pluripotency

多能性
  • 文章类型: Journal Article
    Li-Fraumeni综合征(LFS)是一种罕见的常染色体显性遗传遗传疾病,大大增加了患几种癌症的风险,包括幼儿和年轻人。LFS主要由肿瘤抑制基因TP53中的特定突变引起。在这项研究中,我们成功地从诊断为LFS的患者中产生了两个人诱导多能干细胞(iPSC)系,每个在TP53基因中携带不同的杂合突变。这些LFS患者来源的iPSC细胞系表现出关键多能性标志物的稳健表达,证明了分化为所有三个胚层(内胚层,中胚层,和外胚层),保持正常的核型.这些iPSC细胞系的建立为体外LFS建模提供了有价值的工具,使研究人员能够在各种细胞类型和组织中研究与疾病相关的潜在病理机制。
    Li-Fraumeni syndrome (LFS) is a rare autosomal dominant inherited genetic disorder that greatly increases the risk of developing several types of cancer, including young children and young adults. LFS is primarily caused by specific mutations in the tumor suppressor gene TP53. In this study, we successfully generated two human induced pluripotent stem cell (iPSC) lines derived from patients diagnosed with LFS, each carrying a distinct heterozygous mutation in the TP53 gene. These LFS patient-derived iPSC lines exhibited robust expression of key pluripotency markers, demonstrated the capacity to differentiate into all three germ layers (endoderm, mesoderm, and ectoderm), and maintained a normal karyotype. The establishment of these iPSC lines provides a valuable tool for modeling LFS in vitro, enabling researchers to investigate the underlying pathological mechanisms associated with the disease across various cell types and tissues.
    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:建立稳定的猪胚胎干细胞(pESCs)有助于基础和生物医学研究,包括比较发育生物学,以及评估干细胞疗法的安全性。尽管有这些优势,从体外囊胚获得的大多数pESCs需要复杂的培养基和饲养层,常规使用,基因改造,和分化为特定的细胞类型困难。我们旨在建立具有单细胞传代能力的pESCs,高增殖潜能,并且使用简化的无血清培养基从体外来源的胚泡长期培养中稳定。
    方法:我们使用各种基础培养基(DMEM/F10(1:1),DMEM/F12和a-MEM)和因子(FGF2,IWR-1,CHIR99021和WH-4-023)。在饲养或无饲养条件下分析建立的pESC的多能性和自我更新能力。最终,我们在无血清条件下开发了由FGF2,IWR-1和WH-4-023组成的简化培养基(FIW)。
    结果:pESC-FIW细胞系能够以短细胞倍增时间进行单细胞传代,并表达多能性标记POU5F1,SOX2和NANOG,以及细胞表面标记SSEA1、SSEA4和TRA-1-60。pESC-FIW显示稳定的增殖速率和正常的核型,即使经过50个通道。转录组分析显示,pESC-FIW与报道的在复杂培养基中维持的pESC相似,并显示出胃泌素上胚细胞特征。使用mTeSR™在纤连蛋白包被的平板上在无饲养条件下维持pESC-FIW多次传代,用于无饲养培养的商业培养基,表现出与在饲养条件下观察到的特征相似的特征。
    结论:这些结果表明,WNT和SRC的抑制足以建立能够在无血清条件下进行单细胞传代和无饲养细胞扩增的pESC。pESCs易于维护,有利于其在农业和生物医学基因编辑技术中的应用。以及血统承诺研究。
    BACKGROUND: The establishment of stable porcine embryonic stem cells (pESCs) can contribute to basic and biomedical research, including comparative developmental biology, as well as assessing the safety of stem cell-based therapies. Despite these advantages, most pESCs obtained from in vitro blastocysts require complex media and feeder layers, making routine use, genetic modification, and differentiation into specific cell types difficult. We aimed to establish pESCs with a single cell-passage ability, high proliferative potency, and stable in long-term culture from in vitro-derived blastocysts using a simplified serum-free medium.
    METHODS: We evaluated the establishment efficiency of pESCs from in vitro blastocysts using various basal media (DMEM/F10 (1:1), DMEM/F12, and a-MEM) and factors (FGF2, IWR-1, CHIR99021, and WH-4-023). The pluripotency and self-renewal capacity of the established pESCs were analyzed under feeder or feeder-free conditions. Ultimately, we developed a simplified culture medium (FIW) composed of FGF2, IWR-1, and WH-4-023 under serum-free conditions.
