino acids influence pluripotency and stem cell fate, serving as substrates for biosynthesis and facilitating chemical modifications182. Specific amino acids such as threonine, methionine, and proline are key regulators of pluripotency. mESCs rely heavily on threonine metabolism to sustain pluripotency and self-renewal192. Threonine supports naive pluripotency by supplying S-adenosyl methionine (SAM) for the trimethylation of histone H3 lysine 4 (H3K4me3)193. As an essential amino acid, threonine can be broken down by threonine dehydrogenase (TDH) into glycine and acetyl-coenzyme A. Disruption of threonine metabolism in mESCs, either through inhibiting TDH or depriving cells of threonine, leads to a loss of stemness, diminished proliferation, apoptosis, and cell cycle arrest192,194. hPSCs do not depend on threonine as the TDH gene in humans is a non-functional pseudogene195. In primed hPSCs, methionine is essential for synthesizing SAM, with its depletion leading to differentiation and apoptosis196. Conversely, in naïve hPSCs, low levels of SAM, maintained by nicotinamide N-methyltransferase, are necessary to preserve the naïve state187. Proline also plays a vital role, particularly in mESCs, where its addition to the culture media enhances proliferation and facilitates the transition to EpiSCs197,198. The implantation process involves breaking down a proline-rich extracellular matrix, suggesting that the released proline may support the epiblast transition during peri-implantation development199,200.

3.2. Functional Hallmarks

A range of assays has been developed to assess the developmental potential of cultured PSCs128 (Figure 3). These include universal assays, such as in vitro embryoid body differentiation and the teratoma assays, which are suitable for all PSCs. Additionally, there are assays designed to differentiate between various pluripotency states, briefly outlined below.

Blastocyst chimera formation and germline transmission.

Assessing pluripotency in PSCs often involves testing their ability to form chimeras by combining them with preimplantation embryos and transferring these into surrogate mothers128. PSCs with high developmental potential not only support normal development but also produce high-grade chimeras capable of generating offspring entirely derived from the donor PSCs via germline transmission. In contrast, PSCs with low developmental potential may result in modest to no chimerism. Mouse and rat ESCs are known for their ability to form blastocyst chimeras and achieve germline transmission. Chimeras are formed not only by naïve PSCs but also by several intermediate and formative PSC types, such as ASCs98, RSCs101, FTW-ESCs/INTPSCs35,100, FS cells102, fPSCs103, Oct4-GFP+ EpiSCs and IWP2-EpiSCs. ASCs, RSCs, and FTW-ESCs/INTPSCs have also demonstrated germline transmission, a trait not observed in fPSCs, Oct4-GFP+ EpiSCs75, IWP2-EpiSCs85 and not tested in FS cells102. Generally, the extent of chimera contribution from intermediate/formative PSCs tends to be lower than from mESCs. Blastocyst chimera assays have also been applied to NHPs, with varied success. For instance, cynomolgus monkey PSCs (cyPSCs) cultured under a modified NHSM condition (NHSM supplemented with Vitamin C) showed modest chimeric contribution of less than 20% into two fetuses at around 100 days of gestation67. Importantly, cyPSCs cultured in 4CL63 medium resulted in an aborted chimeric fetus and a live neonatal chimeric monkey, with the latter showing a high donor cell contribution of up to 90%69. However, this monkey developed severe health problems and was euthanized shortly after birth, preventing assessment of its germline transmission capabilities.

Blastocyst chimera formation is not suitable for human research due to ethical issues and is challenging for species with long gestation or poorly understood reproductive biology. As an alternative, interspecies chimeras are used, but this approach is often unreliable due to low success rates and xenogeneic barriers between distant species201 (Figure 3). Consequently, teratoma formation and in vitro differentiation of embryoid bodies are broadly used for accessing pluripotency in humans. Notably, some naïve hPSCs require an adaptation period in primed media to respond effectively to differentiation signals202, which may explain their limited ability to generate teratomas203.

Tetraploid complementation.

Tetraploid complementation, the most stringent test for pluripotency, involves introducing donor PSCs into a tetraploid blastocyst7, created by fusing two-cell stage embryos. In this process, tetraploid cells predominantly contribute to extraembryonic placenta tissue, whereas the diploid donor PSCs develop the embryo proper. High-quality mESCs and early passages of rESCs43 have demonstrated the ability to complement tetraploid blastocysts, leading to animals entirely derived from donor cells. Additionally, intermediate ASCs have been used to produce full-term offspring in tetraploid hosts98. However, tetraploid complementation has yet to be tested for RSCs and FTW-ESCs.

Post-implantation epiblast chimera formation.

EpiSCs show limited ability to generate blastocyst chimeras but can effectively integrate into the post-implantation epiblast, contributing to ex vivo epiblast chimeric embryos74,79,204,205. This outcome differs from mESCs, which are limited in contributing to chimera formation in post-implantation epiblasts204. Notably, primed hPSCs and rhesus monkey PSCs can engraft into E6.5 or E7.5 mouse epiblasts and differentiate into cells from all three germ layers79,206,207. Grafting to the post-implantation epiblast thus serves as a functional test for primed pluripotency. The capability of intermediate and formative PSCs to generate chimeras through post-implantation epiblast grafting remains to be tested.

Primordial germ cell specification.

Germ cells, vital for species continuity, originate from precursor cells known as PGCs, which arise from competent formative epiblast cells around E5.5-E6.25 in response to signals such as BMPs in mice. These PGCs proliferate and migrate through the developing hindgut before colonizing the nascent gonads, where they differentiate into oocytes or sperm104. Notably, mESCs require a transient differentiation into formative EpiLCs to generate PGC-LCs in vitro208. This process is also mirrored in rat EpiLCs, which can be induced from rESCs to form functional PGC-LCs in spherical aggregates209. Although previously considered inefficient, we now know that mEpiSCs can produce PGC-LCs and fully functional germ cells210. Rat EpiSCs cultured in a WNT-inhibited condition AFI2XY (Activin-A, FGF2, IWP2, XAV and Y27632) demonstrate germline competency, producing functional PGC-LCs in vitro and showing molecular traits akin to pre-gastrulating pluripotent epiblasts, thus representing an intermediate pluripotency state89. Consistently, WNT-inhibited cynomolgus monkey and rabbit PSCs also exhibit robust PGC-LC differentiation94,95. Contrary to mESCs, formative PSCs such as FTW-ESCs, EpiLSCs, FS cells, and fPSCs, directly specify PGC-LCs35,102,103,105. Intermediate or formative hPSCs also show similar PGC responsiveness, especially when grown on laminin-111115, cultured in FTW medium35, or propagated in a simplified NHSM condition, termed '4i'211. Intriguingly, direct PGC-LC induction from primed hPSCs is achievable either through incipient mesoderm-like cells83 or using a microfluidic device84. However, species-specific differences in PGC specification mechanisms exist between primates and rodents, including varied developmental stages and origins from distinct progenitor cells (epiblast and/or amnion)212.

4. Dynamic Totipotent Stem Cell States

Zygotes and early blastomeres, capable of forming the entire organism and producing all embryonic and extraembryonic tissues, exhibit totipotency, the highest level of developmental potency. Unlike stem cells, early blastomeres, derived from zygote cleavage, lack self-renewal ability213. Consequently, there's interest in whether totipotent stem cells, recapitulating some features of zygotes/early blastomeres, can be effectively derived and maintained in vitro. Totipotent or totipotent-like stem cells, though present in low proportions in pluripotent cultures, can be isolated and even stably maintained under specific conditions, sparking significant research interest. This summary highlights recent advancements in generating and characterizing these so-called totipotent stem cells, acknowledging their currently limited understanding compared to PSCs.

4.1. Metastable Mouse 2-Cell-Like Cells and Human 8 Cell-Like Cells

Using 2C::tdTomato+ reporter mESCs, generated based on the MuERV-L-LTR retrotransposon expressed in mouse 2-cell blastomeres at the ZGA stage, a study found a small subpopulation (about 0.1–1%) could transiently activate 2-cell specific genes, such as Zscan4 and Zfp352. These cells, known as 2-cell-like cells (2CLCs), quickly revert to tdTomato-, suggesting a metastable state with fluctuating entry into and exit from the 2-cell totipotent-like state214. Currently, no study has confirmed that a single 2CLC can generate both embryonic and extra-embryonic tissues in chimeric mice when injected into blastocysts (or earlier stages). More research is needed to fully validate the functional totipotent potency of these cells215.

Recent advancements have led to the generation of metastable human 8 cell-like cells (8CLCs) under naïve hPSC culture conditions. Similar to mouse 2CLCs, human 8CLCs activate a range of ZGA genes, such as ZSCAN4/5B, DUX4, TPRX1, and LEUTX, and constitute about 1.6% of all cells cultured in PXGL medium216. In a second study using the e4CL (enhanced 4 chemicals + LIF) medium, 8CLCs were transiently induced to make up 11.9% of the cell population for less than a week, though they could not be passaged. These isolated 8CLCs, identified using TPRX1 as a reporter, demonstrated both embryonic and extraembryonic developmental potential in teratomas and interspecies chimeras in mice63. In a third study, 8CLCs were identified from prEpiSCs and PXGL cultured naïve hPSCs using single-cell RNA-sequencing (scRNA-seq). The researchers created a LEUTX reporter to aid in isolating 8CLCs and developed a culture condition to increase the proportion of 8CLCs from prEpiSCs. In this culture, 8CLCs were able to be sustained at a relatively high percentage for 2–3 weeks65.

The discovery of metastable 2CLCs and 8CLCs63,65,216, which mirror the ZGA stage blastomeres in mice and humans, respectively, hints at the potential to culture totipotent-like cells in vitro. Subsequent research, following these initial findings, has identified several regulatory factors, including DUX4/Dux and Dppa2/4, that promote the shift from pluripotent to totipotent states217220. Despite these advances, none of these efforts have succeeded in achieving the stable maintenance and long-term passaging of totipotent stem cells.

4.2. Stable Culture of Mouse Totipotent Stem Cells

Notably, a type of so-called mouse and human expanded/extended pluripotent stem cells (EPSCs) has been cultured and maintained in specific culture media for long-term passages by two separate research groups32,33. Interestingly, these cells don’t express any typical totipotency related genes, but instead still highly express pluripotency markers, such as Oct4, Nanog, Sox2 and Klf4/5. Surprisingly, functional assays have suggested that they are able to generate extraembryonic and embryonic tissues in vivo and in vitro. However, the totipotent features of these EPSCs have been challenged at molecular and functional levels34. EPSCs are more appropriately considered as a type of intermediate PSCs rather than totipotent cells, given their closer transcriptional resemblance to early post-implantation epiblasts in mice and humans.

In a surprising development, a mouse totipotent stem cell line was successfully isolated and stably maintained by inhibiting spliceosome activity221. Spliceosomes, essential for mRNA splicing and maturation, are typically expressed at low levels in zygotes and totipotent blastomeres, with their activation occurring around the ZGA stage, a pattern observed across various mammals. The success of this method suggests that splicing might influence the transition from totipotency to pluripotency. Suppression of splicing factors in mESCs triggers the activation of totipotent genes while repressing pluripotent genes, steering cells towards totipotency. Remarkably, adding Pladienolide B (PlaB), a splicing inhibitor, to the S/L medium, reprograms mESCs into totipotent blastomere-like cells (TBLCs) and enables their stable maintenance over long-term passages. Molecular analyses indicate that these TBLCs closely resemble 2-/4-cell blastomeres at the transcriptomic and epigenomic levels. In chimeric mice, single mouse TBLCs contribute to both embryonic and extraembryonic tissues, including 6 trophoblast and 1 yolk sac lineage221.

Mouse TBLCs are different from 2CLCs. TBLCs show a wider and more pronounced activation of totipotent genes, such as Zfp365, Ddit4l, Plk2 and Btg2, which are predominantly present in zygotes and early to middle 2-cell blastomeres at the pre-ZGA stage. In contrast, key pluripotent genes, such as Sox2, Nanog, Pou5f1, Zfp42, Esrrb, Klf2, and Tdgf1, which remain highly expressed in 2CLCs, are completely silenced in TBLCs. Epigenetically, 2CLCs are similar to mESCs, including chromatin accessibility, H3K4me3, H3K27me3, and H3K27ac marks, especially at pluripotent and totipotent loci, unlike in vivo 2-cell blastomeres222. Conversely, TBLCs, induced through spliceosome repression, show epigenetic features closely mirroring 2-/4-cell blastomeres, including DNA methylation, chromatin accessibility, and histone modifications, both at specific loci and globally. Therefore, although 2CLCs exhibit both pluripotent and totipotent characteristics, fitting their metastable nature, TBLCs are stable totipotent stem cells, having shed all pluripotent regulatory networks. scRNA-seq analysis indicates that TBLCs uniformly activate totipotent genes and suppress pluripotent genes, suggesting a relatively homogenous cell population.

Subsequently, several research groups have reported the successful isolation of other types of mouse totipotent stem cells, primarily through epigenetic modulation. These cell types include totipotent potential stem cells (TPSCs), totipotent-like stem cells (TLSCs), and chemically induced totipotent stem cells (ciTotiSCs) (Figure 4A)222224, which are derived using chemical compounds that target epigenetic regulators, such as KDM5B, DOT1L, and HDAC1/2, influencing histone modifications, such as H3 acetylation and H3K79 methylation, as well as activation of RARγ signaling. Similar to mouse TBLCs, TPSCs, TLSCs, and ciTotiSCs show extensive activation of totipotent genes and repression of pluripotent genes at the molecular level. Functionally, these cells contribute to a range of embryonic and extraembryonic lineages (Table 1).

