Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 4.343
Filter
1.
Nat Commun ; 15(1): 5055, 2024 Jun 13.
Article in English | MEDLINE | ID: mdl-38871742

ABSTRACT

The anterior-posterior axis of the mammalian embryo is laid down by the anterior visceral endoderm (AVE), an extraembryonic signaling center that is specified within the visceral endoderm. Current models posit that AVE differentiation is promoted globally by epiblast-derived Nodal signals, and spatially restricted by a BMP gradient established by the extraembryonic ectoderm. Here, we report spatially restricted AVE differentiation in bilayered embryo-like aggregates made from mouse embryonic stem cells that lack an extraembryonic ectoderm. Notably, clusters of AVE cells also form in pure visceral endoderm cultures upon activation of Nodal signaling, indicating that tissue-intrinsic factors can restrict AVE differentiation. We identify ß-catenin activity as a tissue-intrinsic factor that antagonizes AVE-inducing Nodal signals. Together, our results show how an AVE-like population can arise through interactions between epiblast and visceral endoderm alone. This mechanism may be a flexible solution for axis patterning in a wide range of embryo geometries, and provide robustness to axis patterning when coupled with signal gradients.


Subject(s)
Body Patterning , Cell Differentiation , Endoderm , Nodal Protein , Signal Transduction , beta Catenin , Animals , Endoderm/cytology , Endoderm/metabolism , Endoderm/embryology , beta Catenin/metabolism , Mice , Nodal Protein/metabolism , Nodal Protein/genetics , Germ Layers/metabolism , Germ Layers/cytology , Mouse Embryonic Stem Cells/metabolism , Mouse Embryonic Stem Cells/cytology , Gene Expression Regulation, Developmental , Embryo, Mammalian/cytology
2.
Nat Cell Biol ; 26(6): 868-877, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38849542

ABSTRACT

Despite a distinct developmental origin, extraembryonic cells in mice contribute to gut endoderm and converge to transcriptionally resemble their embryonic counterparts. Notably, all extraembryonic progenitors share a non-canonical epigenome, raising several pertinent questions, including whether this landscape is reset to match the embryonic regulation and if extraembryonic cells persist into later development. Here we developed a two-colour lineage-tracing strategy to track and isolate extraembryonic cells over time. We find that extraembryonic gut cells display substantial memory of their developmental origin including retention of the original DNA methylation landscape and resulting transcriptional signatures. Furthermore, we show that extraembryonic gut cells undergo programmed cell death and neighbouring embryonic cells clear their remnants via non-professional phagocytosis. By midgestation, we no longer detect extraembryonic cells in the wild-type gut, whereas they persist and differentiate further in p53-mutant embryos. Our study provides key insights into the molecular and developmental fate of extraembryonic cells inside the embryo.


Subject(s)
Apoptosis , Cell Lineage , DNA Methylation , Endoderm , Gene Expression Regulation, Developmental , Animals , Endoderm/cytology , Endoderm/metabolism , Tumor Suppressor Protein p53/metabolism , Tumor Suppressor Protein p53/genetics , Phagocytosis , Mice, Inbred C57BL , Mice , Cell Differentiation , Female , Embryonic Development , Embryo, Mammalian/cytology , Embryo, Mammalian/metabolism , Mice, Transgenic , Gastrointestinal Tract/cytology , Gastrointestinal Tract/embryology , Gastrointestinal Tract/metabolism
3.
Development ; 151(11)2024 Jun 01.
Article in English | MEDLINE | ID: mdl-38752427

ABSTRACT

Bone morphogenic protein (BMP) signaling plays an essential and highly conserved role in embryo axial patterning in animal species. However, in mammalian embryos, which develop inside the mother, early development includes a preimplantation stage, which does not occur in externally developing embryos. During preimplantation, the epiblast is segregated from extra-embryonic lineages that enable implantation and development in utero. Yet, the requirement for BMP signaling is imprecisely defined in mouse early embryos. Here, we show that, in contrast to previous reports, BMP signaling (SMAD1/5/9 phosphorylation) is not detectable until implantation when it is detected in the primitive endoderm - an extra-embryonic lineage. Moreover, preimplantation development appears to be normal following deletion of maternal and zygotic Smad4, an essential effector of canonical BMP signaling. In fact, mice lacking maternal Smad4 are viable. Finally, we uncover a new requirement for zygotic Smad4 in epiblast scaling and cavitation immediately after implantation, via a mechanism involving FGFR/ERK attenuation. Altogether, our results demonstrate no role for BMP4/SMAD4 in the first lineage decisions during mouse development. Rather, multi-pathway signaling among embryonic and extra-embryonic cell types drives epiblast morphogenesis postimplantation.


