Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 1.176
Filtrar
1.
Genome Res ; 34(3): 484-497, 2024 Apr 25.
Artigo em Inglês | MEDLINE | ID: mdl-38580401

RESUMO

Transcriptional regulation controls cellular functions through interactions between transcription factors (TFs) and their chromosomal targets. However, understanding the fate conversion potential of multiple TFs in an inducible manner remains limited. Here, we introduce iTF-seq as a method for identifying individual TFs that can alter cell fate toward specific lineages at a single-cell level. iTF-seq enables time course monitoring of transcriptome changes, and with biotinylated individual TFs, it provides a multi-omics approach to understanding the mechanisms behind TF-mediated cell fate changes. Our iTF-seq study in mouse embryonic stem cells identified multiple TFs that trigger rapid transcriptome changes indicative of differentiation within a day of induction. Moreover, cells expressing these potent TFs often show a slower cell cycle and increased cell death. Further analysis using bioChIP-seq revealed that GCM1 and OTX2 act as pioneer factors and activators by increasing gene accessibility and activating the expression of lineage specification genes during cell fate conversion. iTF-seq has utility in both mapping cell fate conversion and understanding cell fate conversion mechanisms.


Assuntos
Diferenciação Celular , Fatores de Transcrição , Animais , Camundongos , Fatores de Transcrição/metabolismo , Fatores de Transcrição/genética , Diferenciação Celular/genética , Análise de Célula Única/métodos , Células-Tronco Embrionárias Murinas/metabolismo , Células-Tronco Embrionárias Murinas/citologia , Linhagem da Célula/genética , Transcriptoma , Análise de Sequência de RNA/métodos , RNA-Seq/métodos , Perfilação da Expressão Gênica/métodos , RNA Citoplasmático Pequeno/genética , RNA Citoplasmático Pequeno/metabolismo , Multiômica , Análise da Expressão Gênica de Célula Única
2.
Cell Rep ; 43(4): 114024, 2024 Apr 23.
Artigo em Inglês | MEDLINE | ID: mdl-38581679

RESUMO

Mouse embryonic stem cells (mESCs) in the primed pluripotency state, which resembles the post-implantation epiblast, can be de-differentiated in culture to a naive state that resembles the pre-implantation inner cell mass. We report that primed-to-naive mESC transition entails a significant slowdown of DNA replication forks and the compensatory activation of dormant origins. Using isolation of proteins on nascent DNA coupled to mass spectrometry, we identify key changes in replisome composition that are responsible for these effects. Naive mESC forks are enriched in MRE11 nuclease and other DNA repair proteins. MRE11 is recruited to newly synthesized DNA in response to transcription-replication conflicts, and its inhibition or genetic downregulation in naive mESCs is sufficient to restore the fork rate of primed cells. Transcriptomic analyses indicate that MRE11 exonuclease activity is required for the complete primed-to-naive mESC transition, demonstrating a direct link between DNA replication dynamics and the mESC de-differentiation process.


Assuntos
Replicação do DNA , Proteína Homóloga a MRE11 , Animais , Camundongos , Proteína Homóloga a MRE11/metabolismo , Células-Tronco Embrionárias Murinas/metabolismo , Células-Tronco Embrionárias Murinas/citologia , Desdiferenciação Celular , Proteínas de Ligação a DNA/metabolismo
3.
Cell Rep ; 43(4): 114090, 2024 Apr 23.
Artigo em Inglês | MEDLINE | ID: mdl-38607915

RESUMO

Gene repression by the Polycomb pathway is essential for metazoan development. Polycomb domains, characterized by trimethylation of histone H3 lysine 27 (H3K27me3), carry the memory of repression and hence need to be maintained to counter the dilution of parental H3K27me3 with unmodified H3 during replication. Yet, how locus-specific H3K27me3 is maintained through replication is unclear. To understand H3K27me3 recovery post-replication, we first define nucleation sites within each Polycomb domain in mouse embryonic stem cells. To map dynamics of H3K27me3 domains across the cell cycle, we develop CUT&Flow (coupling cleavage under target and tagmentation with flow cytometry). We show that post-replication recovery of Polycomb domains occurs by nucleation and spreading, using the same nucleation sites used during de novo domain formation. By using Polycomb repressive complex 2 (PRC2) subunit-specific inhibitors, we find that PRC2 targets nucleation sites post-replication independent of pre-existing H3K27me3. Thus, competition between H3K27me3 deposition and nucleosome turnover drives both de novo domain formation and maintenance during every cell cycle.


