human embryonic stem cells

人胚胎干细胞
  • 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

    求助全文

  • 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

    求助全文

  • 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

    求助全文

  • 文章类型: Journal Article
    以前的回顾性队列研究发现,与子宫和输卵管中的氧气张力(2%-8%)相比,着床前胚胎在辅助生殖技术(ART)期间暴露于大气氧分压(AtmO2,20%)会影响胚胎质量,妊娠结局和后代健康。然而,目前关于AtmO2对胚胎和后代发育的影响和机制的研究主要局限于动物实验。人胚胎干细胞(hESCs)在人类早期胚胎发育研究中发挥着特殊而不可替代的作用。在这项研究中,我们使用hESCs作为模型来阐明AtmO2暴露对人胚胎发育的可能影响和机制.我们发现暴露于AtmO2可以降低细胞活力,产生氧化应激,增加DNA损伤,启动DNA修复,激活自噬,增加细胞凋亡。我们还注意到大约50%的hESC存活,通过自我更新和多能性调节因子的高表达适应和增殖,并影响胚状体的分化。这些数据表明hESCs经历氧化应激,DNA损伤的积累,并在AtmO2的选择压力下激活DNA损伤反应。一些hESC经历细胞死亡,而其他hESC通过增加自我更新基因的表达来适应和增殖。目前的发现提供了体外证据,表明在植入前早期阶段暴露于AtmO2会对hESC产生负面影响。
    Previous retrospective cohort studies have found that, compared with oxygen tension in the uterus and fallopian tubes (2 %-8 %), exposure of pre-implantation embryos to atmospheric oxygen tension (AtmO2, 20 %) during assisted reproductive technology(ART) can affect embryo quality, pregnancy outcomes and offspring health. However, current research on the effects and mechanisms of AtmO2 on the development of embryos and offspring is mainly limited to animal experiments. Human embryonic stem cells (hESCs) play a special and irreplaceable role in the study of early human embryonic development. In this study, we used hESCs as a model to elucidate the possible effects and mechanisms of AtmO2 exposure on human embryonic development. We found that exposure to AtmO2 can reduce cell viability, produce oxidative stress, increase DNA damage, initiate DNA repair, activate autophagy, and increase cell apoptosis. We also noticed that approximately 50 % of hESCs survived, adapted and proliferated through high expression of self-renewal and pluripotency regulatory factors, and affected embryoid body differentiation. These data indicate that hESCs experience oxidative stress, accumulation of DNA damage, and activate DNA damage response under the selective pressure of AtmO2.Some hESCs undergo cell death, whereas other hESCs adapt and proliferate through increased expression of self-renewal genes. The current findings provide in vitro evidence that exposure to AtmO2 during the early preimplantation stage negatively affects hESCs.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

    求助全文

  • 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)