Subject(s)
Embryo Implantation , Germ Layers , Morphogenesis , Signal Transduction , Smad4 Protein , Animals , Smad4 Protein/metabolism , Smad4 Protein/genetics , Germ Layers/metabolism , Embryo Implantation/genetics , Mice , Morphogenesis/genetics , Female , Bone Morphogenetic Protein 4/metabolism , Bone Morphogenetic Protein 4/genetics , Gene Expression Regulation, Developmental , Embryonic Development/genetics , Mice, Knockout , Embryo, Mammalian/metabolism , Endoderm/metabolism , Endoderm/embryology , Blastocyst/metabolism , Blastocyst/cytology
4.
Cell Commun Signal ; 22(1): 300, 2024 May 30.
Article in English | MEDLINE | ID: mdl-38816763

ABSTRACT

Optimizing the efficiency of definitive endoderm (DE) differentiation is necessary for the generation of diverse organ-like structures. In this study, we used the small molecule inhibitor saracatinib (SAR) to enhance DE differentiation of human embryonic stem cells and induced pluripotent stem cells. SAR significantly improved DE differentiation efficiency at low concentrations. The interaction between SAR and Focal Adhesion Kinase (FAK) was explored through RNA-seq and molecular docking simulations, which further supported the inhibition of DE differentiation by p-FAK overexpression in SAR-treated cells. In addition, we found that SAR inhibited the nuclear translocation of Yes-associated protein (YAP), a downstream effector of FAK, which promoted DE differentiation. Moreover, the addition of SAR enabled a significant reduction in activin A (AA) from 50 to 10 ng/mL without compromising DE differentiation efficiency. For induction of the pancreatic lineage, 10 ng/ml AA combined with SAR at the DE differentiation stage yielded a comparative number of PDX1+/NKX6.1+ pancreatic progenitor cells to those obtained by 50 ng/ml AA treatment. Our study highlights SAR as a potential modulator that facilitates the cost-effective generation of DE cells and provides insight into the orchestration of cell fate determination.


Subject(s)
Benzodioxoles , Cell Differentiation , Endoderm , Quinazolines , Signal Transduction , Humans , Cell Differentiation/drug effects , Endoderm/drug effects , Endoderm/cytology , Endoderm/metabolism , Benzodioxoles/pharmacology , Signal Transduction/drug effects , Quinazolines/pharmacology , Transcription Factors/metabolism , Induced Pluripotent Stem Cells/drug effects , Induced Pluripotent Stem Cells/metabolism , Induced Pluripotent Stem Cells/cytology , Adaptor Proteins, Signal Transducing/metabolism , YAP-Signaling Proteins/metabolism , Focal Adhesion Kinase 1/metabolism , Focal Adhesion Kinase 1/genetics , Human Embryonic Stem Cells/drug effects , Human Embryonic Stem Cells/metabolism , Human Embryonic Stem Cells/cytology , Activins/metabolism , Molecular Docking Simulation
5.
Toxicol In Vitro ; 98: 105836, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38702034

ABSTRACT

Nanoparticles have unique properties that make them useful in biomedicine. However, their extensive use raises concerns about potential hazards to the body. Therefore, it is crucial to establish effective and robust toxicology models to evaluate the developmental and functional toxicity of nanoparticles on the body. This article discusses the use of stem cells to study the developmental and functional toxicity of organs of endodermal origin due to nanoparticles. The study discovered that various types of nanoparticles have varying effects on stem cells. The application of stem cell models can provide a possibility for studying the effects of nanoparticles on organ development and function, as they can more accurately reflect the toxic mechanisms of different types of nanoparticles. However, stem cell toxicology systems currently cannot fully reflect the effects of nanoparticles on entire organs. Therefore, the establishment of organoid models and other advanced assessment models is expected to address this issue.


