Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 24
Filter
Add more filters










Publication year range
1.
Adv Sci (Weinh) ; 10(15): e2204741, 2023 05.
Article in English | MEDLINE | ID: mdl-36998105

ABSTRACT

Multicellular patterning of stem-cell-derived tissue models is commonly achieved via self-organizing activities triggered by exogenous morphogenetic stimuli. However, such tissue models are prone to stochastic behavior, limiting the reproducibility of cellular composition and forming non-physiological architectures. To enhance multicellular patterning in stem cell-derived tissues, a method for creating complex tissue microenvironments endowed with programmable multimodal mechano-chemical cues, including conjugated peptides, proteins, morphogens, and Young's moduli defined over a range of stiffnesses is developed. The ability of these cues to spatially guide tissue patterning processes, including mechanosensing and the biochemically driven differentiation of selected cell types, is demonstrated. By rationally designing niches, the authors engineered a bone-fat assembly from stromal mesenchyme cells and regionalized germ layer tissues from pluripotent stem cells. Through defined niche-material interactions, mechano-chemically microstructured niches enable the spatial programming of tissue patterning processes. Mechano-chemically microstructured cell niches thereby offer an entry point for enhancing the organization and composition of engineered tissues, potentiating structures that better recapitulate their native counterparts.


Subject(s)
Pluripotent Stem Cells , Tissue Engineering , Reproducibility of Results , Tissue Engineering/methods , Morphogenesis , Bone and Bones
2.
Methods Mol Biol ; 2490: 3-9, 2022.
Article in English | MEDLINE | ID: mdl-35486234

ABSTRACT

Epiblast stem cells are made from the epiblast of mouse post-implantation embryo. They have been critical in the understanding of mammalian pluripotent stem cells as they share similar properties, such as their incapability to contribute to the formation of an embryo after injection into blastocyst. The epiblast stem cells (EpiSC) have delineated a novel status of pluripotency called "primed." How to establish EpiSC from mouse embryo is described in detail in this chapter.


Subject(s)
Germ Layers , Pluripotent Stem Cells , Animals , Blastocyst , Embryo, Mammalian , Embryonic Development , Mammals , Mice
3.
Methods Mol Biol ; 2490: 93-100, 2022.
Article in English | MEDLINE | ID: mdl-35486242

ABSTRACT

The assay for transposase-accessible chromatin using sequencing (ATAC-seq) is used to identify open chromatin regions in cells. This can be used to identify putative regulatory regions, determine dynamics and mechanisms of transcription factors when coupled with ChIP-seq and predict interactions between proteins and chromatin. Compared to previous methods, MNase-seq and DNase-seq, ATAC-seq requires only 50,000 cells, orders of magnitude fewer cells. In addition, the ATAC-seq protocol takes one day to progress from cells to sequencing ready libraries.


Subject(s)
Chromatin Immunoprecipitation Sequencing , Chromatin , Animals , Chromatin/genetics , Germ Layers , High-Throughput Nucleotide Sequencing/methods , Mice , Stem Cells
4.
Methods Mol Biol ; 2490: 269-279, 2022.
Article in English | MEDLINE | ID: mdl-35486252

ABSTRACT

Here we describe a method to engraft epiblast stem cells (EpiSC) into the epiblast of gastrulation-stage mouse embryo to test the lineage propensity acquired by the EpiSCs during in vitro culture under different signaling conditions. After dissection and grafting, the recipient embryos can be grown in whole-embryo culture for up to 48 h and the contribution of the EpiSC-derived cells to tissues in the recipient embryo is assessed by light sheet 3D microscopy.


Subject(s)
Embryo, Mammalian , Germ Layers , Animals , Cell Differentiation , Gastrulation , Mice , Stem Cells
5.
Genesis ; 60(1-2): e23466, 2022 02.
Article in English | MEDLINE | ID: mdl-35104045

