hESCs

hESCs
  • 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
    STAMBP的突变已被确定为导致先天性人类小头畸形-毛细血管畸形(MIC-CAP)综合征,一种以全球发育迟缓为特征的罕见遗传疾病,严重的小头畸形,毛细血管畸形,等。先前的生化研究和小鼠功能丧失研究提供了对STAMBP机制的见解,然而,STAMBP缺乏如何导致患者受影响组织畸形仍存在争议.在这项研究中,我们研究了STAMBP在人胚胎干细胞(hESCs)神经分化过程中的功能和潜在机制。我们发现STAMBP对于hESC的多能性维持或神经分化是不必要的。然而,来自STAMBP缺陷型hESC的神经祖细胞(NPC)无法在体外长期维持/扩增。我们发现抗凋亡蛋白CFLAR在那些受影响的NPCs中下调,CFLAR的异位表达挽救了由STAMBP缺乏症引起的NPC缺陷。我们的研究不仅为STAMBP突变患者神经缺陷的机制提供了新的见解,这也表明死亡受体介导的细胞凋亡是体外NPCs长期维持/扩增的障碍,因此抵消这种细胞死亡途径可能有利于体外NPCs的产生。
    Mutations in STAMBP have been well-established to cause congenital human microcephaly-capillary malformation (MIC-CAP) syndrome, a rare genetic disorder characterized by global developmental delay, severe microcephaly, capillary malformations, etc. Previous biochemical investigations and loss-of-function studies in mice have provided insights into the mechanism of STAMBP, however, it remains controversial how STAMBP deficiency leads to malformation of those affected tissues in patients. In this study, we investigated the function and underlying mechanism of STAMBP during neural differentiation of human embryonic stem cells (hESCs). We found that STAMBP is dispensable for the pluripotency maintenance or neural differentiation of hESCs. However, neural progenitor cells (NPCs) derived from STAMBP-deficient hESCs fail to be long-term maintained/expanded in vitro. We identified the anti-apoptotic protein CFLAR is down-regulated in those affected NPCs and ectopic expression of CFLAR rescues NPC defects induced by STAMBP-deficiency. Our study not only provides novel insight into the mechanism of neural defects in STAMBP mutant patients, it also indicates that the death receptor mediated apoptosis is an obstacle for long-term maintenance/expansion of NPCs in vitro thus counteracting this cell death pathway could be beneficial to the generation of NPCs in vitro.
    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
    胚胎干细胞(ESC)被定义为具有自我更新和分化能力的干细胞。这些独特的特性受到复杂的遗传和分子机制的严格调控和控制,其理解对于基础研究和转化研究都至关重要。大量的研究主要集中在理解ESCs多能性和分化的分子机制。而对扩散的监管受到的关注相对较少。这里,我们研究了ZZZ3(含锌指ZZ型3)在人类ESCs稳态中的作用。我们发现ZZZ3的敲低对核糖体生物发生有负面影响,翻译,和mTOR信号,导致细胞增殖的显著减少。这个过程不会影响多能性,表明ZZZ3耗尽的ESC进入“休眠样”状态,并且在人类ESC中增殖和多能性也可以解偶联。
    Embryonic stem cells (ESCs) are defined as stem cells with self-renewing and differentiation capabilities. These unique properties are tightly regulated and controlled by complex genetic and molecular mechanisms, whose understanding is essential for both basic and translational research. A large number of studies have mostly focused on understanding the molecular mechanisms governing pluripotency and differentiation of ESCs, while the regulation of proliferation has received comparably less attention. Here, we investigate the role of ZZZ3 (zinc finger ZZ-type containing 3) in human ESCs homeostasis. We found that knockdown of ZZZ3 negatively impacts ribosome biogenesis, translation, and mTOR signaling, leading to a significant reduction in cell proliferation. This process occurs without affecting pluripotency, suggesting that ZZZ3-depleted ESCs enter a \"dormant-like\" state and that proliferation and pluripotency can be uncoupled also in human ESCs.
    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
    体外培养小鼠和人胚胎干细胞(ESCs)是干细胞生物学领域的重大突破。这些模型很快就普及了,主要是由于它们的多能性。显然,小鼠和人类来源的ESC由于其多能性而具有典型的表型反应,如自我更新能力和效力。核心转录因子的保守网络调节这些反应。然而,显著不同的信号通路和上游转录网络调节这些核心多能性因子在两个物种的ESC中的表达和活性。事实上,充足的证据表明,一条途径,在小鼠胚胎干细胞中保持多能性,促进人ESC的分化。在这次审查中,我们讨论了典型信号通路在多能性和分化调控中的作用,特别是在小鼠和人ESCs中.我们认为,了解这些独特的,有时相反的机制,对于干细胞生物学和再生医学领域的进展至关重要。
    Culturing of mouse and human embryonic stem cells (ESCs) in vitro was a major breakthrough in the field of stem cell biology. These models gained popularity very soon mainly due to their pluripotency. Evidently, the ESCs of mouse and human origin share typical phenotypic responses due to their pluripotent nature, such as self-renewal capacity and potency. The conserved network of core transcription factors regulates these responses. However, significantly different signaling pathways and upstream transcriptional networks regulate expression and activity of these core pluripotency factors in ESCs of both the species. In fact, ample evidence shows that a pathway, which maintains pluripotency in mouse ESCs, promotes differentiation in human ESCs. In this review, we discuss the role of canonical signaling pathways implicated in regulation of pluripotency and differentiation particularly in mouse and human ESCs. We believe that understanding these distinct and at times-opposite mechanisms-is critical for the progress in the field of stem cell biology and regenerative medicine.
    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
    人类胚胎干细胞(hESC)类似于在早期移植后人类胚胎中发现的多能外胚层细胞,代表多能性的“启动”状态。有助于引发的多能细胞保持多能性并制备用于分化的基因的一个因子是转录因子TCF7L1,其是被称为T细胞因子/淋巴增强因子(TCF/LEF)的小蛋白家族的成员,其充当WNT信号传导途径的下游组分。WNT途径的转录输出是受调控的,在某种程度上,通过TCF/LEF的活性与WNT途径的另一个组成部分相结合,β-catenin.因为TCF7L1在调节多能性中起着重要作用,我们开始使用内源性蛋白的快速免疫沉淀(RIME)表征与hESC中染色质结合时与TCF7L1相关的蛋白复合物.数据可通过ProteomeXchange获得,标识符为PXD047582。这些数据确定了TCF7L1在染色质上的已知和新的伴侣,并提供了关于TCF7L1和多能性本身如何被调节的新见解。
    Human embryonic stem cells (hESCs) resemble the pluripotent epiblast cells found in the early postimplantation human embryo and represent the \"primed\" state of pluripotency. One factor that helps primed pluripotent cells retain pluripotency and prepare genes for differentiation is the transcription factor TCF7L1, a member of a small family of proteins known as T cell factors/Lymphoid enhancer factors (TCF/LEF) that act as downstream components of the WNT signaling pathway. Transcriptional output of the WNT pathway is regulated, in part, by the activity of TCF/LEFs in conjunction with another component of the WNT pathway, β-CATENIN. Because TCF7L1 plays an important role in regulating pluripotency, we began to characterize the protein complex associated with TCF7L1 when bound to chromatin in hESCs using rapid immunoprecipitation of endogenous proteins (RIME).  Data are available via ProteomeXchange with identifier PXD047582. These data identify known and new partners of TCF7L1 on chromatin and provide novel insights into how TCF7L1 and pluripotency itself might be regulated.
    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: Systematic Review
    背景:视网膜退行性疾病(RDDs)通过损害视网膜神经元和光感受器而导致视力丧失,影响所有年龄段的人。基于细胞的疗法已成为治疗RDD的有效方法,并取得了有希望的结果。这项荟萃分析旨在全面评估细胞疗法治疗年龄相关性黄斑变性(AMD)的疗效。视网膜色素变性(RP),Stargardt黄斑变性(SMD)是最常见的RDD。
    方法:PubMed,Scopus,WebofScience,和Embase使用与各种视网膜疾病和细胞治疗相关的关键词进行搜索,直到11月25日,2023年。使用JoannaBriggs研究所(JBI)的准实验研究清单评估研究质量。以LogMAR评分测量的视力被用作我们的主要结果。使用三级随机效应荟萃分析来探索接受基于细胞的治疗的患者的视力。使用亚组和敏感性分析评估纳入研究之间的异质性。此外,细胞类型的元回归,出版年份,和参与者的平均年龄进行。
    结果:总体而言,通过搜索检索了8345项研究,39人符合资格标准,其中18项研究共224只眼纳入荟萃分析.有12项关于AMD的研究,7在SMD上,2在RP上。AMD的细胞疗法显示LogMAR的显著改善(p<0.05)。此外,细胞治疗降低了SMD和RP的LogMAR评分(分别为p<0.01和p<0.0001)。在所有条件下,未检测到实质性发表偏倚(p<0.05).
    结论:研究结果突出表明,细胞疗法的应用可以提高AMD的视力,SMD,和RP。
    Retinal degenerative disorders (RDDs) cause vision loss by damaging retinal neurons and photoreceptors, affecting individuals of all ages. Cell-based therapy has emerged as an effective approach for the treatment of RDDs with promising results. This meta-analysis aims to comprehensively evaluate the efficacy of cell therapy in treating age-related macular degeneration (AMD), retinitis pigmentosa (RP), and Stargardt macular degeneration (SMD) as the most prevalent RDDs.
    PubMed, Scopus, Web of Science, and Embase were searched using keywords related to various retinal diseases and cell therapy treatments until November 25th, 2023. The studies\' quality was evaluated using the Joanna Briggs Institute\'s (JBI) checklist for quasi-experimental studies. Visual acuity measured as LogMAR score was used as our main outcome. A three-level random-effect meta-analysis was used to explore the visual acuity in patients who received cell-based therapy. Heterogeneity among the included studies was evaluated using subgroup and sensitivity analyses. Moreover, meta-regression for the type of cells, year of publication, and mean age of participants were performed.
    Overall, 8345 studies were retrieved by the search, and 39 met the eligibility criteria, out of which 18 studies with a total of 224 eyes were included in the meta-analysis. There were 12 studies conducted on AMD, 7 on SMD, and 2 on RP. Cell therapy for AMD showed significant improvement in LogMAR (p < 0.05). Also, cell therapy decreased the LogMAR score in SMD and RP (p < 0.01 and p < 0.0001, respectively). Across all conditions, no substantial publication bias was detected (p < 0.05).
    The findings of the study highlight that the application of cell therapy can enhance the visual acuity in AMD, SMD, and RP.
    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:临床上,复发性自然流产(RSA)是一种难以治疗的妊娠疾病。判定化受损是RSA的一个记录在案的原因,但病因和机制尚不清楚。cAMP反应元件结合蛋白5(CREB5)是ATF/CREB家族的成员。据报道,CREB5与病理性妊娠有关,但是在RSA患者中很少有相关的研究,潜在机制尚不清楚。
    方法:我们收集RSA患者和健康孕妇的蜕膜组织,以测量CREB5,PRL,IGFBP1,ATG5,LC3B,和SQSTM/p62。然后,在蜕膜化过程中测量人子宫内膜基质细胞(hESCs)中CREB5表达和自噬水平的变化.在蜕膜化诱导后的sh-CREB5/ov-CREB5hESCs中检测PRL和IGFBP1的表达水平,并且在没有蜕膜化诱导的情况下测量sh-CREB5/ov-CREB5hESCs中的自噬水平。测量用自噬诱导剂或抑制剂处理的sh-CREB5和ov-CREB5hESC的蜕膜化能力。探讨hESCs中CREB5对HTR8/SVneo细胞侵袭和迁移的影响,我们进行了共培养实验。最后,通过构建流产小鼠模型,检测CREB5和自噬关键蛋白在小鼠蜕膜组织中的表达。
    结果:在我们的研究中,我们发现CREB5的表达在RSA患者的子宫蜕膜中异常升高,但是PRL的表达,IGFBP1和自噬均降低。在hESC的蜕膜化过程中,随着自噬的增加,CREB5的表达呈时间依赖性逐渐降低。此外,通过在hESC中敲低或过表达CREB5,发现CREB5可以损害hESCs的蜕膜化和减少自噬。此外,CREB5在蜕膜化方面引起的损伤可以通过添加自噬诱导剂(雷帕霉素)来逆转。此外,CREB5可以增加hESCs中蛋白质(IL-1β和TGF-β1)的分泌,从而抑制滋养细胞的侵袭和迁移。
    结论:我们的数据支持以下假设:CREB5通过抑制自噬干扰子宫内膜基质细胞的蜕膜化和母胎界面的相互作用,其异常的上调和功能障碍可能导致RSA。它可以作为RSA的诊断和治疗靶标。同样,我们发现在自然流产小鼠模型中,流产组蜕膜组织中CREB5的表达显著升高,自噬减少。
    BACKGROUND: Clinically, recurrent spontaneous abortion (RSA) is a pregnancy illness that is difficult to treat. Impaired decidualization is a documented cause of RSA, but the etiology and mechanism are still unknown. cAMP-responsive element binding protein 5 (CREB5) is a member of the ATF/CREB family. CREB5 has been reported to be related to pathological pregnancy, but there are few related studies on this topic in patients with RSA, and the underlying mechanism is unclear.
    METHODS: We collected decidual tissues from RSA patients and healthy pregnant women to measure the expression level of CREB5, PRL, IGFBP1, ATG5, LC3B, and SQSTM/p62. Then, the changes in CREB5 expression and autophagy levels were measured in human endometrial stromal cells (hESCs) during decidualization. The expression levels of PRL and IGFBP1 were tested in sh-CREB5/ov-CREB5 hESCs after decidualization induction, and the autophagy level in sh-CREB5/ov-CREB5 hESCs was measured without decidualization induction. The decidualization ability of sh-CREB5 and ov-CREB5 hESCs treated with an autophagy inducer or inhibitor was measured. To investigate the effect of CREB5 in hESCs on the invasion and migration of HTR8/SVneo cells, we performed a coculture experiment. Finally, we examined the expression of CREB5 and autophagy key proteins in mouse decidual tissues by constructing an abortion mouse model.
    RESULTS: In our study, we found that the expression of CREB5 was unusually elevated in the uterine decidua of RSA patients, but the expression of PRL, IGFBP1, and autophagy were decreased. During the decidualization of hESCs, the expression of CREB5 gradually decreases in a time-dependent manner with increasing autophagy. Moreover, by knocking down or overexpressing CREB5 in hESCs, it was found that CREB5 can impair decidualization and reduce autophagy in hESCs. Furthermore, the damage caused by CREB5 in terms of decidualization can be reversed by the addition of an autophagy inducer (rapamycin). In addition, CREB5 can increase the secretion of proteins (IL-1β and TGF-β1) in hESCs to inhibit trophoblast invasion and migration.
    CONCLUSIONS: Our data support the supposition that CREB5 disturbs the decidualization of endometrial stromal cells and interactions at the maternal-fetal interface by inhibiting autophagy and that its abnormal upregulation and dysfunction may lead to RSA. It may function as a diagnostic and therapeutic target for RSA. Similarly, we found that in the spontaneous abortion mouse model, the expression of CREB5 in the decidual tissue of the abortion group was significantly increased, and autophagy was decreased.
    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
    探索人胚胎干细胞(hESCs)的自我更新和多能性维持机制,在基础研究和临床应用中具有重要意义。但还没有完全阐明。长链非编码RNA(lncRNA)已被证明在hESC的自我更新和多能性维持中起关键作用。我们以前报道过lncRNAESRG,在未分化的hESC中高度表达,可以保持hPSC的自我更新和多能性。RNA下拉质谱显示ESRG可以与其他蛋白质结合,其中异质核核糖核蛋白A1(HNRNPA1)引起了我们的注意。在这项研究中,我们发现HNRNPA1可以维持hESCs的自我更新和多能性。ESRG通过泛素-蛋白酶体途径结合并稳定HNRNPA1蛋白。此外,ESRG或HNRNPA1的敲减导致TCF3的选择性剪接,TCF3最初和主要编码E12,主要编码E47并抑制CDH1表达。HNRNPA1可以挽救ESRG敲低或过表达引起的hESCs生物学功能变化。我们的结果表明,ESRG通过结合和稳定HNRNPA1蛋白来调节TCF3的可变剪接以影响CDH1表达并维持hESCs的自我更新和多能性。本研究为探索ESRG维持hESCs自我更新和多能性的分子调控机制奠定了良好的基础。
    Exploring the mechanism of self-renewal and pluripotency maintenance of human embryonic stem cells (hESCs) is of great significance in basic research and clinical applications, but it has not been fully elucidated. Long non-coding RNAs (lncRNAs) have been shown to play a key role in the self-renewal and pluripotency maintenance of hESCs. We previously reported that the lncRNA ESRG, which is highly expressed in undifferentiated hESCs, can maintain the self-renewal and pluripotency of hPSCs. RNA pull-down mass spectrometry showed that ESRG could bind to other proteins, among which heterogeneous nuclear ribonucleoprotein A1 (HNRNPA1) attracted our attention. In this study, we showed that HNRNPA1 can maintain self-renewal and pluripotency of hESCs. ESRG bound to and stabilized HNRNPA1 protein through the ubiquitin-proteasome pathway. In addition, knockdown of ESRG or HNRNPA1 resulted in alternative splicing of TCF3, which originally and primarily encoded E12, to mainly encode E47 and inhibit CDH1 expression. HNRNPA1 could rescue the biological function changes of hESCs caused by ESRG knockdown or overexpression. Our results suggest that ESRG regulates the alternative splicing of TCF3 to affect CDH1 expression and maintain hESCs self-renewal and pluripotency by binding and stabilizing HNRNPA1 protein. This study lays a good foundation for exploring the new molecular regulatory mechanism by which ESRG maintains hESCs self-renewal and pluripotency.
    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: Preprint
    体外建模是研究驱动人类先天性疾病的病理机制的有力方法。在这里,我们使用人类胚胎干细胞(hESCs)来模拟Nager和Rodriguez综合征,两种颅面疾病的特征是发育不良的神经c衍生的颅面骨,由SF3B4的致病变体引起,SF3B4是剪接体的核心组成部分。我们观察到siRNA介导的SF3B4敲低干扰了hESC衍生的神经c细胞的产生,如神经c基因表达显着减少所见。这种表型与神经c细胞凋亡及其过早神经元分化的增加有关。一起,这些结果表明SF3B4在神经c细胞存活中的作用,维护,和分化为Nager/Rodriguez综合征相关颅面缺损的主要原因,并说明体外人类干细胞模型对理解先天性疾病的益处。
    In vitro modeling is a powerful approach to investigate the pathomechanisms driving human congenital conditions. Here we use human embryonic stem cells (hESCs) to model Nager and Rodriguez syndromes, two craniofacial conditions characterized by hypoplastic neural crest-derived craniofacial bones, caused by pathogenic variants of SF3B4, a core component of the spliceosome. We observed that siRNA-mediated knockdown of SF3B4 interferes with the production of hESC-derived neural crest cells, as seen by a marked reduction in neural crest gene expression. This phenotype is associated with an increase in neural crest cell apoptosis and premature neuronal differentiation. Altogether these results point at a role of SF3B4 in neural crest cell survival, maintenance, and differentiation. We propose that the dysregulation of these processes may contribute to Nager/Rodriguez syndrome associated craniofacial defects.
    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(PSC)是用于再生疗法的干细胞的有希望的来源。干细胞功能取决于端粒维持机制,端粒维持机制为它们提供增殖和再生组织所必需的增殖能力和基因组稳定性。我们在这里表明,建立的人胚胎干细胞(hESC)具有稳定的端粒长度,其依赖于端粒酶,但不依赖于基于同源重组途径的替代机制。这里,我们表明,从体细胞重编程的人诱导的多能干细胞(hiPSCs)显示出逐渐的端粒延长,直到达到与ESC相似的长度。hiPSC还获得ESC的端粒染色质标记,包括三甲基化组蛋白H3K9和H4K20和HP1异色标记的丰度降低,以及shelterin组件TRF2。这些染色质特征伴随着端粒转录物或TERRAs的丰度增加。我们还发现,hESCs和hiPSCs的端粒在端粒延伸过程中都能很好地保护DNA免受损伤,一旦达到完整的端粒长度,并表现出稳定的基因组。总的来说,这项研究强调了hiPSC在重编程过程中获得了ESC特征,并揭示了人类多能干细胞(hPSC)的端粒生物学。
    我们显示已建立的人胚胎干细胞(hESC)具有最大且稳定的端粒长度,该端粒长度取决于端粒酶,但不取决于替代同源重组途径或ALT。从体细胞重编程的人诱导多能干细胞(hiPSCs)显示出进行性端粒延长,直到达到与ESC相似的最大端粒长度,表明端粒长度受人类细胞表观遗传机制的调节。在这方面,hiPSC获得“开放染色质”特征的端粒染色质标记,包括端粒转录物或TERRA的丰度增加。hESC和hiPSC的端粒在端粒延伸过程中得到很好的保护并且表现出稳定的基因组。总的来说,这项研究强调了hiPSCs在重编程过程中获得了ESC特征,并揭示了人类多能干细胞(hiPSCs)的端粒生物学。
    Pluripotent stem cells (PSCs) are a promising source of stem cells for regenerative therapies. Stem cell function depends on telomere maintenance mechanisms that provide them with the proliferative capacity and genome stability necessary to multiply and regenerate tissues. We show here that established human embryonic stem cells (hESCs) have stable telomere length that is dependent on telomerase but not on alternative mechanisms based on homologous recombination pathways. Here, we show that human-induced pluripotent stem cells (hiPSCs) reprogrammed from somatic cells show progressive telomere lengthening until reaching a length similar to ESCs. hiPSCs also acquire telomeric chromatin marks of ESCs including decreased abundance of tri-methylated histone H3K9 and H4K20 and HP1 heterochromatic marks, as well as of the shelterin component TRF2. These chromatin features are accompanied with increased abundance of telomere transcripts or TERRAs. We also found that telomeres of both hESCs and hiPSCs are well protected from DNA damage during telomere elongation and once full telomere length is achieved, and exhibit stable genomes. Collectively, this study highlights that hiPSCs acquire ESC features during reprogramming and reveals the telomere biology in human pluripotent stem cells (hPSCs).
    We show that established human embryonic stem cells (hESCs) have a maximum and stable telomere length that is dependent on telomerase but not on the alternative homologous recombination pathway or ALT. Human-induced pluripotent stem cells (hiPSCs) reprogrammed from somatic cells show progressive telomere lengthening until reaching a length similar maximum telomere length than ESCs, suggesting that telomere length is regulated by epigenetic mechanisms in human cells. In this regard, hiPSCs acquire telomeric chromatin marks characteristic of an “open chromatin” including increased abundance of telomere transcripts or TERRAs. Telomeres of both hESCs and hiPSCs are well protected during telomere elongation and exhibit stable genomes. Collectively, this study highlights that hiPSCs acquire ESC features during reprogramming and reveals the telomere biology in human pluripotent stem cells (hiPSCs).
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

公众号