human embryonic stem cells

人胚胎干细胞
  • 文章类型: Journal Article
    背景:咽内胚层(PE)是一种极其相关的发育组织,作为食道的祖先,甲状旁腺,甲状腺,肺,还有胸腺.虽然一些研究强调了PE细胞的重要性,这一重要发育阶段的详细转录和表观遗传特征仍然缺失,尤其是在人类中,由于其早期形成的技术和道德限制。
    结果:在这里,我们通过开发从人类胚胎干细胞(hESC)衍生PE样细胞的体外方案,并通过提供综合的多组学表征来填补这一知识空白。我们的PE样细胞强健地表达PE标志物,并且在转录上是同质的,并且与体内小鼠PE细胞相似。此外,我们通过结合ATAC-Seq和ChIP-Seq组蛋白修饰来定义它们的表观遗传景观和响应维甲酸的动态变化。多个高通量数据集的整合导致新的推定调控区的鉴定和以视黄酸为中心的转录因子网络的推断,从而协调PE样细胞的发育。
    结论:通过将hESCs分化与计算基因组学相结合,我们的工作揭示了人类PE分化过程中发生的表观遗传动力学,为专注于PE衍生物的开发及其在遗传综合征中的发育缺陷建模的研究提供了坚实的资源和基础。
    BACKGROUND: The Pharyngeal Endoderm (PE) is an extremely relevant developmental tissue, serving as the progenitor for the esophagus, parathyroids, thyroids, lungs, and thymus. While several studies have highlighted the importance of PE cells, a detailed transcriptional and epigenetic characterization of this important developmental stage is still missing, especially in humans, due to technical and ethical constraints pertaining to its early formation.
    RESULTS: Here we fill this knowledge gap by developing an in vitro protocol for the derivation of PE-like cells from human Embryonic Stem Cells (hESCs) and by providing an integrated multi-omics characterization. Our PE-like cells robustly express PE markers and are transcriptionally homogenous and similar to in vivo mouse PE cells. In addition, we define their epigenetic landscape and dynamic changes in response to Retinoic Acid by combining ATAC-Seq and ChIP-Seq of histone modifications. The integration of multiple high-throughput datasets leads to the identification of new putative regulatory regions and to the inference of a Retinoic Acid-centered transcription factor network orchestrating the development of PE-like cells.
    CONCLUSIONS: By combining hESCs differentiation with computational genomics, our work reveals the epigenetic dynamics that occur during human PE differentiation, providing a solid resource and foundation for research focused on the development of PE derivatives and the modeling of their developmental defects in genetic syndromes.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Journal Article
    OSGEP基因编码O-唾液酸糖蛋白内肽酶,高度保守的KEOPS复合物的催化单元(激酶,内肽酶,和其他小尺寸蛋白质),可调节N-6-苏酰基氨基甲酰基腺苷(t6A)形成中的第二个生物合成步骤。KEOPS突变导致Galloway-Mowat综合征(GAMOS),其在哺乳动物中的细胞功能和潜在的分子机制尚不清楚。在这项研究中,我们利用慢病毒介导的OSGEP敲低产生OSGEP缺陷型人胚胎干细胞(hESCs).OSGEP敲低hESC表现出干性因子表达降低和G2/M期阻滞,表明OSGEP在调节hESC命运中的潜在作用。此外,OSGEP沉默导致蛋白质合成增强和蛋白质聚集增加,这进一步诱导了不适当的自噬,如P62的表达改变和LC3-I向LC3-II的转化所证明的。上述发现揭示了OSGEP在调节hESCs多能性和分化中的潜在参与,同时强调了其在维持蛋白质平衡和自噬中的关键作用。这可能对人类疾病有影响。
    The OSGEP gene encodes O-sialoglycoprotein endopeptidase, a catalytic unit of the highly conserved KEOPS complex (Kinase, Endopeptidase, and Other Proteins of small Size) that regulates the second biosynthetic step in the formation of N-6-threonylcarbamoyladenosine (t6A). Mutations in KEOPS cause Galloway-Mowat syndrome (GAMOS), whose cellular function in mammals and underlying molecular mechanisms are not well understood. In this study, we utilized lentivirus-mediated OSGEP knockdown to generate OSGEP-deficient human embryonic