Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 26
Filter
Add more filters











Publication year range
1.
Nature ; 628(8007): 391-399, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38408487

ABSTRACT

The human nervous system is a highly complex but organized organ. The foundation of its complexity and organization is laid down during regional patterning of the neural tube, the embryonic precursor to the human nervous system. Historically, studies of neural tube patterning have relied on animal models to uncover underlying principles. Recently, models of neurodevelopment based on human pluripotent stem cells, including neural organoids1-5 and bioengineered neural tube development models6-10, have emerged. However, such models fail to recapitulate neural patterning along both rostral-caudal and dorsal-ventral axes in a three-dimensional tubular geometry, a hallmark of neural tube development. Here we report a human pluripotent stem cell-based, microfluidic neural tube-like structure, the development of which recapitulates several crucial aspects of neural patterning in brain and spinal cord regions and along rostral-caudal and dorsal-ventral axes. This structure was utilized for studying neuronal lineage development, which revealed pre-patterning of axial identities of neural crest progenitors and functional roles of neuromesodermal progenitors and the caudal gene CDX2 in spinal cord and trunk neural crest development. We further developed dorsal-ventral patterned microfluidic forebrain-like structures with spatially segregated dorsal and ventral regions and layered apicobasal cellular organizations that mimic development of the human forebrain pallium and subpallium, respectively. Together, these microfluidics-based neurodevelopment models provide three-dimensional lumenal tissue architectures with in vivo-like spatiotemporal cell differentiation and organization, which will facilitate the study of human neurodevelopment and disease.


Subject(s)
Body Patterning , Microfluidics , Neural Tube , Humans , Cell Culture Techniques, Three Dimensional , Cell Differentiation , Neural Crest/cytology , Neural Crest/embryology , Neural Tube/cytology , Neural Tube/embryology , Pluripotent Stem Cells/cytology , Prosencephalon/cytology , Prosencephalon/embryology , Spinal Cord/cytology , Spinal Cord/embryology
2.
Reproduction ; 165(3): R75-R89, 2023 03 01.
Article in English | MEDLINE | ID: mdl-36449538

ABSTRACT

In brief: Understanding the establishment of post-fertilization totipotency has broad implications for modern biotechnologies. This review summarizes the current knowledge of putative egg components governing this process following natural fertilization and after somatic cell nuclear transfer. Abstract: The mammalian oocyte is a unique cell, and comprehending its physiology and biology is essential for understanding fertilization, totipotency and early events of embryogenesis. Consequently, research in these areas influences the outcomes of various technologies, for example, the production and conservation of laboratory and large animals with rare and valuable genotypes, the rescue of the species near extinction, as well as success in human assisted reproduction. Nevertheless, even the most advanced and sophisticated reproductive technologies of today do not always guarantee a favorable outcome. Elucidating the interactions of oocyte components with its natural partner cell - the sperm or an 'unnatural' somatic nucleus, when the somatic cell nucleus transfer is used is essential for understanding how totipotency is established and thus defining the requirements for normal development. One of the crucial aspects is the stoichiometry of different reprogramming and remodeling factors present in the oocyte and their balance. Here, we discuss how these factors, in combination, may lead to the formation of a new organism. We focus on the laboratory mouse and its genetic models, as this species has been instrumental in shaping our understanding of early post-fertilization events.