    RESULTS: The pESC-FIW lines were capable of single-cell passaging with short cell doubling times and expressed the pluripotency markers POU5F1, SOX2, and NANOG, as well as cell surface markers SSEA1, SSEA4, and TRA-1-60. pESC-FIW showed a stable proliferation rate and normal karyotype, even after 50 passages. Transcriptome analysis revealed that pESC-FIW were similar to reported pESC maintained in complex media and showed gastrulating epiblast cell characteristics. pESC-FIW were maintained for multiple passages under feeder-free conditions on fibronectin-coated plates using mTeSR™, a commercial medium used for feeder-free culture, exhibiting characteristics similar to those observed under feeder conditions.
    CONCLUSIONS: These results indicated that inhibition of WNT and SRC was sufficient to establish pESCs capable of single-cell passaging and feeder-free expansion under serum-free conditions. The easy maintenance of pESCs facilitates their application in gene editing technology for agriculture and biomedicine, as well as lineage commitment studies.
    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
    细胞命运决定是一个复杂的过程,由多个调控层协调,包括信号通路,转录因子,表观遗传修饰,和代谢重新布线。在复杂的表观遗传调制中,抑制标记H3K27me3由PRC2(多梳状抑制复合物2)沉积,并由去甲基酶KDM6去除,通过其动态和精确的改变在介导细胞身份转换中起关键作用。在这里,我们概述并讨论了H3K27me3及其修饰剂如何调节多能性维持和早期谱系分化。我们主要强调以下四个方面:1)两个亚复合物PRC2.1和PRC2.2以及基因组H3K27甲基化的分布;2)PRC2作为多能性维持和退出的关键调节因子;3)橡皮擦KDM6在早期分化中的新兴作用;4)新发现的影响H3K27me3的其他因素。我们对H3K27me3动态调节的分子原理以及这种表观遗传标记如何参与以多能干细胞为中心的细胞命运决定进行了全面的了解。
    Cell fate determination is an intricate process which is orchestrated by multiple regulatory layers including signal pathways, transcriptional factors, epigenetic modifications, and metabolic rewiring. Among the sophisticated epigenetic modulations, the repressive mark H3K27me3, deposited by PRC2 (polycomb repressive complex 2) and removed by demethylase KDM6, plays a pivotal role in mediating the cellular identity transition through its dynamic and precise alterations. Herein, we overview and discuss how H3K27me3 and its modifiers regulate pluripotency maintenance and early lineage differentiation. We primarily highlight the following four aspects: 1) the two subcomplexes PRC2.1 and PRC2.2 and the distribution of genomic H3K27 methylation; 2) PRC2 as a critical regulator in pluripotency maintenance and exit; 3) the emerging role of the eraser KDM6 in early differentiation; 4) newly identified additional factors influencing H3K27me3. We present a comprehensive insight into the molecular principles of the dynamic regulation of H3K27me3, as well as how this epigenetic mark participates in pluripotent stem cell-centered cell fate determination.
    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
    哺乳动物胚胎非常容易受到环境毒物(ET)的影响。双酚A(BPA),最扩散的ET之一,通过雌激素模拟和激素样特性发挥内分泌干扰作用,有害的健康影响,包括繁殖。然而,其在围植入期的影响尚不清楚.这项研究,使用gastruloids作为基于3D干细胞的胚胎发育体外模型,表明,在Wnt/β-catenin途径激活期间,BPA暴露会通过β-catenin蛋白减少而阻止其轴向伸长。Snail的下调可能阻碍了Gastroid的重塑,子弹和扭曲,已知抑制E-cadherin表达并激活N-cadherin基因,和N-cadherin蛋白的低表达。此外,缺乏腺样体伸长可能与暴露于BPA的细胞从多能性条件中退出的改变及其随后的分化有关。总之,在这里,我们表明,由BPA的gastruloids\'轴向伸长的抑制可能是伴随的Wnt/β-连环蛋白扰动的结果,N-cadherin表达降低,Oct4、T/Bra和Cdx2表达改变,所有这些都与小鼠类动物的发育受损一致。
    Mammalian embryos are very vulnerable to environmental toxicants (ETs) exposure. Bisphenol A (BPA), one of the most diffused ETs, exerts endocrine-disrupting effects through estro-gen-mimicking and hormone-like properties, with detrimental health effects, including on reproduction. However, its impact during the peri-implantation stages is still unclear. This study, using gastruloids as a 3D stem cell-based in vitro model of embryonic development, showed that BPA exposure arrests their axial elongation when present during the Wnt/β-catenin pathway activation period by β-catenin protein reduction. Gastruloid reshaping might have been impeded by the downregulation of Snail, Slug and Twist, known to suppress E-cadherin expression and to activate the N-cadherin gene, and by the low expression of the N-cadherin protein. Also, the lack of gastruloids elongation might be related to altered exit of BPA-exposed cells from the pluripotency condition and their following differentiation. In conclusion, here we show that the inhibition of gastruloids\' axial elongation by BPA might be the result of the concomitant Wnt/β-catenin perturbation, reduced N-cadherin expression and Oct4, T/Bra and Cdx2 altered patter expression, which all together concur in the impaired development of mouse gastruloids.
