Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 38
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Nature ; 615(7954): 900-906, 2023 03.
Artigo em Inglês | MEDLINE | ID: mdl-36922585

RESUMO

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.


Assuntos
Engenharia Genética , Técnicas In Vitro , Oócitos , Cromossomo X , Animais , Feminino , Masculino , Camundongos , Oócitos/metabolismo , Oócitos/fisiologia , Cromossomo X/genética , Cromossomo Y/genética , Células-Tronco Pluripotentes/metabolismo , Síndrome de Down/genética , Síndrome de Down/terapia , Fertilização , Infertilidade/terapia , Homossexualidade Masculina , Transtornos dos Cromossomos Sexuais/complicações , Transtornos dos Cromossomos Sexuais/genética , Transtornos dos Cromossomos Sexuais/terapia , Engenharia Genética/métodos
2.
EMBO J ; 42(9): e112962, 2023 05 02.
Artigo em Inglês | MEDLINE | ID: mdl-36929479

RESUMO

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.


Assuntos
Oócitos , Oogênese , Animais , Feminino , Humanos , Macaca fascicularis , Oogênese/fisiologia , Ovário , Células-Tronco Embrionárias
3.
EMBO J ; 41(13): e110600, 2022 07 04.
Artigo em Inglês | MEDLINE | ID: mdl-35703121

RESUMO

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.


Assuntos
Epigênese Genética , Células Germinativas , Animais , Cromatina/genética , Cromatina/metabolismo , Metilação de DNA , Epigenômica , Feminino , Células Germinativas/metabolismo , Masculino , Mamíferos/genética , Camundongos , Espermatogônias
4.
Science ; 374(6570): eabd8887, 2021 Nov 19.
Artigo em Inglês | MEDLINE | ID: mdl-34793202

RESUMO

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.


Assuntos
Blastocisto/fisiologia , Compensação de Dosagem (Genética) , Macaca fascicularis/embriologia , Macaca fascicularis/genética , Trofoblastos/fisiologia , Inativação do Cromossomo X , Cromossomo X/genética , Animais , Desenvolvimento Embrionário , Feminino , Regulação da Expressão Gênica no Desenvolvimento , Genes Ligados ao Cromossomo X , Células Germinativas/fisiologia , Histonas/metabolismo , Metilação , RNA Longo não Codificante/genética , RNA Longo não Codificante/metabolismo , Regulação para Cima , Cromossomo X/metabolismo , Cromossomo X/ultraestrutura
5.
Cell Rep ; 35(5): 109075, 2021 05 04.
Artigo em Inglês | MEDLINE | ID: mdl-33951437

RESUMO

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.


Assuntos
Células-Tronco Adultas/metabolismo , Gônadas/fisiologia , Animais , Diferenciação Celular , Macaca fascicularis , Masculino , Camundongos
6.
Cell Stem Cell ; 28(6): 1023-1039.e13, 2021 06 03.
Artigo em Inglês | MEDLINE | ID: mdl-33831365

RESUMO

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.


Assuntos
Células-Tronco Pluripotentes , Trofoblastos , Diferenciação Celular , Feminino , Humanos , Placenta , Gravidez
7.
Life Sci Alliance ; 4(5)2021 05.
Artigo em Inglês | MEDLINE | ID: mdl-33608411

RESUMO

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.


Assuntos
Células Germinativas/metabolismo , Fatores de Transcrição SOXF/metabolismo , Fator de Transcrição AP-2/metabolismo , Animais , Diferenciação Celular/genética , Linhagem da Célula/genética , Feminino , Fatores de Transcrição GATA/genética , Fatores de Transcrição GATA/metabolismo , Células Germinativas/fisiologia , Humanos , Células-Tronco Pluripotentes Induzidas/metabolismo , Macaca fascicularis , Camundongos , Camundongos Endogâmicos ICR , Fatores de Transcrição SOXF/genética , Transdução de Sinais/genética , Fator de Transcrição AP-2/genética , Fatores de Transcrição/metabolismo
8.
Sci Rep ; 10(1): 6827, 2020 04 22.
Artigo em Inglês | MEDLINE | ID: mdl-32321940