Assuntos
Ciclo Celular , Histonas , Complexo Repressor Polycomb 2 , Animais , Camundongos , Histonas/metabolismo , Complexo Repressor Polycomb 2/metabolismo , Metilação , Células-Tronco Embrionárias Murinas/metabolismo , Células-Tronco Embrionárias Murinas/citologia , Proteínas do Grupo Polycomb/metabolismo , Proteínas do Grupo Polycomb/genética , Domínios Proteicos , Nucleossomos/metabolismo
4.
Dev Cell ; 59(8): 979-990.e5, 2024 Apr 22.
Artigo em Inglês | MEDLINE | ID: mdl-38458189

RESUMO

Argonaute (AGO) proteins are evolutionarily conserved RNA-binding proteins that control gene expression through the small RNAs they interact with. Whether AGOs have regulatory roles independent of RNAs, however, is unknown. Here, we show that AGO1 controls cell fate decisions through facilitating protein folding. We found that in mouse embryonic stem cells (mESCs), while AGO2 facilitates differentiation via the microRNA (miRNA) pathway, AGO1 controls stemness independently of its binding to small RNAs. We determined that AGO1 specifically interacts with HOP, a co-chaperone for the HSP70 and HSP90 chaperones, and enhances the folding of a set of HOP client proteins with intrinsically disordered regions. This AGO1-mediated facilitation of protein folding is important for maintaining stemness in mESCs. Our results demonstrate divergent functions between AGO1 and AGO2 in controlling cellular states and identify an RNA-independent function of AGO1 in controlling gene expression and cell fate decisions.


Assuntos
Proteínas Argonautas , Diferenciação Celular , Células-Tronco Embrionárias Murinas , Dobramento de Proteína , Animais , Proteínas Argonautas/metabolismo , Proteínas Argonautas/genética , Camundongos , Células-Tronco Embrionárias Murinas/metabolismo , Células-Tronco Embrionárias Murinas/citologia , MicroRNAs/genética , MicroRNAs/metabolismo , Fatores de Iniciação em Eucariotos/metabolismo , Fatores de Iniciação em Eucariotos/genética , Proteínas de Choque Térmico HSP90/metabolismo , Proteínas de Choque Térmico HSP90/genética , Proteínas de Ligação a RNA/metabolismo , Proteínas de Ligação a RNA/genética , Linhagem da Célula
5.
FEBS Lett ; 598(8): 915-934, 2024 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-38408774

RESUMO

The development of embryonic stem (ES) cells to extraembryonic trophectoderm and primitive endoderm lineages manifests distinct steady-state expression patterns of two key transcription factors-Oct4 and Nanog. How dynamically such kind of steady-state expressions are maintained remains elusive. Herein, we demonstrate that steady-state dynamics involving two bistable switches which are interlinked via a stepwise (Oct4) and a mushroom-like (Nanog) manner orchestrate the fate specification of ES cells. Our hypothesis qualitatively reconciles various experimental observations and elucidates how different feedback and feedforward motifs orchestrate the extraembryonic development and stemness maintenance of ES cells. Importantly, the model predicts strategies to optimize the dynamics of self-renewal and differentiation of embryonic stem cells that may have therapeutic relevance in the future.


Assuntos
Diferenciação Celular , Células-Tronco Embrionárias , Proteína Homeobox Nanog , Fator 3 de Transcrição de Octâmero , Proteína Homeobox Nanog/metabolismo , Proteína Homeobox Nanog/genética , Animais , Fator 3 de Transcrição de Octâmero/metabolismo , Fator 3 de Transcrição de Octâmero/genética , Células-Tronco Embrionárias/metabolismo , Células-Tronco Embrionárias/citologia , Camundongos , Linhagem da Célula/genética , Modelos Biológicos , Proteínas de Homeodomínio/metabolismo , Proteínas de Homeodomínio/genética , Regulação da Expressão Gênica no Desenvolvimento , Células-Tronco Embrionárias Murinas/metabolismo , Células-Tronco Embrionárias Murinas/citologia
6.
Cell Mol Biol Lett ; 28(1): 67, 2023 Aug 17.
Artigo em Inglês | MEDLINE | ID: mdl-37592237