    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
    胚胎滞育是一种生殖适应,使一些哺乳动物物种能够停止胚胎发育的其他连续步伐。在这个休眠状态下,胚胎利用知之甚少的调节机制来长时间保持其发育潜力。这里,使用小鼠胚胎和单细胞RNA测序,我们在单细胞分辨率下分子定义了胚胎滞育,揭示转录动力学,而胚胎似乎处于悬浮动画状态。此外,我们发现,休眠的多能细胞依赖于整合素受体来感知其微环境,并通过Yap/Taz介导的促存活信号维持其生存能力.
    Embryonic diapause is a reproductive adaptation that enables some mammalian species to halt the otherwise continuous pace of embryonic development. In this dormant state, the embryo exploits poorly understood regulatory mechanisms to preserve its developmental potential for prolonged periods of time. Here, using mouse embryos and single-cell RNA sequencing, we molecularly defined embryonic diapause at single-cell resolution, revealing transcriptional dynamics while the embryo seemingly resides in a state of suspended animation. Additionally, we found that the dormant pluripotent cells rely on integrin receptors to sense their microenvironment and preserve their viability via Yap/Taz-mediated prosurvival signaling.
    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
    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
    人多能干细胞(hPSC),包括人类胚胎干细胞(hESCs)和人类诱导多能干细胞(hiPSCs),在再生医学中拥有巨大的潜力,为个性化细胞治疗提供新的机会。然而,它们的临床翻译受到文化环境中对异种成分的不可避免的依赖。这项研究通过设计一种完全合成的,无异种培养基质,其表面组成被系统地定制用于hPSC的无异种培养。功能性聚合物表面,pGC2(聚(甲基丙烯酸缩水甘油酯-接枝-胍-共-羧酸丙烯酸酯)),提供优异的细胞粘附特性以及非细胞毒性,实现强劲的hESC和hiPSC增长,同时与Matrigel相比具有成本竞争力和可扩展性。这项调查包括在不同的实验条件下对pGC2的全面评估,证明了其对各种多能干细胞系的广泛适应性,文化媒介,和基底。至关重要的是,pGC2支持长期的hESC和hiPSC扩增,多达十个通道,而不损害其干性和多能性。值得注意的是,与Matrigel相比,这项研究首次证实了在聚合物底物上无异种培养hiPSC十次传代后相同的蛋白质组学谱。创新的基质弥合了实验室研究和临床翻译之间的差距,为推进基于干细胞的疗法提供了新的有希望的途径。
    Human pluripotent stem cells (hPSCs), encompassing human embryonic stem cells (hESCs) and human induced pluripotent stem cells (hiPSCs), hold immense potential in regenerative medicine, offering new opportunities for personalized cell therapies. However, their clinical translation is hindered by the inevitable reliance on xenogeneic components in culture environments. This study addresses this challenge by engineering a fully synthetic, xeno-free culture substrate, whose surface composition is tailored systematically for xeno-free culture of hPSCs. A functional polymer surface, pGC2 (poly(glycidyl methacrylate-grafting-guanidine-co-carboxylic acrylate)), offers excellent cell-adhesive properties as well as non-cytotoxicity, enabling robust hESCs and hiPSCs growth while presenting cost-competitiveness and scalability over Matrigel. This investigation includes comprehensive evaluations of pGC2 across diverse experimental conditions, demonstrating its wide adaptability with various pluripotent stem cell lines, culture media, and substrates. Crucially, pGC2 supports long-term hESCs and hiPSCs expansion, up to ten passages without compromising their stemness and pluripotency. Notably, this study is the first to confirm an identical proteomic profile after ten passages of xeno-free cultivation of hiPSCs on a polymeric substrate compared to Matrigel. The innovative substrate bridges the gap between laboratory research and clinical translation, offering a new promising avenue for advancing stem cell-based therapies.