ABSTRACT

Allocation of cells to an endodermal fate in the gastrulating embryo is driven by Nodal signaling and consequent activation of TGFß pathway. In vitro methodologies striving to recapitulate the process of endoderm differentiation, however, use TGFß family member Activin in place of Nodal. This is despite Activin not known to have an in vivo role in endoderm differentiation. In this study, five epiblast stem cell lines were subjected to directed differentiation using both Activin A and Nodal to induce endodermal fate. A reporter line harboring endoderm markers FoxA2 and Sox17 was further analyzed for TGFß pathway activation and WNT response. We demonstrated that Activin A-treated cells remain more primitive streak-like when compared to Nodal-treated cells that have a molecular profile suggestive of more advanced differentiation. Activin A elicited a robust TGFß/SMAD activity, enhanced WNT signaling activity and promoted the generation of DE precursors. Nodal treatment resulted in lower TGFß/SMAD activity, and a weaker, sustained WNT response, and ultimately failed to upregulate endoderm markers. This is despite signaling response resembling more closely the activity seen in vivo. These findings emphasize the importance of understanding the downstream activities of Activin A and Nodal signaling in directing in vitro endoderm differentiation of primed-state epiblast stem cells.


Subject(s)
Endoderm , Nodal Protein , Activins/metabolism , Activins/pharmacology , Cell Differentiation/physiology , Endoderm/metabolism , Germ Layers , Nodal Protein/genetics , Nodal Protein/metabolism , Stem Cells/metabolism , Transforming Growth Factor beta
6.
Elife ; 102021 02 08.
Article in English | MEDLINE | ID: mdl-33554859

ABSTRACT

Protein interaction is critical molecular regulatory activity underlining cellular functions and precise cell fate choices. Using TWIST1 BioID-proximity-labeling and network propagation analyses, we discovered and characterized a TWIST-chromatin regulatory module (TWIST1-CRM) in the neural crest cells (NCC). Combinatorial perturbation of core members of TWIST1-CRM: TWIST1, CHD7, CHD8, and WHSC1 in cell models and mouse embryos revealed that loss of the function of the regulatory module resulted in abnormal differentiation of NCCs and compromised craniofacial tissue patterning. Following NCC delamination, low level of TWIST1-CRM activity is instrumental to stabilize the early NCC signatures and migratory potential by repressing the neural stem cell programs. High level of TWIST1 module activity at later phases commits the cells to the ectomesenchyme. Our study further revealed the functional interdependency of TWIST1 and potential neurocristopathy factors in NCC development.


Shaping the head and face during development relies on a complex ballet of molecular signals that orchestrates the movement and specialization of various groups of cells. In animals with a backbone for example, neural crest cells (NCCs for short) can march long distances from the developing spine to become some of the tissues that form the skull and cartilage but also the pigment cells and nervous system. NCCs mature into specific cell types thanks to a complex array of factors which trigger a precise sequence of binary fate decisions at the right time and place. Amongst these factors, the protein TWIST1 can set up a cascade of genetic events that control how NCCs will ultimately form tissues in the head. To do so, the TWIST1 protein interacts with many other molecular actors, many of which are still unknown. To find some of these partners, Fan et al. studied TWIST1 in the NCCs of mice and cells grown in the lab. The experiments showed that TWIST1 interacted with CHD7, CHD8 and WHSC1, three proteins that help to switch genes on and off, and which contribute to NCCs moving across the head during development. Further work by Fan et al. then revealed that together, these molecular actors are critical for NCCs to form cells that will form facial bones and cartilage, as opposed to becoming neurons. This result helps to show that there is a trade-off between NCCs forming the face or being part of the nervous system. One in three babies born with a birth defect shows anomalies of the head and face: understanding the exact mechanisms by which NCCs contribute to these structures may help to better predict risks for parents, or to develop new approaches for treatment.


Subject(s)
Cell Differentiation , Chromatin/metabolism , Neural Crest/cytology , Neural Crest/metabolism , Twist-Related Protein 1/metabolism , Animals , Chromatin/genetics , Embryonic Stem Cells/cytology , Embryonic Stem Cells/metabolism , Mice , Neural Crest/embryology , Twist-Related Protein 1/genetics
7.
Int J Mol Sci ; 21(19)2020 Oct 02.
Article in English | MEDLINE | ID: mdl-33023104

