Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 38
Filter
1.
Nature ; 615(7954): 900-906, 2023 03.
Article in English | MEDLINE | ID: mdl-36922585

ABSTRACT

Sex chromosome disorders severely compromise gametogenesis in both males and females. In oogenesis, the presence of an additional Y chromosome or the loss of an X chromosome disturbs the robust production of oocytes1-5. Here we efficiently converted the XY chromosome set to XX without an additional Y chromosome in mouse pluripotent stem (PS) cells. In addition, this chromosomal alteration successfully eradicated trisomy 16, a model of Down's syndrome, in PS cells. Artificially produced euploid XX PS cells differentiated into mature oocytes in culture with similar efficiency to native XX PS cells. Using this method, we differentiated induced pluripotent stem cells from the tail of a sexually mature male mouse into fully potent oocytes, which gave rise to offspring after fertilization. This study provides insights that could ameliorate infertility caused by sex chromosome or autosomal disorders, and opens the possibility of bipaternal reproduction.


Subject(s)
Genetic Engineering , In Vitro Techniques , Oocytes , X Chromosome , Animals , Female , Male , Mice , Oocytes/metabolism , Oocytes/physiology , X Chromosome/genetics , Y Chromosome/genetics , Pluripotent Stem Cells/metabolism , Down Syndrome/genetics , Down Syndrome/therapy , Fertilization , Infertility/therapy , Homosexuality, Male , Sex Chromosome Disorders/complications , Sex Chromosome Disorders/genetics , Sex Chromosome Disorders/therapy , Genetic Engineering/methods
2.
EMBO J ; 42(9): e112962, 2023 05 02.
Article in English | MEDLINE | ID: mdl-36929479

ABSTRACT

Human in vitro oogenesis provides a framework for clarifying the mechanism of human oogenesis. To create its benchmark, it is vital to promote in vitro oogenesis using a model physiologically close to humans. Here, we establish a foundation for in vitro oogenesis in cynomolgus (cy) monkeys (Macaca fascicularis): cy female embryonic stem cells harboring one active and one inactive X chromosome (Xa and Xi, respectively) differentiate robustly into primordial germ cell-like cells, which in xenogeneic reconstituted ovaries develop efficiently into oogonia and, remarkably, further into meiotic oocytes at the zygotene stage. This differentiation entails comprehensive epigenetic reprogramming, including Xi reprogramming, yet Xa and Xi remain epigenetically asymmetric with, as partly observed in vivo, incomplete Xi reactivation. In humans and monkeys, the Xi epigenome in pluripotent stem cells functions as an Xi-reprogramming determinant. We further show that developmental pathway over-activations with suboptimal up-regulation of relevant meiotic genes impede in vitro meiotic progression. Cy in vitro oogenesis exhibits critical homology with the human system, including with respect to bottlenecks, providing a salient model for advancing human in vitro oogenesis.


Subject(s)
Oocytes , Oogenesis , Animals , Female , Humans , Macaca fascicularis , Oogenesis/physiology , Ovary , Embryonic Stem Cells
3.
EMBO J ; 41(13): e110600, 2022 07 04.
Article in English | MEDLINE | ID: mdl-35703121

ABSTRACT

Germ cells are unique in engendering totipotency, yet the mechanisms underlying this capacity remain elusive. Here, we perform comprehensive and in-depth nucleome analysis of mouse germ-cell development in vitro, encompassing pluripotent precursors, primordial germ cells (PGCs) before and after epigenetic reprogramming, and spermatogonia/spermatogonial stem cells (SSCs). Although epigenetic reprogramming, including genome-wide DNA de-methylation, creates broadly open chromatin with abundant enhancer-like signatures, the augmented chromatin insulation safeguards transcriptional fidelity. These insulatory constraints are then erased en masse for spermatogonial development. Notably, despite distinguishing epigenetic programming, including global DNA re-methylation, the PGCs-to-spermatogonia/SSCs development entails further euchromatization. This accompanies substantial erasure of lamina-associated domains, generating spermatogonia/SSCs with a minimal peripheral attachment of chromatin except for pericentromeres-an architecture conserved in primates. Accordingly, faulty nucleome maturation, including persistent insulation and improper euchromatization, leads to impaired spermatogenic potential. Given that PGCs after epigenetic reprogramming serve as oogenic progenitors as well, our findings elucidate a principle for the nucleome programming that creates gametogenic progenitors in both sexes, defining a basis for nuclear totipotency.