RESUMO

BACKGROUND: Prenatal infection has been implicated in the development of neuropsychiatric disorders in children. We hypothesised that exposure to lipopolysaccharide during prenatal development could induce anxiety-like behaviour and sensorineural hearing loss in offspring, as well as disrupt neural differentiation during embryonic neural development. METHODS: We simulated prenatal infection in FVB mice and mouse embryonic stem cell (ESC) lines, specifically 46C and E14Tg2a, through lipopolysaccharide treatment. Gene expression profiling analyses and behavioural tests were utilized to study the effects of lipopolysaccharide on the offspring and alterations in toll-like receptor (TLR) 2-positive and TLR4-positive cells during neural differentiation in the ESCs. RESULTS: Exposure to lipopolysaccharide (25 µg/kg) on gestation day 9 resulted in anxiety-like behaviour specifically in male offspring, while no effects were detected in female offspring. We also found significant increases in the expression of GFAP and CNPase, as well as higher numbers of GFAP + astrocytes and O4+ oligodendrocytes in the prefrontal cortex of male offspring. Furthermore, increased scores for genes related to oligodendrocyte and lipid metabolism, particularly ApoE, were observed in the prefrontal cortex regions. Upon exposure to lipopolysaccharide during the ESC-to-neural stem cell (NSC) transition, Tuj1, Map2, Gfap, O4, and Oligo2 mRNA levels increased in the differentiated neural cells on day 14. In vitro experiments demonstrated that lipopolysaccharide exposure induced inflammatory responses, as evidenced by increased expression of IL1b and ApoB mRNA. CONCLUSIONS: Our findings suggest that prenatal infection at different stages of neural differentiation may result in distinct disturbances in neural differentiation during ESC-NSC transitions. Furthermore, early prenatal challenges with lipopolysaccharide selectively induce anxiety-like behaviour in male offspring. This behaviour may be attributed to the abnormal differentiation of astrocytes and oligodendrocytes in the brain, potentially mediated by ApoB/E signalling pathways in response to inflammatory stimuli.


Assuntos
Ansiedade , Células-Tronco Embrionárias Murinas , Células-Tronco Neurais , Feminino , Animais , Camundongos , Lipopolissacarídeos/toxicidade , Gravidez , Células-Tronco Embrionárias Murinas/citologia , Ansiedade/induzido quimicamente , Células-Tronco Neurais/citologia , Diferenciação Celular , Masculino , Comportamento Animal
7.
Sci Rep ; 12(1): 11482, 2022 07 07.
Artigo em Inglês | MEDLINE | ID: mdl-35798792

RESUMO

Epithelial development starts with stem cell commitment to ectoderm followed by differentiation to the basal keratinocytes. The basal keratinocytes, first committed in embryogenesis, constitute the basal layer of the epidermis. They have robust proliferation and differentiation potential and are responsible for epidermal expansion, maintenance and regeneration. We generated basal epithelial cells in vitro through differentiation of mouse embryonic stem cells (mESCs). Early on in differentiation, the expression of stem cell markers, Oct4 and Nanog, decreased sharply along with increased ectoderm marker keratin (Krt) 18. Later on, Krt 18 expression was subdued when cells displayed basal keratinocyte characteristics, including regular polygonal shape, adherent and tight junctions and Krt 14 expression. These cells additionally expressed abundant Sca-1, Krt15 and p63, suggesting epidermal progenitor characteristics. Using Map3k1 mutant mESCs and environmental dioxin, we examined the gene and environment effects on differentiation. Neither Map3k1 mutation nor dioxin altered mESC differentiation to ectoderm and basal keratinocytes, but they, individually and in combination, potentiated Krt 1 expression and basal to spinous differentiation. Similar gene-environment effects were observed in vivo where dioxin exposure increased Krt 1 more substantially in the epithelium of Map3k1+/- than wild type embryos. Thus, the in vitro model of epithelial differentiation can be used to investigate the effects of genetic and environmental factors on epidermal development.


Assuntos
Dioxinas , Queratinócitos , MAP Quinase Quinase Quinase 1 , Células-Tronco Embrionárias Murinas , Animais , Diferenciação Celular , Dioxinas/farmacologia , Células Epidérmicas , Epiderme/metabolismo , Queratinócitos/citologia , Queratinócitos/efeitos dos fármacos , MAP Quinase Quinase Quinase 1/genética , Camundongos , Células-Tronco Embrionárias Murinas/citologia , Mutação
8.
Pancreas ; 51(4): 330-337, 2022 04 01.
Artigo em Inglês | MEDLINE | ID: mdl-35695761

RESUMO

OBJECTIVES: Embryonic stem cells (ESCs)-derived pancreatic precursor cells have great potential for pancreas repair. Expression of pancreatic duodenal homeobox 1 (Pdx1) in definitive endoderm (DE) cells is the premise that DE cells differentiate into pancreatic cells. To achieve the required number of Pdx1-expressing DE cells for cell transplantation therapy, a valid model must be established. Using this model, researchers investigated how Pdx1 regulates ESC differentiation into pancreatic cells. METHODS: Tet-On inducible lentiviral vector encoding Pdx1 or mock vector was transduced into mouse ESC (ES-E14TG2a). The mouse ESCs were divided into 3 groups: control (ESC), mock vector (Pdx1 - -ESC), and vector encoding Pdx1 (Pdx1 + -ESC). All groups were separately cocultured with the DE cells sorted by immune beads containing CXCR-4 + (C-X-C chemokine receptor type-4) antibody. Doxycycline induced the expression of Pdx1 on the Pdx1 + -ESC cells. The markers of cell differentiation and Notch pathway were examined. RESULTS: Significantly increased expression levels of Ptf1a, CK19, and amylase on day (d) 3 and d7, Neuro-D1 on d10 and d14, Pax6 and insulin on d14, as well as Notch1, Notch2, Hes1, and Hes5 on d3 and thereafter declined on d14 were observed in Pdx1 + -ESC group. CONCLUSIONS: Pdx1 + -ESC could differentiate into pancreatic-like cells with involvement of the Notch pathway.