  • 文章类型: Journal Article
    尽管目前的干细胞疗法显示出有希望的潜力,使用自体细胞的扩展过程以及供体-宿主匹配以避免移植细胞排斥的必要性极大地限制了这些治疗的广泛适用性。这将是非常有利的,产生多能通用供体干细胞系,是免疫规避和,因此,不受个人免疫系统的限制,在细胞替代疗法中实现无限应用。在这种免疫逃避性干细胞进入临床试验之前,在有免疫能力的动物中通过移植实验进行体内测试将是临床前测试之前的有利方法。通过在有免疫能力的动物中使用人类干细胞,结果将更容易转化为临床环境,因为免疫系统的任何部分都没有改变,尽管是在异种环境中。这样,免疫逃避,细胞存活,和不需要的增殖作用可以在人类临床试验之前进行评估。目前的研究提出了三种人胚胎干细胞系(hESC)的产生和表征,用于免疫活性小鼠的异种移植。主要的组织相容性复合物I和II编码基因,B2M和CIITA,已使用CRISPR-Cas9靶向基因替换策略和敲除从hESC中删除。通过插入鼠CD47敲除B2M。将人分泌的胚胎碱性磷酸酶(hSEAP)插入安全港位点以跟踪体内细胞。编辑的hESC保持了他们的多能性,核型正态,小鼠CD47和hSEAP在体外稳定表达。通过测量血液样品中的hSEAP,成功地监测了hESC向免疫活性BALB/c小鼠的体内移植。然而,免疫逃避性hESCs移植在11天内导致完全排斥,在第8天具有明确的T细胞免疫浸润。我们的结果表明,B2M和CIITA的敲除以及CD47的物种特异性表达不足以防止免疫活性和异种环境中的排斥反应。
    Although current stem cell therapies exhibit promising potential, the extended process of employing autologous cells and the necessity for donor-host matching to avert the rejection of transplanted cells significantly limit the widespread applicability of these treatments. It would be highly advantageous to generate a pluripotent universal donor stem cell line that is immune-evasive and, therefore, not restricted by the individual\'s immune system, enabling unlimited application within cell replacement therapies. Before such immune-evasive stem cells can be moved forward to clinical trials, in vivo testing via transplantation experiments in immune-competent animals would be a favorable approach preceding preclinical testing. By using human stem cells in immune competent animals, results will be more translatable to a clinical setting, as no parts of the immune system have been altered, although in a xenogeneic setting. In this way, immune evasiveness, cell survival, and unwanted proliferative effects can be assessed before clinical trials in humans. The current study presents the generation and characterization of three human embryonic stem cell lines (hESCs) for xenogeneic transplantation in immune-competent mice. The major histocompatibility complexes I- and II-encoding genes, B2M and CIITA, have been deleted from the hESCs using CRISPR-Cas9-targeted gene replacement strategies and knockout. B2M was knocked out by the insertion of murine CD47. Human-secreted embryonic alkaline phosphatase (hSEAP) was inserted in a safe harbor site to track cells in vivo. The edited hESCs maintained their pluripotency, karyotypic normality, and stable expression of murine CD47 and hSEAP in vitro. In vivo transplantation of hESCs into immune-competent BALB/c mice was successfully monitored by measuring hSEAP in blood samples. Nevertheless, transplantation of immune-evasive hESCs resulted in complete rejection within 11 days, with clear immune infiltration of T-cells on day 8. Our results reveal that knockout of B2M and CIITA together with species-specific expression of CD47 are insufficient to prevent rejection in an immune-competent and xenogeneic context.
    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
    叉头盒蛋白J1(FOXJ1),叉头家族的一员,是调节多纤毛细胞分化和活动纤毛程序的重要转录因子。这里,我们通过使用CRISPR/Cas9系统插入FOXJ1的P2A-EGFP基因盒,建立了FOXJ1-EGFP敲入人胚胎干细胞(hESC)系。报告细胞系保留了正常的核型,表达可比较的多能标记基因,并保持差异化潜力。该报告细胞系能够在一般肺分化过程中对多纤毛细胞进行活鉴定,并且将是研究多纤毛细胞分化的有价值的工具。纤毛发生及相关肺部疾病的机制。
    Forkhead box protein J1 (FOXJ1), a member of the forkhead family, is an important transcription factor regulating multiciliated cell differentiation and motile ciliogenic program. Here, we established a FOXJ1- EGFP knock-in human embryonic stem cell (hESC) line by inserting a P2A-EGFP gene cassette of FOXJ1 using CRISPR/Cas9 system. The reporter cell line retained a normal karyotype, expressed comparable pluripotent marker genes, and maintained differentiation potential. This reporter cell line enables live identification of multiciliated cells during the general lung differentiation and will be a valuable tool for studying the multiciliated cell differentiation, ciliogenesis and mechanism of related pulmonary diseases.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

    求助全文

  • 文章类型: Journal Article
    优化定形内胚层(DE)分化的效率对于产生不同的器官样结构是必要的。在这项研究中,我们使用小分子抑制剂saracatinib(SAR)增强人胚胎干细胞的DE分化和诱导多能干细胞。SAR在低浓度下显着提高了DE分化效率。通过RNA-seq和分子对接模拟探讨了SAR与粘着斑激酶(FAK)的相互作用,这进一步支持在SAR处理的细胞中通过p-FAK过表达抑制DE分化。此外,我们发现SAR抑制Yes相关蛋白(YAP)的核易位,FAK的下游效应器,这促进了DE分化。此外,SAR的添加使得活化素A(AA)从50ng/mL显著降低至10ng/mL,而不损害DE分化效率。为了诱导胰腺谱系,在DE分化阶段,10ng/mlAA与SAR结合产生的PDX1/NKX6.1胰腺祖细胞数量与通过50ng/mlAA处理获得的细胞数量相当。我们的研究强调了SAR作为一种潜在的调节剂,可以促进DE细胞的成本效益产生,并提供了对细胞命运决定的编排的见解。
    Optimizing the efficiency of definitive endoderm (DE) differentiation is necessary for the generation of diverse organ-like structures. In this study, we used the small molecule inhibitor saracatinib (SAR) to enhance DE differentiation of human embryonic stem cells and induced pluripotent stem cells. SAR significantly improved DE differentiation efficiency at low concentrations. The interaction between SAR and Focal Adhesion Kinase (FAK) was explored through RNA-seq and molecular docking simulations, which further supported the inhibition of DE differentiation by p-FAK overexpression in SAR-treated cells. In addition, we found that SAR inhibited the nuclear translocation of Yes-associated protein (YAP), a downstream effector of FAK, which promoted DE differentiation. Moreover, the addition of SAR enabled a significant reduction in activin A (AA) from 50 to 10 ng/mL without compromising DE differentiation efficiency. For induction of the pancreatic lineage, 10 ng/ml AA combined with SAR at the DE differentiation stage yielded a comparative number of PDX1+/NKX6.1+ pancreatic progenitor cells to those obtained by 50 ng/ml AA treatment. Our study highlights SAR as a potential modulator that facilitates the cost-effective generation of DE cells and provides insight into the orchestration of cell fate determination.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

公众号