ABSTRACT

Induced pluripotent stem cells (iPSCs) are obtained by genetically reprogramming adult somatic cells via the overexpression of specific pluripotent genes. The resulting cells possess the same differentiation properties as blastocyst-stage embryonic stem cells (ESCs) and can be used to produce new individuals by embryonic complementation, nuclear transfer cloning, or in vitro fertilization after differentiation into male or female gametes. Therefore, iPSCs are highly valuable for preserving biodiversity and, together with somatic cells, can enlarge the pool of reproductive samples for cryobanking. In this study, we subjected rabbit iPSCs (rbiPSCs) and rabbit ear tissues to several cryopreservation conditions with the aim of defining safe and non-toxic slow-freezing protocols. We compared a commercial synthetic medium (STEM ALPHA.CRYO3) with a biological medium based on fetal bovine serum (FBS) together with low (0-5%) and high (10%) concentrations of dimethyl sulfoxide (DMSO). Our data demonstrated the efficacy of a CRYO3-based medium containing 4% DMSO for the cryopreservation of skin tissues and rbiPSCs. Specifically, this medium provided similar or even better biological results than the commonly used freezing medium composed of FBS and 10% DMSO. The results of this study therefore represent an encouraging first step towards the use of iPSCs for species preservation.


Subject(s)
Cell Differentiation/genetics , Cryopreservation , Induced Pluripotent Stem Cells/cytology , Pluripotent Stem Cells/cytology , Animals , Biological Specimen Banks , Blastocyst/drug effects , Blastocyst/metabolism , Cell Differentiation/drug effects , Cryoprotective Agents/pharmacology , Ear/growth & development , Male , Rabbits
8.
J Bioinform Comput Biol ; 18(1): 2040003, 2020 02.
Article in English | MEDLINE | ID: mdl-32336246

ABSTRACT

Assays for transposase-accessible chromatin sequencing (ATAC-seq) provides an innovative approach to study chromatin status in multiple cell types. Moreover, it is also possible to efficiently extract differentially accessible chromatin (DACs) regions by using state-of-the-art algorithms (e.g. DESeq2) to predict gene activity in specific samples. Furthermore, it has recently been shown that small dips in sequencing peaks can be attributed to the binding of transcription factors. These dips, also known as footprints, can be used to identify trans-regulating interactions leading to gene expression. Current protocols used to identify footprints (e.g. pyDNAse and HINT-ATAC) have shown limitations resulting in the discovery of many false positive footprints. We generated a novel approach to identify genuine footprints within any given ATAC-seq dataset. Herein, we developed a new pipeline embedding DACs together with bona fide footprints resulting in the generation of a Predictive gene regulatory Network (PreNet) simply from ATAC-seq data. We further demonstrated that PreNet can be used to unveil meaningful molecular regulatory pathways in a given cell type.


Subject(s)
Chromatin/genetics , Computational Biology/methods , Software , Transposases/genetics , Animals , Chromatin Immunoprecipitation Sequencing , Gene Regulatory Networks , Mice , Mouse Embryonic Stem Cells/physiology , Promoter Regions, Genetic , Reproducibility of Results , Transcription Factors/genetics
9.
Mol Cell Biol ; 40(11)2020 05 14.
Article in English | MEDLINE | ID: mdl-32179550

ABSTRACT

The extensive array of basic helix-loop-helix (bHLH) transcription factors and their combinations as dimers underpin the diversity of molecular function required for cell type specification during embryogenesis. The bHLH factor TWIST1 plays pleiotropic roles during development. However, which combinations of TWIST1 dimers are involved and what impact each dimer imposes on the gene regulation network controlled by TWIST1 remain elusive. In this work, proteomic profiling of human TWIST1-expressing cell lines and transcriptome analysis of mouse cranial mesenchyme have revealed that TWIST1 homodimers and heterodimers with TCF3, TCF4, and TCF12 E-proteins are the predominant dimer combinations. Disease-causing mutations in TWIST1 can impact dimer formation or shift the balance of different types of TWIST1 dimers in the cell, which may underpin the defective differentiation of the craniofacial mesenchyme. Functional analyses of the loss and gain of TWIST1-E-protein dimer activity have revealed previously unappreciated roles in guiding lineage differentiation of embryonic stem cells: TWIST1-E-protein heterodimers activate the differentiation of mesoderm and neural crest cells, which is accompanied by the epithelial-to-mesenchymal transition. At the same time, TWIST1 homodimers maintain the stem cells in a progenitor state and block entry to the endoderm lineage.