Assuntos
Endoderma , Proteínas de Homeodomínio , Células-Tronco Embrionárias Murinas , Pâncreas , Transativadores , Animais , Diferenciação Celular , Endoderma/citologia , Proteínas de Homeodomínio/genética , Proteínas de Homeodomínio/metabolismo , Camundongos , Células-Tronco Embrionárias Murinas/citologia , Pâncreas/citologia , Receptores Notch/metabolismo , Transativadores/genética , Transativadores/metabolismo
9.
Differentiation ; 125: 18-26, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35349880

RESUMO

Mouse embryonic stem cells (mESCs) are characterized by self-renewal and pluripotency and can undergo differentiation into the three germ layers (ectoderm, mesoderm, and endoderm). Melanoma-associated antigen D1 (Maged1), which is expressed in all developing and adult tissues, modulates tissue regeneration and development. In the present study, we examined the expression and function of Maged1 in mESCs. Maged1 protein and mRNA expression increased during mESC differentiation. The pluripotency of mESCs was significantly reduced through extracellular signal-regulated kinase 1/2 phosphorylation upon knockdown of Maged1, and through G1 cell cycle arrest during cell division, resulting in significantly reduced mESC proliferation. Moreover, the diameter of the embryoid bodies was significantly reduced, accompanied by increased levels of ectodermal differentiation markers and decreased levels of mesodermal and endodermal differentiation markers. Maged1-knockdown mESC lines showed significantly reduced teratoma volumes and inhibition of teratoma formation in nude mice. Additionally, we observed increased ectodermal markers but decreased mesodermal and endodermal markers in teratoma tissues. These findings show that Maged1 affects mESC pluripotency, proliferation, cell cycle, and differentiation, thereby contributing to our understanding of the basic molecular biological mechanisms and potential roles of Maged1 as a regulator of various mESC properties.


Assuntos
Células-Tronco Embrionárias Murinas , Animais , Antígenos de Diferenciação/metabolismo , Ciclo Celular/genética , Morte Celular , Diferenciação Celular/genética , Divisão Celular , Camundongos , Camundongos Nus , Células-Tronco Embrionárias Murinas/citologia , Células-Tronco Embrionárias Murinas/metabolismo , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/metabolismo , Teratoma/genética , Teratoma/metabolismo , Teratoma/patologia
10.
Development ; 149(4)2022 02 15.
Artigo em Inglês | MEDLINE | ID: mdl-35175328

RESUMO

Signal transduction networks generate characteristic dynamic activities to process extracellular signals and guide cell fate decisions such as to divide or differentiate. The differentiation of pluripotent cells is controlled by FGF/ERK signaling. However, only a few studies have addressed the dynamic activity of the FGF/ERK signaling network in pluripotent cells at high time resolution. Here, we use live cell sensors in wild-type and Fgf4-mutant mouse embryonic stem cells to measure dynamic ERK activity in single cells, for defined ligand concentrations and differentiation states. These sensors reveal pulses of ERK activity. Pulsing patterns are heterogeneous between individual cells. Consecutive pulse sequences occur more frequently than expected from simple stochastic models. Sequences become more prevalent with higher ligand concentration, but are rarer in more differentiated cells. Our results suggest that FGF/ERK signaling operates in the vicinity of a transition point between oscillatory and non-oscillatory dynamics in embryonic stem cells. The resulting heterogeneous dynamic signaling activities add a new dimension to cellular heterogeneity that may be linked to divergent fate decisions in stem cell cultures.


Assuntos
MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Fatores de Crescimento de Fibroblastos/metabolismo , Animais , Caderinas/metabolismo , Ciclo Celular , Fator 4 de Crescimento de Fibroblastos/genética , Fator 4 de Crescimento de Fibroblastos/metabolismo , Camundongos , Quinases de Proteína Quinase Ativadas por Mitógeno/antagonistas & inibidores , Quinases de Proteína Quinase Ativadas por Mitógeno/metabolismo , Células-Tronco Embrionárias Murinas/citologia , Células-Tronco Embrionárias Murinas/metabolismo , Proteínas Recombinantes/biossíntese , Proteínas Recombinantes/isolamento & purificação , Proteínas Recombinantes/farmacologia , Transdução de Sinais/efeitos dos fármacos
11.
Int J Mol Sci ; 23(3)2022 Jan 20.
Artigo em Inglês | MEDLINE | ID: mdl-35163031