Subject(s)
Cell Nucleus , Semen , Humans , Animals , Mice , Male , Cell Nucleus/physiology , Spermatozoa/physiology , Embryonic Development , Oocytes/physiology , Mammals
3.
Nature ; 607(7919): 540-547, 2022 07.
Article in English | MEDLINE | ID: mdl-35794482

ABSTRACT

Gonadal development is a complex process that involves sex determination followed by divergent maturation into either testes or ovaries1. Historically, limited tissue accessibility, a lack of reliable in vitro models and critical differences between humans and mice have hampered our knowledge of human gonadogenesis, despite its importance in gonadal conditions and infertility. Here, we generated a comprehensive map of first- and second-trimester human gonads using a combination of single-cell and spatial transcriptomics, chromatin accessibility assays and fluorescent microscopy. We extracted human-specific regulatory programmes that control the development of germline and somatic cell lineages by profiling equivalent developmental stages in mice. In both species, we define the somatic cell states present at the time of sex specification, including the bipotent early supporting population that, in males, upregulates the testis-determining factor SRY and sPAX8s, a gonadal lineage located at the gonadal-mesonephric interface. In females, we resolve the cellular and molecular events that give rise to the first and second waves of granulosa cells that compartmentalize the developing ovary to modulate germ cell differentiation. In males, we identify human SIGLEC15+ and TREM2+ fetal testicular macrophages, which signal to somatic cells outside and inside the developing testis cords, respectively. This study provides a comprehensive spatiotemporal map of human and mouse gonadal differentiation, which can guide in vitro gonadogenesis.


Subject(s)
Cell Lineage , Germ Cells , Ovary , Sex Differentiation , Single-Cell Analysis , Testis , Animals , Chromatin/genetics , Chromatin/metabolism , Female , Germ Cells/cytology , Germ Cells/metabolism , Granulosa Cells/cytology , Granulosa Cells/metabolism , Humans , Immunoglobulins , Macrophages/metabolism , Male , Membrane Glycoproteins , Membrane Proteins , Mice , Microscopy, Fluorescence , Ovary/cytology , Ovary/embryology , PAX8 Transcription Factor , Pregnancy , Pregnancy Trimester, First , Pregnancy Trimester, Second , Receptors, Immunologic , Sex Differentiation/genetics , Testis/cytology , Testis/embryology , Transcriptome
4.
Stem Cell Reports ; 17(5): 1019-1022, 2022 05 10.
Article in English | MEDLINE | ID: mdl-35395176

ABSTRACT

Greater transcultural and transdisciplinary engagement within Muslim contexts and deliberate inclusion of diverse Muslim voices in the development of international guidelines is required to improve understanding of the state of stem cell science, strengthen thinking about attendant ethical complexities, enhance compliance, deepen public deliberation, increase trust, and strengthen practice standards.


Subject(s)
Islam , Stem Cells
6.
Stem Cell Reports ; 16(6): 1416-1424, 2021 06 08.
Article in English | MEDLINE | ID: mdl-34048690

ABSTRACT

The ISSCR Guidelines for Stem Cell Research and Clinical Translation were last revised in 2016. Since then, rapid progress has been made in research areas related to in vitro culture of human embryos, creation of stem cell-based embryo models, and in vitro gametogenesis. Therefore, a working group of international experts was convened to review the oversight process and provide an update to the guidelines. This report captures the discussion and summarizes the major recommendations made by this working group, with a specific emphasis on updating the categories of review and engagement with the specialized scientific and ethical oversight process.


Subject(s)
Embryo Research/ethics , Embryonic Stem Cells , Practice Guidelines as Topic , Societies, Scientific/ethics , Societies, Scientific/standards , Stem Cell Research/ethics , Embryo, Mammalian , Gametogenesis , Humans , Models, Biological
7.
Stem Cell Reports ; 16(6): 1398-1408, 2021 06 08.
Article in English | MEDLINE | ID: mdl-34048692

ABSTRACT

The International Society for Stem Cell Research has updated its Guidelines for Stem Cell Research and Clinical Translation in order to address advances in stem cell science and other relevant fields, together with the associated ethical, social, and policy issues that have arisen since the last update in 2016. While growing to encompass the evolving science, clinical applications of stem cells, and the increasingly complex implications of stem cell research for society, the basic principles underlying the Guidelines remain unchanged, and they will continue to serve as the standard for the field and as a resource for scientists, regulators, funders, physicians, and members of the public, including patients. A summary of the key updates and issues is presented here.