Figure 4. Summary of in vitro cultured totipotent stem cells.

Figure 4.

(A) Developmental Timeline of In vitro Totipotent Stem Cell Cultures. The reference to 'similar to (embryo stage)' relates specifically to similarities at the transcriptomic level.

(B) Heatmap showing the comparative expression of representative totipotent genes across different cell types.

(C) PCA analysis of transcriptomes of TBLC221, TLSC222, TPSC223, EPSC32,33,223, ciTotiSC224, 2CLC 222and their corresponding mESCs, as well as mouse pre-implantation embryos based on the global transcriptome. Stars represent TBLC data, triangles represent TLSC data, solid squares represent TPSC data, hollow squares represent EPSC data, rounded rectangles represent 2CLC data, and hexagons represent ciTotiSC data.

Table 1.

Dynamic Totipotent Stem Cells

Feature Developmental potentials
Chimera assay (in vivo) Teratoma (in vivo) Blastoid-formation assay (in vitro) Trophoblast stem cell (TSC) reprogramming
Contribute to embryonic tissue Contribute to extra-embryonic tissue Chimeric mice Embryonic lineages Extra-embryonic lineages
mESC (for comparison) Yes No Yes Yes No No No
D-EPSC32 Yes Conflicting results: Yes32 and No34 Yes Three germ layers No Conflicting results: Yes230 and No231 Conflicting results: Yes32 and No34
L-EPSC33 Yes Conflicting results: Yes33 and No34 Yes Three germ layers No Conflicting results: Yes230 and No231 Conflicting results: Yes33 and No34
2CLC214 No single cell chimera tested No single cell chimera tested Not tested Not tested Not tested Not tested Not tested
TBLC221 Yes Yes Yes Yes Yes (scRNA-seq) Yes246 Yes
TLSC222 Yes Yes Not tested Not tested Not tested Yes Not tested
TPSC223 Yes Yes Yes Mesoderm, ectoderm Yes (scRNA-seq) Yes Yes
TotiSC224 Yes Yes Yes Three germ layers TGCs (IHC) Not tested Yes
Feature Transcriptomic profiles
Global comparison with mouse early embryonic states Totipotency or 2C marker Pluripotency marker Transposon element (MERVL, MT2-Mm) Maternal
mESC Similar to epiblast + +++ + +
D-EPSC Similar to E5.534 + +++ + +
L-EPSC Similar to E3.5-E4.534 + +++ + +
2CLC Similar to blastocyst ++ +++ ++ +
TBLC Similar to 2- and 4-cell +++ + +++ +++
TLSC Similar to 2- and 4-cell +++ + +++ +++
TPSC Similar to 2-cell stages +++ + +++ Not tested
TotiSC Similar to 2-cell stages +++ + +++ +++
Feature Genomic landscape
Chromatin accessibility DNA methylome Histone modification
bdH3K4me3 H3K4me3 H3K27me3 H3K9me3 acetylated H3 acetylated H4
mESC Low High -
D-EPSC Not tested Not tested Not tested similar to mESC increasing compared with mESC Not tested Not tested Not tested
L-EPSC Not tested Not tested Not tested similar to mESC No clear conclusion Not tested No clear conclusion Not tested
2CLC Middle, resembled mESCs, but not 2-cell Middle247 Not tested Higher level Not tested Not tested similar to mESCs similar to mESCs
TBLC High, open peaks in 2-,4-cell Low Not tested
TLSC High, open peaks in 2-,4-cell Not tested retain many oocyte/2C-specific bdH3K4me3 domains similar to 2-cell No clear conclusion No clear conclusion Not tested Not tested
TPSCs High, open peaks in 2-cell Low Not tested Not tested Not tested Not tested Higher levels Not tested
TotiSC High, open peaks in 2-,8-cell and ICM Low Not tested
Feature In vitro culture Medium Target
Cell culture passages# in vitro Resource Karyotype Self-renewal Cell cycle chemical compounds
mESC Yes ICM Normal Yes G2 LIF
D-EPSC P10 mESC blastocysts Normal (P10, female) Yes Not tested hLIF, CHIR99021 (Wnt agonist), DiM, MiH (PARP1 inhibitor)
L-EPSC P10 mESC, iPSC, 4-/8-cell Normal (P10, female) Yes Not tested hLIF, CHIR99021(Wnt agonist), PD0325901(Mek1 inhibitor), VIII (JNK Inhibitor), SB203580(p38 inhibitor), A-419259(Src kinase), XAV939(TNKS1/2 inhibitor and stabilize AXIN)
2CLC Transient mESC Not tested No Longer G2 No need
TBLC P18 mESC Normal (P12, male) Yes Not tested Pladienolide B (splicing inhibitor)
TLSC P21 mESC,2-/4-/8-cell Normal (P21, male; P16, female) Yes Longer G2 SGC09469 (DOT1L inhibition), AS8351 (KDM5B inhibitor), A366 (G9a inhibitor), sIL-6R, IL6
TPSCs P15 EPS, 2-cell Normal (P15, male) Yes Not tested CD1530 (RARγ agonist), VPA (HDAC inhibitor), EPZ004777 (Dot1L inhibitor), CHIR99021 (Wnt agonist)
TotiSC P8 mESC Normal (passage not indicated, male) Yes Longer G2 TTNPB (RARγ agonist), 1-azakenpaullone (GSK-3β inhibitor), WS6

In this table, the classifications of high/middle/low and yes/no are based on comparisons with mESCs.

(-)

means mESC serving as the baseline.

The subjective ratings (+) are employed to visually present the extent of the observed differences.

#

represents the highest passage mentioned in the article.

In summary, various methods, including spliceosome repression, epigenetic manipulation, and RARγ signaling modulation, successfully capture and culture mouse totipotent stem cells in vitro. Despite the diversity of these approaches, they interestingly converge to induce a similar totipotent state in cultured stem cells. This convergence points to the existence of complex regulatory networks crucial for establishing and maintaining a stable homeostatic state in totipotent stem cells. However, it remains unknown whether these approaches can be effectively translated to humans, as stable maintenance of human totipotent stem cells has not yet been achieved.

5. Assessing and Evaluating Emerging Totipotent Stem Cells: Challenges and Criteria

Although various mouse totipotent stem cells have been identified, challenges persist in fully understanding their totipotency and potential applications. A critical step is a thorough comparison and characterization of these distinct totipotent stem cells. For example, all the stably cultured cells closely resemble 2-/4-cell blastomeres and show induction of classical totipotent markers such as Zscan4, Gm8300, and Rab43, compared to mESCs. However, some still lack expression of other key totipotent genes, such as Zfp365, Mmp19, and Ddr2, particularly ciTotiSCs and TPSCs224 (Figure 4B and 4C). Although scRNA-seq analyses have been conducted on some of these cells, assessing the heterogeneity of different totipotent stem cells across passages remains complex and warrants cautious evaluation.

The growth rate of all reported totipotent stem cells so far appears slow, or at least not as fast as that of PSCs. This is particularly evident in ciTotiSCs, where certain totipotent markers are transiently induced only in early passages (1–4) and not at later stages, such as passage 8 (Figure 4B). This transient induction raises concerns about the stability of these cells over long-term culture, questioning the claimed self-renewal capabilities. Given that zygotes and totipotent blastomeres naturally lack proliferative capacity, the self-renewal of these in vitro totipotent stem cells might depend on artificially activated cell signaling pathways. An important question arises: can we manipulate specific pathways to achieve rapid and stable expansion of totipotent stem cells, akin to PSCs?

Researchers have started to uncover the epigenetic characteristics of totipotent stem cells, such as histone modifications, DNA methylation, parental imprinting, and X-inactivation. For instance, mouse TBLCs, TPSCs and ciTotiSCs exhibit notably lower DNA methylation levels compared to PSCs, contrasting with in vivo blastomeres that have higher methylation than blastocysts221225. Moreover, the 3D chromosomal structures in totipotent stem cells are relatively less compact, indicating that their organization is still being established. These distinct epigenetic features could lead to genetic and epigenetic aberrations, impacting the stability and developmental potential of these cells during prolonged in vitro culture.

Based on the stringent definition, a single totipotent stem cell should be capable of generating an entire conceptus. This high standard has not yet been met in the functional verification of reported totipotent stem cells. Currently, the functional proof of totipotency often involves assessing the cells' ability to contribute to extraembryonic tissues in chimeric mice in vivo. In this regard, scRNA-seq analysis emerges as a superior approach, enabling the identification of all embryonic and extraembryonic lineages and elucidating the developmental trajectories of various totipotent stem cells. However, it is important to note that false positives can be introduced during fluorescence-activated cell sorting of cells derived from the chimeric conceptus. Accurate identification of “true” chimeric contribution necessitates verifying the expression of genes (e.g. fluorescent marker genes) that are specifically present in the donor cells, a step sometimes overlooked in some research.

Moreover, the limitations of in vivo chimeric assays, particularly for human totipotent-like cells, make in vitro differentiation systems directed towards extraembryonic tissues both invaluable and essential for assessing totipotency. However, artificial manipulation of signaling pathways and inclusion of undetermined factors in cell culture can inadvertently induce transdifferentiation of the plastic hPSCs, leading to the formation of extraembryonic lineages. Such manipulations can skew the assessment of stem cell potency, potentially leading to incorrect conclusions. For instance, the reported generation of trophoblast stem cells (TSCs) from naïve66,226228 and primed229 hPSCs involves complex signaling manipulations and prolonged culturing, which could be seen as artificial transdifferentiation events that do not align with transient embryonic development in vivo. To effectively develop differentiation systems that can assess the developmental potency of totipotent stem cells, two key principles should be followed: firstly, the formation of extraembryonic trophectoderm or primitive endoderm lineages should occur within a time frame comparable to in vivo development, avoiding long-term culturing; secondly, external factors that could induce cell fate transdifferentiation should be minimized. Adhering to these principles is crucial to ensure that the differentiation potential into extraembryonic lineages truly reflects the inherent totipotent characteristics of these stem cells. In this regard, in vitro embryo-like structures, for instance blastoids, could be potential models to test the developmental potency of a single totipotent stem cell. It is noteworthy that although single EPSCs, aided by helper cells, have been shown to form blastoids230, there remains conflicting evidence on the capability of mouse EPSCs to generate blastoids on their own230,231.

6. Emerging Concepts and Conclusion

Totipotent and pluripotent stem cells have been acclimated to artificial culture environments that differ significantly from their in vivo developmental niches. These cultures typically include supporting feeder cells, non-physiological biological matrices, excessive amounts of glucose and nutrients, and, notably for PSCs, a lack of interaction with extraembryonic cells. Recognizing these discrepancies, efforts have been made to optimize stem cell culture niches in vitro to better mimic in vivo conditions. These efforts have led to advancements in several areas, such as refining and identifying signaling pathways critical for the maintenance of totipotent and pluripotent stem cells, applying mechanobiological principles (such as substrate topography and stiffness), using 3D encapsulation and microcarriers232, and co-culturing with extraembryonic stem cells233. The potential benefits of these improvements for stem cell differentiation remain largely unexplored. Surprisingly, a study revealed that S/L culture is more effective than other chemical-based mESC cultures (2i/L, a2i/L and LCDM) in producing complete mice through tetraploid complementation, with higher survival rates to adulthood. Mice generated using 2/L culture exhibit no visible abnormalities for up to 2 years, unlike those from extended chemical-based cultures, which develop atypical teratomas or leiomyomas234. This success may be linked to the downregulation of metabolic pathways in S/L-mESCs compared to those in chemical-based mESC cultures234. Therefore, optimizing PSC cultures to reduce energy activity could be key to preserving and prolonging their developmental potential in vitro.

Interestingly, primed and intermediate/formative cultures are generally more universally applicable compared to naïve and totipotent conditions, allowing for the maintenance of PSCs across a broader range of species. This universal applicability could be due to stabler environmental conditions experienced by intermediate/formative and primed cells after embryo implantation, as opposed to the uterine environments to which naïve and totipotent cells are exposed, which differ among species. In line with this observation, changes in the transcriptomic profile of primate epiblasts have been reported upon implantation. Following implantation, the epiblast enters a phase of relative transcriptomic stability, which persists for approximately five days96,120.

Further studies have introduced an intriguing concept: some PSC cultures are effective in deriving and maintaining extraembryonic stem cells. For instance, one study showed that a defined medium containing Activin-A, CHIR, and LIF (ACL medium) enables the establishment of ACL-ESCs and ACL-Extraembryonic Endoderm (XEN) cells from a single blastocyst235. Additionally, another study found that an EPSC culture medium, LCDM, initially developed for mice and humans32, not only produced a similar type of PSCs in bovines236, but also supported the derivation and long-term culture of bovine TSCs237. Strikingly, the FTW medium, known for supporting the derivation of intermediate/formative FTW-ESCs from mouse and horse blastocysts35, enables de novo derivation and culture of ESCs, TSCs, and XENs from mouse and monkey blastocysts233. This advancement allows for the co-culture of embryonic and extraembryonic stem cells and reveals a growth inhibition effect exerted by extraembryonic endoderm cells on pluripotent cells, partly mediated by extracellular matrix signaling233.