RESUMO

Organic semiconductors are constantly gaining interest in regenerative medicine. Their tunable physico-chemical properties, including electrical conductivity, are very promising for the control of stem-cell differentiation. However, their use for combined material-based and electrical stimulation remains largely underexplored. Therefore, we carried out a study on whether a platform based on the conductive polymer poly(3,4-ethylenedioxythiophene):polystyrene sulfonate (PEDOT:PSS) can be beneficial to the differentiation of mouse embryonic stem cells (mESCs). The platform was prepared using the layout of a standard 24-well cell-culture plate. Polyethylene naphthalate foil served as the substrate for the preparation of interdigitated gold electrodes by physical vapor deposition. The PEDOT:PSS pattern was fabricated by precise screen printing over the gold electrodes. The PEDOT:PSS platform was able to produce higher electrical current with the pulsed-direct-current (DC) electrostimulation mode (1 Hz, 200 mV/mm, 100 ms pulse duration) compared to plain gold electrodes. There was a dominant capacitive component. In proof-of-concept experiments, mESCs were able to respond to such electrostimulation by membrane depolarization and elevation of cytosolic calcium. Further, the PEDOT:PSS platform was able to upregulate cardiomyogenesis and potentially inhibit early neurogenesis per se with minor contribution of electrostimulation. Hence, the present work highlights the large potential of PEDOT:PSS in regenerative medicine.


Assuntos
Compostos Bicíclicos Heterocíclicos com Pontes/química , Diferenciação Celular , Condutividade Elétrica , Células-Tronco Embrionárias Murinas/citologia , Polímeros/farmacologia , Poliestirenos/química , Animais , Técnicas de Cultura de Células , Eletrodos , Camundongos , Células-Tronco Embrionárias Murinas/efeitos dos fármacos , Polímeros/química
12.
Biochem Biophys Res Commun ; 599: 156-163, 2022 04 09.
Artigo em Inglês | MEDLINE | ID: mdl-35202849

RESUMO

Primitive Endoderm (PrE) is an extraembryonic structure derived from inner cell mass (ICM) in the blastocysts. Its interaction with the epiblast is critical to sustain embryonic growth and embryonic pattern. In this study, we reported a simple and efficient method to induce the differentiation of mouse Embryonic Stem Cells (mESCs) into PrE cells. In the process of ESC monolayer adherent culture, 1 µM atRA and 10 µM CHIR inducers were used to activate RA and Wnt signaling pathways respectively. After 9 days of differentiation, the proportion of PrE cells was up to 85%. Further studies indicated that Wnt signaling pathway acted as a switch that RA induces mESCs differentiation between SMC and PrE cell. In the presence of only RA signaling, mESCs adopted the fate of smooth muscle cells (SMCs); Simultaneous activation of the Wnt signaling pathway changed the differentiation fate of mESCs into PrE cells. This efficient induction method can provide new cellular resources and models for relevant studies of PrE.


Assuntos
Diferenciação Celular/efeitos dos fármacos , Endoderma/citologia , Células-Tronco Embrionárias Murinas/citologia , Animais , Diferenciação Celular/fisiologia , Células Cultivadas , Regulação da Expressão Gênica/efeitos dos fármacos , Camundongos , Células-Tronco Embrionárias Murinas/fisiologia , Piridinas/farmacologia , Pirimidinas/farmacologia , Tretinoína/farmacologia , Via de Sinalização Wnt/efeitos dos fármacos
13.
Development ; 149(3)2022 02 01.
Artigo em Inglês | MEDLINE | ID: mdl-35147187

RESUMO

Corticogenesis consists of a series of synchronised events, including fate transition of cortical progenitors, neuronal migration, specification and connectivity. NeuroD1, a basic helix-loop-helix (bHLH) transcription factor (TF), contributes to all of these events, but how it coordinates these independently is still unknown. Here, we demonstrate that NeuroD1 expression is accompanied by a gain of active chromatin at a large number of genomic loci. Interestingly, transcriptional activation of these loci relied on a high local density of adjacent bHLH TFs motifs, including, predominantly, Tcf12. We found that activity and expression levels of Tcf12 were high in cells with induced levels of NeuroD1 that spanned the transition of cortical progenitors from proliferative to neurogenic divisions. Moreover, Tcf12 forms a complex with NeuroD1 and co-occupies a subset of NeuroD1 target loci. This Tcf12-NeuroD1 cooperativity is essential for gaining active chromatin and targeted expression of genes involved in cell migration. By functional manipulation in vivo, we further show that Tcf12 is essential during cortical development for the correct migration of newborn neurons and, hence, for proper cortical lamination.