Subject(s)
Bioethical Issues/standards , Policy , Practice Guidelines as Topic , Societies, Scientific/standards , Stem Cell Research/ethics , Stem Cells , Humans , Societies, Scientific/ethics
8.
Nat Cell Biol ; 21(6): 687-699, 2019 06.
Article in English | MEDLINE | ID: mdl-31160711

ABSTRACT

We recently derived mouse expanded potential stem cells (EPSCs) from individual blastomeres by inhibiting the critical molecular pathways that predispose their differentiation. EPSCs had enriched molecular signatures of blastomeres and possessed developmental potency for all embryonic and extra-embryonic cell lineages. Here, we report the derivation of porcine EPSCs, which express key pluripotency genes, are genetically stable, permit genome editing, differentiate to derivatives of the three germ layers in chimeras and produce primordial germ cell-like cells in vitro. Under similar conditions, human embryonic stem cells and induced pluripotent stem cells can be converted, or somatic cells directly reprogrammed, to EPSCs that display the molecular and functional attributes reminiscent of porcine EPSCs. Importantly, trophoblast stem-cell-like cells can be generated from both human and porcine EPSCs. Our pathway-inhibition paradigm thus opens an avenue for generating mammalian pluripotent stem cells, and EPSCs present a unique cellular platform for translational research in biotechnology and regenerative medicine.


Subject(s)
Cell Differentiation/genetics , Cellular Reprogramming/genetics , Induced Pluripotent Stem Cells/cytology , Pluripotent Stem Cells/cytology , Animals , Blastomeres/cytology , Blastomeres/metabolism , Cell Lineage/genetics , Embryonic Stem Cells/cytology , Germ Layers/growth & development , Germ Layers/metabolism , Humans , Mice , Regenerative Medicine , Signal Transduction/genetics , Swine , Trophoblasts/cytology , Trophoblasts/metabolism
9.
Trends Mol Med ; 25(3): 165-167, 2019 03.
Article in English | MEDLINE | ID: mdl-30738627

ABSTRACT

The implications of scientific breakthroughs are rarely faced up to in advance of their realization. Stem cell-derived human gametes, a disruptive technology in waiting, are likely to recapitulate this historic pattern absent active intervention. Herein we call for the conduct of thoughtful ante hoc deliberations on the prospect of stem cell-derived human gametes with an eye toward minimizing potential untoward post hoc regulatory or statutory impositions.


Subject(s)
Gametogenesis , Germ Cells/cytology , Stem Cell Research , Stem Cells/cytology , Animals , Cell Differentiation , Embryonic Stem Cells , Germ Cells/metabolism , Humans , Induced Pluripotent Stem Cells , Reproductive Techniques, Assisted , Stem Cell Research/ethics , Stem Cell Research/legislation & jurisprudence , Stem Cell Transplantation , Stem Cells/metabolism
10.
Nat Commun ; 8(1): 1297, 2017 11 03.
Article in English | MEDLINE | ID: mdl-29101321

ABSTRACT

X-chromosome inactivation is established during early development. In mice, transcriptional repression of the paternal X-chromosome (Xp) and enrichment in epigenetic marks such as H3K27me3 is achieved by the early blastocyst stage. X-chromosome inactivation is then reversed in the inner cell mass. The mechanisms underlying Xp reactivation remain enigmatic. Using in vivo single-cell approaches (allele-specific RNAseq, nascent RNA-fluorescent in situ hybridization and immunofluorescence), we show here that different genes are reactivated at different stages, with more slowly reactivated genes tending to be enriched in H3meK27. We further show that in UTX H3K27 histone demethylase mutant embryos, these genes are even more slowly reactivated, suggesting that these genes carry an epigenetic memory that may be actively lost. On the other hand, expression of rapidly reactivated genes may be driven by transcription factors. Thus, some X-linked genes have minimal epigenetic memory in the inner cell mass, whereas others may require active erasure of chromatin marks.