RESUMO

The placenta forms a maternal-fetal junction that supports many physiological functions such as the supply of nutrition and exchange of gases and wastes. Establishing an in vitro culture model of human and non-human primate trophoblast stem/progenitor cells is important for investigating the process of early placental development and trophoblast differentiation. In this study, we have established five trophoblast stem cell (TSC) lines from cynomolgus monkey blastocysts, named macTSC #1-5. Fibroblast growth factor 4 (FGF4) enhanced proliferation of macTSCs, while other exogenous factors were not required to maintain their undifferentiated state. macTSCs showed a trophoblastic gene expression profile and trophoblast-like DNA methylation status and also exhibited differentiation capacity towards invasive trophoblast cells and multinucleated syncytia. In a xenogeneic chimera assay, these stem cells contributed to trophectoderm (TE) development in the chimeric blastocysts. macTSC are the first primate trophoblast cell lines whose proliferation is promoted by FGF4. These cell lines provide a valuable in vitro culture model to analyze the similarities and differences in placental development between human and non-human primates.


Assuntos
Técnicas de Cultura de Células/métodos , Células-Tronco/citologia , Trofoblastos/citologia , Animais , Bucladesina/farmacologia , Diferenciação Celular/efeitos dos fármacos , Linhagem Celular , Quimera , Cromossomos de Mamíferos/genética , Metilação de DNA/genética , Ectoderma/citologia , Regulação da Expressão Gênica/efeitos dos fármacos , Células Gigantes/citologia , Macaca fascicularis , Camundongos , MicroRNAs/genética , MicroRNAs/metabolismo , Especificidade da Espécie , Células-Tronco/efeitos dos fármacos , Trofoblastos/efeitos dos fármacos
9.
Biol Reprod ; 102(3): 620-638, 2020 03 13.
Artigo em Inglês | MEDLINE | ID: mdl-31724030

RESUMO

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.


Assuntos
Diferenciação Celular/fisiologia , Células-Tronco Pluripotentes/citologia , Animais , Células-Tronco Pluripotentes Induzidas/citologia , Macaca fascicularis , Transcriptoma
10.
Nat Struct Mol Biol ; 26(5): 350-360, 2019 05.
Artigo em Inglês | MEDLINE | ID: mdl-30962582

RESUMO

Gene-regulatory networks control the establishment and maintenance of alternative gene-expression states during development. A particular challenge is the acquisition of opposing states by two copies of the same gene, as in the case of the long non-coding RNA Xist in mammals at the onset of random X-chromosome inactivation (XCI). The regulatory principles that lead to stable mono-allelic expression of Xist remain unknown. Here, we uncover the minimal regulatory network that can ensure female-specific and mono-alleleic upregulation of Xist, by combining mathematical modeling and experimental validation of central model predictions. We identify a symmetric toggle switch as the basis for random mono-allelic upregulation of Xist, which reproduces data from several mutant, aneuploid and polyploid mouse cell lines with various Xist expression patterns. Moreover, this toggle switch explains the diversity of strategies employed by different species at the onset of XCI. In addition to providing a unifying conceptual framework with which to explore XCI across mammals, our study sets the stage for identifying the molecular mechanisms needed to initiate random XCI.