Assuntos
Fatores de Transcrição Hélice-Alça-Hélice Básicos/metabolismo , Córtex Cerebral/crescimento & desenvolvimento , Animais , Fatores de Transcrição Hélice-Alça-Hélice Básicos/antagonistas & inibidores , Fatores de Transcrição Hélice-Alça-Hélice Básicos/genética , Movimento Celular , Córtex Cerebral/metabolismo , Cromatina/metabolismo , Desenvolvimento Embrionário/genética , Feminino , Histonas/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Células-Tronco Embrionárias Murinas/citologia , Células-Tronco Embrionárias Murinas/metabolismo , Neurogênese , Neurônios/citologia , Neurônios/metabolismo , Interferência de RNA , RNA Interferente Pequeno/metabolismo
14.
Protein Cell ; 13(8): 580-601, 2022 08.
Artigo em Inglês | MEDLINE | ID: mdl-35147915

RESUMO

Chemically defined medium is widely used for culturing mouse embryonic stem cells (mESCs), in which N2B27 works as a substitution for serum, and GSK3ß and MEK inhibitors (2i) help to promote ground-state pluripotency. However, recent studies suggested that MEKi might cause irreversible defects that compromise the developmental potential of mESCs. Here, we demonstrated the deficient bone morphogenetic protein (BMP) signal in the chemically defined condition is one of the main causes for the impaired pluripotency. Mechanistically, activating the BMP signal pathway by BMP4 could safeguard the chromosomal integrity and proliferation capacity of mESCs through regulating downstream targets Ube2s and Chmp4b. More importantly, BMP4 promotes a distinct in vivo developmental potential and a long-term pluripotency preservation. Besides, the pluripotent improvements driven by BMP4 are superior to those by attenuating MEK suppression. Taken together, our study shows appropriate activation of BMP signal is essential for regulating functional pluripotency and reveals that BMP4 should be applied in the serum-free culture system.


Assuntos
Proteína Morfogenética Óssea 4 , Células-Tronco Embrionárias Murinas , Células-Tronco Pluripotentes , Animais , Proteína Morfogenética Óssea 4/metabolismo , Diferenciação Celular , Instabilidade Cromossômica , Complexos Endossomais de Distribuição Requeridos para Transporte , Camundongos , Quinases de Proteína Quinase Ativadas por Mitógeno/metabolismo , Células-Tronco Embrionárias Murinas/citologia , Células-Tronco Pluripotentes/citologia , Transdução de Sinais , Enzimas de Conjugação de Ubiquitina
15.
Sci Rep ; 12(1): 2364, 2022 02 11.
Artigo em Inglês | MEDLINE | ID: mdl-35149723

RESUMO

We have previously reported that RING1 and YY1 binding protein (RYBP) is important for central nervous system development in mice and that Rybp null mutant (Rybp-/-) mouse embryonic stem (ES) cells form more progenitors and less terminally differentiated neural cells than the wild type cells in vitro. Accelerated progenitor formation coincided with a high level of Pax6 expression in the Rybp-/- neural cultures. Since Pax6 is a retinoic acid (RA) inducible gene, we have analyzed whether altered RA signaling contributes to the accelerated progenitor formation and impaired differentiation ability of the Rybp-/- cells. Results suggested that elevated Pax6 expression was driven by the increased activity of the RA signaling pathway in the Rybp-/- neural cultures. RYBP was able to repress Pax6 through its P1 promoter. The repression was further attenuated when RING1, a core member of ncPRC1s was also present. According to this, RYBP and PAX6 were rarely localized in the same wild type cells during in vitro neural differentiation. These results suggest polycomb dependent regulation of Pax6 by RYBP during in vitro neural differentiation. Our results thus provide novel insights on the dynamic regulation of Pax6 and RA signaling by RYBP during mouse neural development.


Assuntos
Células-Tronco Embrionárias Murinas/metabolismo , Tecido Nervoso/embriologia , Tecido Nervoso/metabolismo , Neurogênese , Fator de Transcrição PAX6/metabolismo , Proteínas Repressoras/metabolismo , Animais , Regulação da Expressão Gênica no Desenvolvimento , Camundongos , Camundongos Knockout , Células-Tronco Embrionárias Murinas/citologia , Fator de Transcrição PAX6/genética , Proteínas Repressoras/genética , Tretinoína/metabolismo
16.
Arterioscler Thromb Vasc Biol ; 42(3): 326-342, 2022 03.
Artigo em Inglês | MEDLINE | ID: mdl-35021856