Subject(s)
Blastocyst Inner Cell Mass/metabolism , Epigenesis, Genetic , Transcription Factors/pharmacokinetics , X Chromosome Inactivation/genetics , Animals , Female , Genes, X-Linked , Histones/metabolism , In Situ Hybridization, Fluorescence , Male , Mice , Mice, Inbred C57BL , Mice, Inbred DBA , Models, Genetic , Pregnancy , RNA, Long Noncoding/genetics , Sequence Analysis, RNA , Single-Cell Analysis
11.
Nat Struct Mol Biol ; 24(3): 226-233, 2017 03.
Article in English | MEDLINE | ID: mdl-28134930

ABSTRACT

The long noncoding RNA Xist is expressed from only the paternal X chromosome in mouse preimplantation female embryos and mediates transcriptional silencing of that chromosome. In females, absence of Xist leads to postimplantation lethality. Here, through single-cell RNA sequencing of early preimplantation mouse embryos, we found that the initiation of imprinted X-chromosome inactivation absolutely requires Xist. Lack of paternal Xist leads to genome-wide transcriptional misregulation in the early blastocyst and to failure to activate the extraembryonic pathway that is essential for postimplantation development. We also demonstrate that the expression dynamics of X-linked genes depends on the strain and parent of origin as well as on the location along the X chromosome, particularly at the first 'entry' sites of Xist. This study demonstrates that dosage-compensation failure has an effect as early as the blastocyst stage and reveals genetic and epigenetic contributions to orchestrating transcriptional silencing of the X chromosome during early embryogenesis.


Subject(s)
Embryonic Development/genetics , Genomic Imprinting , RNA, Long Noncoding/genetics , X Chromosome Inactivation/genetics , Alleles , Animals , Blastocyst/cytology , Blastocyst/metabolism , Cell Differentiation/genetics , Dosage Compensation, Genetic , Embryo Implantation/genetics , Embryo, Mammalian/metabolism , Female , Gene Expression Regulation, Developmental , Gene Knockout Techniques , Gene Silencing , Genes, X-Linked , Kinetics , Male , Mice, Inbred C57BL , Mutation/genetics , RNA, Long Noncoding/metabolism , Sequence Analysis, RNA , Single-Cell Analysis , Time Factors , X Chromosome/genetics
12.
Chromosoma ; 126(3): 399-415, 2017 06.
Article in English | MEDLINE | ID: mdl-27165042

ABSTRACT

Meiosis is a critical phase in the life cycle of sexually reproducing organisms. Chromosome numbers are halved during meiosis, which requires meiosis-specific modification of chromosome behaviour. Furthermore, suppression of transposons is particularly important during meiosis to allow the transmission of undamaged genomic information between generations. Correspondingly, specialized genome defence mechanisms and nuclear structures characterize the germ line during meiosis. Survival of mammalian spermatocytes requires that the sex chromosomes form a distinct silenced chromatin domain, called the sex body. An enigmatic spherical DNA-negative structure, called the meiotic dense body, forms in association with the sex body. The dense body contains small non-coding RNAs including microRNAs and PIWI-associated RNAs. These observations gave rise to speculations that the dense body may be involved in sex body formation and or small non-coding RNA functions, e.g. the silencing of transposons. Nevertheless, the function of the dense body has remained mysterious because no protein essential for dense body formation has been reported yet. We discovered that the polycomb-related sex comb on midleg-like 1 (SCML1) is a meiosis-specific protein and is an essential component of the meiotic dense body. Despite abolished dense body formation, Scml1-deficient mice are fertile and proficient in sex body formation, transposon silencing and in timely progression through meiosis and gametogenesis. Thus, we conclude that dense body formation is not an essential component of the gametogenetic program in the mammalian germ line.


