Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 64
Filter
1.
Cell ; 165(4): 1012-26, 2016 May 05.
Article in English | MEDLINE | ID: mdl-27062923

ABSTRACT

Mouse studies have been instrumental in forming our current understanding of early cell-lineage decisions; however, similar insights into the early human development are severely limited. Here, we present a comprehensive transcriptional map of human embryo development, including the sequenced transcriptomes of 1,529 individual cells from 88 human preimplantation embryos. These data show that cells undergo an intermediate state of co-expression of lineage-specific genes, followed by a concurrent establishment of the trophectoderm, epiblast, and primitive endoderm lineages, which coincide with blastocyst formation. Female cells of all three lineages achieve dosage compensation of X chromosome RNA levels prior to implantation. However, in contrast to the mouse, XIST is transcribed from both alleles throughout the progression of this expression dampening, and X chromosome genes maintain biallelic expression while dosage compensation proceeds. We envision broad utility of this transcriptional atlas in future studies on human development as well as in stem cell research.


Subject(s)
Blastocyst/metabolism , Chromosomes, Human, X , Single-Cell Analysis , Blastocyst Inner Cell Mass/metabolism , Dosage Compensation, Genetic , Female , Humans , Male , RNA, Long Noncoding/genetics , Sequence Analysis, RNA , Sex Characteristics , Transcriptome
2.
Nature ; 622(7983): 562-573, 2023 Oct.
Article in English | MEDLINE | ID: mdl-37673118

ABSTRACT

The ability to study human post-implantation development remains limited owing to ethical and technical challenges associated with intrauterine development after implantation1. Embryo-like models with spatially organized morphogenesis and structure of all defining embryonic and extra-embryonic tissues of the post-implantation human conceptus (that is, the embryonic disc, the bilaminar disc, the yolk sac, the chorionic sac and the surrounding trophoblast layer) remain lacking1,2. Mouse naive embryonic stem cells have recently been shown to give rise to embryonic and extra-embryonic stem cells capable of self-assembling into post-gastrulation structured stem-cell-based embryo models with spatially organized morphogenesis (called SEMs)3. Here we extend those findings to humans using only genetically unmodified human naive embryonic stem cells (cultured in human enhanced naive stem cell medium conditions)4. Such human fully integrated and complete SEMs recapitulate the organization of nearly all known lineages and compartments of post-implantation human embryos, including the epiblast, the hypoblast, the extra-embryonic mesoderm and the trophoblast layer surrounding the latter compartments. These human complete SEMs demonstrated developmental growth dynamics that resemble key hallmarks of post-implantation stage embryogenesis up to 13-14 days after fertilization (Carnegie stage 6a). These include embryonic disc and bilaminar disc formation, epiblast lumenogenesis, polarized amniogenesis, anterior-posterior symmetry breaking, primordial germ-cell specification, polarized yolk sac with visceral and parietal endoderm formation, extra-embryonic mesoderm expansion that defines a chorionic cavity and a connecting stalk, and a trophoblast-surrounding compartment demonstrating syncytium and lacunae formation. This SEM platform will probably enable the experimental investigation of previously inaccessible windows of human early post implantation up to peri-gastrulation development.


Subject(s)
Embryo Implantation , Embryo, Mammalian , Embryonic Development , Human Embryonic Stem Cells , Humans , Embryo, Mammalian/cytology , Embryo, Mammalian/embryology , Fertilization , Gastrulation , Germ Layers/cytology , Germ Layers/embryology , Human Embryonic Stem Cells/cytology , Trophoblasts/cytology , Yolk Sac/cytology , Yolk Sac/embryology , Giant Cells/cytology
3.
Cell ; 152(5): 1008-20, 2013 Feb 28.
Article in English | MEDLINE | ID: mdl-23452850