Subject(s)
Epigenesis, Genetic , Germ Cells , Animals , Chromatin/genetics , Chromatin/metabolism , DNA Methylation , Epigenomics , Female , Germ Cells/metabolism , Male , Mammals/genetics , Mice , Spermatogonia
4.
Nature ; 537(7618): 57-62, 2016 09 01.
Article in English | MEDLINE | ID: mdl-27556940

ABSTRACT

The epiblast (EPI) is the origin of all somatic and germ cells in mammals, and of pluripotent stem cells in vitro. To explore the ontogeny of human and primate pluripotency, here we perform comprehensive single-cell RNA sequencing for pre- and post-implantation EPI development in cynomolgus monkeys (Macaca fascicularis). We show that after specification in the blastocysts, EPI from cynomolgus monkeys (cyEPI) undergoes major transcriptome changes on implantation. Thereafter, while generating gastrulating cells, cyEPI stably maintains its transcriptome over a week, retains a unique set of pluripotency genes and acquires properties for 'neuron differentiation'. Human and monkey pluripotent stem cells show the highest similarity to post-implantation late cyEPI, which, despite co-existing with gastrulating cells, bears characteristics of pre-gastrulating mouse EPI and epiblast-like cells in vitro. These findings not only reveal the divergence and coherence of EPI development, but also identify a developmental coordinate of the spectrum of pluripotency among key species, providing a basis for better regulation of human pluripotency in vitro.


Subject(s)
Embryo, Mammalian/cytology , Embryo, Mammalian/embryology , Embryonic Development , Macaca fascicularis/embryology , Pluripotent Stem Cells/cytology , Animals , Blastocyst/cytology , Blastocyst/metabolism , Cell Differentiation/genetics , Embryo, Mammalian/metabolism , Embryonic Development/genetics , Female , Gastrulation/genetics , Gene Expression Regulation, Developmental , Germ Layers/cytology , Germ Layers/embryology , Germ Layers/metabolism , Humans , Macaca fascicularis/genetics , Mice , Neural Stem Cells/cytology , Neural Stem Cells/metabolism , Neurons/cytology , Neurons/metabolism , Pluripotent Stem Cells/metabolism , Sequence Analysis, RNA , Single-Cell Analysis , Species Specificity , Transcriptome
5.
Genes Dev ; 28(5): 463-78, 2014 Mar 01.
Article in English | MEDLINE | ID: mdl-24589776

ABSTRACT

Many loci maintain parent-of-origin DNA methylation only briefly after fertilization during mammalian development: Whether this form of transient genomic imprinting can impact the early embryonic transcriptome or even have life-long consequences on genome regulation and possibly phenotypes is currently unknown. Here, we report a maternal germline differentially methylated region (DMR) at the mouse Gpr1/Zdbf2 (DBF-type zinc finger-containing protein 2) locus, which controls the paternal-specific expression of long isoforms of Zdbf2 (Liz) in the early embryo. This DMR loses parental specificity by gain of DNA methylation at implantation in the embryo but is maintained in extraembryonic tissues. As a consequence of this transient, tissue-specific maternal imprinting, Liz expression is restricted to the pluripotent embryo, extraembryonic tissues, and pluripotent male germ cells. We found that Liz potentially functions as both Zdbf2-coding RNA and cis-regulatory RNA. Importantly, Liz-mediated events allow a switch from maternal to paternal imprinted DNA methylation and from Liz to canonical Zdbf2 promoter use during embryonic differentiation, which are stably maintained through somatic life and conserved in humans. The Gpr1/Zdbf2 locus lacks classical imprinting histone modifications, but analysis of mutant embryonic stem cells reveals fine-tuned regulation of Zdbf2 dosage through DNA and H3K27 methylation interplay. Together, our work underlines the developmental and evolutionary need to ensure proper Liz/Zdbf2 dosage as a driving force for dynamic genomic imprinting at the Gpr1/Zdbf2 locus.