Subject(s)
Gametogenesis , Germ Cells/cytology , Meiosis , Mice/metabolism , Polycomb-Group Proteins/metabolism , Animals , Female , Fertility , Germ Cells/metabolism , Male , Mice/genetics , Polycomb-Group Proteins/genetics
14.
Mol Biosyst ; 8(3): 744-52, 2012 Mar.
Article in English | MEDLINE | ID: mdl-22222461

ABSTRACT

Analysis of transcription at the level of single cells in prokaryotes and eukaryotes has revealed the existence of heterogeneities in the expression of individual genes within genetically homogeneous populations. This variation is an emerging hallmark of populations of Embryonic Stem (ES) cells and has been ascribed to the stochasticity associated with the biochemical events that mediate gene expression. It has been suggested that these heterogeneities play a role in the maintenance of pluripotency. However, for the most part, studies have focused on individual genes in large cell populations. Here we use an existing dataset on the expression of eight genes involved in pluripotency in eighty-three ES cells to create Gene Regulatory Networks (GRNs) at the single cell level. We observe widespread heterogeneities in the expression of the eight genes, but analysis of correlations within individual cells reveals three distinct classes centered on the expression of Nanog, a marker of pluripotency, and Fgf5, a gene associated with differentiation: high levels of Nanog and low levels of Fgf5, low levels of Nanog and high levels of Fgf5, and low levels of both. Each of these classes is associated with a collection of active sub-networks, with differing degrees of connectivity between their elements, which define a cellular state: self-renewal, primed for differentiation or transition between the two. Though every cell should be governed by the same network, the active sub-networks may emerge due to considerations such as variation in (i) the expression level of active transcription factors (e.g. through post-translational modification or ligand/co-factor availability) or (ii) access to the target gene locus (e.g. via changes in chromatin status or epigenetic modifications). We conclude that heterogeneities in gene expression should not be interpreted as representing different states of a single unique network, but as a reflection of the activity of different sub-networks in sub-populations of cells.


Subject(s)
Embryonic Stem Cells/cytology , Pluripotent Stem Cells/cytology , Animals , Cell Differentiation , Cells, Cultured , Embryonic Stem Cells/metabolism , Gene Regulatory Networks , Mice , Octamer Transcription Factor-3/genetics , Octamer Transcription Factor-3/metabolism , Pluripotent Stem Cells/metabolism , Protein Processing, Post-Translational
15.
Cell ; 139(6): 1032-7, 2009 Dec 11.
Article in English | MEDLINE | ID: mdl-20005794

ABSTRACT

Given the explosion of research on induced pluripotent stem (iPS) cells, it is timely to consider the various ethical, legal, and social issues engaged by this fast-moving field. Here, we review issues associated with the procurement, basic research, and clinical translation of iPS cells.


Subject(s)
Biomedical Research , Induced Pluripotent Stem Cells/cytology , Stem Cell Transplantation , Humans , Public Policy , Tissue Donors
16.
Differentiation ; 78(2-3): 116-23, 2009.
Article in English | MEDLINE | ID: mdl-19683852

ABSTRACT

Embryonic stem (ES) cells, derived from pre-implantation embryo, embryonic germ (EG) cells, derived from embryonic precursors of gametes, primordial germ cells (PGCs), can differentiate into any cell type in the body. Moreover, ES cells have the capacity to differentiate into PGCs in vitro. In the present study we have shown the differentiation capacity of six EG cell lines to form PGCs in vitro, in comparison to ES cells. Cell lines were differentiated via embryoid body (EB) formation using the co-expression of mouse vasa homolog (Mvh) and Oct-4 to identify newly formed PGCs in vitro. We found an increase of PGC numbers in almost all analysed cell lines in 5-day-old EBs, thus suggesting that EG and ES cells have similar efficiency to generate PGCs. The addition of retinoic acid confirmed that the cultures had attained a PGC-like identity and continued to proliferate. Furthermore we have shown that the expression pattern of Prmt5 and H3K27me3 in newly formed PGCs is similar to that observed in embryonic day E11.5 PGCs in vivo. By co-culturing EBs with Chinese hamster ovary (CHO) cells some of the PGCs entered into meiosis, as judged by Scp3 expression. The derivation of germ cells from pluripotent stem cells in vitro could provide an invaluable model system to study both the genetic and epigenetic programming of germ cell development in vivo.