The development of diverse totipotent and pluripotent stem cell cultures has greatly advanced the field of mammalian embryo modeling, especially for humans. It has led to the generation of pre-implantation and post-implantation embryo-like structures that mimic different aspects of early human development84,238245. To create human embryo models from cultured hPSCs, two methods are used: either aggregating cells of embryonic and extraembryonic origins243,244 or inducing differentiation and self-organization from a single PSC type84,241,242,245. The latter approach potentially provides a superior assay for functionally evaluating human pluripotency than the pre-existing embryoid body and teratoma assays. For instance, the generation of human blastoids238240, peri-gastruloids242 and gastruloids241 from naïve hPSCs, hEPSCs, and primed hPSCs, respectively, serves as a functional assessment of their developmental potency, which facilitates the staging of these hPSCs along the developmental axis. The differentiation of epiblast-like cells in these models occurs in conditions that, while not identical, are more akin to natural embryonic environments than random in vitro differentiation. They offer valuable insights into defining the human epiblast developmental trajectory. The success of this exciting intersection of stem cell biology with embryology requires a deeper understanding of the self-organization capabilities of spatiotemporally distinct early embryonic cells. In-depth insights are now more attainable due to the increasing variety of culture adaptations.

While artificially maintained, the dynamic totipotent and pluripotent stem cells offer an invaluable resource for studying early mammalian development, focusing on cell potency and cell plasticity, environmental and experimental perturbations, and in vitro state transitions. These cells also hold great potential to transform the fields of regenerative medicine and reproductive biology.

Acknowledgements

We would like to thank Hui Shen from the Du laboratory and Carlos A Pinzón-Arteaga from the Wu laboratory for their assistance in preparing the tables and figures. J.W. is a New York Stem Cell Foundation - Robertson Investigator and Virginia Murchison Linthicum Scholar in Medical Research. Research in the Wu laboratory is supported by National Institutes of Health (UM1HG011996, R01HD103627, and R01GM138565), NYSCF, ARSM, and The Welch Foundation (I-2088). Research in the Du laboratory is supported by the National Natural Science Foundation of China (32225017, 32050214 and 32090012) and the National Key Research and Development Program of China (2021YFA1100100 and 2019YFA0110000).

Footnotes

Declaration of Interests

Jun Wu is a member of Cell Stem Cell advisory board.

Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.

References:

  • 1.Plusa B, and Piliszek A. (2020). Common principles of early mammalian embryo self-organisation. Development 147. 10.1242/dev.183079. [DOI] [PubMed] [Google Scholar]
  • 2.Tam PP, and Loebel DA (2007). Gene function in mouse embryogenesis: get set for gastrulation. Nat Rev Genet 8, 368–381. 10.1038/nrg2084. [DOI] [PubMed] [Google Scholar]
  • 3.Baker CL, and Pera MF (2018). Capturing Totipotent Stem Cells. Cell Stem Cell 22, 25–34. 10.1016/j.stem.2017.12.011. [DOI] [PubMed] [Google Scholar]
  • 4.Wu J, and Belmonte JCI (2015). Dynamic Pluripotent Stem Cell States and Their Applications. Cell Stem Cell 17, 509–525. 10.1016/j.stem.2015.10.009. [DOI] [PubMed] [Google Scholar]
  • 5.Boroviak T, Loos R, Bertone P, Smith A, and Nichols J. (2014). The ability of inner-cell-mass cells to self-renew as embryonic stem cells is acquired following epiblast specification. Nature Cell Biology 16, 513-+. 10.1038/ncb2965. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6.Bradley A, Evans M, Kaufman MH, and Robertson E. (1984). Formation of germ-line chimaeras from embryo-derived teratocarcinoma cell lines. Nature 309, 255–256. 10.1038/309255a0. [DOI] [PubMed] [Google Scholar]
  • 7.Nagy A, Rossant J, Nagy R, Abramow-Newerly W, and Roder JC (1993). Derivation of completely cell culture-derived mice from early-passage embryonic stem cells. Proc Natl Acad Sci U S A 90, 8424–8428. 10.1073/pnas.90.18.8424. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.Evans MJ, and Kaufman MH (1981). Establishment in culture of pluripotential cells from mouse embryos. Nature 292, 154–156. 10.1038/292154a0. [DOI] [PubMed] [Google Scholar]
  • 9.Martin GR (1981). Isolation of a pluripotent cell line from early mouse embryos cultured in medium conditioned by teratocarcinoma stem cells. Proc Natl Acad Sci U S A 78, 7634–7638. 10.1073/pnas.78.12.7634. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.Smith AG, Heath JK, Donaldson DD, Wong GG, Moreau J, Stahl M, and Rogers D. (1988). Inhibition of pluripotential embryonic stem cell differentiation by purified polypeptides. Nature 336, 688–690. 10.1038/336688a0. [DOI] [PubMed] [Google Scholar]
  • 11.Williams RL, Hilton DJ, Pease S, Willson TA, Stewart CL, Gearing DP, Wagner EF, Metcalf D, Nicola NA, and Gough NM (1988). Myeloid leukaemia inhibitory factor maintains the developmental potential of embryonic stem cells. Nature 336, 684–687. 10.1038/336684a0. [DOI] [PubMed] [Google Scholar]
  • 12.Ying QL, Nichols J, Chambers I, and Smith A. (2003). BMP induction of Id proteins suppresses differentiation and sustains embryonic stem cell self-renewal in collaboration with STAT3. Cell 115, 281–292. 10.1016/s0092-8674(03)00847-x. [DOI] [PubMed] [Google Scholar]
  • 13.Ying QL, 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. 10.1038/nature06968. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.Burdon T, Stracey C, Chambers I, Nichols J, and Smith A. (1999). Suppression of SHP-2 and ERK signalling promotes self-renewal of mouse embryonic stem cells. Dev Biol 210, 30–43. 10.1006/dbio.1999.9265. [DOI] [PubMed] [Google Scholar]
  • 15.Toyooka Y, Shimosato D, Murakami K, Takahashi K, and Niwa H. (2008). Identification and characterization of subpopulations in undifferentiated ES cell culture. Development 135, 909–918. 10.1242/dev.017400. [DOI] [PubMed] [Google Scholar]
  • 16.Chambers I, Silva J, Colby D, Nichols J, Nijmeijer B, Robertson M, Vrana J, Jones K, Grotewold L, and Smith A. (2007). Nanog safeguards pluripotency and mediates germline development. Nature 450, 1230–1234. 10.1038/nature06403. [DOI] [PubMed] [Google Scholar]
  • 17.Marks H, Kalkan T, Menafra R, Denissov S, Jones K, Hofemeister H, Nichols J, Kranz A, Stewart AF, Smith A, and Stunnenberg HG (2012). The transcriptional and epigenomic foundations of ground state pluripotency. Cell 149, 590–604. 10.1016/j.cell.2012.03.026. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Hayashi K, de Sousa Lopes SMC, Tang F, Lao K, and Surani MA (2008). Dynamic equilibrium and heterogeneity of mouse pluripotent stem cells with distinct functional and epigenetic states. Cell Stem Cell 3, 391–401. 10.1016/j.stem.2008.07.027. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.Skelly DA, Czechanski A, Byers C, Aydin S, Spruce C, Olivier C, Choi K, Gatti DM, Raghupathy N, Keele GR, et al. (2020). Mapping the Effects of Genetic Variation on Chromatin State and Gene Expression Reveals Loci That Control Ground State Pluripotency. Cell Stem Cell 27, 459–469 e458. 10.1016/j.stem.2020.07.005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.Nichols J, Jones K, Phillips JM, Newland SA, Roode M, Mansfield W, Smith A, and Cooke A. (2009). Validated germline-competent embryonic stem cell lines from nonobese diabetic mice. Nat Med 15, 814–818. 10.1038/nm.1996. [DOI] [PubMed] [Google Scholar]
  • 21.Hanna J, Markoulaki S, Mitalipova M, Cheng AW, Cassady JP, Staerk J, Carey BW, Lengner CJ, Foreman R, Love J, et al. (2009). Metastable pluripotent states in NOD-mouse-derived ESCs. Cell Stem Cell 4, 513–524. 10.1016/j.stem.2009.04.015. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Ortmann D, Brown S, Czechanski A, Aydin S, Muraro D, Huang Y, Tomaz RA, Osnato A, Canu G, Wesley BT, et al. (2020). Naive Pluripotent Stem Cells Exhibit Phenotypic Variability that Is Driven by Genetic Variation. Cell Stem Cell 27, 470-+. 10.1016/j.stem.2020.07.019. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.Hackett JA, Kobayashi T, Dietmann S, and Surani MA (2017). Activation of Lineage Regulators and Transposable Elements across a Pluripotent Spectrum. Stem Cell Reports 8, 1645–1658. 10.1016/j.stemcr.2017.05.014. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Choi J, Huebner AJ, Clement K, Walsh RM, Savol A, Lin K, Gu H, Di Stefano B,Brumbaugh J, Kim SY, et al. (2017). Prolonged Mek1/2 suppression impairs the developmental potential of embryonic stem cells. Nature 548, 219–223. 10.1038/nature23274. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Yagi M, Kishigami S, Tanaka A, Semi K, Mizutani E, Wakayama S, Wakayama T, Yamamoto T, and Yamada Y. (2017). Derivation of ground-state female ES cells maintaining gamete-derived DNA methylation. Nature 548, 224–227. 10.1038/nature23286. [DOI] [PubMed] [Google Scholar]
  • 26.Chen H, Guo R, Zhang Q, Guo H, Yang M, Wu Z, Gao S, Liu L, and Chen L. (2015). Erk signaling is indispensable for genomic stability and self-renewal of mouse embryonic stem cells. Proc Natl Acad Sci U S A 112, E5936–5943. 10.1073/pnas.1516319112. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Shimizu T, Ueda J, Ho JC, Iwasaki K, Poellinger L, Harada I, and Sawada Y. (2012). Dual inhibition of Src and GSK3 maintains mouse embryonic stem cells, whose differentiation is mechanically regulated by Src signaling. Stem Cells 30, 1394–1404. 10.1002/stem.1119. [DOI] [PubMed] [Google Scholar]
  • 28.Li X, Zhu L, Yang A, Lin J, Tang F, Jin S, Wei Z, Li J, and Jin Y. (2011). Calcineurin-NFAT signaling critically regulates early lineage specification in mouse embryonic stem cells and embryos. Cell Stem Cell 8, 46–58. 10.1016/j.stem.2010.11.027. [DOI] [PubMed] [Google Scholar]
  • 29.Blaschke K, Ebata KT, Karimi MM, Zepeda-Martinez JA, Goyal P, Mahapatra S, Tam A, Laird DJ, Hirst M, Rao A, et al. (2013). Vitamin C induces Tet-dependent DNA demethylation and a blastocyst-like state in ES cells. Nature 500, 222–226. 10.1038/nature12362. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.Dutta D, Ray S, Home P, Larson M, Wolfe MW, and Paul S. (2011). Self-Renewal Versus Lineage Commitment of Embryonic Stem Cells: Protein Kinase C Signaling Shifts the Balance. Stem Cells 29, 618–628. 10.1002/stem.605. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Lynch CJ, Bernad R, Martínez-Val A, Shahbazi MN, Nóbrega-Pereira S, Calvo I, Blanco-Aparicio C, Tarantino C, Garreta E, Richart-Ginés L, et al. (2020). Global hyperactivation of enhancers stabilizes human and mouse naive pluripotency through inhibition of CDK8/19 Mediator kinases. Nature Cell Biology 22, 1223-+. 10.1038/s41556-020-0573-1. [DOI] [PubMed] [Google Scholar]
  • 32.Yang Y, Liu B, Xu J, Wang J, Wu J, Shi C, Xu Y, Dong J, Wang C, Lai W, et al. (2017). Derivation of Pluripotent Stem Cells with In Vivo Embryonic and Extraembryonic Potency. Cell 169, 243–257 e225. 10.1016/j.cell.2017.02.005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Yang J, Ryan DJ, Wang W, Tsang JC, Lan G, Masaki H, Gao X, Antunes L, Yu Y, Zhu Z, et al. (2017). Establishment of mouse expanded potential stem cells. Nature 550, 393–397. 10.1038/nature24052. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Posfai E, Schell JP, 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. 10.1038/s41556-020-00609-2. [DOI] [PubMed] [Google Scholar]
  • 35.Yu L, Wei Y, Sun HX, Mahdi AK, Pinzon Arteaga CA, Sakurai M, Schmitz DA, Zheng C, Ballard ED, Li J, et al. (2021). Derivation of Intermediate Pluripotent Stem Cells Amenable to Primordial Germ Cell Specification. Cell Stem Cell 28, 550–567 e512. 10.1016/j.stem.2020.11.003. [DOI] [PubMed] [Google Scholar]
  • 36.Buehr M, Meek S, Blair K, Yang J, Ure J, Silva J, McLay R, Hall J, Ying QL, and Smith A. (2008). Capture of authentic embryonic stem cells from rat blastocysts. Cell 135, 1287–1298. 10.1016/j.cell.2008.12.007. [DOI] [PubMed] [Google Scholar]
  • 37.Li P, Tong C, Mehrian-Shai R, Jia L, Wu N, Yan Y, Maxson RE, Schulze EN, Song H, Hsieh CL, et al. (2008). Germline competent embryonic stem cells derived from rat blastocysts. Cell 135, 1299–1310. 10.1016/j.cell.2008.12.006. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.Tong C, Li P, Wu NL, Yan Y, and Ying QL (2010). Production of p53 gene knockout rats by homologous recombination in embryonic stem cells. Nature 467, 211–213. 10.1038/nature09368. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.Lee J, Wang J, Ally R, Trzaska S, Hickey J, Mujica A, Miloscio L, Mastaitis J, Morse B, Smith J, et al. (2023). Production of large, defined genome modifications in rats by targeting rat embryonic stem cells. Stem Cell Reports 18, 394–409. 10.1016/j.stemcr.2022.11.012. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.Kawamata M, and Ochiya T. (2010). Generation of genetically modified rats from embryonic stem cells. Proc Natl Acad Sci U S A 107, 14223–14228. 10.1073/pnas.1009582107. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41.Chen YY, Blair K, and Smith A. (2013). Robust Self-Renewal of Rat Embryonic Stem Cells Requires Fine-Tuning of Glycogen Synthase Kinase-3 Inhibition. Stem Cell Reports 1, 209–217. 10.1016/j.stemcr.2013.07.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42.Meek S, Wei J, Sutherland L, Nilges B, Buehr M, Tomlinson SR, Thomson AJ, and Burdon T. (2013). Tuning of β-catenin activity is required to stabilize self-renewal of rat embryonic stem cells. Stem Cells 31, 2104–2115. 10.1002/stem.1466. [DOI] [PubMed] [Google Scholar]
  • 43.Li TD, Feng GH, Li YF, Wang M, Mao JJ, Wang JQ, Li X, Wang XP, Qu B, Wang LY, et al. (2017). Rat embryonic stem cells produce fertile offspring through tetraploid complementation. P Natl Acad Sci USA 114, 11974–11979. 10.1073/pnas.1708710114. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44.Rajendran G, Dutta D, Hong J, Paul A, Saha B, Mahato B, Ray S, Home P, Ganguly A, Weiss ML, and Paul S. (2013). Inhibition of protein kinase C signaling maintains rat embryonic stem cell pluripotency. J Biol Chem 288, 24351–24362. 10.1074/jbc.M113.455725. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45.Li W, Wei W, Zhu S, Zhu J, Shi Y, Lin T, Hao E, Hayek A, Deng H, and Ding S. (2009). Generation of rat and human induced pluripotent stem cells by combining genetic reprogramming and chemical inhibitors. Cell Stem Cell 4, 16–19. 10.1016/j.stem.2008.11.014. [DOI] [PubMed] [Google Scholar]
  • 46.Buecker C, Chen HH, Polo JM, Daheron L, Bu L, Barakat TS, Okwieka P, Porter A, Gribnau J, Hochedlinger K, and Geijsen N. (2010). A murine ESC-like state facilitates transgenesis and homologous recombination in human pluripotent stem cells. Cell Stem Cell 6, 535–546. 10.1016/j.stem.2010.05.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47.Valamehr B, Robinson M, Abujarour R, Rezner B, Vranceanu F, Le T, Medcalf A, Lee TT, Fitch M, Robbins D, and Flynn P. (2014). Platform for induction and maintenance of transgene-free hiPSCs resembling ground state pluripotent stem cells. Stem Cell Reports 2, 366–381. 10.1016/j.stemcr.2014.01.014. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.Hanna J, Cheng AW, Saha K, Kim J, Lengner CJ, Soldner F, Cassady JP, Muffat J, Carey BW, and Jaenisch R. (2010). Human embryonic stem cells with biological and epigenetic characteristics similar to those of mouse ESCs. P Natl Acad Sci USA 107, 9222–9227. 10.1073/pnas.1004584107. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49.Theunissen TW, Powell BE, Wang H, Mitalipova M, Faddah DA, Reddy J, Fan ZP, Maetzel D, Ganz K, Shi L, et al. (2014). Systematic identification of culture conditions for induction and maintenance of naive human pluripotency. Cell Stem Cell 15, 471–487. 10.1016/j.stem.2014.07.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50.Takashima Y, Guo G, Loos R, Nichols J, Ficz G, Krueger F, Oxley D, Santos F, Clarke J, Mansfield W, et al. (2014). Resetting transcription factor control circuitry toward ground-state pluripotency in human. Cell 158, 1254–1269. 10.1016/j.cell.2014.08.029. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51.Chen H, Aksoy I, Gonnot F, Osteil P, Aubry M, Hamela C, Rognard C, Hochard A, Voisin S, Fontaine E, et al. (2015). Reinforcement of STAT3 activity reprogrammes human embryonic stem cells to naive-like pluripotency. Nat Commun 6, 7095. 10.1038/ncomms8095. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52.Qin H, Hejna M, Liu Y, Percharde M, Wossidlo M, Blouin L, Durruthy-Durruthy J, Wong P, Qi Z, Yu J, et al. (2016). YAP Induces Human Naive Pluripotency. Cell Rep 14, 2301–2312. 10.1016/j.celrep.2016.02.036. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53.Gafni O, Weinberger L, Mansour AA, Manor YS, Chomsky E, Ben-Yosef D, Kalma Y, Viukov S, Maza I, Zviran A, et al. (2013). Derivation of novel human ground state naive pluripotent stem cells. Nature 504, 282–286. 10.1038/nature12745. [DOI] [PubMed] [Google Scholar]