stem cells (hESCs). OSGEP-knockdown hESCs exhibited reduced stemness factor expression and G2/M phase arrest, indicating a potential role of OSGEP in the regulation of hESC fate. Additionally, OSGEP silencing led to enhanced protein synthesis and increased aggregation of proteins, which further induced inappropriate autophagy, as evidenced by the altered expression of P62 and the conversion of LC3-I to LC3-II. The above findings shed light on the potential involvement of OSGEP in regulating pluripotency and differentiation in hESCs while simultaneously highlighting its crucial role in maintaining proteostasis and autophagy, which may have implications for human disease.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Journal Article
    选择性5-羟色胺再摄取抑制剂(SSRIs),包括西酞普兰,是怀孕期间广泛使用的抗抑郁药。然而,产前暴露于西酞普兰对神经发育的影响仍然知之甚少。我们旨在使用多组学方法研究西酞普兰暴露对人胚胎干细胞早期神经元分化的影响。西酞普兰对参与神经发育过程或与抑郁症相关的基因表达和DNA甲基化的时间和剂量依赖性影响,比如BDNF,GDF11、CCL2、STC1、DDIT4和GAD2。单细胞RNA测序分析揭示了不同的干细胞簇,神经元祖细胞和成神经细胞,暴露于西酞普兰会微妙地影响祖先亚型。假颞叶分析显示神经元分化增强。我们的发现表明,早期神经元分化过程中的西酞普兰暴露会影响与神经发育和抑郁症相关的基因表达模式。提供对其潜在神经发育影响的见解,并强调进一步研究以了解产前SSRI暴露的长期后果的重要性。
    Selective serotonin reuptake inhibitors (SSRIs), including citalopram, are widely used antidepressants during pregnancy. However, the effects of prenatal exposure to citalopram on neurodevelopment remain poorly understood. We aimed to investigate the impact of citalopram exposure on early neuronal differentiation of human embryonic stem cells using a multi-omics approach. Citalopram induced time- and dose-dependent effects on gene expression and DNA methylation of genes involved in neurodevelopmental processes or linked to depression, such as BDNF, GDF11, CCL2, STC1, DDIT4 and GAD2. Single-cell RNA-sequencing analysis revealed distinct clusters of stem cells, neuronal progenitors and neuroblasts, where exposure to citalopram subtly influenced progenitor subtypes. Pseudotemporal analysis showed enhanced neuronal differentiation. Our findings suggest that citalopram exposure during early neuronal differentiation influences gene expression patterns associated with neurodevelopment and depression, providing insights into its potential neurodevelopmental impact and highlighting the importance of further research to understand the long-term consequences of prenatal SSRI exposure.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Journal Article
    目的:本研究旨在研究声振动对人胚胎干细胞(hESCs)多能性的影响,并评估治疗后细胞的增殖和自我更新能力。
    方法:实验使用人ES细胞系H1。用声振动装置处理hESC。随后使用集落形成测定法检测它们的增殖能力,而通过免疫荧光染色检测到多能性相关标志物的表达。最后,在适当的引物存在下,使用定量聚合酶链反应(qPCR)检测基因表达水平的变化.
    结果:与对照组的正常细胞相比,受声振动作用的实验细胞形态无明显变化。相反,实验细胞的集落形成效率显着提高。免疫荧光染色结果显示实验组细胞多能性标志物NANOG阳性,八聚体结合转录因子4基因(OCT4),和SRY(性别决定区Y)-框2(SOX2)。此外,多能性基因NANOG的表达水平,OCT4,SOX2和Yes相关蛋白(YAP)相关基因在声振动后上调。
    结论:我们的结果表明,声振动增强了hESCs的增殖能力,并增加了NANOG的表达水平,OCT4、SOX2和YAP相关基因,表明声振动可以优化hESCs的自我更新能力,YAP信号通路可能在声振动的功能过程中起关键作用。
    OBJECTIVE: This study aimed to investigate the effect of acoustic vibration on the pluripotency of human embryonic stem cells (hESCs) and evaluate cell proliferation and self-renewal ability post-treatment.