Subject(s)
DNA Methylation , Genomic Imprinting/genetics , Mammals/genetics , Receptors, G-Protein-Coupled/genetics , Receptors, G-Protein-Coupled/metabolism , Animals , Embryonic Stem Cells/metabolism , Evolution, Molecular , Female , Gene Expression Regulation, Developmental , Histones/metabolism , Humans , Male , Mammals/embryology , Mammals/metabolism , Mice , Promoter Regions, Genetic , Spermatogenesis/genetics
6.
EMBO J ; 36(13): 1888-1907, 2017 07 03.
Article in English | MEDLINE | ID: mdl-28559416

ABSTRACT

The expansion of primordial germ cells (PGCs), the precursors for the oocytes and spermatozoa, is a key challenge in reproductive biology/medicine. Using a chemical screening exploiting PGC-like cells (PGCLCs) induced from mouse embryonic stem cells (ESCs), we here identify key signaling pathways critical for PGCLC proliferation. We show that the combinatorial application of Forskolin and Rolipram, which stimulate cAMP signaling via different mechanisms, expands PGCLCs up to ~50-fold in culture. The expanded PGCLCs maintain robust capacity for spermatogenesis, rescuing the fertility of infertile mice. Strikingly, during expansion, PGCLCs comprehensively erase their DNA methylome, including parental imprints, in a manner that precisely recapitulates genome-wide DNA demethylation in gonadal germ cells, while essentially maintaining their identity as sexually uncommitted PGCs, apparently through appropriate histone modifications. By establishing a paradigm for PGCLC expansion, our system reconstitutes the epigenetic "blank slate" of the germ line, an immediate precursory state for sexually dimorphic differentiation.


Subject(s)
Cell Differentiation , Cell Proliferation , Embryonic Stem Cells/physiology , Epigenesis, Genetic , Germ Cells/growth & development , Animals , Colforsin/metabolism , Germ Cells/drug effects , Mice , Rolipram/metabolism , Signal Transduction
7.
Biol Reprod ; 102(3): 620-638, 2020 03 13.
Article in English | MEDLINE | ID: mdl-31724030

ABSTRACT

In vitro reconstitution of germ-cell development from pluripotent stem cells (PSCs) has created key opportunities to explore the fundamental mechanisms underlying germ-cell development, particularly in mice and humans. Importantly, such investigations have clarified critical species differences in the mechanisms regulating mouse and human germ-cell development, highlighting the necessity of establishing an in vitro germ-cell development system in other mammals, such as non-human primates. Here, we show that multiple lines of embryonic stem cells (ESCs) and induced pluripotent stem cells (iPSCs) in cynomolgus monkeys (Macaca fascicularis; cy) can be maintained stably in an undifferentiated state under a defined condition with an inhibitor for WNT signaling, and such PSCs are induced efficiently into primordial germ cell-like cells (PGCLCs) bearing a transcriptome similar to early cyPGCs. Interestingly, the induction kinetics of cyPGCLCs from cyPSCs is faster than that of human (h) PGCLCs from hPSCs, and while the transcriptome dynamics during cyPGCLC induction is relatively similar to that during hPGCLC induction, it is substantially divergent from that during mouse (m) PGCLC induction. Our findings delineate common as well as species-specific traits for PGC specification, creating a foundation for parallel investigations into the mechanism for germ-cell development in mice, monkeys, and humans.


