Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Dev Cell ; 59(4): 529-544.e5, 2024 Feb 26.
Artículo en Inglés | MEDLINE | ID: mdl-38295793

RESUMEN

During human fetal development, sex differentiation occurs not only in the gonads but also in the adjacent developing reproductive tract. However, while the cellular composition of male and female human fetal gonads is well described, that of the adjacent developing reproductive tract remains poorly characterized. Here, we performed single-cell transcriptomics on male and female human fetal gonads together with the adjacent developing reproductive tract from first and second trimesters, highlighting the morphological and molecular changes during sex differentiation. We validated different cell populations of the developing reproductive tract and gonads and compared the molecular signatures between the first and second trimesters, as well as between sexes, to identify conserved and sex-specific features. Together, our study provides insights into human fetal sex-specific gonadogenesis and development of the reproductive tract beyond the gonads.


Asunto(s)
Gónadas , Testículo , Humanos , Masculino , Femenino , Ovario , Diferenciación Sexual , Perfilación de la Expresión Génica
2.
Front Cell Dev Biol ; 10: 854740, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35359445

RESUMEN

The study of human intestinal biology in healthy and diseased conditions has always been challenging. Primary obstacles have included limited tissue accessibility, inadequate in vitro maintenance and ethical constrains. The development of three-dimensional organoid cultures has transformed this entirely. Intestinal organoids are self-organized three-dimensional structures that partially recapitulate the identity, cell heterogeneity and cell behaviour of the original tissue in vitro. This includes the capacity of stem cells to self-renew, as well as to differentiate towards major intestinal lineages. Therefore, over the past decade, the use of human organoid cultures has been instrumental to model human intestinal development, homeostasis, disease, and regeneration. Intestinal organoids can be derived from pluripotent stem cells (PSC) or from adult somatic intestinal stem cells (ISC). Both types of organoid sources harbour their respective strengths and weaknesses. In this mini review, we describe the applications of human intestinal organoids, discussing the differences, advantages, and disadvantages of PSC-derived and ISC-derived organoids.

3.
Cells ; 10(8)2021 08 09.
Artículo en Inglés | MEDLINE | ID: mdl-34440801

RESUMEN

Male human fetal germ cells (hFGCs) give rise to spermatogonial stem cells (SSCs), which are the adult precursors of the male gametes. Human SSCs are a promising (autologous) source of cells for male fertility preservation; however, in contrast to mouse SSCs, we are still unable to culture them in the long term. Here, we investigated the effect of two different culture media and four substrates (laminin, gelatin, vitronectin and matrigel) in the culture of dissociated second trimester testes, enriched for hFGCs. After 6 days in culture, we quantified the presence of POU5F1 and DDX4 expressing hFGCs. We observed a pronounced difference in hFGC number in different substrates. The combination of gelatin-coated substrate and medium containing GDNF, LIF, FGF2 and EGF resulted in the highest percentage of hFGCs (10% of the total gonadal cells) after 6 days of culture. However, the vitronectin-coated substrate resulted in a comparable percentage of hFGCs regardless of the media used (3.3% of total cells in Zhou-medium and 4.8% of total cells in Shinohara-medium). We provide evidence that not only the choices of culture medium but also choices of the adequate substrate are crucial for optimizing culture protocols for male hFGCs. Optimizing culture conditions in order to improve the expansion of hFGCs will benefit the development of gametogenesis assays in vitro.


Asunto(s)
Técnicas de Cultivo de Célula/métodos , Medios de Cultivo/farmacología , Células Germinativas/efectos de los fármacos , Células Madre/efectos de los fármacos , Testículo/citología , Diferenciación Celular/efectos de los fármacos , Diferenciación Celular/genética , Células Cultivadas , Colágeno/metabolismo , Medios de Cultivo/química , ARN Helicasas DEAD-box/genética , ARN Helicasas DEAD-box/metabolismo , Combinación de Medicamentos , Gelatina/metabolismo , Perfilación de la Expresión Génica/métodos , Células Germinativas/citología , Células Germinativas/metabolismo , Humanos , Laminina/metabolismo , Masculino , Factor 3 de Transcripción de Unión a Octámeros/genética , Factor 3 de Transcripción de Unión a Octámeros/metabolismo , Proteoglicanos/metabolismo , RNA-Seq/métodos , Análisis de la Célula Individual/métodos , Células Madre/citología , Células Madre/metabolismo , Testículo/embriología , Vitronectina/metabolismo
4.
Cells ; 10(5)2021 05 16.
Artículo en Inglés | MEDLINE | ID: mdl-34065661

RESUMEN

The second trimester of human development is marked by asynchronous gonadal development hampering the isolation of homogenous populations of early and late fetal germ cells (FGCs). We evaluated the feasibility of using surface markers TNAP, PDPN, EPCAM and ITGA6 to isolate FGCs as well as human primordial germ cell-like cells (hPGCLCs) derived from embryonic stem cells (hESCs) from both sexes by fluorescence-activated cell sorting (FACS). Our results suggest that a combination of TNAP and PDPN was sufficient to separate populations of premeiotic FGCs and hPGCLCs in both sexes. This combination of antibodies also proved efficient in separating 'mitotic' from 'retinoic-acid responsive' female FGCs. Furthermore, we report that the differentiation efficiency of TNAP+PDPN+ hPGCLCs from hESCs was sex-independent, but the ability to propagate differed considerably between the sexes. In contrast to male, female hPGCLCs retained their characteristics and exhibited robust colony-forming ability when cultured for five days in medium containing LIF, forskolin and FGF2. We conclude that marked sex differences exist in the isolation and propagation of human FGCs and hPGCLCs. Our study provides novel insights relevant for the optimization of in vitro gametogenesis in humans.


Asunto(s)
Gametogénesis , Técnicas de Cultivo de Célula , Femenino , Feto , Gónadas , Células Madre Embrionarias Humanas , Humanos , Masculino
5.
Stem Cells ; 39(5): 551-563, 2021 05.
Artículo en Inglés | MEDLINE | ID: mdl-33470497

RESUMEN

Protocols for specifying human primordial germ cell-like cells (hPGCLCs) from human embryonic stem cells (hESCs) remain hindered by differences between hESC lines, their derivation methods, and maintenance culture conditions. This poses significant challenges for establishing reproducible in vitro models of human gametogenesis. Here, we investigated the influence of activin A (ActA) during derivation and maintenance on the propensity of hESCs to differentiate into PGCLCs. We show that continuous ActA supplementation during hESC derivation (from blastocyst until the formation of the post-inner cell mass intermediate [PICMI]) and supplementation (from the first passage of the PICMI onwards) is beneficial to differentiate hESCs to PGCLCs subsequently. Moreover, comparing isogenic primed and naïve states prior to differentiation, we showed that conversion of hESCs to the 4i-state improves differentiation to (TNAP [tissue nonspecific alkaline phosphatase]+/PDPN [podoplanin]+) PGCLCs. Those PGCLCs expressed several germ cell markers, including TFAP2C (transcription factor AP-2 gamma), SOX17 (SRY-box transcription factor 17), and NANOS3 (nanos C2HC-type zinc finger 3), and markers associated with germ cell migration, CXCR4 (C-X-C motif chemokine receptor 4), LAMA4 (laminin subunit alpha 4), ITGA6 (integrin subunit alpha 6), and CDH4 (cadherin 4), suggesting that the large numbers of PGCLCs obtained may be suitable to differentiate further into more mature germ cells. Finally, hESCs derived in the presence of ActA showed higher competence to differentiate to hPGCLC, in particular if transiently converted to the 4i-state. Our work provides insights into the differences in differentiation propensity of hESCs and delivers an optimized protocol to support efficient human germ cell derivation.


Asunto(s)
Activinas/genética , Diferenciación Celular/genética , Células Germinativas/citología , Células Madre Embrionarias Humanas/citología , Blastocisto/citología , Cadherinas/genética , Células Cultivadas , Regulación del Desarrollo de la Expresión Génica/genética , Células Germinativas/crecimiento & desarrollo , Células Madre Embrionarias Humanas/metabolismo , Humanos , Integrina alfa6/genética , Laminina/genética , Proteínas de Unión al ARN/genética , Receptores CXCR4/genética , Factores de Transcripción SOXF/genética , Transducción de Señal/genética , Factor de Transcripción AP-2/genética
6.
Sci Rep ; 9(1): 17240, 2019 11 21.
Artículo en Inglés | MEDLINE | ID: mdl-31754138

RESUMEN

Recent progress has enabled the conversion of primed human embryonic stem cells (hESCs) to the naive state of pluripotency, resembling the well-characterized naive mouse ESCs (mESCs). However, a thorough histone epigenetic characterization of this conversion process is currently lacking, while its likeness to the mouse model has not been clearly established. Here, we profile the histone epigenome of hESCs during conversion in a time-resolved experimental design, using an untargeted mass spectrometry-based approach. In total, 23 histone post-translational modifications (hPTMs) changed significantly over time. H3K27Me3 was the most prominently increasing marker hPTM in naive hESCs. This is in line with previous reports in mouse, prompting us to compare all the shared hPTM fold changes between mouse and human, revealing a set of conserved hPTM markers for the naive state. Principally, we present the first roadmap of the changing human histone epigenome during the conversion of hESCs from the primed to the naive state. This further revealed similarities with mouse, which hint at a conserved mammalian epigenetic signature of the ground state of pluripotency.


Asunto(s)
Biomarcadores/metabolismo , Histonas/metabolismo , Células Madre Embrionarias Humanas/metabolismo , Animales , Diferenciación Celular/fisiología , Células Cultivadas , Epigenoma/fisiología , Humanos , Ratones , Células Madre Pluripotentes/metabolismo , Transducción de Señal/fisiología
7.
Mol Hum Reprod ; 25(9): 519-526, 2019 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-31211841

RESUMEN

Our current knowledge of the mechanisms leading to human primordial germ cell (PGC) specification stems solely from differentiation experiments starting from human pluripotent stem cells. However, information regarding the origin of PGCs in vivo remains obscure. Here we apply an improved system for extended in vitro culture of human embryos to investigate the presence of PGC-like cells (PGCLCs) 12 days post fertilization (dpf). Good quality blastocysts (n = 141) were plated at 6 dpf and maintained in hypoxia, in medium supplemented with Activin A until 12 dpf. We primarily reveal that 12 dpf outgrowths recapitulate human peri-implantation events and demonstrate that blastocyst quality significantly impacts both embryo viability at 12 dpf, as well as the presence of POU5F1+ cells within viable outgrowths. Moreover, detailed examination of 12 dpf blastocyst outgrowths revealed a population of POU5F1+, SOX2- and SOX17+ cells that may correspond to PGCLCs, alongside POU5F1+ epiblast-like cells and GATA6+ endoderm-like cells. Our findings suggest that, in human, PGC precursors may become specified within the epiblast and migrate either transiently to the extra-embryonic mesoderm or directly to the dorsal part of the yolk sac endoderm around 12 dpf. This is a descriptive analysis and as such the conclusion that POU5F1+ and SOX17+ cells represent bona fide PGCs can only be considered as preliminary. In the future, other PGC markers may be used to further validate the observed cell populations. Overall, our findings provide insights into the origin of the human germline and may serve as a foundation to further unravel the molecular mechanisms governing PGC specification in human.


Asunto(s)
Blastocisto/citología , Blastocisto/fisiología , Linaje de la Célula/fisiología , Células Germinativas/citología , Células Germinativas/fisiología , Diferenciación Celular , Supervivencia Celular , Células Cultivadas , Técnicas de Cultivo de Embriones , Implantación del Embrión/fisiología , Embrión de Mamíferos , Estratos Germinativos/citología , Estratos Germinativos/fisiología , Humanos , Factor 3 de Transcripción de Unión a Octámeros/metabolismo , Células Madre Pluripotentes/citología , Células Madre Pluripotentes/fisiología , Seudópodos/fisiología
8.
Stem Cells Dev ; 28(9): 579-592, 2019 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-30827199

RESUMEN

Human embryonic stem cells (hESCs) hold great value for future clinical applications. However, standard culture conditions maintain hESCs in a primed state, which bears heterogeneity in pluripotency and a tendency for spontaneous differentiation. To counter these drawbacks, primed hESCs have been converted to a naive state, but this has restricted the efficiency of existing directed differentiation protocols. In mouse, WNT inhibition by inhibitor of WNT production-2, together with a higher dose of fibroblast growth factor 2 (12 ng/mL) in DMEM/F12 basal medium (DhiFI), markedly improved derivation and maintenance of primed mouse epiblast stem cells. In this study, we show that DhiFI conditions similarly improved primed hESC traits, such as conferring a primed transcriptional signature with high levels of pluripotency markers and reduced levels of differentiation markers. When triggered to differentiate to neuronal and cardiac lineages, DhiFI hESCs and isogenic primed hESCs progressed similarly. Moreover, DhiFI conditions supported the derivation of hESC lines from a post-inner cell mass intermediate (PICMI). DhiFI-derived hESCs showed less spontaneous differentiation and expressed significantly lower levels of lineage-specific markers, compared to primed-derived lines from the same PICMI. Overall, DhiFI hESCs retained advantages of both primed and naive pluripotency and may ultimately represent a more favorable starting point for differentiation toward clinically desired cell types.


Asunto(s)
Diferenciación Celular/efectos de los fármacos , Factor 2 de Crecimiento de Fibroblastos/farmacología , Células Madre Embrionarias Humanas/efectos de los fármacos , Proteínas Wnt/antagonistas & inhibidores , Benzotiazoles/farmacología , Blastocisto/citología , Células Cultivadas , Técnicas de Cultivo de Embriones , Embrión de Mamíferos , Células Madre Embrionarias Humanas/fisiología , Humanos , Transducción de Señal/efectos de los fármacos , Regulación hacia Arriba/efectos de los fármacos , Vía de Señalización Wnt/efectos de los fármacos
9.
Cell Reprogram ; 17(3): 170-80, 2015 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-26053517

RESUMEN

In mice, inhibition of both the fibroblast growth factor (FGF) mitogen-activated protein kinase kinase/extracellular-signal regulated kinase (MEK/Erk) and the Wnt signaling inhibitor glycogen synthase-3ß (GSK3ß) enables the derivation of mouse embryonic stem cells (mESCs) from nonpermissive strains in the presence of leukemia inhibitory factor (LIF). Whereas mESCs are in an uncommitted naïve state, human embryonic stem cells (hESCs) represent a more advanced state, denoted as primed pluripotency. This burdens hESCs with a series of characteristics, which, in contrast to naïve ESCs, makes them not ideal for key applications such as cell-based clinical therapies and human disease modeling. In this study, different small molecule combinations were applied during human ESC derivation. Hereby, we aimed to sustain the naïve pluripotent state, by interfering with various key signaling pathways. First, we tested several combinations on existing, 2i (PD0325901 and CHIR99021)-derived mESCs. All combinations were shown to be equally adequate to sustain the expression of naïve pluripotency markers. Second, these conditions were tested during hESC derivation. Overall, the best results were observed in the presence of medium supplemented with 2i, LIF, and the noncanonical Wnt signaling agonist Wnt5A, alone and combined with epinephrine. In these conditions, outgrowths repeatedly showed an ESC progenitor-like morphology, starting from day 3. Culturing these "progenitor cells" did not result in stable, naïve hESC lines in the current conditions. Although Wnt5A could not promote naïve hESC derivation, we found that it was sustaining the conversion of established hESCs toward a more naïve state. Future work should aim to distinct the effects of the various culture formulations, including our Wnt5A-supplemented medium, reported to promote stable naïve pluripotency in hESCs.


Asunto(s)
Benzamidas/farmacología , Difenilamina/análogos & derivados , Células Madre Embrionarias Humanas/efectos de los fármacos , Células Madre Embrionarias Humanas/fisiología , Células Madre Pluripotentes/fisiología , Piridinas/farmacología , Pirimidinas/farmacología , Animales , Difenilamina/farmacología , Combinación de Medicamentos , Epinefrina/farmacología , Humanos , Factor Inhibidor de Leucemia/farmacología , Ratones , Cultivo Primario de Células , Proteínas Proto-Oncogénicas/farmacología , Transducción de Señal , Proteínas Wnt/farmacología , Proteína Wnt-5a
10.
Mol Hum Reprod ; 21(5): 410-23, 2015 May.
Artículo en Inglés | MEDLINE | ID: mdl-25634576

RESUMEN

Human embryonic stem cells (hESCs) derived in the presence of Activin A (ActA) demonstrate an increased differentiation propensity toward the germ cell lineage. In addition, mouse epiblast stem cells and mouse epiblast-like cells are poised toward germ cell differentiation and are derived in the presence of ActA. We therefore investigated whether supplementation with ActA enhances in vitro hESC differentiation toward germ cell lineage. ActA up-regulated early primordial germ cell (PGC) genes STELLA/DPPA3 (developmental pluripotency associated 3) and tyrosine kinase receptor cKIT in both ActA-derived and standard-derived hESCs indicating its role in priming hESCs toward the PGC lineage. Indeed, ActA plus bone morphogenic protein 4 (BMP4) strongly increased germ cell differentiation potential of hESCs based on the high expression of late PGC markers DAZL (deleted in azoospermia-like) and VASA/DDX4 (DEAD-box polypeptide 4) at mRNA and protein level. Hence, the combination of ActA with BMP4 provides an additional boost for hESCs to develop into postmigratory germ cells. Together with increased VASA expression in the presence of ActA and BMP4, we also observed up-regulation of endoderm-specific genes GATA4 (GATA binding protein 4) and GATA6. Finally, we were able to further mature these in vitro-derived PGC-like cells (PGCLCs) by culturing them in in vitro maturation (IVM) medium, resulting in the formation of germ cell-like clusters and induction of meiotic gene expression. In conclusion, we demonstrate for the first time a synergism between ActA and BMP4 in facilitating germ cell-directed differentiation of hESCs, which is enhanced by extended culture in IVM medium, as shown by cytoplasmic VASA-expressing PGCLCs. We propose a novel relationship between the endoderm and germ cell lineage during hESC differentiation.


Asunto(s)
Activinas/metabolismo , Regulación del Desarrollo de la Expresión Génica , Células Germinativas/citología , Células Madre Embrionarias Humanas/citología , Bélgica , Biomarcadores/metabolismo , Proteína Morfogenética Ósea 4/metabolismo , Diferenciación Celular , Línea Celular , Linaje de la Célula , Células Cultivadas , Proteínas Cromosómicas no Histona , ARN Helicasas DEAD-box/genética , ARN Helicasas DEAD-box/metabolismo , Cuerpos Embrioides/citología , Cuerpos Embrioides/metabolismo , Factor de Transcripción GATA4/genética , Factor de Transcripción GATA4/metabolismo , Factor de Transcripción GATA6/genética , Factor de Transcripción GATA6/metabolismo , Células Germinativas/metabolismo , Células Madre Embrionarias Humanas/metabolismo , Humanos , Proteínas/genética , Proteínas/metabolismo , Proteínas Proto-Oncogénicas c-kit/genética , Proteínas Proto-Oncogénicas c-kit/metabolismo , Proteínas de Unión al ARN/genética , Proteínas de Unión al ARN/metabolismo , Regulación hacia Arriba
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...