Subject(s)
Endoderm , Nanoparticles , Stem Cells , Animals , Nanoparticles/toxicity , Humans , Stem Cells/drug effects , Endoderm/drug effects , Endoderm/cytology
6.
Cell Commun Signal ; 22(1): 242, 2024 Apr 26.
Article in English | MEDLINE | ID: mdl-38664733

ABSTRACT

BACKGROUND: Paired box 1 (PAX1) is a transcription factor and essential for the development of pharyngeal pouches-derived tissues, including thymus. PAX1 mutations are identified in Severe Combined Immunodeficiency (SCID) patients with Otofaciocervical Syndrome Type 2 (OTFCS2). However, despite the critical roles of PAX1 in embryonic development and diseases, detailed insights into its molecular mode of action are critically missing. METHODS: The repressing roles of PAX1 and SCID associated mutants on Wnt signaling pathway were investigated by luciferase reporter assays, qRT-PCR and in situ hybridization in HEK293FT, HCT116 cells and zebrafish embryos, respectively. Co-immunoprecipitation (co-IP) and western blotting assays were carried out to identify the molecular mechanisms underlying PAX1's role on Wnt signaling pathway. hESC based endoderm differentiation, flow cytometry, high-throughput sequencing data analysis, and qRT-PCR assays were utilized to determine the roles of PAX1 during endoderm differentiation. RESULTS: Here, we show that PAX1 represses canonical Wnt signaling pathway in vertebrate cells. Mechanically, PAX1 competes with SUMO E3 ligase PIASy to bind to TCF7L2, thus perturbing TCF7L2 SUMOylation level, further reducing its transcriptional activity and protein stability. Moreover, we reveal that PAX1 plays dual roles in hESC-derived definitive and foregut/pharyngeal endoderm cells, which give rise to the thymus epithelium, by inhibiting Wnt signaling. Importantly, our data show PAX1 mutations found in SCID patients significantly compromise the suppressing ability of PAX1 on Wnt signaling. CONCLUSIONS: Our study presents a novel molecular mode of action of PAX1 in regulation of canonical Wnt signaling and endoderm differentiation, thus providing insights for the molecular basis of PAX1 associated SCID, offering better understanding of the behavior of PAX1 in embryogenesis.


Subject(s)
Cell Differentiation , Endoderm , Wnt Signaling Pathway , Zebrafish , Humans , Wnt Signaling Pathway/genetics , Cell Differentiation/genetics , Endoderm/metabolism , Endoderm/cytology , Animals , Zebrafish/genetics , HEK293 Cells , Transcription Factor 7-Like 2 Protein/metabolism , Transcription Factor 7-Like 2 Protein/genetics , HCT116 Cells , Paired Box Transcription Factors/metabolism , Paired Box Transcription Factors/genetics
7.
Development ; 151(8)2024 Apr 15.
Article in English | MEDLINE | ID: mdl-38563517

ABSTRACT

The lineage decision that generates the epiblast and primitive endoderm from the inner cell mass (ICM) is a paradigm for cell fate specification. Recent mathematics has formalized Waddington's landscape metaphor and proven that lineage decisions in detailed gene network models must conform to a small list of low-dimensional stereotypic changes called bifurcations. The most plausible bifurcation for the ICM is the so-called heteroclinic flip that we define and elaborate here. Our re-analysis of recent data suggests that there is sufficient cell movement in the ICM so the FGF signal, which drives the lineage decision, can be treated as spatially uniform. We thus extend the bifurcation model for a single cell to the entire ICM by means of a self-consistently defined time-dependent FGF signal. This model is consistent with available data and we propose additional dynamic experiments to test it further. This demonstrates that simplified, quantitative and intuitively transparent descriptions are possible when attention is shifted from specific genes to lineages. The flip bifurcation is a very plausible model for any situation where the embryo needs control over the relative proportions of two fates by a morphogen feedback.