RESUMO

BACKGROUND: Endothelial cells (ECs) play a critical role in angiogenesis and vascular remodeling. The heterogeneity of ECs has been reported at adult stages, yet it has not been fully investigated. This study aims to assess the transcriptional heterogeneity of developmental ECs at spatiotemporal level and to reveal the changes of embryonic ECs clustering when endothelium-enriched microRNA-126 (miR-126) was specifically knocked out. METHODS: C57BL/6J mice embryos at day 14.5 were harvested and digested, followed by fluorescence-activated cell sorting to enrich ECs. Then, single-cell RNA sequencing was applied to enriched embryonic ECs. Tie2 (Tek receptor tyrosine kinase)-cre-mediated ECs-specific miR-126 knockout mice were constructed, and ECs from Tie2-cre-mediated ECs-specific miR-126 knockout embryos were subjected to single-cell RNA sequencing. RESULTS: Embryonic ECs were clustered into 11 groups corresponding to anatomic characteristics. The vascular bed (arteries, capillaries, veins, lymphatics) exhibited transcriptomic similarity across the developmental stage. Embryonic ECs had higher proliferative potential than adult ECs. Integrating analysis showed that 3 ECs populations (hepatic, mesenchymal transition, and pulmonary ECs) were apparently disorganized after miR-126 being knocked out. Gene ontology analysis revealed that disrupted ECs were mainly related to hypoxia, glycometabolism, and vascular calcification. Additionally, in vivo experiment showed that Tie2-cre-mediated ECs-specific miR-126 knockout mice exhibited excessive intussusceptive angiogenesis; reductive glucose and pyruvate tolerance; and excessive accumulation of calcium. Agonist miR-126-3p agomir significantly rescued the phenotype of glucose metabolic dysfunction in Tie2-cre-mediated ECs-specific miR-126 knockout mice. CONCLUSIONS: The heterogeneity of ECs is established as early as the embryonic stage. The deficiency of miR-126 disrupts the differentiation and diversification of embryonic ECs, suggesting that miR-126 plays an essential role in the maintenance of ECs heterogeneity.


Assuntos
Células Endoteliais/citologia , Células Endoteliais/metabolismo , MicroRNAs/genética , Células-Tronco Embrionárias Murinas/citologia , Células-Tronco Embrionárias Murinas/metabolismo , Animais , Apoptose/genética , Hipóxia Celular/genética , Linhagem da Célula/genética , Plasticidade Celular/genética , Proliferação de Células/genética , Células Endoteliais/classificação , Perfilação da Expressão Gênica , Regulação da Expressão Gênica no Desenvolvimento , Idade Gestacional , Glucose/metabolismo , Fígado/irrigação sanguínea , Fígado/embriologia , Fígado/metabolismo , Redes e Vias Metabólicas/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , MicroRNAs/antagonistas & inibidores , MicroRNAs/metabolismo , Células-Tronco Embrionárias Murinas/classificação , Neovascularização Fisiológica/genética , Análise de Célula Única , Análise Espaço-Temporal , Calcificação Vascular/genética , Calcificação Vascular/metabolismo , Calcificação Vascular/patologia
17.
PLoS One ; 17(1): e0261950, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-34995303

RESUMO

Mouse embryonic stem cells (mESCs) can be manipulated in vitro to recapitulate the process of erythropoiesis, during which multipotent cells undergo lineage specification, differentiation and maturation to produce erythroid cells. Although useful for identifying specific progenitors and precursors, this system has not been fully exploited as a source of cells to analyse erythropoiesis. Here, we establish a protocol in which characterised erythroblasts can be isolated in a scalable manner from differentiated embryoid bodies (EBs). Using transcriptional and epigenetic analysis, we demonstrate that this system faithfully recapitulates normal primitive erythropoiesis and fully reproduces the effects of natural and engineered mutations seen in primary cells obtained from mouse models. We anticipate this system to be of great value in reducing the time and costs of generating and maintaining mouse lines in a number of research scenarios.


Assuntos
Diferenciação Celular , Corpos Embrioides/metabolismo , Eritroblastos/metabolismo , Eritropoese , Modelos Biológicos , Células-Tronco Embrionárias Murinas/metabolismo , Animais , Linhagem Celular , Corpos Embrioides/citologia , Eritroblastos/citologia , Camundongos , Células-Tronco Embrionárias Murinas/citologia
18.
Stem Cell Reports ; 17(2): 427-442, 2022 02 08.
Artigo em Inglês | MEDLINE | ID: mdl-35090587

RESUMO

Elucidating regulatory relationships between transcription factors (TFs) and target genes is fundamental to understanding how cells control their identity and behavior. Unfortunately, existing computational gene regulatory network (GRN) reconstruction methods are imprecise, computationally burdensome, and fail to reveal dynamic regulatory topologies. Here, we present Epoch, a reconstruction tool that uses single-cell transcriptomics to accurately infer dynamic networks. We apply Epoch to identify the dynamic networks underpinning directed differentiation of mouse embryonic stem cells (ESCs) guided by multiple signaling pathways, and we demonstrate that modulating these pathways drives topological changes that bias cell fate potential. We also find that Peg3 rewires the pluripotency network to favor mesoderm specification. By integrating signaling pathways with GRNs, we trace how Wnt activation and PI3K suppression govern mesoderm and endoderm specification, respectively. Finally, we identify regulatory circuits of patterning and axis formation that distinguish in vitro and in vivo mesoderm specification.