Subject(s)
Cell Differentiation , Nuclear Proteins/metabolism , Protein Multimerization , Twist-Related Protein 1/metabolism , Animals , Cell Line , Dogs , Epithelial-Mesenchymal Transition , Gene Expression Regulation, Developmental , Humans , Madin Darby Canine Kidney Cells , Mesoderm/cytology , Mesoderm/metabolism , Mice, Inbred C57BL , Mutation , Neural Crest/cytology , Neural Crest/metabolism , Nuclear Proteins/chemistry , Nuclear Proteins/genetics , Transcriptome , Twist-Related Protein 1/chemistry , Twist-Related Protein 1/genetics
10.
Nucleic Acids Res ; 48(4): 1828-1842, 2020 02 28.
Article in English | MEDLINE | ID: mdl-31853542

ABSTRACT

The developmental potential of cells, termed pluripotency, is highly dynamic and progresses through a continuum of naive, formative and primed states. Pluripotency progression of mouse embryonic stem cells (ESCs) from naive to formative and primed state is governed by transcription factors (TFs) and their target genes. Genomic techniques have uncovered a multitude of TF binding sites in ESCs, yet a major challenge lies in identifying target genes from functional binding sites and reconstructing dynamic transcriptional networks underlying pluripotency progression. Here, we integrated time-resolved 'trans-omic' datasets together with TF binding profiles and chromatin conformation data to identify target genes of a panel of TFs. Our analyses revealed that naive TF target genes are more likely to be TFs themselves than those of formative TFs, suggesting denser hierarchies among naive TFs. We also discovered that formative TF target genes are marked by permissive epigenomic signatures in the naive state, indicating that they are poised for expression prior to the initiation of pluripotency transition to the formative state. Finally, our reconstructed transcriptional networks pinpointed the precise timing from naive to formative pluripotency progression and enabled the spatiotemporal mapping of differentiating ESCs to their in vivo counterparts in developing embryos.


Subject(s)
Embryonic Development/genetics , Mouse Embryonic Stem Cells/metabolism , Pluripotent Stem Cells/metabolism , Transcription Factors/genetics , Animals , Binding Sites/genetics , Cell Differentiation/genetics , Chromatin/genetics , Gene Expression Regulation, Developmental/genetics , Gene Regulatory Networks/genetics , Genome/genetics , Mice
11.
Dev Growth Differ ; 61(5): 327-336, 2019 Jun.
Article in English | MEDLINE | ID: mdl-31111476

ABSTRACT

Development of an embryo is driven by a series of molecular instructions that control the differentiation of tissue precursor cells and shape the tissues into major body parts. LIM homeobox 1 (LHX1) is a transcription factor that plays a major role in the development of the embryonic head of the mouse. Loss of LHX1 function disrupts the morphogenetic movement of head tissue precursors and impacts on the function of molecular factors in modulating the activity of the WNT signaling pathway. LHX1 acts with a transcription factor complex to regulate the transcription of target genes in multiple phases of development and in a range of embryonic tissues of the mouse and Xenopus. Determining the interacting factors and transcriptional targets of LHX1 will be key to unraveling the ensemble of factors involved in head development and building a head gene regulatory network.


Subject(s)
Embryo, Mammalian/embryology , Embryo, Mammalian/metabolism , Gene Regulatory Networks , Head/embryology , LIM-Homeodomain Proteins/metabolism , Animals , Gene Regulatory Networks/genetics , Humans , LIM-Homeodomain Proteins/deficiency , LIM-Homeodomain Proteins/genetics , Transcription Factors/deficiency , Transcription Factors/genetics , Transcription Factors/metabolism
12.
Development ; 146(7)2019 04 05.
Article in English | MEDLINE | ID: mdl-30890572

ABSTRACT

During embryogenesis, the stringent regulation of Wnt activity is crucial for the morphogenesis of the head and brain. The loss of function of the Wnt inhibitor Dkk1 results in elevated Wnt activity, loss of ectoderm lineage attributes from the anterior epiblast, and the posteriorisation of anterior germ layer tissue towards the mesendoderm. The modulation of Wnt signalling may therefore be crucial for the allocation of epiblast cells to ectoderm progenitors during gastrulation. To test this hypothesis, we examined the lineage characteristics of epiblast stem cells (EpiSCs) that were derived and maintained under different signalling conditions. We showed that suppression of Wnt activity enhanced the ectoderm propensity of the EpiSCs. Neuroectoderm differentiation of these EpiSCs was further empowered by the robust re-activation of Wnt activity. Therefore, during gastrulation, the tuning of the signalling activities that mediate mesendoderm differentiation is instrumental for the acquisition of ectoderm potency in the epiblast.