Subject(s)
Blastocyst , Cell Differentiation , Cell Lineage , Models, Biological , Animals , Mice , Blastocyst/metabolism , Blastocyst/cytology , Signal Transduction , Fibroblast Growth Factors/metabolism , Gene Expression Regulation, Developmental , Endoderm/cytology , Endoderm/metabolism , Germ Layers/cytology , Germ Layers/metabolism
8.
In Vitro Cell Dev Biol Anim ; 60(5): 535-543, 2024 May.
Article in English | MEDLINE | ID: mdl-38656570

ABSTRACT

Gastrulation is the first major differentiation process in animal embryos. However, the dynamics of human gastrulation remain mostly unknown owing to the ethical limitations. We studied the dynamics of the mesoderm and endoderm cell differentiation from human pluripotent stem cells for insight into the cellular dynamics of human gastrulation. Human pluripotent stem cells have properties similar to those of the epiblast, which gives rise to the three germ layers. The mesoderm and endoderm were induced with more than 75% purity from human induced pluripotent stem cells. Single-cell dynamics of pluripotent stem cell-derived mesoderm and endoderm cells were traced using time-lapse imaging. Both mesoderm and endoderm cells migrate randomly, accompanied by short-term directional persistence. No substantial differences were detected between mesoderm and endoderm migration. Computer simulations created using the measured parameters revealed that random movement and external force, such as the spread out of cells from the primitive streak area, mimicked the homogeneous discoidal germ layer formation. These results were consistent with the development of amniotes, which suggests the effectiveness of human pluripotent stem cells as a good model for studying human embryogenesis.


Subject(s)
Cell Differentiation , Cell Movement , Endoderm , Mesoderm , Pluripotent Stem Cells , Humans , Endoderm/cytology , Mesoderm/cytology , Pluripotent Stem Cells/cytology , Computer Simulation
9.
Int J Mol Sci ; 25(5)2024 Feb 26.
Article in English | MEDLINE | ID: mdl-38473939

ABSTRACT

Embryonic stem cells (ESCs) favor glycolysis over oxidative phosphorylation for energy production, and glycolytic metabolism is critical for pluripotency establishment, maintenance, and exit. However, an understanding of how glycolysis regulates the self-renewal and differentiation of ESCs remains elusive. Here, we demonstrated that protein lactylation, regulated by intracellular lactate, contributes to the self-renewal of ESCs. We further showed that Esrrb, an orphan nuclear receptor involved in pluripotency maintenance and extraembryonic endoderm stem cell (XEN) differentiation, is lactylated on K228 and K232. The lactylation of Esrrb enhances its activity in promoting ESC self-renewal in the absence of the LIF and XEN differentiation of ESCs by increasing its binding at target genes. Our studies reveal the importance of protein lactylation in the self-renewal and XEN differentiation of ESCs, and the underlying mechanism of glycolytic metabolism regulating cell fate choice.


Subject(s)
Embryonic Stem Cells , Endoderm , Endoderm/metabolism , Cell Differentiation/genetics
10.
Results Probl Cell Differ ; 72: 27-60, 2024.
Article in English | MEDLINE | ID: mdl-38509251

ABSTRACT

As epiblast cells initiate development into various somatic cells, they undergo a large-scale reorganization, called gastrulation. The gastrulation of the epiblast cells produces three groups of cells: the endoderm layer, the collection of miscellaneous mesodermal tissues, and the ectodermal layer, which includes the neural, epidermal, and associated tissues. Most studies of gastrulation have focused on the formation of the tissues that provide the primary route for cell reorganization, that is, the primitive streak, in the chicken and mouse. In contrast, how gastrulation alters epiblast-derived cells has remained underinvestigated. This chapter highlights the regulation of cell and tissue fate via the gastrulation process. The roles and regulatory functions of neuromesodermal progenitors (NMPs) in the gastrulation process, elucidated in the last decade, are discussed in depth to resolve points of confusion. Chicken and mouse embryos, which form a primitive streak as the site of mesoderm precursor ingression, have been investigated extensively. However, primitive streak formation is an exception, even among amniotes. The roles of gastrulation processes in generating various somatic tissues will be discussed broadly.