ABSTRACT

Metazoan evolution involves increasing protein domain complexity, but how this relates to control of biological decisions remains uncertain. The Ras guanine nucleotide exchange factor (RasGEF) Sos1 and its adaptor Grb2 are multidomain proteins that couple fibroblast growth factor (FGF) signaling to activation of the Ras-Erk pathway during mammalian development and drive embryonic stem cells toward the primitive endoderm (PrE) lineage. We show that the ability of Sos1/Grb2 to appropriately regulate pluripotency and differentiation factors and to initiate PrE development requires collective binding of multiple Sos1/Grb2 domains to their protein and phospholipid ligands. This provides a cooperative system that only allows lineage commitment when all ligand-binding domains are occupied. Furthermore, our results indicate that the interaction domains of Sos1 and Grb2 have evolved so as to bind ligands not with maximal strength but with specificities and affinities that maintain cooperativity. This optimized system ensures that PrE lineage commitment occurs in a timely and selective manner during embryogenesis.


Subject(s)
Embryo, Mammalian/metabolism , Embryonic Stem Cells/metabolism , GRB2 Adaptor Protein/metabolism , SOS1 Protein/metabolism , Amino Acid Sequence , Animals , Cell Lineage , Endoderm/metabolism , Eukaryota/genetics , Eukaryota/metabolism , Humans , Models, Molecular , Molecular Sequence Data , Protein Structure, Tertiary , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/metabolism , Sequence Alignment , ras Guanine Nucleotide Exchange Factors/metabolism
5.
Stem Cells ; 41(2): 105-110, 2023 03 02.
Article in English | MEDLINE | ID: mdl-36153824

ABSTRACT

Ovaries are central to development, fertility, and reproduction of women. A particularly interesting feature of ovaries is their accelerated aging compared to other tissues, leading to loss of function far before other organs senesce. The limited pool of ovarian follicles is generated before birth and once exhausted, menopause will inevitably commence around the age of 50 years marking the end of fertility. Yet, there are reports suggesting the presence of germline stem cells and neo-oogenesis in adult human ovaries. These observations have fueled a long debate, created experimental fertility treatments, and opened business opportunities. Our recent analysis of cell types in the ovarian cortex of women of fertile age could not find evidence of germline stem cells. Like before, our work has been met with critique suggesting methodological shortcomings. We agree that excellence starts with methods and welcome discussion on the pros and cons of different protocols. In this commentary, we discuss the recent re-interpretation of our work.


Subject(s)
Oogenesis , Ovary , Adult , Female , Humans , Middle Aged , Oogenesis/physiology , Ovarian Follicle , Germ Cells , Stem Cells/metabolism
6.
Cytotherapy ; 26(4): 340-350, 2024 04.
Article in English | MEDLINE | ID: mdl-38349309

ABSTRACT

BACKGROUND AIMS: Age-related macular degeneration (AMD) is the most common cause of blindness in elderly patients within developed countries, affecting more than 190 million worldwide. In AMD, the retinal pigment epithelial (RPE) cell layer progressively degenerates, resulting in subsequent loss of photoreceptors and ultimately vision. There is currently no cure for AMD, but therapeutic strategies targeting the complement system are being developed to slow the progression of the disease. METHODS: Replacement therapy with pluripotent stem cell-derived (hPSC) RPEs is an alternative treatment strategy. A cell therapy product must be produced in accordance with Good Manufacturing Practices at a sufficient scale to facilitate extensive pre-clinical and clinical testing. Cryopreservation of the final cell product is therefore highly beneficial, as the manufacturing, pre-clinical and clinical testing can be separated in time and location. RESULTS: We found that mature hPSC-RPE cells do not survive conventional cryopreservation techniques. However, replating the cells 2-5 days before cryopreservation facilitates freezing. The replated and cryopreserved hPSC-RPE cells maintained their identity, purity and functionality as characteristic RPEs, shown by cobblestone morphology, pigmentation, transcriptional profile, RPE markers, transepithelial resistance and pigment epithelium-derived factor secretion. Finally, we showed that the optimal replating time window can be tracked noninvasively by following the change in cobblestone morphology. CONCLUSIONS: The possibility of cryopreserving the hPSC-RPE product has been instrumental in our efforts in manufacturing and performing pre-clinical testing with the aim for clinical translation.