Subject(s)
Cell Differentiation/physiology , Embryonic Stem Cells/cytology , Germ Cells/cytology , Pluripotent Stem Cells/cytology , Animals , CHO Cells , Cell Cycle Proteins , Cell Proliferation , Cells, Cultured , Cricetinae , Cricetulus , DNA-Binding Proteins , Lewis X Antigen/metabolism , Meiosis , Mice , Nuclear Proteins/metabolism , Octamer Transcription Factor-3/metabolism , Protein Methyltransferases/metabolism , Protein-Arginine N-Methyltransferases
17.
Development ; 136(8): 1295-303, 2009 Apr.
Article in English | MEDLINE | ID: mdl-19279135

ABSTRACT

Steel factor is an essential survival and proliferation factor for primordial germ cells (PGCs) during their migration in the early mouse embryo. PGCs arise during gastrulation, and migrate into the posterior endoderm that becomes the hindgut. Previous reports have suggested that PGCs become dependent on Steel factor when they colonize the hindgut. However, in the absence of a good marker for living PGCs, their behavior before hindgut colonization has not been previously studied. We report here the normal behavior of PGCs in live embryos before hindgut colonization, and the roles of Steel factor, using a reporter line in which GFP is driven by the promoter of the Stella gene, whose activation accompanies the initial specification of PGCs. We show first that PGCs are surrounded by Steel factor-expressing cells from their first appearance in the allantois to the time they enter the genital ridges. Second, fewer PGCs are found in the allantois in Steel-null embryos, but this is not due to a failure of PGC specification. Third, the analysis of cultured Steel-null early embryos shows that Steel factor is required for normal PGC motility, both in the allantois and in the hindgut. Germ cells migrate actively in the allantois, and move directionally from the allantois into the proximal epiblast. In the absence of Steel factor, caused by either null mutation or antibody blockade, PGC motility is dramatically decreased, but directionality is maintained, demonstrating a primary role for Steel factor in PGC motility. This was found both before and after colonization of the hindgut. These data, together with previously published data, show that PGCs are Steel factor dependent from their initial specification until they colonize the genital ridges, and suggest the existence of a ;spatio-temporal niche' that travels with this important pluripotential cell population in the embryo.


Subject(s)
Allantois/cytology , Allantois/metabolism , Germ Cells/cytology , Germ Cells/metabolism , Stem Cell Factor/metabolism , Stem Cell Niche/cytology , Stem Cell Niche/metabolism , Allantois/embryology , Animals , Cell Death , Cell Movement , Cell Survival , Digestive System/cytology , Digestive System/embryology , Digestive System/metabolism , Embryo, Mammalian/cytology , Embryo, Mammalian/embryology , Embryo, Mammalian/metabolism , Gene Expression Regulation, Developmental , Mice , Mice, Knockout , Protein-Tyrosine Kinases/metabolism , Signal Transduction , Stem Cell Factor/genetics , Time Factors
18.
Mech Dev ; 125(11-12): 963-73, 2008.
Article in English | MEDLINE | ID: mdl-18789387

ABSTRACT

Imprinting control regions (ICRs) are domains within imprinted loci that are essential for their establishment and maintenance. Imprinted loci can extend over several megabases, encompass both maternally and paternally-expressed genes and exhibit multiple and complex epigenetic modifications including large regions of allele-specific DNA methylation. Differential chromatin organisation has also been observed within imprinted loci but is restricted to the ICRs. In this study we report the identification of a novel imprinting control region for the mouse Neuronatin gene. This biochemically defined putative ICR, present within its 250 bp second intron, functions as transcriptional activator in Drosophila. This is unlike other known ICRs which have been shown to function as transcriptional silencers. Furthermore, at the endogenous locus, the activating signal from the ICR extends to the Neuronatin promoter via allele-specific unidirectional nucleosomal positioning. Our results support the proposal that the Neuronatin locus employs the most basic mechanism for establishing allele-specific gene expression and could provide the foundation for the multiplex arrangements reported at more complex loci.