Assuntos
Mamíferos/genética , Inativação do Cromossomo X , Alelos , Animais , Feminino , Redes Reguladoras de Genes , Humanos , Masculino , RNA Longo não Codificante , Especificidade da Espécie , Biologia de Sistemas
11.
Science ; 362(6412): 356-360, 2018 10 19.
Artigo em Inglês | MEDLINE | ID: mdl-30237246

RESUMO

Human in vitro gametogenesis may transform reproductive medicine. Human pluripotent stem cells (hPSCs) have been induced into primordial germ cell-like cells (hPGCLCs); however, further differentiation to a mature germ cell has not been achieved. Here, we show that hPGCLCs differentiate progressively into oogonia-like cells during a long-term in vitro culture (approximately 4 months) in xenogeneic reconstituted ovaries with mouse embryonic ovarian somatic cells. The hPGCLC-derived oogonia display hallmarks of epigenetic reprogramming-genome-wide DNA demethylation, imprint erasure, and extinguishment of aberrant DNA methylation in hPSCs-and acquire an immediate precursory state for meiotic recombination. Furthermore, the inactive X chromosome shows a progressive demethylation and reactivation, albeit partially. These findings establish the germline competence of hPSCs and provide a critical step toward human in vitro gametogenesis.


Assuntos
Técnicas de Reprogramação Celular/métodos , Células-Tronco Pluripotentes Induzidas/citologia , Oogênese , Oogônios/citologia , Ovário/crescimento & desenvolvimento , Metilação de DNA , Epigênese Genética , Feminino , Humanos
12.
Methods Mol Biol ; 1861: 149-159, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30218366

RESUMO

Transcriptional and epigenetic dynamics of the genome occur during early development in mammals. It has been difficult to study these dynamics due to the limitation of materials and the difficulty of handling. In this chapter, we describe our attempt to apply a combination of immunofluorescence (IF), and RNA and DNA fluorescent in situ hybridization (FISH) in preimplantation mouse embryos. We have concentrated on refining these techniques to study the dynamics of X chromosome inactivation in mouse embryos. The techniques and general underlying principles described here should be applicable to other mammals of interest.


Assuntos
Blastocisto/metabolismo , DNA/análise , Hibridização in Situ Fluorescente/métodos , RNA Longo não Codificante/análise , Inativação do Cromossomo X , Animais , Blastocisto/química , Epigenômica/métodos , Regulação da Expressão Gênica no Desenvolvimento , Camundongos
13.
Nat Commun ; 8(1): 1297, 2017 11 03.
Artigo em Inglês | MEDLINE | ID: mdl-29101321

RESUMO

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.


Assuntos
Massa Celular Interna do Blastocisto/metabolismo , Epigênese Genética , Fatores de Transcrição/farmacocinética , Inativação do Cromossomo X/genética , Animais , Feminino , Genes Ligados ao Cromossomo X , Histonas/metabolismo , Hibridização in Situ Fluorescente , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos DBA , Modelos Genéticos , Gravidez , RNA Longo não Codificante/genética , Análise de Sequência de RNA , Análise de Célula Única
14.
EMBO J ; 36(13): 1888-1907, 2017 07 03.
Artigo em Inglês | MEDLINE | ID: mdl-28559416

RESUMO

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.


Assuntos
Diferenciação Celular , Proliferação de Células , Células-Tronco Embrionárias/fisiologia , Epigênese Genética , Células Germinativas/crescimento & desenvolvimento , Animais , Colforsina/metabolismo , Células Germinativas/efeitos dos fármacos , Camundongos , Rolipram/metabolismo , Transdução de Sinais
15.
Sci Data ; 4: 170067, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28649393

RESUMO

In mammals, the development of pluripotency and specification of primordial germ cells (PGCs) have been studied predominantly using mice as a model organism. However, divergences among mammalian species for such processes have begun to be recognized. Between humans and mice, pre-implantation development appears relatively similar, but the manner and morphology of post-implantation development are significantly different. Nevertheless, the embryogenesis just after implantation in primates, including the specification of PGCs, has been unexplored due to the difficulties in analyzing the embryos at relevant developmental stages. Here, we present a comprehensive single-cell transcriptome dataset of pre- and early post-implantation embryo cells, PGCs and embryonic stem cells (ESCs) of cynomolgus monkeys as a model of higher primates. The identities of each transcriptome were also validated rigorously by other way such as immunofluorescent analysis. The information reported here will serve as a foundation for our understanding of a wide range of processes in the developmental biology of primates, including humans.