Subject(s)
Cell Differentiation/physiology , Pluripotent Stem Cells/cytology , Animals , Induced Pluripotent Stem Cells/cytology , Macaca fascicularis , Transcriptome
8.
Nature ; 485(7398): 381-5, 2012 Apr 11.
Article in English | MEDLINE | ID: mdl-22495304

ABSTRACT

In eukaryotes transcriptional regulation often involves multiple long-range elements and is influenced by the genomic environment. A prime example of this concerns the mouse X-inactivation centre (Xic), which orchestrates the initiation of X-chromosome inactivation (XCI) by controlling the expression of the non-protein-coding Xist transcript. The extent of Xic sequences required for the proper regulation of Xist remains unknown. Here we use chromosome conformation capture carbon-copy (5C) and super-resolution microscopy to analyse the spatial organization of a 4.5-megabases (Mb) region including Xist. We discover a series of discrete 200-kilobase to 1 Mb topologically associating domains (TADs), present both before and after cell differentiation and on the active and inactive X. TADs align with, but do not rely on, several domain-wide features of the epigenome, such as H3K27me3 or H3K9me2 blocks and lamina-associated domains. TADs also align with coordinately regulated gene clusters. Disruption of a TAD boundary causes ectopic chromosomal contacts and long-range transcriptional misregulation. The Xist/Tsix sense/antisense unit illustrates how TADs enable the spatial segregation of oppositely regulated chromosomal neighbourhoods, with the respective promoters of Xist and Tsix lying in adjacent TADs, each containing their known positive regulators. We identify a novel distal regulatory region of Tsix within its TAD, which produces a long intervening RNA, Linx. In addition to uncovering a new principle of cis-regulatory architecture of mammalian chromosomes, our study sets the stage for the full genetic dissection of the X-inactivation centre.


Subject(s)
RNA, Untranslated/genetics , X Chromosome Inactivation/genetics , X Chromosome/genetics , Animals , Cell Differentiation , DNA, Intergenic/genetics , Embryonic Stem Cells/cytology , Embryonic Stem Cells/metabolism , Epigenesis, Genetic , Epigenomics , Female , Fibroblasts , Gene Expression Regulation , Histones/metabolism , In Situ Hybridization, Fluorescence , Male , Methylation , Mice , Molecular Sequence Data , Promoter Regions, Genetic/genetics , RNA, Long Noncoding , Transcriptome , X Chromosome/chemistry
9.
Nature ; 472(7343): 370-4, 2011 Apr 21.
Article in English | MEDLINE | ID: mdl-21471966

ABSTRACT

X-chromosome inactivation (XCI) in female mammals allows dosage compensation for X-linked gene products between the sexes. The developmental regulation of this process has been extensively investigated in mice, where the X chromosome of paternal origin (Xp) is silenced during early embryogenesis owing to imprinted expression of the regulatory RNA, Xist (X-inactive specific transcript). Paternal XCI is reversed in the inner cell mass of the blastocyst and random XCI subsequently occurs in epiblast cells. Here we show that other eutherian mammals have very different strategies for initiating XCI. In rabbits and humans, the Xist homologue is not subject to imprinting and XCI begins later than in mice. Furthermore, Xist is upregulated on both X chromosomes in a high proportion of rabbit and human embryo cells, even in the inner cell mass. In rabbits, this triggers XCI on both X chromosomes in some cells. In humans, chromosome-wide XCI has not initiated even by the blastocyst stage, despite the upregulation of XIST. The choice of which X chromosome will finally become inactive thus occurs downstream of Xist upregulation in both rabbits and humans, unlike in mice. Our study demonstrates the remarkable diversity in XCI regulation and highlights differences between mammals in their requirement for dosage compensation during early embryogenesis.