    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
    细胞通讯协调发育过程,保持稳态,并有助于疾病。因此,理解共享环境中细胞之间的关系至关重要。在这里,我们介绍了用于识别邻居的正超亮荧光融合(PUFFFIN),一种基于分泌和摄取带正电的荧光蛋白s36GFP的细胞邻居标记系统。我们将s36GFP融合到mNeonGreen或HaloTag上,促进超明亮,敏感,颜色的选择标签。保管员细胞将PUFFFIN转移给邻居,同时保留核mCherry,进行识别,隔离,直截了当地调查活着的邻居。PUFFFIN可以传送到细胞,组织,或胚胎上的可定制的单质粒构建体,该构建体由可互换的成分组成,可以选择掺入任何转基因。这种多功能性使细胞属性的操纵,同时标记周围的细胞,在细胞培养或体内。我们使用PUFFFIN来询问多能细胞在从幼稚多能性退出期间是否调整分化速度以与它们的邻居同步。PUFFFIN提供了一个简单的,敏感,可定制的方法来分析细胞对细胞身份或行为的自然或诱导变化的非细胞自主反应。
    Cell communication coordinates developmental processes, maintains homeostasis, and contributes to disease. Therefore, understanding the relationship between cells in a shared environment is crucial. Here we introduce Positive Ultra-bright Fluorescent Fusion For Identifying Neighbours (PUFFFIN), a cell neighbour-labelling system based upon secretion and uptake of positively supercharged fluorescent protein s36GFP. We fused s36GFP to mNeonGreen or to a HaloTag, facilitating ultra-bright, sensitive, colour-of-choice labelling. Secretor cells transfer PUFFFIN to neighbours while retaining nuclear mCherry, making identification, isolation, and investigation of live neighbours straightforward. PUFFFIN can be delivered to cells, tissues, or embryos on a customisable single-plasmid construct composed of interchangeable components with the option to incorporate any transgene. This versatility enables the manipulation of cell properties, while simultaneously labelling surrounding cells, in cell culture or in vivo. We use PUFFFIN to ask whether pluripotent cells adjust the pace of differentiation to synchronise with their neighbours during exit from naïve pluripotency. PUFFFIN offers a simple, sensitive, customisable approach to profile non-cell-autonomous responses to natural or induced changes in cell identity or behaviour.
    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
    在这次审查中,我们考虑神经c细胞(NCC)的多能性,雪旺氏细胞前体(SCP),基于遗传追踪和敲除模型动物和单细胞转录组学分析,以及它们在胚胎发生中的作用。特别是,我们总结和分析了NCCs和SCP对腺体发育和功能的贡献。
    In this review, we consider the multipotency of neural crest cells (NCCs), Schwann cell precursors (SCPs), and their role in embryogenesis base on genetic tracing and knock out model animals and single cell transcriptomic analysis. In particular, we summarize and analyze data on the contribution of NCCs and SCPs to the gland development and functions.
    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
    在植入过程中,胚胎经历非极化到极化的转变,以启动植入后的形态发生。然而,潜在的分子机制是未知的。这里,我们确定了在植入过程中控制胚胎形态发生和多能性转变的瞬时转录激活。在幼稚多能胚胎干细胞(ESC)中,代表着床前胚胎,我们发现,微处理器成分DGCR8可以识别新生mRNAs内的茎环结构,以隔离转录共激活子FLII,从而直接抑制转录。当mESC从幼稚多能性退出时,ERK/RSK/P70S6K通路快速激活,导致FLII磷酸化和DGCR8/FLII相互作用的破坏。磷酸化FLII可以与转录因子JUN结合,激活细胞迁移相关基因以建立类似于植入胚胎的平衡多能性。DGCR8对FLII的重新测序驱动平衡的ESC进入形成性多能性。总之,我们确定了DGCR8/FLII/JUN介导的瞬时转录激活机制。这种机制的破坏抑制了胚胎植入过程中幼稚形成的多能性转变和相应的非极化到极化的转变,在小鼠和人类中都是保守的。
    During implantation, embryos undergo an unpolarized-to-polarized transition to initiate postimplantation morphogenesis. However, the underlying molecular mechanism is unknown. Here, we identify a transient transcriptional activation governing embryonic morphogenesis and pluripotency transition during implantation. In naive pluripotent embryonic stem cells (ESCs), which represent preimplantation embryos, we find that the microprocessor component DGCR8 can recognize stem-loop structures within nascent mRNAs to sequester transcriptional coactivator FLII to suppress transcription directly. When mESCs exit from naive pluripotency, the ERK/RSK/P70S6K pathway rapidly activates, leading to FLII phosphorylation and disruption of DGCR8/FLII interaction. Phosphorylated FLII can bind to transcription factor JUN, activating cell migration-related genes to establish poised pluripotency akin to implanting embryos. Resequestration of FLII by DGCR8 drives poised ESCs into formative pluripotency. In summary, we identify a DGCR8/FLII/JUN-mediated transient transcriptional activation mechanism. Disruption of this mechanism inhibits naive-poised-formative pluripotency transition and the corresponding unpolarized-to-polarized transition during embryo implantation, which are conserved in mice and humans.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

    求助全文

公众号