Subject(s)
Macular Degeneration , Pluripotent Stem Cells , Humans , Aged , Cell Differentiation , Macular Degeneration/therapy , Cryopreservation , Epithelial Cells , Retinal Pigments
7.
Nature ; 600(7888): 223-224, 2021 12.
Article in English | MEDLINE | ID: mdl-34789887
8.
Dev Biol ; 447(1): 3-13, 2019 03 01.
Article in English | MEDLINE | ID: mdl-29391166

ABSTRACT

The journey of embryonic development starts at oocyte fertilization, which triggers a complex cascade of events and cellular pathways that guide early embryogenesis. Recent technological advances have greatly expanded our knowledge of cleavage-stage embryo development, which is characterized by an increased rate of whole-chromosome losses and gains, mixoploidy, and atypical cleavage morphokinetics. Embryonic aneuploidy significantly contributes to implantation failure, spontaneous miscarriage, stillbirth or congenital birth defects in both natural and assisted human reproduction. Essentially, early embryo development is strongly determined by maternal factors. Owing to considerable limitations associated with human oocyte and embryo research, the use of animal models is inevitable. However, cellular and molecular mechanisms driving the error-prone early stages of development are still poorly described. In this review, we describe known events that lead to aneuploidy in mammalian oocytes and preimplantation embryos. As the processes of oocyte and embryo development are rigorously regulated by multiple signal-transduction pathways, we explore the putative role of signaling pathways in genomic integrity maintenance. Based on the existing evidence from human and animal data, we investigate whether critical early developmental pathways, like Wnt, Hippo and MAPK, together with distinct DNA damage response and DNA repair pathways can be associated with embryo genomic instability, a question that has, so far, remained largely unexplored.


Subject(s)
Aneuploidy , Blastocyst , Embryonic Development , Genomic Instability , Pregnancy Complications , Animals , Blastocyst/metabolism , Blastocyst/pathology , Female , Humans , Oocytes/metabolism , Oocytes/pathology , Pregnancy , Pregnancy Complications/genetics , Pregnancy Complications/metabolism , Pregnancy Complications/pathology
9.
Development ; 144(1): 3-7, 2017 01 01.
Article in English | MEDLINE | ID: mdl-28049687

ABSTRACT

Developmental biologists have become increasingly aware that the wealth of knowledge generated through genetic studies of pre-implantation mouse development might not easily be translated to the human embryo. Comparative studies have been fueled by recent technological advances in single-cell analysis, allowing in-depth analysis of the human embryo. This field could shortly gain more momentum as novel genome editing technologies might, for the first time, also allow functional genetic studies in the human embryo. In this Spotlight article, we summarize the CRISPR-Cas9 genome editing system and discuss its potential applications and limitations in human pre-implantation embryos, and the ethical considerations thereof.


Subject(s)
CRISPR-Cas Systems/genetics , Embryo Research , Embryonic Development/genetics , Gene Editing/ethics , Gene Editing/statistics & numerical data , Gene Editing/trends , Animals , Embryo Research/ethics , Embryo, Mammalian , Humans
10.
Genome Res ; 26(10): 1342-1354, 2016 10.
Article in English | MEDLINE | ID: mdl-27486082

ABSTRACT

Pluripotency, differentiation, and X Chromosome inactivation (XCI) are key aspects of embryonic development. However, the underlying relationship and mechanisms among these processes remain unclear. Here, we systematically dissected these features along developmental progression using mouse embryonic stem cells (mESCs) and single-cell RNA sequencing with allelic resolution. We found that mESCs grown in a ground state 2i condition displayed transcriptomic profiles diffused from preimplantation mouse embryonic cells, whereas EpiStem cells closely resembled the post-implantation epiblast. Sex-related gene expression varied greatly across distinct developmental states. We also identified novel markers that were highly enriched in each developmental state. Moreover, we revealed that several novel pathways, including PluriNetWork and Focal Adhesion, were responsible for the delayed progression of female EpiStem cells. Importantly, we "digitalized" XCI progression using allelic expression of active and inactive X Chromosomes and surprisingly found that XCI states exhibited profound variability in each developmental state, including the 2i condition. XCI progression was not tightly synchronized with loss of pluripotency and increase of differentiation at the single-cell level, although these processes were globally correlated. In addition, highly expressed genes, including core pluripotency factors, were in general biallelically expressed. Taken together, our study sheds light on the dynamics of XCI progression and the asynchronicity between pluripotency, differentiation, and XCI.