  • 54.Ware CB, Nelson AM, Mecham B, Hesson J, Zhou W, Jonlin EC, Jimenez-Caliani AJ, Deng X, Cavanaugh C, Cook S, et al. (2014). Derivation of naive human embryonic stem cells. Proc Natl Acad Sci U S A 111, 4484–4489. 10.1073/pnas.1319738111. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55.Chan YS, Göke J, Ng JH, Lu XY, Gonzales KAU, Tan CP, Tng WQ, Hong ZZ, Lim YS, and Ng HH (2013). Induction of a Human Pluripotent State with Distinct Regulatory Circuitry that Resembles Preimplantation Epiblast. Cell Stem Cell 13, 663–675. 10.1016/j.stem.2013.11.015. [DOI] [PubMed] [Google Scholar]
  • 56.Duggal G, Warrier S, Ghimire S, Broekaert D, Van der Jeught M, Lierman S, Deroo T, Peelman L, Van Soom A, Cornelissen R, et al. (2015). Alternative Routes to Induce Naïve Pluripotency in Human Embryonic Stem Cells. Stem Cells 33, 2686–2698. 10.1002/stem.2071. [DOI] [PubMed] [Google Scholar]
  • 57.Gao X, Nowak-Imialek M, Chen X, Chen D, Herrmann D, Ruan D, Chen ACH, Eckersley-Maslin MA, Ahmad S, Lee YL, et al. (2019). Establishment of porcine and human expanded potential stem cells. Nat Cell Biol 21, 687–699. 10.1038/s41556-019-0333-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58.Bredenkamp N, Yang J, Clarke J, Stirparo GG, von Meyenn F, Dietmann S, Baker D, Drummond R, Ren Y, Li D, et al. (2019). Wnt Inhibition Facilitates RNA-Mediated Reprogramming of Human Somatic Cells to Naive Pluripotency. Stem Cell Reports 13, 1083–1098. 10.1016/j.stemcr.2019.10.009. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59.Bayerl J, Ayyash M, Shani T, Manor YS, Gafni O, Massarwa R, Kalma Y, Aguilera-Castrejon A, Zerbib M, Amir H, et al. (2021). Principles of signaling pathway modulation for enhancing human naive pluripotency induction. Cell Stem Cell 28, 1549–1565 e1512. 10.1016/j.stem.2021.04.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60.Di Stefano B, Ueda M, Sabri S, Brumbaugh J, Huebner AJ, Sahakyan A, Clement K, Clowers KJ, Erickson AR, Shioda K, et al. (2018). Reduced MEK inhibition preserves genomic stability in naive human embryonic stem cells. Nat Methods 15, 732–740. 10.1038/s41592-018-0104-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61.Khan SA, Park KM, Fischer LA, Dong C, Lungjangwa T, Jimenez M, Casalena D, Chew B, Dietmann S, Auld DS, et al. (2021). Probing the signaling requirements for naive human pluripotency by high-throughput chemical screening. Cell Rep 35, 109233. 10.1016/j.celrep.2021.109233. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62.Hu Z, Li H, Jiang H, Ren Y, Yu X, Qiu J, Stablewski AB, Zhang B, Buck MJ, and Feng J. (2020). Transient inhibition of mTOR in human pluripotent stem cells enables robust formation of mouse-human chimeric embryos. Sci Adv 6, eaaz0298. 10.1126/sciadv.aaz0298. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63.Mazid MA, Ward C, Luo Z, Liu C, Li Y, Lai Y, Wu L, Li J, Jia W, Jiang Y, et al. (2022). Rolling back human pluripotent stem cells to an eight-cell embryo-like stage. Nature 605, 315–324. 10.1038/s41586-022-04625-0. [DOI] [PubMed] [Google Scholar]
  • 64.Ai Z, Niu B, Yin Y, Xiang L, Shi G, Duan K, Wang S, Hu Y, Zhang C, Zhang C, et al. (2023). Dissecting peri-implantation development using cultured human embryos and embryo-like assembloids. Cell Res 33, 661–678. 10.1038/s41422-023-00846-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65.Yu X, Liang S, Chen M, Yu H, Li R, Qu Y, Kong X, Guo R, Zheng R, Izsvak Z, et al. (2022). Recapitulating early human development with 8C-like cells. Cell Rep 39, 110994. 10.1016/j.celrep.2022.110994. [DOI] [PubMed] [Google Scholar]
  • 66.Guo G, Stirparo GG, Strawbridge SE, Spindlow D, Yang J, Clarke J, Dattani A, Yanagida A, Li MA, Myers S, et al. (2021). Human naive epiblast cells possess unrestricted lineage potential. Cell Stem Cell 28, 1040–1056 e1046. 10.1016/j.stem.2021.02.025. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 67.Chen Y, Niu Y, Li Y, Ai Z, Kang Y, Shi H, Xiang Z, Yang Z, Tan T, Si W, et al. (2015). Generation of Cynomolgus Monkey Chimeric Fetuses using Embryonic Stem Cells. Cell Stem Cell 17, 116–124. 10.1016/j.stem.2015.06.004. [DOI] [PubMed] [Google Scholar]
  • 68.Aksoy I, Rognard C, Moulin A, Marcy G, Masfaraud E, Wianny F, Cortay V, Bellemin-Menard A, Doerflinger N, Dirheimer M, et al. (2021). Apoptosis, G1 Phase Stall, and Premature Differentiation Account for Low Chimeric Competence of Human and Rhesus Monkey Naive Pluripotent Stem Cells. Stem Cell Reports 16, 56–74. 10.1016/j.stemcr.2020.12.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 69.Cao J, Li W, Li J, Mazid MA, Li C, Jiang Y, Jia W, Wu L, Liao Z, Sun S, et al. (2023). Live birth of chimeric monkey with high contribution from embryonic stem cells. Cell 186, 4996–5014 e4924. 10.1016/j.cell.2023.10.005. [DOI] [PubMed] [Google Scholar]
  • 70.Fang R, Liu K, Zhao Y, Li H, Zhu D, Du Y, Xiang C, Li X, Liu H, Miao Z, et al. (2014). Generation of naive induced pluripotent stem cells from rhesus monkey fibroblasts. Cell Stem Cell 15, 488–497. 10.1016/j.stem.2014.09.004. [DOI] [PubMed] [Google Scholar]
  • 71.Brons IG, Smithers LE, Trotter MW, Rugg-Gunn P, Sun B, Chuva de Sousa Lopes SM, Howlett SK, Clarkson A, Ahrlund-Richter L, Pedersen RA, and Vallier L. (2007). Derivation of pluripotent epiblast stem cells from mammalian embryos. Nature 448, 191–195. 10.1038/nature05950. [DOI] [PubMed] [Google Scholar]
  • 72.Tesar PJ, Chenoweth JG, Brook FA, Davies TJ, Evans EP, Mack DL, Gardner RL, and McKay RD (2007). New cell lines from mouse epiblast share defining features with human embryonic stem cells. Nature 448, 196–199. 10.1038/nature05972. [DOI] [PubMed] [Google Scholar]
  • 73.Najm FJ, Chenoweth JG, Anderson PD, Nadeau JH, Redline RW, McKay RD, and Tesar PJ (2011). Isolation of epiblast stem cells from preimplantation mouse embryos. Cell Stem Cell 8, 318–325. 10.1016/j.stem.2011.01.016. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 74.Kojima Y, Kaufman-Francis K, Studdert JB, Steiner KA, Power MD, Loebel DA, Jones V, Hor A, de Alencastro G, Logan GJ, et al. (2014). The transcriptional and functional properties of mouse epiblast stem cells resemble the anterior primitive streak. Cell Stem Cell 14, 107–120. 10.1016/j.stem.2013.09.014. [DOI] [PubMed] [Google Scholar]
  • 75.Han DW, Tapia N, Joo JY, Greber B, Arauzo-Bravo MJ, Bernemann C, Ko K, Wu G, Stehling M, Do JT, and Scholer HR (2010). Epiblast stem cell subpopulations represent mouse embryos of distinct pregastrulation stages. Cell 143, 617–627. 10.1016/j.cell.2010.10.015. [DOI] [PubMed] [Google Scholar]
  • 76.Bernemann C, Greber B, Ko K, Sterneckert J, Han DW, Arauzo-Bravo MJ, and Scholer HR (2011). Distinct developmental ground states of epiblast stem cell lines determine different pluripotency features. Stem Cells 29, 1496–1503. 10.1002/stem.709. [DOI] [PubMed] [Google Scholar]
  • 77.Tsakiridis A, Huang Y, Blin G, Skylaki S, Wymeersch F, Osorno R, Economou C, Karagianni E, Zhao S, Lowell S, and Wilson V. (2014). Distinct Wnt-driven primitive streak-like populations reflect in vivo lineage precursors. Development 141, 1209–1221. 10.1242/dev.101014. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 78.Sumi T, Oki S, Kitajima K, and Meno C. (2013). Epiblast ground state is controlled by canonical Wnt/beta-catenin signaling in the postimplantation mouse embryo and epiblast stem cells. PLoS One 8, e63378. 10.1371/journal.pone.0063378. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 79.Wu J, Okamura D, Li M, Suzuki K, Luo C, Ma L, He Y, Li Z, Benner C, Tamura I, et al. (2015). An alternative pluripotent state confers interspecies chimaeric competency. Nature 521, 316–321. 10.1038/nature14413. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 80.Ludwig TE, Bergendahl V, Levenstein ME, Yu J, Probasco MD, and Thomson JA (2006). Feeder-independent culture of human embryonic stem cells. Nat Methods 3, 637–646. 10.1038/nmeth902. [DOI] [PubMed] [Google Scholar]
  • 81.Chia NY, Chan YS, Feng B, Lu X, Orlov YL, Moreau D, Kumar P, Yang L, Jiang J, Lau MS, et al. (2010). A genome-wide RNAi screen reveals determinants of human embryonic stem cell identity. Nature 468, 316–320. 10.1038/nature09531. [DOI] [PubMed] [Google Scholar]
  • 82.Chen D, Sun N, Hou L, Kim R, Faith J, Aslanyan M, Tao Y, Zheng Y, Fu J, Liu W, et al. (2019). Human Primordial Germ Cells Are Specified from Lineage-Primed Progenitors. Cell Rep 29, 4568–4582 e4565. 10.1016/j.celrep.2019.11.083. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 83.Sasaki K, Yokobayashi S, Nakamura T, Okamoto I, Yabuta Y, Kurimoto K, Ohta H, Moritoki Y, Iwatani C, Tsuchiya H, et al. (2015). Robust In Vitro Induction of Human Germ Cell Fate from Pluripotent Stem Cells. Cell Stem Cell 17, 178–194. 10.1016/j.stem.2015.06.014. [DOI] [PubMed] [Google Scholar]
  • 84.Zheng Y, Xue X, Shao Y, Wang S, Esfahani SN, Li Z, Muncie JM, Lakins JN, Weaver VM, Gumucio DL, and Fu J. (2019). Controlled modelling of human epiblast and amnion development using stem cells. Nature 573, 421–425. 10.1038/s41586-019-1535-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 85.Kurek D, Neagu A, Tastemel M, Tuysuz N, Lehmann J, van de Werken HJG, Philipsen S, van der Linden R, Maas A, van IWFJ, et al. (2015). Endogenous WNT signals mediate BMP-induced and spontaneous differentiation of epiblast stem cells and human embryonic stem cells. Stem Cell Reports 4, 114–128. 10.1016/j.stemcr.2014.11.007. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 86.Ben-Nun IF, Montague SC, Houck ML, Tran HT, Garitaonandia I, Leonardo TR, Wang YC, Charter SJ, Laurent LC, Ryder OA, and Loring JF (2011). Induced pluripotent stem cells from highly endangered species. Nature Methods 8, 829–U889. 10.1038/Nmeth.1706. [DOI] [PubMed] [Google Scholar]
  • 87.Déjosez M, Marin A, Hughes GM, Morales AE, Godoy-Parejo C, Gray JL, Qin YR, Singh AA, Xu H, Juste J, et al. (2023). Bat pluripotent stem cells reveal unusual entanglement between host and viruses. Cell 186, 957-+. 10.1016/j.cell.2023.01.011. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 88.Zheng C, Hu Y, Sakurai M, Pinzon-Arteaga CA, Li J, Wei Y, Okamura D, Ravaux B, Barlow HR, Yu L, et al. (2021). Cell competition constitutes a barrier for interspecies chimerism. Nature 592, 272–276. 10.1038/s41586-021-03273-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 89.Iwatsuki K, Oikawa M, Kobayashi H, Penfold CA, Sanbo M, Yamamoto T, Hochi S, Kurimoto K, Hirabayashi M, and Kobayashi T. (2023). Rat post-implantation epiblast-derived pluripotent stem cells produce functional germ cells. Cell Rep Methods 3, 100542. 10.1016/j.crmeth.2023.100542. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 90.Bogliotti YS, Wu J, Vilarino M, Okamura D, Soto DA, Zhong C, Sakurai M, Sampaio RV, Suzuki K, Izpisua Belmonte JC, and Ross PJ (2018). Efficient derivation of stable primed pluripotent embryonic stem cells from bovine blastocysts. Proc Natl Acad Sci U S A 115, 2090–2095. 10.1073/pnas.1716161115. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 91.Soto DA, Navarro M, Zheng C, Halstead MM, Zhou C, Guiltinan C, Wu J, and Ross PJ (2021). Simplification of culture conditions and feeder-free expansion of bovine embryonic stem cells. Sci Rep 11, 11045. 10.1038/s41598-021-90422-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 92.Kinoshita M, Kobayashi T, Planells B, Klisch D, Spindlow D, Masaki H, Bornelov S, Stirparo GG, Matsunari H, Uchikura A, et al. (2021). Pluripotent stem cells related to embryonic disc exhibit common self-renewal requirements in diverse livestock species. Development 148. 10.1242/dev.199901. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 93.Vilarino M, Alba Soto D, Soledad Bogliotti Y, Yu L, Zhang Y, Wang C, Paulson E, Zhong C, Jin M, Carlos Izpisua Belmonte J, et al. (2020). Derivation of sheep embryonic stem cells under optimized conditions. Reproduction 160, 761–772. 10.1530/REP-19-0606. [DOI] [PubMed] [Google Scholar]
  • 94.Kobayashi T, Castillo-Venzor A, Penfold CA, Morgan M, Mizuno N, Tang WWC, Osada Y, Hirao M, Yoshida F, Sato H, et al. (2021). Tracing the emergence of primordial germ cells from bilaminar disc rabbit embryos and pluripotent stem cells. Cell Rep 37, 109812. 10.1016/j.celrep.2021.109812. [DOI] [PubMed] [Google Scholar]
  • 95.Sakai Y, Nakamura T, Okamoto I, Gyobu-Motani S, Ohta H, Yabuta Y, Tsukiyama T, Iwatani C, Tsuchiya H, Ema M, et al. (2020). Induction of the germ cell fate from pluripotent stem cells in cynomolgus monkeysdagger. Biol Reprod 102, 620–638. 10.1093/biolre/ioz205. [DOI] [PubMed] [Google Scholar]
  • 96.Pera MF, and Rossant J. (2021). The exploration of pluripotency space: Charting cell state transitions in peri-implantation development. Cell Stem Cell 28, 1896–1906. 10.1016/j.stem.2021.10.001. [DOI] [PubMed] [Google Scholar]
  • 97.Chang KH, and Li M. (2013). Clonal isolation of an intermediate pluripotent stem cell state. Stem Cells 31, 918–927. 10.1002/stem.1330. [DOI] [PubMed] [Google Scholar]