Subject(s)
DNA/genetics , Drosophila melanogaster/genetics , Genomic Imprinting , Introns , Membrane Proteins/genetics , Membrane Proteins/physiology , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/physiology , Transcriptional Activation , Alleles , Animals , Chromatin/chemistry , DNA Methylation , Epigenesis, Genetic , Female , Male , Mice , Nucleosomes/metabolism
19.
Curr Protoc Stem Cell Biol ; Chapter 1: Unit1A.3, 2008 Apr.
Article in English | MEDLINE | ID: mdl-18770625

ABSTRACT

In this unit we describe the derivation of pluripotent embryonic germ (EG) cells from mouse primordial germ cells (PGCs) isolated from both 8.5- and 11.5-days post-coitum (dpc) embryos. Once EG cells are derived we explain how to propagate and characterize the cell lines. We introduce readers to PGCs and explain differences between PGCs and their in vitro derivatives EG cells. Finally, we also compare mouse EG cells with ES cells. This unit will be of great interest to anyone interested in PGCs or studying the behavior of cultured PGCs or the derivation of new EG cell lines.


Subject(s)
Cell Culture Techniques/methods , Cellular Reprogramming , Embryo, Mammalian/cytology , Germ Cells/cytology , Stem Cells/cytology , Alkaline Phosphatase/metabolism , Animals , Cell Line , Cell Separation , Cryopreservation , Fluorescent Antibody Technique , Lewis X Antigen/metabolism , Magnetics , Mice , Microspheres , Mitosis , Octamer Transcription Factor-3/metabolism , Polymerase Chain Reaction , Sex Determination Analysis
20.
Dev Biol ; 313(2): 674-81, 2008 Jan 15.
Article in English | MEDLINE | ID: mdl-18062950

ABSTRACT

Pluripotent stem cells, termed embryonic germ (EG) cells, have been generated from both human and mouse primordial germ cells (PGCs). Like embryonic stem (ES) cells, EG cells have the potential to differentiate into all germ layer derivatives and may also be important for any future clinical applications. The development of PGCs in vivo is accompanied by major epigenetic changes including DNA demethylation and imprint erasure. We have investigated the DNA methylation pattern of several imprinted genes and repetitive elements in mouse EG cell lines before and after differentiation. Analysed cell lines were derived soon after PGC specification, "early", in comparison with EG cells derived after PGC colonisation of the genital ridge, "late" and embryonic stem (ES) cell lines, derived from the inner cell mass (ICM). Early EG cell lines showed strikingly heterogeneous DNA methylation patterns, in contrast to the uniformity of methylation pattern seen in somatic cells (control), late EG cell and ES cell lines. We also observed that all analysed XX cell lines exhibited less methylation than XY. We suggest that this heterogeneity may reflect the changes in DNA methylation taking place in the germ cell lineage soon after specification.


Subject(s)
DNA Methylation , Genetic Heterogeneity , Genomic Imprinting , Germ Cells/metabolism , Pluripotent Stem Cells/metabolism , Alkaline Phosphatase/metabolism , Animals , Cell Differentiation , Cell Line , Crosses, Genetic , Embryo, Mammalian , Female , Karyotyping , Male , Mice , Mice, Inbred C57BL , Time Factors , Transgenes
SELECTION OF CITATIONS
SEARCH DETAIL