    METHODS: The human ES cell line H1 was used for the experiments. hESCs were treated with an acoustic vibration device. Their proliferative ability was subsequently detected using a colony formation assay, while the expression of pluripotency-related markers was detected via immunofluorescence staining. Finally, changes in gene expression levels were examined using quantitative polymerase chain reaction (qPCR) in the presence of appropriate primers.
    RESULTS: Compared with normal cells in the control group, the morphology of experimental cells subjected to acoustic vibration did not significantly change. Contrastingly, the colony-forming efficiency of the experimental cells significantly increased. Immunofluorescence staining results showed the cells in experimental group were positive for the pluripotency markers NANOG, octamer-binding transcription factor 4 gene (OCT4), and SRY (sex determining region Y)-box 2 (SOX2). In addition, the expression levels of pluripotency genes NANOG, OCT4, SOX2, and Yes-associated protein (YAP)-related genes were up-regulated following acoustic vibration.
    CONCLUSIONS: Our results revealed that acoustic vibration enhanced the proliferative ability of hESCs and increased the expression levels of NANOG, OCT4, SOX2, and YAP-related genes, indicating that acoustic vibration can optimize the self-renewal ability of hESCs and that the YAP signaling pathway may play a critical role in the functional process of acoustic vibration.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Journal Article
    超级增强子(SE)是基因组DNA的扩展区域,可调节与细胞身份和细胞命运有关的基因的表达。我们最近在鼠Vsx2SE中确定了发育阶段和细胞类型特异性模块。这里,我们表明,在报告基因测定中,人VSX2SE模块具有相似的发育阶段和细胞类型特异性活性。通过将一个VSX2SE模块的人类序列插入患有小眼症的小鼠中,眼睛大小获救。为了了解这些SE模块在人类视网膜发育过程中的功能,我们删除了人类胚胎干细胞中的单个模块,并生成了视网膜类器官.删除一个模块会导致小的类器官,概述小眼症小鼠的小眼表型,而另一个模块的删除导致双极神经元发育中断。这种典型的SE用作理解具有复杂表达模式的神经源性转录因子的发育阶段和细胞类型特异性效应的模型。此外,通过阐明基因调控机制,我们可以开始研究这些机制的失调是如何导致表型多样性和疾病的。
    Super-enhancers (SEs) are expansive regions of genomic DNA that regulate the expression of genes involved in cell identity and cell fate. We recently identified developmental stage- and cell type-specific modules within the murine Vsx2 SE. Here, we show that the human VSX2 SE modules have similar developmental stage- and cell type-specific activity in reporter gene assays. By inserting the human sequence of one VSX2 SE module into a mouse with microphthalmia, eye size was rescued. To understand the function of these SE modules during human retinal development, we deleted individual modules in human embryonic stem cells and generated retinal organoids. Deleting one module results in small organoids, recapitulating the small-eyed phenotype of mice with microphthalmia, while deletion of the other module led to disruptions in bipolar neuron development. This prototypical SE serves as a model for understanding developmental stage- and cell type-specific effects of neurogenic transcription factors with complex expression patterns. Moreover, by elucidating the gene regulatory mechanisms, we can begin to examine how dysregulation of these mechanisms contributes to phenotypic diversity and disease.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Journal Article