  • 98.Bao S, Tang WW, Wu B, Kim S, Li J, Li L, Kobayashi T, Lee C, Chen Y, Wei M, et al. (2018). Derivation of hypermethylated pluripotent embryonic stem cells with high potency. Cell Res 28, 22–34. 10.1038/cr.2017.134. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 99.Du P, Pirouz M, Choi J, Huebner AJ, Clement K, Meissner A, Hochedlinger K, and Gregory RI (2018). An Intermediate Pluripotent State Controlled by MicroRNAs Is Required for the Naive-to-Primed Stem Cell Transition. Cell Stem Cell 22, 851–864 e855. 10.1016/j.stem.2018.04.021. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 100.Tsukiyama T, and Ohinata Y. (2014). A Modified EpiSC Culture Condition Containing a GSK3 Inhibitor Can Support Germline-Competent Pluripotency in Mice. Plos One 9. ARTN e95329 10.1371/journal.pone.0095329. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 101.Neagu A, van Genderen E, Escudero I, Verwegen L, Kurek D, Lehmann J, Stel J, Dirks RAM, van Mierlo G, Maas A, et al. (2020). In vitro capture and characterization of embryonic rosette-stage pluripotency between naive and primed states. Nat Cell Biol 22, 534–545. 10.1038/s41556-020-0508-x. [DOI] [PubMed] [Google Scholar]
  • 102.Kinoshita M, Barber M, Mansfield W, Cui Y, Spindlow D, Stirparo GG, Dietmann S, Nichols J, and Smith A. (2021). Capture of Mouse and Human Stem Cells with Features of Formative Pluripotency. Cell Stem Cell 28, 453–471 e458. 10.1016/j.stem.2020.11.005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 103.Wang X, Xiang Y, Yu Y, Wang R, Zhang Y, Xu Q, Sun H, Zhao ZA, Jiang X, Wang X, et al. (2021). Formative pluripotent stem cells show features of epiblast cells poised for gastrulation. Cell Res 31, 526–541. 10.1038/s41422-021-00477-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 104.Ohinata Y, Ohta H, Shigeta M, Yamanaka K, Wakayama T, and Saitou M. (2009). A signaling principle for the specification of the germ cell lineage in mice. Cell 137, 571–584. 10.1016/j.cell.2009.03.014. [DOI] [PubMed] [Google Scholar]
  • 105.Luo Q, Pui HP, Chen J, Yu L, Jannig PR, Pei Y, Zhao L, Chen X, Petropoulos S, Ruas JL, et al. (2023). Epiblast-like stem cells established by Wnt/beta-catenin signaling manifest distinct features of formative pluripotency and germline competence. Cell Rep 42, 112021. 10.1016/j.celrep.2023.112021. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 106.Joo JY, Choi HW, Kim MJ, Zaehres H, Tapia N, Stehling M, Jung KS, Do JT, and Scholer HR (2014). Establishment of a primed pluripotent epiblast stem cell in FGF4-based conditions. Sci Rep 4, 7477. 10.1038/srep07477. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 107.Kim H, Wu J, Ye S, Tai CI, Zhou X, Yan H, Li P, Pera M, and Ying QL (2013). Modulation of beta-catenin function maintains mouse epiblast stem cell and human embryonic stem cell self-renewal. Nat Commun 4, 2403. 10.1038/ncomms3403. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 108.Smith A. (2017). Formative pluripotency: the executive phase in a developmental continuum. Development 144, 365–373. 10.1242/dev.142679. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 109.Bedzhov I, and Zernicka-Goetz M. (2014). Self-organizing properties of mouse pluripotent cells initiate morphogenesis upon implantation. Cell 156, 1032–1044. 10.1016/j.cell.2014.01.023. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 110.Mulas C, Kalkan T, and Smith A. (2017). NODAL Secures Pluripotency upon Embryonic Stem Cell Progression from the Ground State. Stem Cell Reports 9, 77–91. 10.1016/j.stemcr.2017.05.033. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 111.Molotkov A, Mazot P, Brewer JR, Cinalli RM, and Soriano P. (2017). Distinct Requirements for FGFR1 and FGFR2 in Primitive Endoderm Development and Exit from Pluripotency. Dev Cell 41, 511–526 e514. 10.1016/j.devcel.2017.05.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 112.Nakanishi M, Mitchell RR, Benoit YD, Orlando L, Reid JC, Shimada K, Davidson KC, Shapovalova Z, Collins TJ, Nagy A, and Bhatia M. (2019). Human Pluripotency Is Initiated and Preserved by a Unique Subset of Founder Cells. Cell 177, 910–924 e922. 10.1016/j.cell.2019.03.013. [DOI] [PubMed] [Google Scholar]
  • 113.Cornacchia D, Zhang C, Zimmer B, Chung SY, Fan Y, Soliman MA, Tchieu J, Chambers SM, Shah H, Paull D, et al. (2019). Lipid Deprivation Induces a Stable, Naive-to-Primed Intermediate State of Pluripotency in Human PSCs. Cell Stem Cell 25, 120–136 e110. 10.1016/j.stem.2019.05.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 114.Lau KX, Mason EA, Kie J, De Souza DP, Kloehn J, Tull D, McConville MJ, Keniry A, Beck T, Blewitt ME, et al. (2020). Unique properties of a subset of human pluripotent stem cells with high capacity for self-renewal. Nat Commun 11, 2420. 10.1038/s41467-020-16214-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 115.Gropp M, Waldhorn I, Gil Y, Steiner D, Turetsky TT, Smith Y, Sabag O, Falick-Michaeli T, Even Ram S, and Reubinoff BE (2022). Laminin111-based defined culture promoting self-renewing human pluripotent stem cells with properties of the early post-implantation epiblast. Stem Cell Reports 17, 2643–2660. 10.1016/j.stemcr.2022.10.010. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 116.Linneberg-Agerholm M, Wong YF, Romero Herrera JA, Monteiro RS, Anderson KGV, and Brickman JM (2019). Naive human pluripotent stem cells respond to Wnt, Nodal and LIF signalling to produce expandable naive extra-embryonic endoderm. Development 146. 10.1242/dev.180620. [DOI] [PubMed] [Google Scholar]
  • 117.Wu J, Platero-Luengo A, Sakurai M, Sugawara A, Gil MA, Yamauchi T, Suzuki K, Bogliotti YS, Cuello C, Morales Valencia M, et al. (2017). Interspecies Chimerism with Mammalian Pluripotent Stem Cells. Cell 168, 473–486 e415. 10.1016/j.cell.2016.12.036. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 118.Liu D, Wang X, He D, Sun C, He X, Yan L, Li Y, Han JJ, and Zheng P. (2018). Single-cell RNA-sequencing reveals the existence of naive and primed pluripotency in pre-implantation rhesus monkey embryos. Genome Res 28, 1481–1493. 10.1101/gr.233437.117. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 119.Mole MA, Coorens THH, Shahbazi MN, Weberling A, Weatherbee BAT, Gantner CW, Sancho-Serra C, Richardson L, Drinkwater A, Syed N, et al. (2021). A single cell characterisation of human embryogenesis identifies pluripotency transitions and putative anterior hypoblast centre. Nat Commun 12, 3679. 10.1038/s41467-021-23758-w. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 120.Nakamura T, Okamoto I, Sasaki K, Yabuta Y, Iwatani C, Tsuchiya H, Seita Y, Nakamura S, Yamamoto T, and Saitou M. (2016). A developmental coordinate of pluripotency among mice, monkeys and humans. Nature 537, 57–62. 10.1038/nature19096. [DOI] [PubMed] [Google Scholar]
  • 121.Nichols J, and Smith A. (2009). Naive and primed pluripotent states. Cell Stem Cell 4, 487–492. 10.1016/j.stem.2009.05.015. [DOI] [PubMed] [Google Scholar]
  • 122.Zhou J, Hu J, Wang Y, and Gao S. (2023). Induction and application of human naive pluripotency. Cell Rep 42, 112379. 10.1016/j.celrep.2023.112379. [DOI] [PubMed] [Google Scholar]
  • 123.Weinberger L, Ayyash M, Novershtern N, and Hanna JH (2016). Dynamic stem cell states: naive to primed pluripotency in rodents and humans. Nat Rev Mol Cell Biol 17, 155–169. 10.1038/nrm.2015.28. [DOI] [PubMed] [Google Scholar]
  • 124.Boroviak T, and Nichols J. (2017). Primate embryogenesis predicts the hallmarks of human naive pluripotency. Development 144, 175–186. 10.1242/dev.145177. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 125.Dong C, Fischer LA, and Theunissen TW (2019). Recent insights into the naive state of human pluripotency and its applications. Exp Cell Res 385. ARTN 111645 10.1016/j.yexcr.2019.111645. [DOI] [PubMed] [Google Scholar]
  • 126.Morgani S, Nichols J, and Hadjantonakis AK (2017). The many faces of Pluripotency: in vitro adaptations of a continuum of in vivo states. BMC Dev Biol 17, 7. 10.1186/s12861-017-0150-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 127.Hackett JA, and Surani MA (2014). Regulatory principles of pluripotency: from the ground state up. Cell Stem Cell 15, 416–430. 10.1016/j.stem.2014.09.015. [DOI] [PubMed] [Google Scholar]
  • 128.De Los Angeles A, Ferrari F, Xi R, Fujiwara Y, Benvenisty N, Deng H, Hochedlinger K, Jaenisch R, Lee S, Leitch HG, et al. (2015). Hallmarks of pluripotency. Nature 525, 469–478. 10.1038/nature15515. [DOI] [PubMed] [Google Scholar]
  • 129.Yilmaz A, and Benvenisty N. (2019). Defining Human Pluripotency. Cell Stem Cell 25, 9–22. 10.1016/j.stem.2019.06.010. [DOI] [PubMed] [Google Scholar]
  • 130.Roode M, Blair K, Snell P, Elder K, Marchant S, Smith A, and Nichols J. (2012). Human hypoblast formation is not dependent on FGF signalling. Dev Biol 361, 358–363. 10.1016/j.ydbio.2011.10.030. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 131.Boroviak T, Loos R, Lombard P, Okahara J, Behr R, Sasaki E, Nichols J, Smith A, and Bertone P. (2015). Lineage-Specific Profiling Delineates the Emergence and Progression of Naive Pluripotency in Mammalian Embryogenesis. Dev Cell 35, 366–382. 10.1016/j.devcel.2015.10.011. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 132.Kuijk EW, van Tol LT, Van de Velde H, Wubbolts R, Welling M, Geijsen N, and Roelen BA (2012). The roles of FGF and MAP kinase signaling in the segregation of the epiblast and hypoblast cell lineages in bovine and human embryos. Development 139, 871–882. 10.1242/dev.071688. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 133.Nichols J, Silva J, Roode M, and Smith A. (2009). Suppression of Erk signalling promotes ground state pluripotency in the mouse embryo. Development 136, 3215–3222. 10.1242/dev.038893. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 134.Osnato A, Brown S, Krueger C, Andrews S, Collier AJ, Nakanoh S, Londoño MQ, Wesley BT, Muraro D, Brumm AS, et al. (2021). TGFβ signalling is required to maintain pluripotency of human naive pluripotent stem cells. Elife 10. ARTN e67259 10.7554/eLife.67259. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 135.Blakeley P, Fogarty NM, del Valle I, Wamaitha SE, Hu TX, Elder K, Snell P, Christie L, Robson P, and Niakan KK (2015). Defining the three cell lineages of the human blastocyst by single-cell RNA-seq. Development 142, 3151–3165. 10.1242/dev.123547. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 136.Petropoulos S, Edsgard D, Reinius B, Deng Q, Panula SP, Codeluppi S, Plaza Reyes A, Linnarsson S, Sandberg R, and Lanner F. (2016). Single-Cell RNA-Seq Reveals Lineage and X Chromosome Dynamics in Human Preimplantation Embryos. Cell 165, 1012–1026. 10.1016/j.cell.2016.03.023. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 137.Mittnenzweig M, Mayshar Y, Cheng S, Ben-Yair R, Hadas R, Rais Y, Chomsky E, Reines N, Uzonyi A, Lumerman L, et al. (2021). A single-embryo, single-cell time-resolved model for mouse gastrulation. Cell 184, 2825–2842 e2822. 10.1016/j.cell.2021.04.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 138.Niu Y, Sun N, Li C, Lei Y, Huang Z, Wu J, Si C, Dai X, Liu C, Wei J, et al. (2019). Dissecting primate early post-implantation development using long-term in vitro embryo culture. Science 366. 10.1126/science.aaw5754. [DOI] [PubMed] [Google Scholar]
  • 139.Ma H, Zhai J, Wan H, Jiang X, Wang X, Wang L, Xiang Y, He X, Zhao ZA, Zhao B, et al. (2019). In vitro culture of cynomolgus monkey embryos beyond early gastrulation. Science 366. 10.1126/science.aax7890. [DOI] [PubMed] [Google Scholar]
  • 140.Xiang L, Yin Y, Zheng Y, Ma Y, Li Y, Zhao Z, Guo J, Ai Z, Niu Y, Duan K, et al. (2020). A developmental landscape of 3D-cultured human pre-gastrulation embryos. Nature 577, 537–542. 10.1038/s41586-019-1875-y. [DOI] [PubMed] [Google Scholar]
  • 141.Zhou F, Wang R, Yuan P, Ren Y, Mao Y, Li R, Lian Y, Li J, Wen L, Yan L, et al. (2019). Reconstituting the transcriptome and DNA methylome landscapes of human implantation. Nature 572, 660–664. 10.1038/s41586-019-1500-0. [DOI] [PubMed] [Google Scholar]
  • 142.Smith ZD, Chan MM, Mikkelsen TS, Gu HC, Gnirke A, Regev A, and Meissner A. (2012). A unique regulatory phase of DNA methylation in the early mammalian embryo. Nature 484, 339–U374. 10.1038/nature10960. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 143.Smallwood SA, Tomizawa S, Krueger F, Ruf N, Carli N, Segonds-Pichon A, Sato S, Hata K, Andrews SR, and Kelsey G. (2011). Dynamic CpG island methylation landscape in oocytes and preimplantation embryos. Nature Genetics 43, 811–U126. 10.1038/ng.864. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 144.Kobayashi H, Sakurai T, Imai M, Takahashi N, Fukuda A, Yayoi O, Sato S, Nakabayashi K, Hata K, Sotomaru Y, et al. (2012). Contribution of intragenic DNA methylation in mouse gametic DNA methylomes to establish oocyte-specific heritable marks. PLoS Genet 8, e1002440. 10.1371/journal.pgen.1002440. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 145.Guo H, Zhu P, Yan L, Li R, Hu B, Lian Y, Yan J, Ren X, Lin S, Li J, et al. (2014). The DNA methylation landscape of human early embryos. Nature 511, 606–610. 10.1038/nature13544. [DOI] [PubMed] [Google Scholar]