Assuntos
Células-Tronco Embrionárias , Macaca fascicularis , Transcriptoma , Animais , Regulação da Expressão Gênica no Desenvolvimento , Macaca fascicularis/embriologia , Macaca fascicularis/genética , Análise de Célula Única
16.
Methods Mol Biol ; 1605: 133-145, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28456962

RESUMO

Characterizing the maternal-to-zygotic transition (MZT) is a central question in embryogenesis, and is critical for our understanding of early post-fertilization events in mammals. High-throughput RNA sequencing (RNA Seq) of mouse oocytes and early embryos has recently revealed that elaborate transcription patterns of genes and repeats are established post-fertilization. This occurs in the context of the gradually depleted maternal pool of RNA provided by the oocyte, which can confound the accurate analysis of the zygotic genome activation when the mRNA population is sequenced. In this context, and given the limited amounts of material available from embryos, particularly when studying mutants, as well as the cost of sequencing, an alternative, complementary single cell approach is RNA FISH. This approach can assay the expression of specific genes or genetic elements during preimplantation development, in particular during the MZT. Here, we describe how RNA FISH can be applied to visualize nascent transcription at specific genomic loci in embryos at different stages of preimplantation development and also discuss possible analytical methods of RNA FISH data.


Assuntos
Embrião de Mamíferos/química , Hibridização in Situ Fluorescente/métodos , RNA/genética , Animais , Análise Citogenética , Regulação da Expressão Gênica no Desenvolvimento , Camundongos , Análise de Célula Única , Ativação Transcricional
17.
Cell Rep ; 17(10): 2789-2804, 2016 12 06.
Artigo em Inglês | MEDLINE | ID: mdl-27926879

RESUMO

The in vitro derivation and propagation of spermatogonial stem cells (SSCs) from pluripotent stem cells (PSCs) is a key goal in reproductive science. We show here that when aggregated with embryonic testicular somatic cells (reconstituted testes), primordial germ cell-like cells (PGCLCs) induced from mouse embryonic stem cells differentiate into spermatogonia-like cells in vitro and are expandable as cells that resemble germline stem cells (GSCs), a primary cell line with SSC activity. Remarkably, GSC-like cells (GSCLCs), but not PGCLCs, colonize adult testes and, albeit less effectively than GSCs, contribute to spermatogenesis and fertile offspring. Whole-genome analyses reveal that GSCLCs exhibit aberrant methylation at vulnerable regulatory elements, including those critical for spermatogenesis, which may restrain their spermatogenic potential. Our study establishes a strategy for the in vitro derivation of SSC activity from PSCs, which, we propose, relies on faithful epigenomic regulation.


Assuntos
Células-Tronco Germinativas Adultas/citologia , Células-Tronco Pluripotentes/citologia , Espermatogênese/genética , Testículo/citologia , Animais , Proliferação de Células/genética , Masculino , Camundongos , Espermatogônias/citologia
18.
Dev Cell ; 39(2): 169-185, 2016 10 24.
Artigo em Inglês | MEDLINE | ID: mdl-27720607

RESUMO

The germ cell lineage ensures reproduction and heredity. The mechanism for germ cell specification in primates, including humans, has remained unknown. In primates, upon implantation the pluripotent epiblast segregates the amnion, an extra-embryonic membrane eventually ensheathing an embryo, and thereafter initiates gastrulation to generate three germ layers. Here, we show that in cynomolgus monkeys, the SOX17/TFAP2C/BLIMP1-positive primordial germ cells (cyPGCs) originate from the dorsal amnion at embryonic day 11 (E11) prior to gastrulation. cyPGCs appear to migrate down the amnion and, through proliferation and recruitment from the posterior amnion, expand in number around the posterior yolk sac by E17. Remarkably, the amnion itself expresses BMP4 and WNT3A, cytokines potentially critical for cyPGC specification, and responds primarily to them. Moreover, human PGC-like cells in vitro exhibit a transcriptome similar to cyPGCs just after specification. Our study identifies the origin of PGCs and a unique function of the nascent amnion in primates.