    YAP在控制各种细胞系的生长和分化中起着至关重要的作用。尽管YAP在小鼠睾丸和生精细胞中的表达表明其在哺乳动物精子发生中的作用,YAP在人类男性生殖细胞发育中的作用尚未确定。使用体外模型和基因编辑方法,我们从人胚胎干细胞(hESCs)产生了人精原细胞干细胞样细胞(hSSLCs),并研究了YAP在人精子发生中的作用.结果表明,在生精分化早期降低YAP表达会增加PLZFhSSLCs和单倍体精子细胞的数量。我们还证明,在生精分化的后期,YAP的上调对于维持生精细胞的存活至关重要。偏离该模式的YAP的表达导致较低数量的hSSLCs和较高水平的生精细胞死亡。一起来看,我们的结果表明,YAP的动态表达模式对人类精子发生至关重要。在人类精子发生过程中调节YAP的水平可以提高源自hESCs的雄性生殖细胞的产量,为体外配子的发生提供了优化方法,深入了解其在男性不育治疗中的应用。
    YAP plays a vital role in controlling growth and differentiation in various cell lineages. Although the expression of YAP in mice testicular and spermatogenic cells suggests its role in mammalian spermatogenesis, the role of YAP in the development of human male germ cells has not yet been determined. Using an in vitro model and a gene editing approach, we generated human spermatogonia stem cell-like cells (hSSLCs) from human embryonic stem cells (hESCs) and investigated the role of YAP in human spermatogenesis. The results showed that reducing YAP expression during the early stage of spermatogenic differentiation increased the number of PLZF+ hSSLCs and haploid spermatid-like cells. We also demonstrated that the up-regulation of YAP is essential for maintaining spermatogenic cell survival during the later stages of spermatogenic differentiation. The expression of YAP that deviates from this pattern results in a lower number of hSSLCs and an increased level of spermatogenic cell death. Taken together, our result demonstrates that the dynamic expression pattern of YAP is essential for human spermatogenesis. Modulating the level of YAP during human spermatogenesis could improve the production yield of male germ cells derived from hESCs, which could provide the optimization method for in vitro gametogenesis and gain insight into the application in the treatment of male infertility.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Journal Article
    发育需要CTCF组织的三维基因组结构。临床鉴定的CTCF突变与不良发育结果有关。然而,潜在的机制仍然难以捉摸。在这次调查中,我们探讨了临床相关的R567W点突变的调节作用,位于CTCF的第11个锌指内,通过将这种突变引入小鼠模型和人类胚胎干细胞来源的皮质类器官模型。具有纯合CTCFR567W突变的小鼠表现出生长障碍,导致产后死亡率,和大脑的偏差,心,和病理和单细胞转录组水平的肺发育。这种突变诱导过早的干细胞样细胞衰竭,加速GABA能神经元的成熟,破坏神经发育和突触通路.此外,它特别阻碍CTCF与核心共识位点上游的外周基序结合,导致局部染色质结构和基因表达的改变,特别是在成簇的protcadherin位点。使用人类皮质类器官的比较分析反映了这种突变引起的后果。总之,这项研究阐明了CTCFR567W突变对人类神经发育障碍的影响,为潜在的治疗干预铺平道路。
    The three-dimensional genome structure organized by CTCF is required for development. Clinically identified mutations in CTCF have been linked to adverse developmental outcomes. Nevertheless, the underlying mechanism remains elusive. In this investigation, we explore the regulatory roles of a clinically relevant R567W point mutation, located within the 11th zinc finger of CTCF, by introducing this mutation into both murine models and human embryonic stem cell-derived cortical organoid models. Mice with homozygous CTCFR567W mutation exhibit growth impediments, resulting in postnatal mortality, and deviations in brain, heart, and lung development at the pathological and single-cell transcriptome levels. This mutation induces premature stem-like cell exhaustion, accelerates the maturation of GABAergic neurons, and disrupts neurodevelopmental and