Subject(s)
Gastrula , Gastrulation , Mice , Animals , Mesoderm , Endoderm , Embryonic Development
11.
Proc Natl Acad Sci U S A ; 121(11): e2315540121, 2024 Mar 12.
Article in English | MEDLINE | ID: mdl-38437561

ABSTRACT

Insects lack acquired immunity and were thought to have no immune memory, but recent studies reported a phenomenon called immune priming, wherein sublethal dose of pathogens or nonpathogenic microbes stimulates immunity and prevents subsequential pathogen infection. Although the evidence for insect immune priming is accumulating, the underlying mechanisms are still unclear. The bean bug Riptortus pedestris acquires its gut microbiota from ambient soil and spatially structures them into a multispecies and variable community in the anterior midgut and a specific, monospecies Caballeronia symbiont population in the posterior region. We demonstrate that a particular Burkholderia strain colonizing the anterior midgut stimulates systemic immunity by penetrating gut epithelia and migrating into the hemolymph. The activated immunity, consisting of a humoral and a cellular response, had no negative effect on the host fitness, but on the contrary protected the insect from subsequent infection by pathogenic bacteria. Interruption of contact between the Burkholderia strain and epithelia of the gut weakened the host immunity back to preinfection levels and made the insects more vulnerable to microbial infection, demonstrating that persistent acquisition of environmental bacteria is important to maintain an efficient immunity.


Subject(s)
Burkholderia , Burkholderiaceae , Animals , Endoderm , Insecta , Soil
12.
Nature ; 627(8004): 636-645, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38418875

ABSTRACT

A hallmark of cancer is the avoidance of immune destruction. This process has been primarily investigated in locally advanced or metastatic cancer1-3; however, much less is known about how pre-malignant or early invasive tumours evade immune detection. Here, to understand this process in early colorectal cancers (CRCs), we investigated how naive colon cancer organoids that were engineered in vitro to harbour Apc-null, KrasG12D and Trp53-null (AKP) mutations adapted to the in vivo native colonic environment. Comprehensive transcriptomic and chromatin analyses revealed that the endoderm-specifying transcription factor SOX17 became strongly upregulated in vivo. Notably, whereas SOX17 loss did not affect AKP organoid propagation in vitro, its loss markedly reduced the ability of AKP tumours to persist in vivo. The small fraction of SOX17-null tumours that grew displayed notable interferon-γ (IFNγ)-producing effector-like CD8+ T cell infiltrates in contrast to the immune-suppressive microenvironment in wild-type counterparts. Mechanistically, in both endogenous Apc-null pre-malignant adenomas and transplanted organoid-derived AKP CRCs, SOX17 suppresses the ability of tumour cells to sense and respond to IFNγ, preventing anti-tumour T cell responses. Finally, SOX17 engages a fetal intestinal programme that drives differentiation away from LGR5+ tumour cells to produce immune-evasive LGR5- tumour cells with lower expression of major histocompatibility complex class I (MHC-I). We propose that SOX17 is a transcription factor that is engaged during the early steps of colon cancer to orchestrate an immune-evasive programme that permits CRC initiation and progression.


Subject(s)
Adenoma , Colorectal Neoplasms , Immune Evasion , SOXF Transcription Factors , Animals , Humans , Mice , Adenoma/immunology , Adenoma/pathology , CD8-Positive T-Lymphocytes/immunology , Chromatin/genetics , Colorectal Neoplasms/immunology , Colorectal Neoplasms/pathology , Gene Expression Profiling , Interferon-gamma/immunology , Organoids/immunology , Organoids/pathology , SOXF Transcription Factors/metabolism , Tumor Microenvironment/immunology , Mutation , Endoderm/metabolism , Disease Progression
13.
Nat Protoc ; 19(4): 1149-1182, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38302732