Subject(s)
Cell Differentiation , Embryonic Stem Cells/cytology , Pluripotent Stem Cells/cytology , X Chromosome Inactivation , Animals , Cells, Cultured , Embryonic Stem Cells/metabolism , Female , Gene Expression Regulation, Developmental , Male , Mice , Mice, Inbred C57BL , Pluripotent Stem Cells/metabolism , Single-Cell Analysis , Transcriptome
11.
Development ; 143(19): 3459-3469, 2016 10 01.
Article in English | MEDLINE | ID: mdl-27578796

ABSTRACT

Leucine twenty homeobox (LEUTX) is a paired (PRD)-like homeobox gene that is expressed almost exclusively in human embryos during preimplantation development. We previously identified a novel transcription start site for the predicted human LEUTX gene based on the transcriptional analysis of human preimplantation embryos. The novel variant encodes a protein with a complete homeodomain. Here, we provide a detailed description of the molecular cloning of the complete homeodomain-containing LEUTX Using a human embryonic stem cell overexpression model we show that the complete homeodomain isoform is functional and sufficient to activate the transcription of a large proportion of the genes that are upregulated in human embryo genome activation (EGA), whereas the previously predicted partial homeodomain isoform is largely inactive. Another PRD-like transcription factor, DPRX, is then upregulated as a powerful repressor of transcription. We propose a two-stage model of human EGA in which LEUTX acts as a transcriptional activator at the 4-cell stage, and DPRX as a balancing repressor at the 8-cell stage. We conclude that LEUTX is a candidate regulator of human EGA.


Subject(s)
Blastocyst/metabolism , Embryonic Stem Cells/metabolism , Homeodomain Proteins/metabolism , Protein Isoforms/metabolism , Animals , Cell Line , Electrophoretic Mobility Shift Assay , Fluorescent Antibody Technique, Indirect , Gene Expression Regulation, Developmental , Homeodomain Proteins/genetics , Humans , Mice , Polymerase Chain Reaction , Protein Isoforms/genetics
12.
Hum Reprod ; 34(11): 2297-2310, 2019 11 01.
Article in English | MEDLINE | ID: mdl-31743397