synaptic pathways. Additionally, it specifically hinders CTCF binding to peripheral motifs upstream to the core consensus site, causing alterations in local chromatin structure and gene expression, particularly at the clustered protocadherin locus. Comparative analysis using human cortical organoids mirrors the consequences induced by this mutation. In summary, this study elucidates the influence of the CTCFR567W mutation on human neurodevelopmental disorders, paving the way for potential therapeutic interventions.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Journal Article
    多能干细胞系之间的遗传差异导致细胞外信号通路的可变活性,定向分化方案的限制性可重复性。在这里,我们使用人胚胎干细胞(hESCs)来询问外源因子如何调节前肠内胚层谱系规范过程中的内源性信号事件。我们发现转化生长因子β1(TGF-β1)激活了推定的人类OTX2/LHX1基因调控网络,该网络通过拮抗内源性Wnt信号来促进前命运。与豪猪抑制相反,TGF-β1的作用不能被外源性Wnt配体逆转,提示SHISA蛋白的诱导和Fzd受体的细胞内积累使TGF-β1处理的细胞对Wnt信号传导难以反应。随后,TGF-β1介导的BMP和Wnt信号抑制抑制肝脏命运并促进胰腺命运。此外,TGF-β1治疗和胰腺特化期间的Wnt抑制联合可重复且稳健地增强hESC细胞系中胰岛素+细胞产量。广泛使用的分化方案的这种修改将提高用于基于细胞的治疗应用的胰腺β细胞产量。
    Genetic differences between pluripotent stem cell lines cause variable activity of extracellular signaling pathways, limiting reproducibility of directed differentiation protocols. Here we used human embryonic stem cells (hESCs) to interrogate how exogenous factors modulate endogenous signaling events during specification of foregut endoderm lineages. We find that transforming growth factor β1 (TGF-β1) activates a putative human OTX2/LHX1 gene regulatory network which promotes anterior fate by antagonizing endogenous Wnt signaling. In contrast to Porcupine inhibition, TGF-β1 effects cannot be reversed by exogenous Wnt ligands, suggesting that induction of SHISA proteins and intracellular accumulation of Fzd receptors render TGF-β1-treated cells refractory to Wnt signaling. Subsequently, TGF-β1-mediated inhibition of BMP and Wnt signaling suppresses liver fate and promotes pancreas fate. Furthermore, combined TGF-β1 treatment and Wnt inhibition during pancreatic specification reproducibly and robustly enhance INSULIN+ cell yield across hESC lines. This modification of widely used differentiation protocols will enhance pancreatic β cell yield for cell-based therapeutic applications.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Journal Article
    了解人类胚胎干细胞(hESCs)多能性的调节对于推进发育生物学和再生医学领域至关重要。尽管最近取得了进展,调节hESC多能性的分子事件,尤其是在幼稚状态和初始状态之间的过渡,仍然不清楚。在这里,我们显示,与引发的hESC相比,幼稚hESC显示更低水平的O-连接的N-乙酰葡糖胺(O-GlcNAcylation)。O-GlcNAcase(OGA),催化从蛋白质中去除O-GlcNAc的关键酶,在幼稚hESC中高度表达,对幼稚多能性很重要。OGA的耗尽加速了从幼稚到引发的多能性转变。OGA由EP300转录调节,并且充当对于维持幼稚多能性重要的基因的转录调节物。此外,我们通过定量蛋白质组学分析了两种多能性状态的蛋白质O-GlcNAcylation。一起,这项研究确定OGA是hESCs幼稚多能性的重要因素,并表明O-GlcNAcylation对hESCs稳态具有广泛影响。
    Understanding the regulation of human embryonic stem cells (hESCs) pluripotency is critical to advance the field of developmental biology and regenerative medicine. Despite the recent progress, molecular events regulating hESC pluripotency, especially the transition between naive and primed states, still remain unclear. Here we show that naive hESCs display lower levels of O-linked N-acetylglucosamine (O-GlcNAcylation) than primed hESCs. O-GlcNAcase (OGA), the key enzyme catalyzing the removal of O-GlcNAc from proteins, is