ABSTRACT

Human primordial germ cells (hPGCs), the precursors of eggs and sperm, start their complex development shortly after specification and during their migration to the primitive gonads. Here, we describe protocols for specifying hPGC-like cells (hPGCLCs) from resetting precursors and progressing them with the support of human hindgut organoids. Resetting hPGCLCs (rhPGCLCs) are specified from human embryonic stem cells (hESCs) transitioning from the primed into the naive state of pluripotency. Hindgut organoids are also derived from hESCs after a sequential differentiation into a posterior endoderm/hindgut fate. Both rhPGCLCs and hindgut organoids are combined and co-cultured for 25 d. The entire procedure takes ~1.5 months and can be successfully implemented by a doctoral or graduate student with basic skills and experience in hESC cultures. The co-culture system supports the progression of rhPGCLCs at a developmental timing analogous to that observed in vivo. Compared with previously developed hPGCLC progression protocols, which depend on co-cultures with mouse embryonic gonadal tissue, our co-culture system represents a developmentally relevant model closer to the environment that hPGCs first encounter after specification. Together with the potential for investigations of events during hPGC specification and early development, these protocols provide a practical approach to designing efficient models for in vitro gametogenesis. Notably, the rhPGCLC-hindgut co-culture system can also be adapted to study failings in hPGC migration, which are associated with the etiology of some forms of infertility and germ cell tumors.


Subject(s)
Endoderm , Semen , Humans , Male , Animals , Mice , Germ Cells , Cell Differentiation , Organoids
14.
FASEB J ; 38(4): e23463, 2024 Feb 29.
Article in English | MEDLINE | ID: mdl-38334393

ABSTRACT

With self-renewal and pluripotency features, embryonic stem cells (ESCs) provide an invaluable tool to investigate early cell fate decisions. Pluripotency exit and lineage commitment depend on precise regulation of gene expression that requires coordination between transcription (TF) and chromatin factors in response to various signaling pathways. SET domain-containing 3 (SETD3) is a methyltransferase that can modify histones in the nucleus and actin in the cytoplasm. Through an shRNA screen, we previously identified SETD3 as an important factor in the meso/endodermal lineage commitment of mouse ESCs (mESC). In this study, we identified SETD3-dependent transcriptomic changes during endoderm differentiation of mESCs using time-course RNA-seq analysis. We found that SETD3 is involved in the timely activation of the endoderm-related gene network. The canonical Wnt signaling pathway was one of the markedly altered signaling pathways in the absence of SETD3. The assessment of Wnt transcriptional activity revealed a significant reduction in Setd3-deleted (setd3∆) mESCs coincident with a decrease in the nuclear pool of the key TF ß-catenin level, though no change was observed in its mRNA or total protein level. Furthermore, a proximity ligation assay (PLA) found an interaction between SETD3 and ß-catenin. We were able to rescue the differentiation defect by stably re-expressing SETD3 or activating the canonical Wnt signaling pathway by changing mESC culture conditions. Our results suggest that alterations in the canonical Wnt pathway activity and subcellular localization of ß-catenin might contribute to the endoderm differentiation defect of setd3∆ mESCs.


Subject(s)
Mouse Embryonic Stem Cells , beta Catenin , Animals , Mice , beta Catenin/metabolism , Cell Differentiation/genetics , Endoderm , Wnt Signaling Pathway/physiology
15.
Cell Biol Int ; 48(6): 835-847, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38419492

ABSTRACT

Pluripotent stem cells (PSCs) hold enormous potential for treating multiple diseases owing to their ability to self-renew and differentiate into any cell type. Albeit possessing such promising potential, controlling their differentiation into a desired cell type continues to be a challenge. Recent studies suggest that PSCs respond to different substrate stiffness and, therefore, can differentiate towards some lineages via Hippo pathway. Human PSCs can also differentiate and self-organize into functional cells, such as organoids. Traditionally, human PSCs are differentiated on stiff plastic or glass plates towards definitive endoderm and then into functional pancreatic progenitor cells in the presence of soluble growth factors. Thus, whether stiffness plays any role in differentiation towards definitive endoderm from human pluripotent stem cells (hPSCs) remains unclear. Our study found that the directed differentiation of human embryonic stem cells towards endodermal lineage on the varying stiffness did not differ from the differentiation on stiff plastic dishes. We also observed no statistical difference between the expression of yes-associated protein (YAP) and phosphorylated YAP. Furthermore, we demonstrate that lysophosphatidic acid, a YAP activator, enhanced definitive endoderm formation, whereas verteporfin, a YAP inhibitor, did not have the significant effect on the differentiation. In summary, our results suggest that human embryonic stem cells may not differentiate in response to changes in stiffness, and that such cues may not have as significant impact on the level of YAP. Our findings indicate that more research is needed to understand the direct relationship between biophysical forces and hPSCs differentiation.