ABSTRACT

STUDY QUESTION: Does the X chromosome inactivation (XCI) of Klinefelter syndrome (KS)-derived human induced pluripotent stem cells (hiPSCs) correspond to female human pluripotent stem cells (hPSCs) and reflect the KS genotype? SUMMARY ANSWER: Our results demonstrate for the first time that KS-derived hiPSCs show similar XCI behavior to female hPSCs in culture and show biological relevance to KS genotype-related clinical features. WHAT IS KNOWN ALREADY: So far, assessment of XCI of KS-derived hiPSCs was based on H3K27me3 staining and X-inactive specific transcript gene expression disregarding the at least three XCI states (XaXi with XIST coating, XaXi lacking XIST coating, and XaXe (partially eroded XCI)) that female hPSCs display in culture. STUDY DESIGN, SIZE, DURATION: The study used hiPSC lines generated from two azoospermic patients with KS and included two healthy male (HM) and one healthy female donor. PARTICIPANTS/MATERIALS, SETTING, METHODS: In this study, we derived hiPSCs by reprograming fibroblasts with episomal plasmids and applying laminin 521 as culture substrate. hiPSCs were characterized by karyotyping, immunocytochemistry, immunohistochemistry, quantitative PCR, teratoma formation, and embryoid body differentiation. XCI and KS hiPSC relevance were assessed by whole genome transcriptomics analysis and immunocytochemistry plus FISH of KS, HM and female fibroblast, and their hiPSC derivatives. MAIN RESULTS AND THE ROLE OF CHANCE: Applying whole genome transcriptomics analysis, we could identify differentially expressed genes (DEGs) between KS and HM donors with enrichment in gene ontology terms associated with fertility, cardiovascular development, ossification, and brain development, all associated with KS genotype-related clinical features. Furthermore, XCI analysis based on transcriptomics data, RNA FISH, and H3K27me3 staining revealed variable XCI states of KS hiPSCs similar to female hiPSCs, showing either normal (XaXi) or eroded (XaXe) XCI. KS hiPSCs with normal XCI showed nevertheless upregulated X-linked genes involved in nervous system development as well as synaptic transmission, supporting the potential use of KS-derived hiPSCs as an in vitro model for KS. LIMITATIONS, REASONS FOR CAUTION: Detailed clinical information for patients included in this study was not available. Although a correlation between DEGs and the KS genotype could be observed, the biological relevance of these cells has to be confirmed with further experiments. In addition, karyotype analysis for two hiPSC lines was performed at passage 12 but not repeated at a later passage. Nevertheless, since all XCI experiments for those lines were performed between passage 11 and 15 the authors expect no karyotypic changes for those experiments. WIDER IMPLICATIONS OF THE FINDINGS: As KS patients have variable clinical phenotypes that are influenced by the grade of aneuploidy, mosaicism, origin of the X chromosome, and XCI 'escapee' genes, which vary not only among individuals but also among different tissues within the same individual, differentiated KS hiPSCs could be used for a better understanding of KS pathogenesis. STUDY FUNDING/COMPETING INTEREST(S): This study was supported by grants from the Knut and Alice Wallenberg Foundation (2016.0121 and 2015.0096), Ming Wai Lau Centre for Reparative Medicine (2-343/2016), Ragnar Söderberg Foundation (M67/13), Swedish Research Council (2013-32485-100360-69), the Centre for Innovative Medicine (2-388/2016-40), Kronprinsessan Lovisas Förening För Barnasjukvård/Stiftelsen Axel Tielmans Minnesfond, Samariten Foundation, Jonasson Center at the Royal Institute of Technology, Sweden, and Initial Training Network Marie Curie Program 'Growsperm' (EU-FP7-PEOPLE-2013-ITN 603568). The authors declare no conflicts of interest.


Subject(s)
Azoospermia/genetics , Chromosomes, Human, X , Klinefelter Syndrome/genetics , Pluripotent Stem Cells/cytology , X Chromosome Inactivation , Adult , Cell Differentiation , Female , Fibroblasts/metabolism , Genotype , Histones/metabolism , Humans , Male , Phenotype , Sex Factors , Teratoma/metabolism , Transcriptome
14.
Nature ; 548(7668): 398-400, 2017 08 24.
Article in English | MEDLINE | ID: mdl-28783721
15.
Development ; 140(14): 2961-71, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23760955

ABSTRACT

In mice and humans the X-chromosomal porcupine homolog (Porcn) gene is required for the acylation and secretion of all 19 Wnt ligands and thus represents a bottleneck for all Wnt signaling. We have generated a mouse line carrying a floxed allele for Porcn and used zygotic, oocyte-specific and visceral endoderm-specific deletions to investigate embryonic and extra-embryonic requirements for Wnt ligand secretion. We show that there is no requirement for Porcn-dependent secretion of Wnt ligands during preimplantation development of the mouse embryo. Porcn-dependent Wnts are first required for the initiation of gastrulation, where Porcn function is required in the epiblast but not the visceral endoderm. Heterozygous female embryos, which are mutant in both trophoblast and visceral endoderm due to imprinted X chromosome inactivation, complete gastrulation but display chorio-allantoic fusion defects similar to Wnt7b mutants. Our studies highlight the importance of Wnt3 and Wnt7b for embryonic and placental development but suggest that endogenous Porcn-dependent Wnt secretion does not play an essential role in either implantation or blastocyst lineage specification.