Assuntos
Redes Reguladoras de Genes/genética , Camadas Germinativas/metabolismo , Animais , Diferenciação Celular , Endoderma/citologia , Endoderma/metabolismo , Camadas Germinativas/citologia , Fatores de Transcrição Kruppel-Like/genética , Fatores de Transcrição Kruppel-Like/metabolismo , Mesoderma/citologia , Mesoderma/metabolismo , Camundongos , Células-Tronco Embrionárias Murinas/citologia , Células-Tronco Embrionárias Murinas/metabolismo , Fosfatidilinositol 3-Quinases/metabolismo , Transdução de Sinais/genética , Análise de Célula Única , Proteínas Wnt/metabolismo
19.
Stem Cell Reports ; 17(2): 231-244, 2022 02 08.
Artigo em Inglês | MEDLINE | ID: mdl-35063128

RESUMO

The formation of the primitive streak (PS) and the subsequent induction of neuroectoderm are hallmarks of gastrulation. Combining an in vitro reconstitution of this process based on mouse embryonic stem cells (mESCs) with a collection of knockouts in reporter mESC lines, we identified retinoic acid (RA) as a critical mediator of early neural induction triggered by TGFß or Wnt signaling inhibition. Single-cell RNA sequencing analysis captured the temporal unfolding of cell type diversification, up to the emergence of somite and neural fates. In the absence of the RA-synthesizing enzyme Aldh1a2, a sensitive RA reporter revealed a hitherto unidentified residual RA signaling that specified neural fate. Genetic evidence showed that the RA-degrading enzyme Cyp26a1 protected PS-like cells from neural induction, even in the absence of TGFß and Wnt antagonists. Overall, we characterized a multi-layered control of RA levels that regulates early neural differentiation in an in vitro PS-like system.


Assuntos
Diferenciação Celular/efeitos dos fármacos , Neurônios/metabolismo , Tretinoína/farmacologia , Família Aldeído Desidrogenase 1/deficiência , Família Aldeído Desidrogenase 1/genética , Animais , Benzamidas/farmacologia , Dioxóis/farmacologia , Ectoderma/citologia , Ectoderma/metabolismo , Camundongos , Células-Tronco Embrionárias Murinas/citologia , Células-Tronco Embrionárias Murinas/efeitos dos fármacos , Células-Tronco Embrionárias Murinas/metabolismo , Neurônios/citologia , Linha Primitiva/citologia , Linha Primitiva/metabolismo , Retinal Desidrogenase/deficiência , Retinal Desidrogenase/genética , Ácido Retinoico 4 Hidroxilase/metabolismo , Transdução de Sinais/efeitos dos fármacos , Tretinoína/metabolismo
20.
Biochem Biophys Res Commun ; 590: 125-131, 2022 01 29.
Artigo em Inglês | MEDLINE | ID: mdl-34974300

RESUMO

Embryonic stem cells (ESCs) are derived from the inner cell mass of developing blastocysts, which have self-renewal ability and have the potential to develop or reconstitute into all embryonic lineages. Selenophosphate synthetase 1 (SEPHS1) is an essential protein in mouse early embryo development. However, the role of SEPHS1 in mouse ESCs remains to be elucidated. In this study, we generated Sephs1 KO ESCs and found that deficiency of SEPSH1 has little effect on pluripotency maintenance and proliferation. Notably, SEPHS1 deficiency impaired differentiation into three germ layers and gastruloid aggregation in vitro. RNA-seq analysis showed SEPHS1 is involved in cardiogenesis, verified by no beating signal in Sephs1 KO embryoid body at d10 and low expression of cardiac-related and contraction markers. Taken together, our results suggest that SPEHS1 is dispensable in ESC self-renewal, but indispensable in subsequent germ layer differentiation especially for functional cardiac lineage.


Assuntos
Diferenciação Celular , Células-Tronco Embrionárias Murinas/citologia , Células-Tronco Embrionárias Murinas/metabolismo , Miocárdio/citologia , Fosfotransferases/metabolismo , Animais , Diferenciação Celular/genética , Corpos Embrioides/citologia , Gastrulação/genética , Regulação da Expressão Gênica no Desenvolvimento , Camadas Germinativas/citologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Fosfotransferases/deficiência , Transcrição Gênica
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...