Subject(s)
Cell Differentiation , Cell Lineage , Endoderm , Human Embryonic Stem Cells , Humans , Cell Differentiation/physiology , Human Embryonic Stem Cells/cytology , Human Embryonic Stem Cells/metabolism , Endoderm/cytology , Endoderm/metabolism , YAP-Signaling Proteins/metabolism , Transcription Factors/metabolism
16.
Nucleic Acids Res ; 52(9): 4935-4949, 2024 May 22.
Article in English | MEDLINE | ID: mdl-38421638

ABSTRACT

TGF-ß signaling family plays an essential role to regulate fate decisions in pluripotency and lineage specification. How the action of TGF-ß family signaling is intrinsically executed remains not fully elucidated. Here, we show that HBO1, a MYST histone acetyltransferase (HAT) is an essential cell intrinsic determinant for TGF-ß signaling in human embryonic stem cells (hESCs). HBO1-/- hESCs fail to response to TGF-ß signaling to maintain pluripotency and spontaneously differentiate into neuroectoderm. Moreover, HBO1 deficient hESCs show complete defect in mesendoderm specification in BMP4-triggered gastruloids or teratomas. Molecularly, HBO1 interacts with SMAD4 and co-binds the open chromatin labeled by H3K14ac and H3K4me3 in undifferentiated hESCs. Upon differentiation, HBO1/SMAD4 co-bind and maintain the mesoderm genes in BMP4-triggered mesoderm cells while lose chromatin occupancy in neural cells induced by dual-SMAD inhibition. Our data reveal an essential role of HBO1, a chromatin factor to determine the action of SMAD in both human pluripotency and mesendoderm specification.


Subject(s)
Cell Differentiation , Histone Acetyltransferases , Mesoderm , Signal Transduction , Smad4 Protein , Humans , Bone Morphogenetic Protein 4/metabolism , Bone Morphogenetic Protein 4/genetics , Cell Line , Chromatin/metabolism , Endoderm/cytology , Endoderm/metabolism , Histone Acetyltransferases/metabolism , Histone Acetyltransferases/genetics , Histones/metabolism , Human Embryonic Stem Cells/metabolism , Human Embryonic Stem Cells/cytology , Mesoderm/metabolism , Mesoderm/cytology , Pluripotent Stem Cells/metabolism , Pluripotent Stem Cells/cytology , Smad4 Protein/metabolism , Smad4 Protein/genetics , Transforming Growth Factor beta/metabolism
17.
Cell Rep ; 43(2): 113701, 2024 Feb 27.
Article in English | MEDLINE | ID: mdl-38277271

ABSTRACT

Human embryo implantation is remarkably inefficient, and implantation failure remains among the greatest obstacles in treating infertility. Gene expression data from human embryos have accumulated rapidly in recent years; however, identification of the subset of genes that determine successful implantation remains a challenge. We leverage clinical morphologic grading-known for decades to correlate with implantation potential-and transcriptome analyses of matched embryonic and abembryonic samples to identify factors and pathways enriched and depleted in human blastocysts of good and poor morphology. Unexpectedly, we discovered that the greatest difference was in the state of extraembryonic primitive endoderm (PrE) development, with relative deficiencies in poor morphology blastocysts. Our results suggest that implantation success is most strongly influenced by the embryonic compartment and that deficient PrE development is common among embryos with decreased implantation potential. Our study provides a valuable resource for those investigating the markers and mechanisms of human embryo implantation.