Subject(s)
Cell Differentiation/physiology , Ectoderm/cytology , Germ Layers/cytology , Animals , Cell Differentiation/genetics , Cells, Cultured , Ectoderm/metabolism , Gastrulation/genetics , Gastrulation/physiology , Gene Expression Regulation, Developmental/genetics , Gene Expression Regulation, Developmental/physiology , Germ Layers/metabolism , Mice , Signal Transduction/genetics , Signal Transduction/physiology
13.
Methods Mol Biol ; 1940: 77-95, 2019.
Article in English | MEDLINE | ID: mdl-30788819

ABSTRACT

Efficient and reliable methods for gene editing are critical for the generation of loss-of-gene function stem cells and genetically modified mice. Here, we outline the application of CRISPR-Cas9 technology for gene editing in mouse embryonic stem cells (mESCs) to generate knockout ESC chimeras for the fast-tracked analysis of gene function. Furthermore, we describe the application of gene editing directly to mouse epiblast stem cells (mEpiSCs) for modelling germ layer differentiation in vitro.


Subject(s)
Frameshift Mutation/genetics , Gene Editing/methods , Gene Knockout Techniques/methods , Germ Layers/cytology , Mouse Embryonic Stem Cells/cytology , Animals , CRISPR-Cas Systems/genetics , Cells, Cultured , Clustered Regularly Interspaced Short Palindromic Repeats/genetics , Mice , Plasmids/genetics , RNA, Guide, Kinetoplastida/genetics
14.
Cell Stem Cell ; 23(4): 586-598.e8, 2018 10 04.
Article in English | MEDLINE | ID: mdl-30290179

ABSTRACT

Cardiac differentiation of human pluripotent stem cells (hPSCs) requires orchestration of dynamic gene regulatory networks during stepwise fate transitions but often generates immature cell types that do not fully recapitulate properties of their adult counterparts, suggesting incomplete activation of key transcriptional networks. We performed extensive single-cell transcriptomic analyses to map fate choices and gene expression programs during cardiac differentiation of hPSCs and identified strategies to improve in vitro cardiomyocyte differentiation. Utilizing genetic gain- and loss-of-function approaches, we found that hypertrophic signaling is not effectively activated during monolayer-based cardiac differentiation, thereby preventing expression of HOPX and its activation of downstream genes that govern late stages of cardiomyocyte maturation. This study therefore provides a key transcriptional roadmap of in vitro cardiac differentiation at single-cell resolution, revealing fundamental mechanisms underlying heart development and differentiation of hPSC-derived cardiomyocytes.


Subject(s)
Cell Differentiation/genetics , Homeodomain Proteins/genetics , Myocytes, Cardiac/cytology , Pluripotent Stem Cells/cytology , Single-Cell Analysis , Transcriptome , Tumor Suppressor Proteins/genetics , Animals , Cells, Cultured , Female , Homeodomain Proteins/metabolism , Humans , Male , Mice , Mice, Inbred C3H , Mice, Inbred C57BL , Mice, Inbred NOD , Mice, Knockout , Mice, Transgenic , Myocytes, Cardiac/metabolism , Pluripotent Stem Cells/metabolism , Tumor Suppressor Proteins/metabolism
15.
Genesis ; 56(9): e23246, 2018 09.
Article in English | MEDLINE | ID: mdl-30114334

ABSTRACT

Development of the embryonic head is driven by the activity of gene regulatory networks of transcription factors. LHX1 is a homeobox transcription factor that plays an essential role in the formation of the embryonic head. The loss of LHX1 function results in anterior truncation of the embryo caused by the disruption of morphogenetic movement of tissue precursors and the dysregulation of WNT signaling activity. Profiling the gene expression pattern in the Lhx1 mutant embryo revealed that tissues in anterior germ layers acquire posterior tissue characteristics, suggesting LHX1 activity is required for the allocation and patterning of head precursor tissues. Here, we used LHX1 as an entry point to delineate its transcriptional targets and interactors and construct a LHX1-anchored gene regulatory network. Using a gain-of-function approach, we identified genes that immediately respond to Lhx1 activation. Meta-analysis of the datasets of LHX1-responsive genes and genes expressed in the anterior tissues of mouse embryos at head-fold stage, in conjunction with published Xenopus embryonic LHX1 (Xlim1) ChIP-seq data, has pinpointed the putative transcriptional targets of LHX1 and an array of genetic determinants functioning together in the formation of the mouse embryonic head.