Subject(s)
Gastrulation , Membrane Proteins/metabolism , Wnt Signaling Pathway , Acyltransferases , Animals , Blastocyst/metabolism , Chorioallantoic Membrane/metabolism , Embryo, Mammalian/metabolism , Endoderm/metabolism , Membrane Proteins/genetics , Mice , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/metabolism , Wnt Proteins/genetics , Wnt Proteins/metabolism , Wnt3 Protein/genetics , Wnt3 Protein/metabolism , Zygote/metabolism
16.
Development ; 140(22): 4533-43, 2013 Nov.
Article in English | MEDLINE | ID: mdl-24131634

ABSTRACT

Ectoderm is one of the three classic germ layers in the early mouse embryo, with the capacity to develop into both the central nervous system and epidermis. Because it is a transient phase of development with few molecular markers, the early ectoderm is the least understood germ layer in mouse embryonic development. In this work, we studied the differentiation potential of isolated ectoderm tissue in response to BMP signaling at various developmental stages (E6.5, E7.0 and E7.5), and identified a transient region in the anterior-proximal side of the embryo at E7.0 that possesses the ability to become neural or epidermal ectoderm in response to the absence or presence of BMP4, respectively. Furthermore, we demonstrated that inhibition of Nodal signaling could direct the pluripotent E6.5 epiblast cells towards ectoderm lineages during differentiation in explants in vitro. Our work not only improves our understanding of ectodermal layer development in early embryos, but also provides a framework for regenerative differentiation towards ectodermal tissues.


Subject(s)
Ectoderm/cytology , Ectoderm/embryology , Embryonic Development , Stem Cells/cytology , Animals , Biomarkers/metabolism , Bone Morphogenetic Protein 4/metabolism , Cell Lineage/genetics , Ectoderm/metabolism , Embryo, Mammalian/cytology , Embryo, Mammalian/metabolism , Embryonic Development/genetics , Epidermal Cells , Gene Expression Regulation, Developmental , Mice , Mice, Inbred ICR , Models, Biological , Nodal Protein/metabolism , Pluripotent Stem Cells/cytology , Pluripotent Stem Cells/metabolism , Signal Transduction/genetics
17.
EMBO J ; 30(9): 1778-89, 2011 May 04.
Article in English | MEDLINE | ID: mdl-21468033

ABSTRACT

Induced pluripotent stem (iPS) cell reprogramming is a gradual epigenetic process that reactivates the pluripotent transcriptional network by erasing and establishing repressive epigenetic marks. In contrast to loci-specific epigenetic changes, heterochromatin domains undergo epigenetic resetting during the reprogramming process, but the effect on the heterochromatin ultrastructure is not known. Here, we characterize the physical structure of heterochromatin domains in full and partial mouse iPS cells by correlative electron spectroscopic imaging. In somatic and partial iPS cells, constitutive heterochromatin marked by H3K9me3 is highly compartmentalized into chromocentre structures of densely packed chromatin fibres. In contrast, chromocentre boundaries are poorly defined in pluripotent embryonic stem and full iPS cells, and are characterized by unusually dispersed 10 nm heterochromatin fibres in high Nanog-expressing cells, including pluripotent cells of the mouse blastocyst before differentiation. This heterochromatin reorganization accompanies retroviral silencing during conversion of partial iPS cells by MEK/GSK3 2i inhibitor treatment. Thus, constitutive heterochromatin is compacted in partial iPS cells but reorganizes into dispersed 10 nm chromatin fibres as the fully reprogrammed iPS cell state is acquired.