Subject(s)
Chromosomes, Mammalian/genetics , Gene Expression Regulation, Developmental/genetics , Mammals/genetics , X Chromosome Inactivation/genetics , X Chromosome/genetics , Animals , Biological Evolution , Blastocyst/metabolism , Dosage Compensation, Genetic/genetics , Embryo, Mammalian/embryology , Embryo, Mammalian/metabolism , Female , Genes, X-Linked/genetics , Genomic Imprinting/genetics , Histones/metabolism , Humans , Hypoxanthine Phosphoribosyltransferase/genetics , Male , Mammals/embryology , Mice , Parthenogenesis , RNA, Long Noncoding , RNA, Untranslated/genetics , Rabbits , Species Specificity , Up-Regulation/genetics
10.
Nucleic Acids Res ; 43(9): e60, 2015 May 19.
Article in English | MEDLINE | ID: mdl-25722368

ABSTRACT

Single-cell mRNA sequencing (RNA-seq) methods have undergone rapid development in recent years, and transcriptome analysis of relevant cell populations at single-cell resolution has become a key research area of biomedical sciences. We here present single-cell mRNA 3-prime end sequencing (SC3-seq), a practical methodology based on PCR amplification followed by 3-prime-end enrichment for highly quantitative, parallel and cost-effective measurement of gene expression in single cells. The SC3-seq allows excellent quantitative measurement of mRNAs ranging from the 10,000-cell to 1-cell level, and accordingly, allows an accurate estimate of the transcript levels by a regression of the read counts of spike-in RNAs with defined copy numbers. The SC3-seq has clear advantages over other typical single-cell RNA-seq methodologies for the quantitative measurement of transcript levels and at a sequence depth required for the saturation of transcript detection. The SC3-seq distinguishes four distinct cell types in the peri-implantation mouse blastocysts. Furthermore, the SC3-seq reveals the heterogeneity in human-induced pluripotent stem cells (hiPSCs) cultured under on-feeder as well as feeder-free conditions, demonstrating a more homogeneous property of the feeder-free hiPSCs. We propose that SC3-seq might be used as a powerful strategy for single-cell transcriptome analysis in a broad range of investigations in biomedical sciences.


Subject(s)
Gene Expression Profiling/methods , High-Throughput Nucleotide Sequencing/methods , Sequence Analysis, RNA/methods , Single-Cell Analysis , Animals , Blastocyst/metabolism , Humans , Induced Pluripotent Stem Cells/metabolism , Mice , Mice, Inbred C57BL
11.
PLoS Genet ; 9(11): e1003791, 2013 11.
Article in English | MEDLINE | ID: mdl-24244175

ABSTRACT

In most mouse tissues, long-interspersed elements-1 (L1s) are silenced via methylation of their 5'-untranslated regions (5'-UTR). A gradual loss-of-methylation in pre-implantation embryos coincides with L1 retrotransposition in blastocysts, generating potentially harmful mutations. Here, we show that Dicer- and Ago2-dependent RNAi restricts L1 accumulation and retrotransposition in undifferentiated mouse embryonic stem cells (mESCs), derived from blastocysts. RNAi correlates with production of Dicer-dependent 22-nt small RNAs mapping to overlapping sense/antisense transcripts produced from the L1 5'-UTR. However, RNA-surveillance pathways simultaneously degrade these transcripts and, consequently, confound the anti-L1 RNAi response. In Dicer(-/-) mESC complementation experiments involving ectopic Dicer expression, L1 silencing was rescued in cells in which microRNAs remained strongly depleted. Furthermore, these cells proliferated and differentiated normally, unlike their non-complemented counterparts. These results shed new light on L1 biology, uncover defensive, in addition to regulatory roles for RNAi, and raise questions on the differentiation defects of Dicer(-/-) mESCs.