  • 146.Smith ZD, Chan MM, Humm KC, Karnik R, Mekhoubad S, Regev A, Eggan K, and Meissner A. (2014). DNA methylation dynamics of the human preimplantation embryo. Nature 511, 611–615. 10.1038/nature13581. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 147.Leitch HG, McEwen KR, Turp A, Encheva V, Carroll T, Grabole N, Mansfield W, Nashun B, Knezovich JG, Smith A, et al. (2013). Naive pluripotency is associated with global DNA hypomethylation. Nat Struct Mol Biol 20, 311–316. 10.1038/nsmb.2510. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 148.Sim YJ, Kim MS, Nayfeh A, Yun YJ, Kim SJ, Park KT, Kim CH, and Kim KS (2017). 2i Maintains a Naive Ground State in ESCs through Two Distinct Epigenetic Mechanisms. Stem Cell Reports 8, 1312–1328. 10.1016/j.stemcr.2017.04.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 149.Guo G, von Meyenn F, Rostovskaya M, Clarke J, Dietmann S, Baker D, Sahakyan A, Myers S, Bertone P, Reik W, et al. (2017). Epigenetic resetting of human pluripotency. Development 144, 2748–2763. 10.1242/dev.146811. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 150.Pastor WA, Chen D, Liu W, Kim R, Sahakyan A, Lukianchikov A, Plath K, Jacobsen SE, and Clark AT (2016). Naive Human Pluripotent Cells Feature a Methylation Landscape Devoid of Blastocyst or Germline Memory. Cell Stem Cell 18, 323–329. 10.1016/j.stem.2016.01.019. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 151.Theunissen TW, Friedli M, He Y, Planet E, O'Neil RC, Markoulaki S, Pontis J, Wang H, Iouranova A, Imbeault M, et al. (2016). Molecular Criteria for Defining the Naive Human Pluripotent State. Cell Stem Cell 19, 502–515. 10.1016/j.stem.2016.06.011. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 152.Bar S, Schachter M, Eldar-Geva T, and Benvenisty N. (2017). Large-Scale Analysis of Loss of Imprinting in Human Pluripotent Stem Cells. Cell Reports 19, 957–968. 10.1016/j.celrep.2017.04.020. [DOI] [PubMed] [Google Scholar]
  • 153.Habibi E, Brinkman AB, Arand J, Kroeze LI, Kerstens HH, Matarese F, Lepikhov K, Gut M, Brun-Heath I, Hubner NC, et al. (2013). Whole-genome bisulfite sequencing of two distinct interconvertible DNA methylomes of mouse embryonic stem cells. Cell Stem Cell 13, 360–369. 10.1016/j.stem.2013.06.002. [DOI] [PubMed] [Google Scholar]
  • 154.Zhang Y, Xiang Y, Yin Q, Du Z, Peng X, Wang Q, Fidalgo M, Xia W, Li Y, Zhao ZA, et al. (2018). Dynamic epigenomic landscapes during early lineage specification in mouse embryos. Nat Genet 50, 96–105. 10.1038/s41588-017-0003-x. [DOI] [PubMed] [Google Scholar]
  • 155.Ficz G, Hore TA, Santos F, Lee HJ, Dean W, Arand J, Krueger F, Oxley D, Paul YL, Walter J, et al. (2013). FGF signaling inhibition in ESCs drives rapid genome-wide demethylation to the epigenetic ground state of pluripotency. Cell Stem Cell 13, 351–359. 10.1016/j.stem.2013.06.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 156.Senner CE, Krueger F, Oxley D, Andrews S, and Hemberger M. (2012). DNA methylation profiles define stem cell identity and reveal a tight embryonic-extraembryonic lineage boundary. Stem Cells 30, 2732–2745. 10.1002/stem.1249. [DOI] [PubMed] [Google Scholar]
  • 157.Veillard AC, Marks H, Bernardo AS, Jouneau L, Laloë D, Boulanger L, Kaan A, Brochard V, Tosolini M, Pedersen R, et al. (2014). Stable Methylation at Promoters Distinguishes Epiblast Stem Cells from Embryonic Stem Cells and the In Vivo Epiblasts. Stem Cells and Development 23, 2014–2029. 10.1089/scd.2013.0639. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 158.Okamoto I, Patrat C, Thepot D, Peynot N, Fauque P, Daniel N, Diabangouaya P, Wolf JP, Renard JP, Duranthon V, and Heard E. (2011). Eutherian mammals use diverse strategies to initiate X-chromosome inactivation during development. Nature 472, 370–374. 10.1038/nature09872. [DOI] [PubMed] [Google Scholar]
  • 159.Shiura H, and Abe K. (2019). Xist/Tsix expression dynamics during mouse peri-implantation development revealed by whole-mount 3D RNA-FISH. Sci Rep 9, 3637. 10.1038/s41598-019-38807-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 160.Sousa EJ, Stuart HT, Bates LE, Ghorbani M, Nichols J, Dietmann S, and Silva JCR (2018). Exit from Naive Pluripotency Induces a Transient X Chromosome Inactivation-like State in Males. Cell Stem Cell 22, 919–928 e916. 10.1016/j.stem.2018.05.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 161.Vallot C, Ouimette JF, and Rougeulle C. (2016). Establishment of X chromosome inactivation and epigenomic features of the inactive X depend on cellular contexts. Bioessays 38, 869–880. 10.1002/bies.201600121. [DOI] [PubMed] [Google Scholar]
  • 162.Teklenburg G, Weimar CH, Fauser BC, Macklon N, Geijsen N, Heijnen CJ, Chuva de Sousa Lopes SM, and Kuijk EW (2012). Cell lineage specific distribution of H3K27 trimethylation accumulation in an in vitro model for human implantation. PLoS One 7, e32701. 10.1371/journal.pone.0032701. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 163.Deglincerti A, Croft GF, Pietila LN, Zernicka-Goetz M, Siggia ED, and Brivanlou AH (2016). Self-organization of the in vitro attached human embryo. Nature 533, 251–254. 10.1038/nature17948. [DOI] [PubMed] [Google Scholar]
  • 164.Shahbazi MN, Jedrusik A, Vuoristo S, Recher G, Hupalowska A, Bolton V, Fogarty NNM, Campbell A, Devito L, Ilic D, et al. (2016). Self-organization of the human embryo in the absence of maternal tissues. Nat Cell Biol 18, 700–708. 10.1038/ncb3347. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 165.Dhara SK, and Benvenisty N. (2004). Gene trap as a tool for genome annotation and analysis of X chromosome inactivation in human embryonic stem cells. Nucleic Acids Res 32, 3995–4002. 10.1093/nar/gkh746. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 166.Hoffman LM, Hall L, Batten JL, Young H, Pardasani D, Baetge EE, Lawrence J, and Carpenter MK (2005). X-inactivation status varies in human embryonic stem cell lines. Stem Cells 23, 1468–1478. 10.1634/stemcells.2004-0371. [DOI] [PubMed] [Google Scholar]
  • 167.Hall LL, Byron M, Butler J, Becker KA, Nelson A, Amit M, Itskovitz-Eldor J, Stein J, Stein G, Ware C, and Lawrence JB (2008). X-inactivation reveals epigenetic anomalies in most hESC but identifies sublines that initiate as expected. J Cell Physiol 216, 445–452. 10.1002/jcp.21411. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 168.Shen Y, Matsuno Y, Fouse SD, Rao N, Root S, Xu R, Pellegrini M, Riggs AD, and Fan G. (2008). X-inactivation in female human embryonic stem cells is in a nonrandom pattern and prone to epigenetic alterations. Proc Natl Acad Sci U S A 105, 4709–4714. 10.1073/pnas.0712018105. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 169.Silva SS, Rowntree RK, Mekhoubad S, and Lee JT (2008). X-chromosome inactivation and epigenetic fluidity in human embryonic stem cells. Proc Natl Acad Sci U S A 105, 4820–4825. 10.1073/pnas.0712136105. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 170.Vallot C, Ouimette JF, Makhlouf M, Feraud O, Pontis J, Come J, Martinat C, Bennaceur-Griscelli A, Lalande M, and Rougeulle C. (2015). Erosion of X Chromosome Inactivation in Human Pluripotent Cells Initiates with XACT Coating and Depends on a Specific Heterochromatin Landscape. Cell Stem Cell 16, 533–546. 10.1016/j.stem.2015.03.016. [DOI] [PubMed] [Google Scholar]
  • 171.Lengner CJ, Gimelbrant AA, Erwin JA, Cheng AW, Guenther MG, Welstead GG, Alagappan R, Frampton GM, Xu P, Muffat J, et al. (2010). Derivation of pre-X inactivation human embryonic stem cells under physiological oxygen concentrations. Cell 141, 872–883. 10.1016/j.cell.2010.04.010. [DOI] [PubMed] [Google Scholar]
  • 172.Patrat C, Ouimette JF, and Rougeulle C. (2020). X chromosome inactivation in human development. Development 147. 10.1242/dev.183095. [DOI] [PubMed] [Google Scholar]
  • 173.Bar S, Seaton LR, Weissbein U, Eldar-Geva T, and Benvenisty N. (2019). Global Characterization of X Chromosome Inactivation in Human Pluripotent Stem Cells. Cell Rep 27, 20–29 e23. 10.1016/j.celrep.2019.03.019. [DOI] [PubMed] [Google Scholar]
  • 174.Sahakyan A, Kim R, Chronis C, Sabri S, Bonora G, Theunissen TW, Kuoy E, Langerman J, Clark AT, Jaenisch R, and Plath K. (2017). Human Naive Pluripotent Stem Cells Model X Chromosome Dampening and X Inactivation. Cell Stem Cell 20, 87–101. 10.1016/j.stem.2016.10.006. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 175.Vallot C, Patrat C, Collier AJ, Huret C, Casanova M, Liyakat Ali TM, Tosolini M, Frydman N, Heard E, Rugg-Gunn PJ, and Rougeulle C. (2017). XACT Noncoding RNA Competes with XIST in the Control of X Chromosome Activity during Human Early Development. Cell Stem Cell 20, 102–111. 10.1016/j.stem.2016.10.014. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 176.An C, Feng G, Zhang J, Cao S, Wang Y, Wang N, Lu F, Zhou Q, and Wang H. (2020). Overcoming Autocrine FGF Signaling-Induced Heterogeneity in Naive Human ESCs Enables Modeling of Random X Chromosome Inactivation. Cell Stem Cell 27, 482–497 e484. 10.1016/j.stem.2020.06.002. [DOI] [PubMed] [Google Scholar]
  • 177.Ezashi T, Das P, and Roberts RM (2005). Low O2 tensions and the prevention of differentiation of hES cells. Proc Natl Acad Sci U S A 102, 4783–4788. 10.1073/pnas.0501283102. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 178.Cho YM, Kwon S, Pak YK, Seol HW, Choi YM, Park DJ, Park KS, and Lee HK (2006). Dynamic changes in mitochondrial biogenesis and antioxidant enzymes during the spontaneous differentiation of human embryonic stem cells. Biochem Biophys Res Commun 348, 1472–1478. 10.1016/j.bbrc.2006.08.020. [DOI] [PubMed] [Google Scholar]
  • 179.Kondoh H, Lleonart ME, Nakashima Y, Yokode M, Tanaka M, Bernard D, Gil J, and Beach D. (2007). A high glycolytic flux supports the proliferative potential of murine embryonic stem cells. Antioxid Redox Signal 9, 293–299. 10.1089/ars.2006.1467. [DOI] [PubMed] [Google Scholar]
  • 180.Varum S, Rodrigues AS, Moura MB, Momcilovic O, Easley CA, Ramalho-Santos J, Van Houten B, and Schatten G. (2011). Energy Metabolism in Human Pluripotent Stem Cells and Their Differentiated Counterparts. Plos One 6. ARTN e20914 10.1371/journal.pone.0020914. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 181.Varum S, Momcilovic O, Castro C, Ben-Yehudah A, Ramalho-Santos J, and Navara CS (2009). Enhancement of human embryonic stem cell pluripotency through inhibition of the mitochondrial respiratory chain. Stem Cell Res 3, 142–156. 10.1016/j.scr.2009.07.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 182.Tsogtbaatar E, Landin C, Minter-Dykhouse K, and Folmes CDL (2020). Energy Metabolism Regulates Stem Cell Pluripotency. Front Cell Dev Biol 8, 87. 10.3389/fcell.2020.00087. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 183.Zhou W, Choi M, Margineantu D, Margaretha L, Hesson J, Cavanaugh C, Blau CA, Horwitz MS, Hockenbery D, Ware C, and Ruohola-Baker H. (2012). HIF1alpha induced switch from bivalent to exclusively glycolytic metabolism during ESC-to-EpiSC/hESC transition. EMBO J 31, 2103–2116. 10.1038/emboj.2012.71. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 184.Wu J, Ocampo A, and Belmonte JCI (2016). Cellular Metabolism and Induced Pluripotency. Cell 166, 1371–1385. 10.1016/j.cell.2016.08.008. [DOI] [PubMed] [Google Scholar]
  • 185.Barbehenn EK, Wales RG, and Lowry OH (1978). Measurement of metabolites in single preimplantation embryos; a new means to study metabolic control in early embryos. J Embryol Exp Morphol 43, 29–46. [PubMed] [Google Scholar]
  • 186.Pantaleon M, and Kaye PL (1998). Glucose transporters in preimplantation development. Rev Reprod 3, 77–81. 10.1530/ror.0.0030077. [DOI] [PubMed] [Google Scholar]
  • 187.Sperber H, Mathieu J, Wang Y, Ferreccio A, Hesson J, Xu Z, Fischer KA, Devi A, Detraux D, Gu H, et al. (2015). The metabolome regulates the epigenetic landscape during naive-to-primed human embryonic stem cell transition. Nat Cell Biol 17, 1523–1535. 10.1038/ncb3264. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 188.Wu J, and Izpisua Belmonte JC (2015). Metabolic exit from naive pluripotency. Nat Cell Biol 17, 1519–1521. 10.1038/ncb3269. [DOI] [PubMed] [Google Scholar]
  • 189.Sturmey RG, Reis A, Leese HJ, and McEvoy TG (2009). Role of fatty acids in energy provision during oocyte maturation and early embryo development. Reprod Domest Anim 44 Suppl 3, 50–58. 10.1111/j.1439-0531.2009.01402.x. [DOI] [PubMed] [Google Scholar]
  • 190.Leese HJ (2012). Metabolism of the preimplantation embryo: 40 years on. Reproduction 143, 417–427. 10.1530/REP-11-0484. [DOI] [PubMed] [Google Scholar]
  • 191.Jiang ZM, Guang L, Li L, and Ng SC (2019). Putting Stem Cells on a Low-Fat Diet Switches Their Pluripotent State. Cell Stem Cell 25, 3–5. 10.1016/j.stem.2019.06.002. [DOI] [PubMed] [Google Scholar]
  • 192.Wang J, Alexander P, Wu L, Hammer R, Cleaver O, and McKnight SL (2009). Dependence of mouse embryonic stem cells on threonine catabolism. Science 325, 435–439. 10.1126/science.1173288. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 193.Shyh-Chang N, Locasale JW, Lyssiotis CA, Zheng Y, Teo RY, Ratanasirintrawoot S, Zhang J, Onder T, Unternaehrer JJ, Zhu H, et al. (2013). Influence of threonine metabolism on S-adenosylmethionine and histone methylation. Science 339, 222–226. 10.1126/science.1226603. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 194.Alexander PB, Wang J, and McKnight SL (2011). Targeted killing of a mammalian cell based upon its specialized metabolic state. P Natl Acad Sci USA 108, 15828–15833. 10.1073/pnas.1111312108. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 195.Edgar AJ (2002). The human L-threonine 3-dehydrogenase gene is an expressed pseudogene. Bmc Genet 3. Artn 18 Doi 10.1186/1471-2156-3-18. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 196.Shiraki N, Shiraki Y, Tsuyama T, Obata F, Miura M, Nagae G, Aburatani H, Kume K, Endo F, and Kume S. (2014). Methionine metabolism regulates maintenance and differentiation of human pluripotent stem cells. Cell Metab 19, 780–794. 10.1016/j.cmet.2014.03.017. [DOI] [PubMed] [Google Scholar]
  • 197.Washington JM, Rathjen J, Felquer F, Lonic A, Bettess MD, Hamra N, Semendric L, Tan BS, Lake JA, Keough RA, et al. (2010). L-Proline induces differentiation of ES cells: a novel role for an amino acid in the regulation of pluripotent cells in culture. Am J Physiol Cell Physiol 298, C982–992. 10.1152/ajpcell.00498.2009. [DOI] [PubMed] [Google Scholar]