Subject(s)
Gene Regulatory Networks , Genes, Homeobox , Head/embryology , LIM-Homeodomain Proteins/metabolism , Transcription Factors/metabolism , Animals , Embryonic Stem Cells/metabolism , Gene Expression Profiling , Germ Cells/physiology , Transcription, Genetic , Xenopus laevis/embryology
16.
Stem Cell Reports ; 11(1): 43-57, 2018 07 10.
Article in English | MEDLINE | ID: mdl-30008328

ABSTRACT

The molecular mechanism underpinning the specification of the ectoderm, a transient germ-layer tissue, during mouse gastrulation was examined here in a stem cell-based model. We captured a self-renewing cell population with enhanced ectoderm potency from mouse epiblast stem cells (EpiSCs) by suppressing Nodal signaling activity. The transcriptome of the Nodal-inhibited EpiSCs resembles that of the anterior epiblast of embryonic day (E)7.0 and E7.5 mouse embryo, which is accompanied by chromatin modifications that reflect the priming of ectoderm lineage-related genes for expression. Nodal-inhibited EpiSCs show enhanced ectoderm differentiation in vitro and contribute to the neuroectoderm and the surface ectoderm in postimplantation chimeras but lose the propensity for mesendoderm differentiation in vitro and in chimeras. Our findings show that specification of the ectoderm progenitors is enhanced by the repression of Nodal signaling activity, and the ectoderm-like stem cells provide an experimental model to investigate the molecular characters of the epiblast-derived ectoderm.


Subject(s)
Cell Differentiation , Ectoderm/cytology , Ectoderm/metabolism , Germ Layers/cytology , Germ Layers/metabolism , Nodal Protein/metabolism , Signal Transduction , Animals , Biomarkers , Cell Lineage , Cells, Cultured , Ectoderm/embryology , Embryonic Development/genetics , Epigenesis, Genetic , Fluorescent Antibody Technique , Gene Expression Profiling , Gene Expression Regulation, Developmental , Mice , Wnt Signaling Pathway
17.
Stem Cell Res ; 24: 106-117, 2017 10.
Article in English | MEDLINE | ID: mdl-28889080

ABSTRACT

Rabbit induced pluripotent stem cells (rbiPSCs) possess the characteristic features of primed pluripotency as defined in rodents and primates. In the present study, we reprogrammed rbiPSCs using human Krüppel-like factors (KLFs) 2 and 4 and cultured them in a medium supplemented with fetal calf serum and leukemia inhibitory factor. These cells (designated rbEKA) were propagated by enzymatic dissociation for at least 30 passages, during which they maintained a normal karyotype. This new culturing protocol resulted in transcriptional and epigenetic reconfiguration, as substantiated by the expression of transcription factors and the presence of histone modifications associated with naïve pluripotency. Furthermore, microarray analysis of rbiPSCs, rbEKA cells, rabbit ICM cells, and rabbit epiblast showed that the global gene expression profile of the reprogrammed rbiPSCs was more similar to that of rabbit ICM and epiblast cells. Injection of rbEKA cells into 8-cell stage rabbit embryos resulted in extensive colonization of ICM in 9% early-blastocysts (E3.5), epiblast in 10% mid-blastocysts (E4.5), and embryonic disk in 1.4% pre-gastrulae (E6). Thus, these results indicate that KLF2 and KLF4 triggered the conversion of rbiPSCs into epiblast-like, embryo colonization-competent PSCs. Our results highlight some of the requirements to achieve bona fide chimeric competency.