Subject(s)
Argonaute Proteins/genetics , DEAD-box RNA Helicases/genetics , Embryonic Stem Cells/metabolism , Long Interspersed Nucleotide Elements/genetics , RNA Interference , Ribonuclease III/genetics , 5' Untranslated Regions , Animals , Cell Differentiation/genetics , Cell Proliferation , DEAD-box RNA Helicases/metabolism , DNA Methylation/genetics , Gene Expression Regulation, Developmental , Mice , Promoter Regions, Genetic , Retroelements/genetics , Ribonuclease III/metabolism
13.
Bull Acad Natl Med ; 197(3): 609-17, 2013 Mar.
Article in French | MEDLINE | ID: mdl-25163344

ABSTRACT

X chromosome inactivation (XCI) is a very good model of epigenetic changes that occur during early development. This essential process occurring in females leads to X-linked gene dosage compensation between the sexes. Recent data suggest that different mammalian species may use different strategies to initiate XCI during early embryogenesis. In mice, XCI occurs in two waves, imprinted during preimplantation then random in the embryo. In humans, XCI is not imprinted and has not yet been triggered at the blastocyst stage. These results highlight the remarkable diversity of XCI mechanisms.


Subject(s)
Chromosomes, Human, X/genetics , Embryonic Development/genetics , X Chromosome Inactivation/genetics , Animals , Female , Genomic Imprinting , Gestational Age , Humans , Mice
14.
Proc Natl Acad Sci U S A ; 106(13): 5198-203, 2009 Mar 31.
Article in English | MEDLINE | ID: mdl-19273861

ABSTRACT

In mammals, X-chromosome dosage compensation is achieved by inactivating one of the two X chromosomes in females. In mice, X inactivation is initially imprinted, with inactivation of the paternal X (Xp) chromosome occurring during preimplantation development. One theory is that the Xp is preinactivated in female embryos, because of its previous silence during meiosis in the male germ line. The extent to which the Xp is active after fertilization and the exact time of onset of X-linked gene silencing have been the subject of debate. We performed a systematic, single-cell transcriptional analysis to examine the activity of the Xp chromosome for a panel of X-linked genes throughout early preimplantation development in the mouse. Rather than being preinactivated, we found the Xp to be fully active at the time of zygotic gene activation, with silencing beginning from the 4-cell stage onward. X-inactivation patterns were, however, surprisingly diverse between genes. Some loci showed early onset (4-8-cell stage) of X inactivation, and some showed extremely late onset (postblastocyst stage), whereas others were never fully inactivated. Thus, we show that silencing of some X-chromosomal regions occurs outside of the usual time window and that escape from X inactivation can be highly lineage specific. These results reveal that imprinted X inactivation in mice is far less concerted than previously thought and highlight the epigenetic diversity underlying the dosage compensation process during early mammalian development.


Subject(s)
Genomic Imprinting , X Chromosome Inactivation , X Chromosome , Animals , Embryonic Development/genetics , Epigenesis, Genetic , Female , Gene Expression Profiling , Genes, X-Linked , Male , Mice , Transcription, Genetic
15.
Nature ; 438(7066): 369-73, 2005 Nov 17.
Article in English | MEDLINE | ID: mdl-16227973

ABSTRACT

In mammals, one of the two X chromosomes is inactivated in females to enable dosage compensation for X-linked gene products. In rodents and marsupials, only the X chromosome of paternal origin (Xp) is silenced during early embryogenesis. This could be due to a carry-over effect of the X chromosome's passage through the male germ line, where it becomes transiently silenced together with the Y chromosome, during meiotic sex chromosome inactivation (MSCI). Here we show that Xist (X inactive specific transcript) transgenes, located on autosomes, do not undergo MSCI in the male germ line of mice and yet can induce imprinted cis-inactivation when paternally inherited, with identical kinetics to the Xp chromosome. This suggests that MSCI is not necessary for imprinted X-chromosome inactivation in mice. We also show that the Xp is transcribed, like autosomes, at zygotic gene activation rather than being 'pre-inactivated'. We propose that expression of the paternal Xist gene at zygotic gene activation is sufficient to trigger cis-inactivation of the X chromosome, or of an autosome carrying a Xist transgene.