Assuntos
Âmnio/citologia , Linhagem da Célula , Células Germinativas/citologia , Animais , Comunicação Autócrina/genética , Movimento Celular , Embrião de Mamíferos/citologia , Feto/citologia , Células Germinativas/metabolismo , Gônadas/citologia , Gônadas/embriologia , Humanos , Macaca fascicularis , Mesoderma/metabolismo , Modelos Biológicos , Células-Tronco Pluripotentes/citologia , Transdução de Sinais/genética , Fatores de Transcrição/metabolismo , Transcrição Gênica , Transcriptoma/genética
19.
Nature ; 537(7618): 57-62, 2016 09 01.
Artigo em Inglês | MEDLINE | ID: mdl-27556940

RESUMO

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.


Assuntos
Embrião de Mamíferos/citologia , Embrião de Mamíferos/embriologia , Desenvolvimento Embrionário , Macaca fascicularis/embriologia , Células-Tronco Pluripotentes/citologia , Animais , Blastocisto/citologia , Blastocisto/metabolismo , Diferenciação Celular/genética , Embrião de Mamíferos/metabolismo , Desenvolvimento Embrionário/genética , Feminino , Gastrulação/genética , Regulação da Expressão Gênica no Desenvolvimento , Camadas Germinativas/citologia , Camadas Germinativas/embriologia , Camadas Germinativas/metabolismo , Humanos , Macaca fascicularis/genética , Camundongos , Células-Tronco Neurais/citologia , Células-Tronco Neurais/metabolismo , Neurônios/citologia , Neurônios/metabolismo , Células-Tronco Pluripotentes/metabolismo , Análise de Sequência de RNA , Análise de Célula Única , Especificidade da Espécie , Transcriptoma
20.
Cell Stem Cell ; 17(2): 178-94, 2015 Aug 06.
Artigo em Inglês | MEDLINE | ID: mdl-26189426

RESUMO

Mechanisms underlying human germ cell development are unclear, partly due to difficulties in studying human embryos and lack of suitable experimental systems. Here, we show that human induced pluripotent stem cells (hiPSCs) differentiate into incipient mesoderm-like cells (iMeLCs), which robustly generate human primordial germ cell-like cells (hPGCLCs) that can be purified using the surface markers EpCAM and INTEGRINα6. The transcriptomes of hPGCLCs and primordial germ cells (PGCs) isolated from non-human primates are similar, and although specification of hPGCLCs and mouse PGCs rely on similar signaling pathways, hPGCLC specification transcriptionally activates germline fate without transiently inducing eminent somatic programs. This includes genes important for naive pluripotency and repression of key epigenetic modifiers, concomitant with epigenetic reprogramming. Accordingly, BLIMP1, which represses somatic programs in mice, activates and stabilizes a germline transcriptional circuit and represses a default neuronal differentiation program. Together, these findings provide a foundation for understanding and reconstituting human germ cell development in vitro.


Assuntos
Linhagem da Célula , Células Germinativas/citologia , Células Germinativas/metabolismo , Células-Tronco Pluripotentes Induzidas/citologia , Animais , Sequência de Bases , Biomarcadores/metabolismo , Diferenciação Celular , Epigênese Genética , Genes Reporter , Gônadas/citologia , Humanos , Macaca fascicularis , Mesoderma/citologia , Camundongos , Dados de Sequência Molecular , Neurônios/citologia , Fator 1 de Ligação ao Domínio I Regulador Positivo , Proteínas Repressoras/metabolismo , Transdução de Sinais , Transcrição Gênica
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...