Subject(s)
Embryonic Development , Endoderm , Humans , Embryonic Development/genetics , Embryo Implantation/genetics , Blastocyst/metabolism , Embryo, Mammalian
18.
Stem Cell Reports ; 19(2): 174-186, 2024 Feb 13.
Article in English | MEDLINE | ID: mdl-38215757

ABSTRACT

In early mammalian development, cleavage stage blastomeres and inner cell mass (ICM) cells co-express embryonic and extra-embryonic transcriptional determinants. Using a protein-based double reporter we identify an embryonic stem cell (ESC) population that co-expresses the extra-embryonic factor GATA6 alongside the embryonic factor SOX2. Based on single cell transcriptomics, we find this population resembles the unsegregated ICM, exhibiting enhanced differentiation potential for endoderm while maintaining epiblast competence. To relate transcription factor binding in these cells to future fate, we describe a complete enhancer set in both ESCs and naive extra-embryonic endoderm stem cells and assess SOX2 and GATA6 binding at these elements in the ICM-like ESC sub-population. Both factors support cooperative recognition in these lineages, with GATA6 bound alongside SOX2 on a fraction of pluripotency enhancers and SOX2 alongside GATA6 more extensively on endoderm enhancers, suggesting that cooperative binding between these antagonistic factors both supports self-renewal and prepares progenitor cells for later differentiation.


Subject(s)
Gene Expression Regulation, Developmental , Transcription Factors , Animals , Cell Lineage/genetics , Transcription Factors/genetics , Transcription Factors/metabolism , Cell Differentiation/genetics , Germ Layers , Endoderm , Blastocyst , Mammals/metabolism
19.
Regen Med ; 19(1): 47-63, 2024 Jan.
Article in English | MEDLINE | ID: mdl-38240144

ABSTRACT

Aim: Cell therapies for diabetes rely on differentiation of stem cells into insulin-producing cells, which is complex and expensive. Our goal was to evaluate production costs and test ways to reduce it. Methods: Cost of Goods (COGs) analysis for differentiation was completed and the effects of replacement or reduction of the most expensive item was tested using qRT-PCR, immunohistochemistry, flow cytometry along with glucose-stimulated insulin release. Results: Activin A (AA) was responsible for significant cost. Replacement with small molecules failed to form definitive endoderm (DE). Reducing AA by 50% did not negatively affect expression of beta cell markers. Conclusion: Reduction of AA concentration is feasible without adversely affecting DE and islet-like cell differentiation, leading to significant cost savings in manufacturing.


Subject(s)
Endoderm , Insulins , Endoderm/metabolism , Cell Differentiation , Activins/metabolism , Activins/pharmacology , Insulins/metabolism
20.
In Vitro Cell Dev Biol Anim ; 60(5): 521-534, 2024 May.
Article in English | MEDLINE | ID: mdl-38169039

ABSTRACT

Trisomy 12 is one of the most frequent chromosomal abnormalities in cultured human pluripotent stem cells (hPSCs). Although potential oncogenic properties and augmented cell cycle caused by trisomy 12 have been reported, the consequences of trisomy 12 in terms of cell differentiation, which is the basis for regenerative medicine, drug development, and developmental biology studies, have not yet been investigated. Here, we report that trisomy 12 compromises the mesendodermal differentiation of hPSCs. We identified sublines of hPSCs carrying trisomy 12 after their prolonged culture. Transcriptome analysis revealed that these hPSC sublines carried abnormal gene expression patterns in specific signaling pathways in addition to cancer-related cell cycle pathways. These hPSC sublines showed a lower propensity for mesendodermal differentiation in embryoid bodies cultured in a serum-free medium. BMP4-induced exit from the self-renewal state was impaired in the trisomy 12 hPSC sublines, with less upregulation of key transcription factor gene expression. As a consequence, the differentiation efficiency of hematopoietic and hepatic lineages was also impaired in the trisomy 12 hPSC sublines. We reveal that trisomy 12 disrupts the genome-wide expression patterns that are required for proper mesendodermal differentiation.


Subject(s)
Cell Differentiation , Chromosomes, Human, Pair 12 , Pluripotent Stem Cells , Trisomy , Humans , Cell Differentiation/genetics , Trisomy/genetics , Chromosomes, Human, Pair 12/genetics , Pluripotent Stem Cells/metabolism , Pluripotent Stem Cells/cytology , Mesoderm/cytology , Endoderm/cytology , Endoderm/metabolism , Bone Morphogenetic Protein 4/metabolism , Bone Morphogenetic Protein 4/genetics , Embryoid Bodies/cytology , Embryoid Bodies/metabolism , Cell Line , Signal Transduction/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...