Subject(s)
Genomic Imprinting/genetics , Meiosis/genetics , X Chromosome Inactivation/genetics , X Chromosome/genetics , Animals , Embryo, Mammalian/metabolism , Female , Gene Silencing , Histones/metabolism , In Situ Hybridization, Fluorescence , Male , Mice , Mice, Transgenic , RNA, Long Noncoding , RNA, Untranslated/genetics , Spermatocytes/metabolism , Transcription, Genetic/genetics , Transgenes/genetics , Y Chromosome/genetics , Zygote/metabolism
16.
Science ; 374(6570): eabd8887, 2021 Nov 19.
Article in English | MEDLINE | ID: mdl-34793202

ABSTRACT

X chromosome dosage compensation ensures balanced gene dosage between the X chromosome and autosomes and between the sexes, involving divergent mechanisms among mammals. We elucidated a distinct mechanism for X chromosome inactivation (XCI) in cynomolgus monkeys, a model for human development. The trophectoderm and cytotrophoblast acquire XCI around implantation through an active intermediate bearing repressive modifications and compacted structure, whereas the amnion, epiblast, and hypoblast maintain such an intermediate protractedly, attaining XCI by a week after implantation. Males achieve X chromosome up-regulation (XCU) progressively, whereas females show XCU coincidentally with XCI, both establishing the X:autosome dosage compensation by 1 week after implantation. Conversely, primordial germ cells undergo X chromosome reactivation by reversing the XCI pathway early during their development. Our findings establish a foundation for clarifying the dosage compensation mechanisms in primates, including humans.


Subject(s)
Blastocyst/physiology , Dosage Compensation, Genetic , Macaca fascicularis/embryology , Macaca fascicularis/genetics , Trophoblasts/physiology , X Chromosome Inactivation , X Chromosome/genetics , Animals , Embryonic Development , Female , Gene Expression Regulation, Developmental , Genes, X-Linked , Germ Cells/physiology , Histones/metabolism , Methylation , RNA, Long Noncoding/genetics , RNA, Long Noncoding/metabolism , Up-Regulation , X Chromosome/metabolism , X Chromosome/ultrastructure
17.
Cell Rep ; 35(5): 109075, 2021 05 04.
Article in English | MEDLINE | ID: mdl-33951437

ABSTRACT

In the early fetal stage, the gonads are bipotent and only later become the ovary or testis, depending on the genetic sex. Despite many studies examining how sex determination occurs from biopotential gonads, the spatial and temporal organization of bipotential gonads and their progenitors is poorly understood. Here, using lineage tracing in mice, we find that the gonads originate from a T+ primitive streak through WT1+ posterior intermediate mesoderm and appear to share origins anteriorly with the adrenal glands and posteriorly with the metanephric mesenchyme. Comparative single-cell transcriptomic analyses in mouse and cynomolgus monkey embryos reveal the convergence of the lineage trajectory and genetic programs accompanying the specification of biopotential gonadal progenitor cells. This process involves sustained expression of epithelial genes and upregulation of mesenchymal genes, thereby conferring an epithelial-mesenchymal hybrid state. Our study provides key resources for understanding early gonadogenesis in mice and primates.


Subject(s)
Adult Stem Cells/metabolism , Gonads/physiology , Animals , Cell Differentiation , Macaca fascicularis , Male , Mice
18.
Life Sci Alliance ; 4(5)2021 05.
Article in English | MEDLINE | ID: mdl-33608411