highly expressed in naive hESCs and is important for naive pluripotency. Depletion of OGA accelerates naive-to-primed pluripotency transition. OGA is transcriptionally regulated by EP300 and acts as a transcription regulator of genes important for maintaining naive pluripotency. Moreover, we profile protein O-GlcNAcylation of the two pluripotency states by quantitative proteomics. Together, this study identifies OGA as an important factor of naive pluripotency in hESCs and suggests that O-GlcNAcylation has a broad effect on hESCs homeostasis.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Journal Article
    背景:端粒由染色体末端的重复DNA序列组成,以保护染色体稳定性,并且主要通过端粒酶或偶尔通过基于重组的机制通过端粒的替代端粒延长(ALT)来维持。可能调节端粒维持的其他机制仍有待探索。同时测量同一人胚胎干细胞(hESC)中的端粒长度和转录组,表明UBQLN1的mRNA表达水平与端粒长度呈线性关系。
    方法:在本研究中,我们首先产生了UBQLN1缺陷型hESCs,并通过RNA-seq和蛋白质组学与野生型(WT)hESCs比较了端粒长度和RNA和蛋白质水平的分子变化.然后,我们使用免疫沉淀-质谱(IP-MS)鉴定了与UBQLN1的潜在相互作用蛋白。此外,分析了UBQLN1缺陷型hESCs端粒缩短的潜在机制.
    结果:我们表明Ubiquilin1(UBQLN1)通过促进线粒体功能对人胚胎干细胞(hESCs)的端粒维持至关重要。UBQLN1缺乏导致氧化应激,失去了蛋白质,线粒体功能障碍,DNA损伤,和端粒磨耗。通过在低氧条件下培养或补充N-乙酰半胱氨酸来减少氧化损伤并促进线粒体功能,部分减轻了UBQLN1缺乏症诱导的端粒磨耗。此外,UBQLN1缺乏/端粒缩短下调神经外胚层谱系分化的基因。
    结论:总而言之,UBQLN1功能清除泛素化蛋白,防止线粒体过载和线粒体自噬升高。UBQLN1通过调节蛋白抑制维持线粒体和端粒,并在神经外胚层分化中起关键作用。
    BACKGROUND: Telomeres consist of repetitive DNA sequences at the chromosome ends to protect chromosomal stability, and primarily maintained by telomerase or occasionally by alternative telomere lengthening of telomeres (ALT) through recombination-based mechanisms. Additional mechanisms that may regulate telomere maintenance remain to be explored. Simultaneous measurement of telomere length and transcriptome in the same human embryonic stem cell (hESC) revealed that mRNA expression levels of UBQLN1 exhibit linear relationship with telomere length.
    METHODS: In this study, we first generated UBQLN1-deficient hESCs and compared with the wild-type (WT) hESCs the telomere length and molecular change at RNA and protein level by RNA-seq and proteomics. Then we identified the potential interacting proteins with UBQLN1 using immunoprecipitation-mass spectrometry (IP-MS). Furthermore, the potential mechanisms underlying the shortened telomeres in UBQLN1-deficient hESCs were analyzed.
    RESULTS: We show that Ubiquilin1 (UBQLN1) is critical for telomere maintenance in human embryonic stem cells (hESCs) via promoting mitochondrial function. UBQLN1 deficiency leads to oxidative stress, loss of proteostasis, mitochondria dysfunction, DNA damage, and telomere attrition. Reducing oxidative damage and promoting mitochondria function by culture under hypoxia condition or supplementation with N-acetylcysteine partly attenuate the telomere attrition induced by UBQLN1 deficiency. Moreover, UBQLN1 deficiency/telomere shortening downregulates genes for neuro-ectoderm lineage differentiation.
    CONCLUSIONS: Altogether, UBQLN1 functions to scavenge ubiquitinated proteins, preventing their overloading mitochondria and elevated mitophagy. UBQLN1 maintains mitochondria and telomeres by regulating proteostasis and plays critical role in neuro-ectoderm differentiation.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

公众号