Subject(s)
Cellular Reprogramming , Germ Layers/cytology , Induced Pluripotent Stem Cells/cytology , Kruppel-Like Transcription Factors/metabolism , Animals , Blastocyst/cytology , Blastocyst/metabolism , Cell Proliferation , Cell Survival , Chimera/metabolism , Epigenesis, Genetic , Gene Expression Profiling , Humans , Kruppel-Like Factor 4 , Mice , MicroRNAs/genetics , MicroRNAs/metabolism , Rabbits , Signal Transduction
18.
Stem Cell Res ; 19: 104-112, 2017 03.
Article in English | MEDLINE | ID: mdl-28126658

ABSTRACT

The diverse cell states and in vitro conditions for the derivation and maintenance of the mammalian embryo-derived pluripotent stem cells raise the questions of whether there are multiple states of pluripotency of the stem cells of each species, and if there are innate species-specific variations in the pluripotency state. We will address these questions by taking a snapshot of our knowledge of the properties of the pluripotent stem cells, focusing on the maintenance of pluripotency and inter-conversion of the different types of pluripotent stem cells from rodents, lagomorphs and primates. We conceptualize pluripotent stem cells acquiring a series of cellular states represented as terraces on a slope of descending gradient of pluripotency. We propose that reprogramming pluripotent stem cells from a primed to a naive state is akin to moving upstream over a steep cliff to a higher terrace.


Subject(s)
Embryonic Stem Cells/cytology , Animals , Cell Differentiation , Cell Lineage , Embryonic Stem Cells/metabolism , Lagomorpha , Mice , Mitogen-Activated Protein Kinases/metabolism , Primates , Protein Kinase C/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism
19.
Stem Cell Reports ; 7(3): 383-398, 2016 09 13.
Article in English | MEDLINE | ID: mdl-27594588

ABSTRACT

Conventional rabbit embryonic stem cell (ESC) lines are derived from the inner cell mass (ICM) of pre-implantation embryos using methods and culture conditions that are established for primate ESCs. In this study, we explored the capacity of the rabbit ICM to give rise to ESC lines using conditions similar to those utilized to generate naive ESCs in mice. On single-cell dissociation and culture in fibroblast growth factor 2 (FGF2)-free, serum-supplemented medium, rabbit ICMs gave rise to ESC lines lacking the DNA-damage checkpoint in the G1 phase like mouse ESCs, and with a pluripotency gene expression profile closer to the rabbit ICM/epiblast profiles. These cell lines can be converted to FGF2-dependent ESCs after culture in conventional conditions. They can also colonize the rabbit pre-implantation embryo. These results indicate that rabbit epiblast cells can be coaxed toward different types of pluripotent stem cells and reveal the dynamics of pluripotent states in rabbit ESCs.


Subject(s)
Cell Differentiation , Embryonic Stem Cells/cytology , Embryonic Stem Cells/metabolism , Animals , Biomarkers , Blastocyst/cytology , Blastocyst/metabolism , Cell Culture Techniques , Cell Cycle , Cell Differentiation/genetics , Cell Line , Cell Self Renewal/genetics , Cells, Cultured , Computational Biology/methods , Gene Expression Profiling , Gene Expression Regulation, Developmental , Janus Kinases/metabolism , Leukemia Inhibitory Factor/metabolism , MicroRNAs/genetics , Rabbits , Signal Transduction , Transcriptome
20.
Differentiation ; 91(4-5): 119-25, 2016.
Article in English | MEDLINE | ID: mdl-26610326

ABSTRACT

Conventionally, mouse epiblast stem cells (EpiSCs) are derived directly from the epiblast or ectoderm germ layer of the post-implantation embryo. Self-renewing and multipotent EpiSC-like stem cells can also be derived by the conversion of embryonic stem cells (ESCs) via the provision of culture conditions that enable the maintenance of the EpiSCs. Here, we outline an experimental procedure for deriving EpiSCs from post-implantation chimeric embryos that are generated using genome-edited ESCs. This strategy enables the production of EpiSCs where (i) no genetically modified animals or ESCs are available, (ii) the impact of the genetic modification on post-implantation development, which may influence the property of the EpiSCs, is requisite knowledge for using the EpiSC for a specific investigation, and (iii) multiple editing of the genome is desirable to modify the biological attributes of the EpiSCs for studying, for example, the gene network activity on the trajectory of lineage differentiation and tissue morphogenesis.


Subject(s)
Cell Differentiation/genetics , Embryonic Development/genetics , Embryonic Stem Cells/cytology , Germ Layers/growth & development , Animals , Chimera/genetics , Chimera/growth & development , Germ Layers/cytology , Mice , Pluripotent Stem Cells/cytology
SELECTION OF CITATIONS
SEARCH DETAIL
...