ABSTRACT

The in vitro reconstitution of human germ-cell development provides a robust framework for clarifying key underlying mechanisms. Here, we explored transcription factors (TFs) that engender the germ-cell fate in their pluripotent precursors. Unexpectedly, SOX17, TFAP2C, and BLIMP1, which act under the BMP signaling and are indispensable for human primordial germ-cell-like cell (hPGCLC) specification, failed to induce hPGCLCs. In contrast, GATA3 or GATA2, immediate BMP effectors, combined with SOX17 and TFAP2C, generated hPGCLCs. GATA3/GATA2 knockouts dose-dependently impaired BMP-induced hPGCLC specification, whereas GATA3/GATA2 expression remained unaffected in SOX17, TFAP2C, or BLIMP1 knockouts. In cynomolgus monkeys, a key model for human development, GATA3, SOX17, and TFAP2C were co-expressed exclusively in early PGCs. Crucially, the TF-induced hPGCLCs acquired a hallmark of bona fide hPGCs to undergo epigenetic reprogramming and mature into oogonia/gonocytes in xenogeneic reconstituted ovaries. By uncovering a TF circuitry driving the germ line program, our study provides a paradigm for TF-based human gametogenesis.


Subject(s)
Germ Cells/metabolism , SOXF Transcription Factors/metabolism , Transcription Factor AP-2/metabolism , Animals , Cell Differentiation/genetics , Cell Lineage/genetics , Female , GATA Transcription Factors/genetics , GATA Transcription Factors/metabolism , Germ Cells/physiology , Humans , Induced Pluripotent Stem Cells/metabolism , Macaca fascicularis , Mice , Mice, Inbred ICR , SOXF Transcription Factors/genetics , Signal Transduction/genetics , Transcription Factor AP-2/genetics , Transcription Factors/metabolism
19.
Cell Stem Cell ; 28(6): 1023-1039.e13, 2021 06 03.
Article in English | MEDLINE | ID: mdl-33831365

ABSTRACT

Trophoblasts are extraembryonic cells that are essential for maintaining pregnancy. Human trophoblasts arise from the morula as trophectoderm (TE), which, after implantation, differentiates into cytotrophoblasts (CTs), syncytiotrophoblasts (STs), and extravillous trophoblasts (EVTs), composing the placenta. Here we show that naïve, but not primed, human pluripotent stem cells (PSCs) recapitulate trophoblast development. Naive PSC-derived TE and CTs (nCTs) recreated human and monkey TE-to-CT transition. nCTs self-renewed as CT stem cells and had the characteristics of proliferating villous CTs and CTs in the cell column of the first trimester. Notably, although primed PSCs differentiated into trophoblast-like cells (BMP4, A83-01, and PD173074 [BAP]-treated primed PSCs [pBAPs]), pBAPs were distinct from nCTs and human placenta-derived CT stem cells, exhibiting properties consistent with the amnion. Our findings establish an authentic paradigm for human trophoblast development, demonstrating the invaluable properties of naive human PSCs. Our system provides a platform to study the molecular mechanisms underlying trophoblast development and related diseases.


Subject(s)
Pluripotent Stem Cells , Trophoblasts , Cell Differentiation , Female , Humans , Placenta , Pregnancy
20.
Chromosome Res ; 17(5): 659-69, 2009.
Article in English | MEDLINE | ID: mdl-19802706

ABSTRACT

In most mammals, X-chromosome inactivation is used as the strategy to achieve dosage compensation between XX females and XY males. This process is developmentally regulated, resulting in the differential treatment of the two X chromosomes in the same nucleus and mitotic heritability of the silent state. A lack of dosage compensation in an XX embryo is believed to result in early lethality, at least in eutherians. Given its fundamental importance, X-chromosome inactivation would be predicted to be a highly conserved process in mammals. However, recent studies have revealed major mechanistic differences in X inactivation between eutherians and marsupials, suggesting that the evolution of the X chromosome as well as developmental differences between mammals have led to diverse evolutionary strategies for dosage compensation.


Subject(s)
Mammals/genetics , X Chromosome Inactivation , Animals , Biological Evolution , Epigenesis, Genetic , Female , Gene Silencing , Humans , Male , RNA, Long Noncoding , RNA, Untranslated/genetics
SELECTION OF CITATIONS
SEARCH DETAIL