Subject(s)
Cell Differentiation/physiology , Cellular Reprogramming/physiology , Epigenesis, Genetic/physiology , Heterochromatin/physiology , Induced Pluripotent Stem Cells/physiology , Animals , Blotting, Western , Cell Line , Chromatin Immunoprecipitation , Flow Cytometry , Gene Silencing , Genetic Vectors/genetics , Glycogen Synthase Kinase 3/metabolism , Homeodomain Proteins/metabolism , Image Processing, Computer-Assisted , Mice , Microarray Analysis , Microscopy, Electron, Transmission , Microscopy, Energy-Filtering Transmission Electron , Microscopy, Fluorescence , Mitogen-Activated Protein Kinases/metabolism , Nanog Homeobox Protein , Retroviridae , Retroviridae Proteins/genetics , Retroviridae Proteins/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Sequence Analysis, DNA
18.
Exp Cell Res ; 321(1): 32-9, 2014 Feb 01.
Article in English | MEDLINE | ID: mdl-24333597

ABSTRACT

Before the mammalian embryo is ready to implant in the uterine wall, the single cell zygote must divide and differentiate into three distinct tissues; trophectoderm (prospective placenta), primitive endoderm (prospective yolk sac), and pluripotent epiblast cells which will form the embryo proper. In this review I will discuss our current understanding of how positional information, cell polarization, signaling pathways, and transcription factor networks converge to drive and regulate the progressive segregation of the first three cell types in the mouse embryo.


Subject(s)
Blastocyst/cytology , Cell Differentiation , Cell Lineage , Embryo, Mammalian/cytology , Embryonic Stem Cells/cytology , Endoderm/cytology , Mice/embryology , Animals , Embryo, Mammalian/metabolism , Morphogenesis , Signal Transduction , Transcription Factors/metabolism
19.
Front Cell Dev Biol ; 12: 1341373, 2024.
Article in English | MEDLINE | ID: mdl-38764741

ABSTRACT

Sex differences in the developing human brain are primarily attributed to hormonal influence. Recently however, genetic differences and their impact on the developing nervous system have attracted increased attention. To understand genetically driven sexual dimorphisms in neurodevelopment, we investigated genome-wide gene expression in an in vitro differentiation model of male and female human embryonic stem cell lines (hESC), independent of the effects of human sex hormones. Four male and four female-derived hESC lines were differentiated into a population of mixed neurons over 37 days. Differential gene expression and gene set enrichment analyses were conducted on bulk RNA sequencing data. While similar differentiation tendencies in all cell lines demonstrated the robustness and reproducibility of our differentiation protocol, we found sex-biased gene expression already in undifferentiated ESCs at day 0, but most profoundly after 37 days of differentiation. Male and female cell lines exhibited sex-biased expression of genes involved in neurodevelopment, suggesting that sex influences the differentiation trajectory. Interestingly, the highest contribution to sex differences was found to arise from the male transcriptome, involving both Y chromosome and autosomal genes. We propose 13 sex-biased candidate genes (10 upregulated in male cell lines and 3 in female lines) that are likely to affect neuronal development. Additionally, we confirmed gene dosage compensation of X/Y homologs escaping X chromosome inactivation through their Y homologs and identified a significant overexpression of the Y-linked demethylase UTY and KDM5D in male hESC during neuron development, confirming previous results in neural stem cells. Our results suggest that genetic sex differences affect neuronal differentiation trajectories, which could ultimately contribute to sex biases during human brain development.

20.
Development ; 137(20): 3351-60, 2010 Oct.
Article in English | MEDLINE | ID: mdl-20876656

ABSTRACT

Fibroblast growth factor (FGF) signaling controls fundamental processes such as proliferation, differentiation and migration throughout mammalian development. Here we discuss recent discoveries that implicate FGF/Erk signaling in the control of pluripotency and lineage specification in several different stem cell states, including the separation of pluripotent epiblast and primitive endoderm in the blastocyst, the lineage priming of embryonic stem (ES) cells, and in the stabilization of the metastable state of mouse epiblast and human ES cells. Understanding how extrinsic signals such as FGF regulate different stem cell states will be crucial to harvest the clinical promise of induced pluripotent and embryo-derived stem cells.


Subject(s)
Cell Differentiation/physiology , Cell Lineage/physiology , Cell Movement/physiology , Embryonic Development/physiology , Extracellular Signal-Regulated MAP Kinases/metabolism , Fibroblast Growth Factors/metabolism , Pluripotent Stem Cells/physiology , Signal Transduction/physiology , Animals , Humans , Mice , Pluripotent Stem Cells/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL