Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Stem Cells ; 39(5): 551-563, 2021 05.
Artigo em Inglês | MEDLINE | ID: mdl-33470497

RESUMO

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.


Assuntos
Ativinas/genética , Diferenciação Celular/genética , Células Germinativas/citologia , Células-Tronco Embrionárias Humanas/citologia , Blastocisto/citologia , Caderinas/genética , Células Cultivadas , Regulação da Expressão Gênica no Desenvolvimento/genética , Células Germinativas/crescimento & desenvolvimento , Células-Tronco Embrionárias Humanas/metabolismo , Humanos , Integrina alfa6/genética , Laminina/genética , Proteínas de Ligação a RNA/genética , Receptores CXCR4/genética , Fatores de Transcrição SOXF/genética , Transdução de Sinais/genética , Fator de Transcrição AP-2/genética
2.
Sci Rep ; 9(1): 17240, 2019 11 21.
Artigo em Inglês | MEDLINE | ID: mdl-31754138

RESUMO

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.


Assuntos
Biomarcadores/metabolismo , Histonas/metabolismo , Células-Tronco Embrionárias Humanas/metabolismo , Animais , Diferenciação Celular/fisiologia , Células Cultivadas , Epigenoma/fisiologia , Humanos , Camundongos , Células-Tronco Pluripotentes/metabolismo , Transdução de Sinais/fisiologia
3.
Mol Hum Reprod ; 25(9): 519-526, 2019 09 01.
Artigo em Inglês | MEDLINE | ID: mdl-31211841

RESUMO

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.


Assuntos
Blastocisto/citologia , Blastocisto/fisiologia , Linhagem da Célula/fisiologia , Células Germinativas/citologia , Células Germinativas/fisiologia , Diferenciação Celular , Sobrevivência Celular , Células Cultivadas , Técnicas de Cultura Embrionária , Implantação do Embrião/fisiologia , Embrião de Mamíferos , Camadas Germinativas/citologia , Camadas Germinativas/fisiologia , Humanos , Fator 3 de Transcrição de Octâmero/metabolismo , Células-Tronco Pluripotentes/citologia , Células-Tronco Pluripotentes/fisiologia , Pseudópodes/fisiologia
4.
Stem Cells Dev ; 28(9): 579-592, 2019 05 01.
Artigo em Inglês | MEDLINE | ID: mdl-30827199

RESUMO

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.


Assuntos
Diferenciação Celular/efeitos dos fármacos , Fator 2 de Crescimento de Fibroblastos/farmacologia , Células-Tronco Embrionárias Humanas/efeitos dos fármacos , Proteínas Wnt/antagonistas & inibidores , Benzotiazóis/farmacologia , Blastocisto/citologia , Células Cultivadas , Técnicas de Cultura Embrionária , Embrião de Mamíferos , Células-Tronco Embrionárias Humanas/fisiologia , Humanos , Transdução de Sinais/efeitos dos fármacos , Regulação para Cima/efeitos dos fármacos , Via de Sinalização Wnt/efeitos dos fármacos
5.
Sci Rep ; 8(1): 5884, 2018 04 12.
Artigo em Inglês | MEDLINE | ID: mdl-29650979

RESUMO

Mouse embryonic stem cells (mESCs) exist in a naive, primed and ground state of pluripotency. While comparative analyses of these pluripotency states have been reported, the mESCs utilized originated from various genetic backgrounds and were derived in different laboratories. mESC derivation in conventional LIF + serum culture conditions is strain dependent, with different genetic backgrounds potentially affecting subsequent stem cell characteristics. In the present study, we performed a comprehensive characterization of naive, primed and ground state mESCs originating from the same genetic background within our laboratory, by comparing their transcriptional profiles. We showed unique transcriptional profiles for naive, primed and ground state mESCs. While naive and ground state mESCs have more similar but not identical profiles, primed state mESCs show a very distinct profile. We further demonstrate that the differentiation propensity of mESCs to specific germ layers is highly dependent on their respective state of pluripotency.


Assuntos
Regulação da Expressão Gênica no Desenvolvimento , Células-Tronco Embrionárias Murinas/metabolismo , Células-Tronco Pluripotentes/metabolismo , Transcriptoma , Animais , Diferenciação Celular , Embrião de Mamíferos , Fator 5 de Crescimento de Fibroblastos/genética , Fator 5 de Crescimento de Fibroblastos/metabolismo , Fator de Transcrição GATA6/genética , Fator de Transcrição GATA6/metabolismo , Ontologia Genética , Patrimônio Genético , Proteínas HMGB/genética , Proteínas HMGB/metabolismo , Queratina-18/genética , Queratina-18/metabolismo , Camundongos , Anotação de Sequência Molecular , Células-Tronco Embrionárias Murinas/citologia , Proteína Homeobox Nanog/genética , Proteína Homeobox Nanog/metabolismo , Fator 3 de Transcrição de Octâmero/genética , Fator 3 de Transcrição de Octâmero/metabolismo , Células-Tronco Pluripotentes/citologia , Fatores de Transcrição SOXB1/genética , Fatores de Transcrição SOXB1/metabolismo , Fatores de Transcrição SOXF/genética , Fatores de Transcrição SOXF/metabolismo , Transdução de Sinais
6.
Reprod Biomed Online ; 36(4): 396-407, 2018 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-29609767

RESUMO

Patients presenting with abnormally high numbers of immature oocytes at retrieval are more likely to exhibit maturation resistant oocytes. However, the clinical relevance of such events remains unknown. We investigated nuclear maturation competence of immature oocytes from patients showing >40% of collected immature oocytes (Study group) and Controls, in which a normal number of mature oocytes (≥60%) was retrieved. Following in-vitro culture, oocytes were classified as maturation resistant or in-vitro matured (IVM). Treatment outcomes were evaluated in Study and Control groups based on presence of maturation resistant oocytes. Overall, similarly high spindle and chromosome abnormality rates were observed in maturation resistant oocytes from both Study and Control groups. IVM oocytes from the Study group revealed significantly higher percentages of misaligned chromosomes compared with Controls (P < 0.05). Remarkably, Study group patients with at least one maturation resistant oocyte showed significantly reduced cumulative pregnancy and live birth rates compared with Control group maturation resistant patients (P < 0.05). When further investigating the aetiology, a maturation resistant mouse model revealed defective Ca2+ signalling of maturation resistant oocytes at germinal vesicular breakdown and parthenogenetic activation. In conclusion, appropriate treatment strategies, including clinical utilization of IVM oocytes from Study group patients, warrant further investigation.


Assuntos
Técnicas de Maturação in Vitro de Oócitos , Meiose/fisiologia , Oócitos/citologia , Indução da Ovulação , Adulto , Animais , Cálcio/metabolismo , Feminino , Humanos , Camundongos , Recuperação de Oócitos , Oócitos/metabolismo , Gravidez , Resultado da Gravidez , Falha de Tratamento
7.
Reprod Fertil Dev ; 30(7): 991-1001, 2018 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-29325603

RESUMO

Inconsistent fertilisation and pregnancy rates have been reported by different laboratories after application of ionomycin as a clinical method of assisted oocyte activation (AOA) to overcome fertilisation failure. Using both mouse and human oocytes, in the present study we investigated the effects of ionomycin and Ca2+ concentrations on the pattern of Ca2+ release and embryonic developmental potential. In the mouse, application of 5µM ionomycin in potassium simplex optimisation medium (KSOM) or 10µM ionomycin in Ca2+-free KSOM significantly reduced the Ca2+ flux and resulted in failure of blastocyst formation compared with 10µM ionomycin in KSOM. Increasing the Ca2+ concentration up to three- or sixfold did not benefit mouse embryonic developmental potential. Similarly, 10µM ionomycin-induced rise in Ca2+ in human oocytes increased with increasing total calcium concentrations in the commercial medium. Remarkably, we observed significantly reduced mouse embryo development when performing AOA over a period of 10min in Quinn's AdvantageTM Fertilisation medium (Cooper Surgical) and IVFTM medium (Vitrolife) compared with Sydney IVF COOK cleavage medium (Cook Ireland), using the same sequential culture system from the post-activation stage to blastocyst formation stage in different AOA groups. In conclusion, concentrations of both ionomycin and Ca2+ in culture media used during AOA can have significant effects on Ca2+ release and further embryonic developmental potential.


Assuntos
Cálcio/metabolismo , Técnicas de Cultura Embrionária , Desenvolvimento Embrionário/fisiologia , Oócitos/citologia , Adulto , Animais , Ionóforos de Cálcio/farmacologia , Meios de Cultura , Desenvolvimento Embrionário/efeitos dos fármacos , Feminino , Humanos , Ionomicina/farmacologia , Camundongos , Oócitos/efeitos dos fármacos , Oócitos/metabolismo , Adulto Jovem
8.
Sci Rep ; 7(1): 16650, 2017 11 30.
Artigo em Inglês | MEDLINE | ID: mdl-29192200

RESUMO

The combination of genome-edited human embryonic stem cells (hESCs) and subsequent neural differentiation is a powerful tool to study neurodevelopmental disorders. Since the naïve state of pluripotency has favourable characteristics for efficient genome-editing, we optimized a workflow for the CRISPR/Cas9 system in these naïve stem cells. Editing efficiencies of respectively 1.3-8.4% and 3.8-19% were generated with the Cas9 nuclease and the D10A Cas9 nickase mutant. Next to this, wildtype and genome-edited naïve hESCs were successfully differentiated to neural progenitor cells. As a proof-of-principle of our workflow, two monoclonal genome-edited naïve hESCs colonies were obtained for TUNA, a long non-coding RNA involved in pluripotency and neural differentiation. In these genome-edited hESCs, an effect was seen on expression of TUNA, although not on neural differentiation potential. In conclusion, we optimized a genome-editing workflow in naïve hESCs that can be used to study candidate genes involved in neural differentiation and/or functioning.


Assuntos
Sistemas CRISPR-Cas , Edição de Genes , Células-Tronco Embrionárias Humanas/metabolismo , Diferenciação Celular/genética , Linhagem Celular , Repetições Palindrômicas Curtas Agrupadas e Regularmente Espaçadas , Sequenciamento de Nucleotídeos em Larga Escala , Humanos , Células-Tronco Neurais/citologia , Células-Tronco Neurais/metabolismo , Fluxo de Trabalho
9.
Methods Mol Biol ; 1516: 13-46, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27044048

RESUMO

Mouse embryonic stem cells are known to represent the naïve state of pluripotency, while human embryonic stem cells typically represented the primed state of pluripotency, characterized by a higher drift toward differentiation and some other disadvantages. Here we describe an efficient method for rapid, transgene free induction of the naïve pluripotent state in human by applying a novel combination of small molecules and growth factors in the culture medium (2i, LIF, basic fibroblast growth factor, ascorbic acid, and forskolin). Conversion of primed human embryonic stem cells towards the naive pluripotent state should be confirmed by a detailed characterization of the cells, as described in this chapter.


Assuntos
Técnicas de Cultura de Células/métodos , Células-Tronco Embrionárias Humanas/efeitos dos fármacos , Células-Tronco Embrionárias Murinas/efeitos dos fármacos , Animais , Diferenciação Celular/efeitos dos fármacos , Humanos , Peptídeos e Proteínas de Sinalização Intercelular/farmacologia , Camundongos , Transdução de Sinais/efeitos dos fármacos , Bibliotecas de Moléculas Pequenas/farmacologia
10.
Hum Reprod Update ; 21(5): 616-26, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26089403

RESUMO

BACKGROUND: Until recently, the temporal events that precede the generation of pluripotent embryonic stem cells (ESCs) and their equivalence with specific developmental stages in vivo was poorly understood. Our group has discovered the existence of a transient epiblast-like structure, coined the post-inner cell mass (ICM) intermediate or PICMI, that emerges before human ESC (hESCs) are established, which supports their primed nature (i.e. already showing some predispositions towards certain cell types) of pluripotency. METHODS: The PICMI results from the progressive epithelialization of the ICM and it expresses a mixture of early and late epiblast markers, as well as some primordial germ cell markers. The PICMI is a closer progenitor of hESCs than the ICM and it can be seen as the first proof of why all existing hESCs, until recently, display a primed state of pluripotency. RESULTS: Even though the pluripotent characteristics of ESCs differ from mouse (naïve) to human (primed), it has recently been shown in mice that a similar process of self-organization at the transition from ICM to (naïve) mouse ESCs (mESCs) transforms the amorphous ICM into a rosette of polarized epiblast cells, a mouse PICMI. The transient PICMI stage is therefore at the origin of both mESCs and hESCs. In addition, several groups have now reported the conversion from primed to the naïve (mESCs-like) hESCs, broadening the pluripotency spectrum and opening new opportunities for the use of pluripotent stem cells. CONCLUSIONS: In this review, we discuss the recent discoveries of mouse and human transient states from ICM to ESCs and their relation towards the state of pluripotency in the eventual stem cells, being naïve or primed. We will now further investigate how these intermediate and/or different pluripotent stages may impact the use of human stem cells in regenerative medicine and assisted reproductive technology.


Assuntos
Massa Celular Interna do Blastocisto/citologia , Camadas Germinativas/citologia , Células-Tronco Pluripotentes/citologia , Técnicas de Reprodução Assistida , Animais , Biomarcadores/metabolismo , Blastocisto/citologia , Células-Tronco Embrionárias/citologia , Humanos , Camundongos
11.
Stem Cells ; 33(9): 2686-98, 2015 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-26108678

RESUMO

Human embryonic stem cells (hESCs) closely resemble mouse epiblast stem cells exhibiting primed pluripotency unlike mouse ESCs (mESCs), which acquire a naïve pluripotent state. Efforts have been made to trigger naïve pluripotency in hESCs for subsequent unbiased lineage-specific differentiation, a common conundrum faced by primed pluripotent hESCs due to heterogeneity in gene expression existing within and between hESC lines. This required either ectopic expression of naïve genes such as NANOG and KLF2 or inclusion of multiple pluripotency-associated factors. We report here a novel combination of small molecules and growth factors in culture medium (2i/LIF/basic fibroblast growth factor + Ascorbic Acid + Forskolin) facilitating rapid induction of transgene-free naïve pluripotency in hESCs, as well as in mESCs, which has not been shown earlier. The converted naïve hESCs survived long-term single-cell passaging, maintained a normal karyotype, upregulated naïve pluripotency genes, and exhibited dependence on signaling pathways similar to naïve mESCs. Moreover, they undergo global DNA demethylation and show a distinctive long noncoding RNA profile. We propose that in our medium, the FGF signaling pathway via PI3K/AKT/mTORC induced the conversion of primed hESCs toward naïve pluripotency. Collectively, we demonstrate an alternate route to capture naïve pluripotency in hESCs that is fast, reproducible, supports naïve mESC derivation, and allows efficient differentiation.


Assuntos
Células-Tronco Embrionárias Humanas/fisiologia , Células-Tronco Pluripotentes/fisiologia , Animais , Diferenciação Celular/efeitos dos fármacos , Diferenciação Celular/fisiologia , Células Cultivadas , Feminino , Células-Tronco Embrionárias Humanas/efeitos dos fármacos , Humanos , Peptídeos e Proteínas de Sinalização Intercelular/farmacologia , Camundongos , Camundongos Endogâmicos C57BL , Células-Tronco Pluripotentes/efeitos dos fármacos
12.
Cell Reprogram ; 17(3): 170-80, 2015 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-26053517

RESUMO

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.


Assuntos
Benzamidas/farmacologia , Difenilamina/análogos & derivados , Células-Tronco Embrionárias Humanas/efeitos dos fármacos , Células-Tronco Embrionárias Humanas/fisiologia , Células-Tronco Pluripotentes/fisiologia , Piridinas/farmacologia , Pirimidinas/farmacologia , Animais , Difenilamina/farmacologia , Combinação de Medicamentos , Epinefrina/farmacologia , Humanos , Fator Inibidor de Leucemia/farmacologia , Camundongos , Cultura Primária de Células , Proteínas Proto-Oncogênicas/farmacologia , Transdução de Sinais , Proteínas Wnt/farmacologia , Proteína Wnt-5a
13.
Mol Hum Reprod ; 21(5): 410-23, 2015 May.
Artigo em Inglês | MEDLINE | ID: mdl-25634576

RESUMO

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.


Assuntos
Ativinas/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Células Germinativas/citologia , Células-Tronco Embrionárias Humanas/citologia , Bélgica , Biomarcadores/metabolismo , Proteína Morfogenética Óssea 4/metabolismo , Diferenciação Celular , Linhagem Celular , Linhagem da Célula , Células Cultivadas , Proteínas Cromossômicas não Histona , RNA Helicases DEAD-box/genética , RNA Helicases DEAD-box/metabolismo , Corpos Embrioides/citologia , Corpos Embrioides/metabolismo , Fator de Transcrição GATA4/genética , Fator de Transcrição GATA4/metabolismo , Fator de Transcrição GATA6/genética , Fator de Transcrição GATA6/metabolismo , Células Germinativas/metabolismo , Células-Tronco Embrionárias Humanas/metabolismo , Humanos , Proteínas/genética , Proteínas/metabolismo , Proteínas Proto-Oncogênicas c-kit/genética , Proteínas Proto-Oncogênicas c-kit/metabolismo , Proteínas de Ligação a RNA/genética , Proteínas de Ligação a RNA/metabolismo , Regulação para Cima
14.
Stem Cells Dev ; 24(4): 497-506, 2015 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-25245024

RESUMO

Early lineage segregation in preimplantation embryos and maintenance of pluripotency in embryonic stem cells (ESCs) are both regulated by specific signaling pathways. Small molecules have been shown to modulate these signaling pathways. We examined the influence of several small molecules and growth factors on second-lineage segregation of the inner cell mass toward hypoblast and epiblast lineage during mouse embryonic preimplantation development. We found that the second-lineage segregation is influenced by activation or inhibition of the transforming growth factor (TGF)ß pathway. Inhibition of the TGFß pathway from the two-cell, four-cell, and morula stages onward up to the blastocyst stage significantly increased the epiblast cell proliferation. The epiblast formed in the embryos in which TGFß signaling was inhibited was fully functional as demonstrated by the potential of these epiblast cells to give rise to pluripotent ESCs. Conversely, activating the TGFß pathway reduced epiblast formation. Inhibition of the glycogen synthase kinase (GSK)3 pathway and activation of bone morphogenetic protein 4 signaling reduced the formation of both epiblast and hypoblast cells. Activation of the protein kinase A pathway and of the Janus kinase/signal transducer and activator of transcription 3 pathway did not influence the second-lineage segregation in mouse embryos. The simultaneous inhibition of three pathways--TGFß, GSK3ß, and the fibroblast growth factor (FGF)/extracellular signal-regulated kinases (Erk)--significantly enhanced the proliferation of epiblast cells than that caused by inhibition of either TGFß pathway alone or by combined inhibition of the GSK3ß and FGF/Erk pathways only.


Assuntos
Linhagem da Célula , Camadas Germinativas/citologia , Transdução de Sinais , Fator de Crescimento Transformador beta/metabolismo , Fator 3 Ativador da Transcrição/metabolismo , Animais , Proteína Morfogenética Óssea 4/metabolismo , Proliferação de Células , Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , Células-Tronco Embrionárias/citologia , Células-Tronco Embrionárias/metabolismo , Células-Tronco Embrionárias/fisiologia , Camadas Germinativas/embriologia , Camadas Germinativas/metabolismo , Quinase 3 da Glicogênio Sintase/metabolismo , Janus Quinases/metabolismo , Camundongos
15.
Hum Reprod ; 29(1): 41-8, 2014 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-24256991

RESUMO

STUDY QUESTION: Is there an effect of the TGFß inhibitor SB431542 (SB) on the epiblast compartment of human blastocysts, and does it affect subsequent human embryonic stem cell (hESC) derivation? SUMMARY ANSWER: SB increases the mean number of NANOG-positive cells in the inner cell mass (ICM), and allows for subsequent hESC derivation. WHAT IS KNOWN ALREADY: It is known that inhibition of TGFß by SB has a positive effect on mouse ESC self-renewal, while active TGFß signalling is needed for self-renewal of primed ESC. STUDY DESIGN, SIZE, DURATION: From December 2011 until March 2012, 263 donated spare embryos were used from patients who had undergone IVF/ICSI in our centre. PARTICIPANTS/MATERIALS, SETTING, METHODS: Donated human embryos were cultured in the presence of SB or Activin A, and immunocytochemistry was performed on Day 6 blastocysts for NANOG and GATA6. Moreover, blastocysts were used for the derivation of hESC, with or without exposure to SB. MAIN RESULTS AND THE ROLE OF CHANCE: Immunocytochemistry revealed a significantly higher number of NANOG-positive ICM cells in the SB group compared with the control (12.0 ± 5.9 versus 6.1 ± 4.7), while no difference was observed in the Activin A group compared with other groups (6.7 ± 3.7). The number of GATA6-positive ICM cells did not differ between the SB, Activin A and control group (8.8 ± 4.3, 8.0 ± 4.6 and 7.2 ± 4.0, respectively). Blocking TGFß signalling did not prevent subsequent hESC line derivation. LIMITATIONS, REASONS FOR CAUTION: The number of human blastocysts available for this study was too low to reveal if the observed increase in NANOG-positive epiblast cells after exposure to SB affected the efficiency of hESC derivation (12.5% compared with 16.7%). WIDER IMPLICATIONS OF THE FINDINGS: This work can contribute to the derivation of naive hESC lines in the future. STUDY FUNDING/COMPETING INTEREST(S): M.V.d.J. is holder of a Ph.D. grant of the Agency for Innovation by Science and Technology (IWT, grant number SB093128), Belgium. G.D. and this research are supported by the Research Foundation Flanders (FWO), grant number FWO-3G062910) and a Concerted Research Actions funding from BOF (Bijzonder Onderzoeksfonds University Ghent, grant number BOF GOA 01G01112). S.M.C.d. S.L. is supported by the Netherlands Organization of Scientific Research (NWO) (ASPASIA 015.007.037) and the Interuniversity Attraction Poles (PAI) (no. P7/07). P.D.S. is holder of a fundamental clinical research mandate by the FWO. We would like to thank Ferring Company (Aalst, Belgium) for financial support of this study. The authors do not have any competing interests to declare. TRIAL REGISTRATION NUMBER: Not applicable.


Assuntos
Benzamidas/farmacologia , Dioxóis/farmacologia , Embrião de Mamíferos/efeitos dos fármacos , Células-Tronco Embrionárias/citologia , Camadas Germinativas/citologia , Fator de Crescimento Transformador beta/antagonistas & inibidores , Ativinas/farmacologia , Linhagem Celular , Proliferação de Células , Técnicas de Cultura Embrionária/métodos , Camadas Germinativas/efeitos dos fármacos , Proteínas de Homeodomínio/biossíntese , Humanos , Proteína Homeobox Nanog , Fator de Crescimento Transformador beta/fisiologia
16.
Stem Cells Dev ; 22(23): 3141-55, 2013 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-23829223

RESUMO

Human embryonic stem cells (hESCs) are more similar to "primed" mouse epiblast stem cells (mEpiSCs). mEpiSCs, which are derived in Activin A, show an increased propensity to form primordial germ cell (PGC)-like cells in response to bone morphogenic protein 4 (BMP4). Hence, we hypothesized that hESCs derived in the presence of Activin A may be more competent in differentiating towards PGC-like cells after supplementation with BMP4 compared to standard hESC lines. We were able to successfully derive two hESC lines in the presence of Activin A, which were pluripotent and showed higher base levels of STELLA and cKIT compared to standard hESC lines derived without Activin A addition. Furthermore, upon differentiation as embryoid bodies in the presence of BMP4, we observed upregulation of VASA at day 7, both at the transcript and protein level compared to standard hESC lines, which appeared to take longer time for PGC specification. Unlike other hESC lines, nuclear pSMAD2/3 presence confirmed that Activin signalling was switched on in Activin A-derived hESC lines. They were also responsive to BMP4 based on nuclear detection of pSMAD1/5/8 and showed endodermal differentiation as a result of GATA-6 expression. Hence, our results provide novel insights into the impact of hESC derivation in the presence of Activin A and its subsequent influence on germ cell differentiation potential in vitro.


Assuntos
Ativinas/farmacologia , Diferenciação Celular/efeitos dos fármacos , Células-Tronco Embrionárias/citologia , Células Germinativas/citologia , Animais , Biomarcadores/metabolismo , Proteína Morfogenética Óssea 4/farmacologia , Linhagem Celular , Corpos Embrioides/citologia , Corpos Embrioides/efeitos dos fármacos , Corpos Embrioides/metabolismo , Células-Tronco Embrionárias/efeitos dos fármacos , Células-Tronco Embrionárias/metabolismo , Endoderma/efeitos dos fármacos , Endoderma/metabolismo , Imunofluorescência , Fator de Transcrição GATA6/metabolismo , Perfilação da Expressão Gênica , Células Germinativas/efeitos dos fármacos , Células Germinativas/metabolismo , Humanos , Camundongos , Células-Tronco Pluripotentes/citologia , Células-Tronco Pluripotentes/efeitos dos fármacos , Células-Tronco Pluripotentes/metabolismo , Transdução de Sinais/efeitos dos fármacos , Fator de Crescimento Transformador beta/metabolismo
17.
Nat Protoc ; 8(2): 254-64, 2013 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-23306459

RESUMO

Little is known about the true developmental origin of human embryonic stem cells (hESCs) or the events that initiate their generation. Recently, we have shown that hESCs originate from a post-inner cell mass (ICM) intermediate (PICMI), a unique transient epiblast-like structure that is different from both its ICM progenitor and its subsequent hESC fate. As a closer progenitor of hESCs than the ICM, the PICMI could be used to provide further insight into the human pluripotent state. Here we provide a detailed (7-d) protocol for the culture of the human preimplantation embryos in order to derive the PICMI. Subsequent identification and cryopreservation of the PICMI are described, in addition to hESC derivation. The initial hESC outgrowth is visible within 2-7 d after PICMI plating. By using the protocol provided, we observed PICMI formation in 21.3% of plated blastocysts with good-quality ICMs. Of the PICMIs used for hESC derivation, 80.6% showed hESC outgrowth after further culture.


Assuntos
Massa Celular Interna do Blastocisto/citologia , Técnicas de Cultura de Células/métodos , Criopreservação/métodos , Técnicas de Cultura Embrionária/métodos , Células-Tronco Embrionárias/citologia , Camadas Germinativas/citologia , Massa Celular Interna do Blastocisto/fisiologia , Camadas Germinativas/embriologia , Humanos
18.
Stem Cells Dev ; 22(2): 296-306, 2013 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-22784186

RESUMO

In embryonic stem cell culture, small molecules can be used to alter key signaling pathways to promote self-renewal and inhibit differentiation. In mice, small-molecule inhibition of both the FGF/MEK/Erk and the GSK3ß pathways during preimplantation development suppresses hypoblast formation, and this results in more pluripotent cells of the inner cell mass (ICM). In this study, we evaluated the effects of different small-molecule inhibitors of the FGF/MEK/Erk and GSK3ß pathway on embryo preimplantation development, early lineage segregation, and subsequent embryonic stem cell derivation in the humans. We did not observe any effect on blastocyst formation, but small-molecule inhibition did affect the number of OCT3/4- and NANOG-positive cells in the human ICM. We found that combined inhibition of the FGF/MEK/Erk and GSK3ß pathways by PD0325901 and CHIR99021, respectively, resulted in ICMs containing significantly more OCT3/4-positive cells. Inhibition of FGF/MEK/Erk alone as well as in combination with inhibition of GSK3ß significantly increased the number of NANOG-positive cells in blastocysts possessing good-quality ICMs. Secondly, we verified the influence of this increased pluripotency after 2i culture on the efficiency of stem cell derivation. Similar human embryonic stem cell (hESC) derivation rates were observed after 2i compared to control conditions, resulting in 2 control hESC lines and 1 hESC line from an embryo cultured in 2i conditions. In conclusion, we demonstrated that FGF/MEK/Erk and GSK3ß signaling increases the number of OCT3/4- and NANOG-positive cells in the human ICM, but does not improve stem cell derivation.


Assuntos
Massa Celular Interna do Blastocisto/metabolismo , Células-Tronco Embrionárias/citologia , Quinase 3 da Glicogênio Sintase/antagonistas & inibidores , Proteínas de Homeodomínio/metabolismo , Fator 3 de Transcrição de Octâmero/metabolismo , Transdução de Sinais , Benzamidas/farmacologia , Massa Celular Interna do Blastocisto/citologia , Massa Celular Interna do Blastocisto/efeitos dos fármacos , Contagem de Células , Técnicas de Cultura de Células , Linhagem da Célula , Difenilamina/análogos & derivados , Difenilamina/farmacologia , Técnicas de Cultura Embrionária , Implantação do Embrião/efeitos dos fármacos , Células-Tronco Embrionárias/metabolismo , Fator de Transcrição GATA6/genética , Fator de Transcrição GATA6/metabolismo , Quinase 3 da Glicogênio Sintase/genética , Quinase 3 da Glicogênio Sintase/metabolismo , Glicogênio Sintase Quinase 3 beta , Proteínas de Homeodomínio/genética , Humanos , Imuno-Histoquímica , Sistema de Sinalização das MAP Quinases , Quinases de Proteína Quinase Ativadas por Mitógeno/antagonistas & inibidores , Quinases de Proteína Quinase Ativadas por Mitógeno/genética , Quinases de Proteína Quinase Ativadas por Mitógeno/metabolismo , Proteína Homeobox Nanog , Fator 3 de Transcrição de Octâmero/genética , Piridinas/farmacologia , Pirimidinas/farmacologia , Fatores de Tempo
19.
Stem Cells Dev ; 20(5): 785-93, 2011 May.
Artigo em Inglês | MEDLINE | ID: mdl-20809773

RESUMO

Despite its prognostic value in in vitro fertilization, early embryo morphology is not reported on in the derivation of human embryonic stem cell (hESC) lines. Standard hESC derivation does rely on blastocyst development and its efficiency is highly correlated to inner cell mass (ICM) quality. Poor-quality embryos (PQEs) donated for hESC derivation may have a range of cleavage-stage abnormalities that are known to compromise further development. This study was implemented to determine whether specific PQEs traits influence the efficiency of good-quality ICMs to derive new hESC lines. We found that although the types of PQEs investigated were all able to make blastocysts with good-quality ICMs, the ICMs were unequal in their ability to derive hESCs. Good-quality ICMs from embryos with multiple poor-quality traits were unable to generate hESC lines, in contrast to good-quality ICMs from embryos with a single poor-quality trait. In addition, our data suggest a direct correlation between the number of ICM cells present in the blastocyst and its capacity to derive new hESC lines. This study is the first to demonstrate that ICM quality alone is an incomplete indicator of hESC derivation and that application of in vitro fertilization-based early embryo scoring can help predict hESC derivation efficiency. Experiments aiming to quantify, improve upon, or compare hESC derivation efficiency should thus take into consideration early embryo morphology scoring for the comparison of groups with equal developmental competence.


Assuntos
Massa Celular Interna do Blastocisto/fisiologia , Blastocisto/citologia , Embrião de Mamíferos/citologia , Células-Tronco Embrionárias/citologia , Projetos de Pesquisa , Massa Celular Interna do Blastocisto/citologia , Diferenciação Celular , Linhagem Celular , Técnicas de Cultura Embrionária , Desenvolvimento Embrionário , Células-Tronco Embrionárias/fisiologia , Feminino , Fertilização in vitro/métodos , Humanos , Cariotipagem , Herança Multifatorial
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...