  • 198.Casalino L, Comes S, Lambazzi G, De Stefano B, Filosa S, De Falco S, De Cesare D, Minchiotti G, and Patriarca EJ (2011). Control of embryonic stem cell metastability by L-proline catabolism. J Mol Cell Biol 3, 108–122. 10.1093/jmcb/mjr001. [DOI] [PubMed] [Google Scholar]
  • 199.McEwan M, Lins RJ, Munro SK, Vincent ZL, Ponnampalam AP, and Mitchell MD (2009). Cytokine regulation during the formation of the fetal-maternal interface: focus on cell-cell adhesion and remodelling of the extra-cellular matrix. Cytokine Growth Factor Rev 20, 241–249. 10.1016/j.cytogfr.2009.05.004. [DOI] [PubMed] [Google Scholar]
  • 200.Lu V, Roy IJ, and Teitell MA (2021). Nutrients in the fate of pluripotent stem cells. Cell Metab 33, 2108–2121. 10.1016/j.cmet.2021.09.013. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 201.Zheng C, Ballard EB, and Wu J. (2021). The road to generating transplantable organs: from blastocyst complementation to interspecies chimeras. Development 148. 10.1242/dev.195792. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 202.Rostovskaya M, Stirparo GG, and Smith A. (2019). Capacitation of human naive pluripotent stem cells for multi-lineage differentiation. Development 146. 10.1242/dev.172916. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 203.Liu X, Nefzger CM, Rossello FJ, Chen J, Knaupp AS, Firas J, Ford E, Pflueger J, Paynter JM, Chy HS, et al. (2017). Comprehensive characterization of distinct states of human naive pluripotency generated by reprogramming. Nat Methods 14, 1055–1062. 10.1038/nmeth.4436. [DOI] [PubMed] [Google Scholar]
  • 204.Huang Y, Osorno R, Tsakiridis A, and Wilson V. (2012). In Vivo differentiation potential of epiblast stem cells revealed by chimeric embryo formation. Cell Rep 2, 1571–1578. 10.1016/j.celrep.2012.10.022. [DOI] [PubMed] [Google Scholar]
  • 205.Aguilera-Castrejon A, Oldak B, Shani T, Ghanem N, Itzkovich C, Slomovich S, Tarazi S, Bayerl J, Chugaeva V, Ayyash M, et al. (2021). Ex utero mouse embryogenesis from pre-gastrulation to late organogenesis. Nature 593, 119–124. 10.1038/s41586-021-03416-3. [DOI] [PubMed] [Google Scholar]
  • 206.Mascetti VL, and Pedersen RA (2016). Human-Mouse Chimerism Validates Human Stem Cell Pluripotency. Cell Stem Cell 18, 67–72. 10.1016/j.stem.2015.11.017. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 207.Wu J, Greely HT, Jaenisch R, Nakauchi H, Rossant J, and Belmonte JCI (2016). Stem cells and interspecies chimaeras. Nature 540, 51–59. 10.1038/nature20573. [DOI] [PubMed] [Google Scholar]
  • 208.Hayashi K, Ohta H, Kurimoto K, Aramaki S, and Saitou M. (2011). Reconstitution of the mouse germ cell specification pathway in culture by pluripotent stem cells. Cell 146, 519–532. 10.1016/j.cell.2011.06.052. [DOI] [PubMed] [Google Scholar]
  • 209.Oikawa M, Kobayashi H, Sanbo M, Mizuno N, Iwatsuki K, Takashima T, Yamauchi K, Yoshida F, Yamamoto T, Shinohara T, et al. (2022). Functional primordial germ cell-like cells from pluripotent stem cells in rats. Science 376, 176–179. 10.1126/science.abl4412. [DOI] [PubMed] [Google Scholar]
  • 210.Hayashi K, and Surani MA (2009). Self-renewing epiblast stem cells exhibit continual delineation of germ cells with epigenetic reprogramming in vitro. Development 136, 3549–3556. 10.1242/dev.037747. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 211.Irie N, Weinberger L, Tang WWC, Kobayashi T, Viukov S, Manor YS, Dietmann S, Hanna JH, and Surani MA (2015). SOX17 Is a Critical Specifier of Human Primordial Germ Cell Fate. Cell 160, 253–268. 10.1016/j.cell.2014.12.013. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 212.Sasaki K, Nakamura T, Okamoto K, Yabuta Y, Iwatani C, Tsuchiya H, Seita Y, Nakamura S, Shiraki N, Takakuwa T, et al. (2016). The Germ Cell Fate of Cynomolgus Monkeys Is Specified in the Nascent Amnion. Dev Cell 39, 169–185. 10.1016/j.devcel.2016.09.007. [DOI] [PubMed] [Google Scholar]
  • 213.Condic ML (2014). Totipotency: what it is and what it is not. Stem Cells Dev 23, 796–812. 10.1089/scd.2013.0364. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 214.Macfarlan TS, Gifford WD, Driscoll S, Lettieri K, Rowe HM, Bonanomi D, Firth A, Singer O, Trono D, and Pfaff SL (2012). Embryonic stem cell potency fluctuates with endogenous retrovirus activity. Nature 487, 57–63. 10.1038/nature11244. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 215.Genet M, and Torres-Padilla ME (2020). The molecular and cellular features of 2-cell-like cells: a reference guide. Development 147. 10.1242/dev.189688. [DOI] [PubMed] [Google Scholar]
  • 216.Taubenschmid-Stowers J, Rostovskaya M, Santos F, Ljung S, Argelaguet R, Krueger F, Nichols J, and Reik W. (2022). 8C-like cells capture the human zygotic genome activation program in vitro. Cell Stem Cell 29, 449–459 e446. 10.1016/j.stem.2022.01.014. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 217.De Iaco A, Planet E, Coluccio A, Verp S, Duc J, and Trono D. (2017). DUX-family transcription factors regulate zygotic genome activation in placental mammals. Nat Genet 49, 941–945. 10.1038/ng.3858. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 218.Eckersley-Maslin M, Alda-Catalinas C, Blotenburg M, Kreibich E, Krueger C, and Reik W. (2019). Dppa2 and Dppa4 directly regulate the Dux-driven zygotic transcriptional program. Genes Dev 33, 194–208. 10.1101/gad.321174.118. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 219.Hendrickson PG, Dorais JA, Grow EJ, Whiddon JL, Lim JW, Wike CL, Weaver BD, Pflueger C, Emery BR, Wilcox AL, et al. (2017). Conserved roles of mouse DUX and human DUX4 in activating cleavage-stage genes and MERVL/HERVL retrotransposons. Nat Genet 49, 925–934. 10.1038/ng.3844. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 220.Whiddon JL, Langford AT, Wong CJ, Zhong JW, and Tapscott SJ (2017). Conservation and innovation in the DUX4-family gene network. Nat Genet 49, 935–940. 10.1038/ng.3846. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 221.Shen H, Yang M, Li S, Zhang J, Peng B, Wang C, Chang Z, Ong J, and Du P. (2021). Mouse totipotent stem cells captured and maintained through spliceosomal repression. Cell 184, 2843–2859 e2820. 10.1016/j.cell.2021.04.020. [DOI] [PubMed] [Google Scholar]
  • 222.Yang M, Yu H, Yu X, Liang S, Hu Y, Luo Y, Izsvak Z, Sun C, and Wang J. (2022). Chemical-induced chromatin remodeling reprograms mouse ESCs to totipotent-like stem cells. Cell Stem Cell 29, 400–418 e413. 10.1016/j.stem.2022.01.010. [DOI] [PubMed] [Google Scholar]
  • 223.Xu Y, Zhao J, Ren Y, Wang X, Lyu Y, Xie B, Sun Y, Yuan X, Liu H, Yang W, et al. (2022). Derivation of totipotent-like stem cells with blastocyst-like structure forming potential. Cell Res 32, 513–529. 10.1038/s41422-022-00668-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 224.Hu Y, Yang Y, Tan P, Zhang Y, Han M, Yu J, Zhang X, Jia Z, Wang D, Yao K, et al. (2023). Induction of mouse totipotent stem cells by a defined chemical cocktail. Nature 617, 792–797. 10.1038/s41586-022-04967-9. [DOI] [PubMed] [Google Scholar]
  • 225.Wang L, Zhang J, Duan J, Gao X, Zhu W, Lu X, Yang L, Zhang J, Li G, Ci W, et al. (2014). Programming and inheritance of parental DNA methylomes in mammals. Cell 157, 979–991. 10.1016/j.cell.2014.04.017. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 226.Dong C, Beltcheva M, Gontarz P, Zhang B, Popli P, Fischer LA, Khan SA, Park KM, Yoon EJ, Xing X, et al. (2020). Derivation of trophoblast stem cells from naive human pluripotent stem cells. Elife 9. 10.7554/eLife.52504. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 227.Cinkornpumin JK, Kwon SY, Guo Y, Hossain I, Sirois J, Russett CS, Tseng HW, Okae H, Arima T, Duchaine TF, et al. (2020). Naive Human Embryonic Stem Cells Can Give Rise to Cells with a Trophoblast-like Transcriptome and Methylome. Stem Cell Reports 15, 198–213. 10.1016/j.stemcr.2020.06.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 228.Io S, Kabata M, Iemura Y, Semi K, Morone N, Minagawa A, Wang B, Okamoto I, Nakamura T, Kojima Y, et al. (2021). Capturing human trophoblast development with naive pluripotent stem cells in vitro. Cell Stem Cell 28, 1023–1039 e1013. 10.1016/j.stem.2021.03.013. [DOI] [PubMed] [Google Scholar]
  • 229.Wei Y, Wang T, Ma L, Zhang Y, Zhao Y, Lye K, Xiao L, Chen C, Wang Z, Ma Y, et al. (2021). Efficient derivation of human trophoblast stem cells from primed pluripotent stem cells. Sci Adv 7. 10.1126/sciadv.abf4416. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 230.Li R, Zhong C, Yu Y, Liu H, Sakurai M, Yu L, Min Z, Shi L, Wei Y, Takahashi Y, et al. (2019). Generation of Blastocyst-like Structures from Mouse Embryonic and Adult Cell Cultures. Cell 179, 687–702 e618. 10.1016/j.cell.2019.09.029. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 231.Liu K, Xu X, Bai D, Li Y, Zhang Y, Jia Y, Guo M, Han X, Liu Y, Sheng Y, et al. (2023). Bilineage embryo-like structure from EPS cells can produce live mice with tetraploid trophectoderm. Protein Cell 14, 262–278. 10.1093/procel/pwac029. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 232.Chan SW, Rizwan M, and Yim EKF (2020). Emerging Methods for Enhancing Pluripotent Stem Cell Expansion. Front Cell Dev Biol 8, 70. 10.3389/fcell.2020.00070. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 233.Wei Y, Zhang E, Yu L, Ci B, Sakurai M, Guo L, Zhang X, Lin S, Takii S, Liu L, et al. (2023). Dissecting embryonic and extraembryonic lineage crosstalk with stem cell co-culture. Cell. 10.1016/j.cell.2023.11.008. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 234.Tian C, Wang J, Ye X, Chen J, Zheng R, Yu H, Li J, Yin G, Liu L, Zhao N, et al. (2023). Culture conditions of mouse ESCs impact the tumor appearance in vivo. Cell Rep 42, 112645. 10.1016/j.celrep.2023.112645. [DOI] [PubMed] [Google Scholar]
  • 235.Wu BJ, Yang ZQ, Liu YJ, Li JW, Chen C, Li XH, and Bao SQ (2023). A chemically defined system supports two distinct types of stem cell from a single blastocyst and their self-assembly to generate blastoid. Cell Proliferat 56. 10.1111/cpr.13396. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 236.Xiang J, Wang H, Zhang Y, Wang J, Liu F, Han X, Lu Z, Li C, Li Z, Gao Y, et al. (2021). LCDM medium supports the derivation of bovine extended pluripotent stem cells with embryonic and extraembryonic potency in bovine-mouse chimeras from iPSCs and bovine fetal fibroblasts. FEBS J 288, 4394–4411. 10.1111/febs.15744. [DOI] [PubMed] [Google Scholar]
  • 237.Wang Y, Ming H, Yu L, Li J, Zhu L, Sun HX, Pinzon-Arteaga CA, Wu J, and Jiang Z. (2023). Establishment of bovine trophoblast stem cells. Cell Rep 42, 112439. 10.1016/j.celrep.2023.112439. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 238.Yu L, Wei Y, Duan J, Schmitz DA, Sakurai M, Wang L, Wang K, Zhao S, Hon GC, and Wu J. (2021). Blastocyst-like structures generated from human pluripotent stem cells. Nature 591, 620–626. 10.1038/s41586-021-03356-y. [DOI] [PubMed] [Google Scholar]
  • 239.Kagawa H, Javali A, Khoei HH, Sommer TM, Sestini G, Novatchkova M, Scholte Op Reimer Y, Castel G, Bruneau A, Maenhoudt N, et al. (2022). Human blastoids model blastocyst development and implantation. Nature 601, 600–605. 10.1038/s41586-021-04267-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 240.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 e1014. 10.1016/j.stem.2021.04.031. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 241.Moris N, Anlas K, van den Brink SC, Alemany A, Schroder J, Ghimire S, Balayo T, van Oudenaarden A, and Martinez Arias A. (2020). An in vitro model of early anteroposterior organization during human development. Nature 582, 410–415. 10.1038/s41586-020-2383-9. [DOI] [PubMed] [Google Scholar]
  • 242.Liu L, Oura S, Markham Z, Hamilton JN, Skory RM, Li L, Sakurai M, Wang L, Pinzon-Arteaga CA, Plachta N, et al. (2023). Modeling post-implantation stages of human development into early organogenesis with stem-cell-derived peri-gastruloids. Cell 186, 3776–3792 e3716. 10.1016/j.cell.2023.07.018. [DOI] [PubMed] [Google Scholar]
  • 243.Weatherbee BAT, Gantner CW, Iwamoto-Stohl LK, Daza RM, Hamazaki N, Shendure J, and Zernicka-Goetz M. (2023). Pluripotent stem cell-derived model of the post-implantation human embryo. Nature 622, 584–593. 10.1038/s41586-023-06368-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 244.Oldak B, Wildschutz E, Bondarenko V, Comar MY, Zhao C, Aguilera-Castrejon A, Tarazi S, Viukov S, Pham TXA, Ashouokhi S, et al. (2023). Complete human day 14 post-implantation embryo models from naive ES cells. Nature 622, 562–573. 10.1038/s41586-023-06604-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 245.Pedroza M, Gassaloglu SI, Dias N, Zhong L, Hou TJ, Kretzmer H, Smith ZD, and Sozen B. (2023). Self-patterning of human stem cells into post-implantation lineages. Nature 622, 574–583. 10.1038/s41586-023-06354-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 246.Zhang P, Zhai X, Huang B, Sun S, Wang W, and Zhang M. (2023). Highly efficient generation of blastocyst-like structures from spliceosomes-repressed mouse totipotent blastomere-like cells. Sci China Life Sci 66, 423–435. 10.1007/s11427-022-2209-3. [DOI] [PubMed] [Google Scholar]
  • 247.Eckersley-Maslin MA, Svensson V, Krueger C, Stubbs TM, Giehr P, Krueger F, Miragaia RJ, Kyriakopoulos C, Berrens RV, Milagre I, et al. (2016). MERVL/Zscan4 Network Activation 1578 Results in Transient Genome-wide DNA Demethylation of mESCs. Cell Rep 17, 179–192. 10.1016/j.celrep.2016.08.087. [DOI] [PMC free article] [PubMed] [Google Scholar]

RESOURCES