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
    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: Journal Article
    人胚胎干细胞(hESC)是大规模和均质的间充质干细胞/基质细胞(MSC)产生的有利来源。然而,由于hESC-MSCs诱导的高效程序的局限性,有关中膜发生和早期MSC发育的系统和详细信息在很大程度上是模糊的。在这项研究中,我们利用已确立的扭曲相关蛋白1(TWIST1)过表达hESCs和两种小分子混合物(CHIR99021,地西他滨)进行高效MSC诱导.为了评估多维生物学和转录组特征,我们转向细胞和分子方法,如流式细胞术(FCM),定量逆转录聚合酶链反应(qRT-PCR),体外三谱系分化,细胞因子分泌分析,体内移植治疗急性肝损伤(ALI),和生物信息学分析(例如,基因本体论-生物过程[GO-BP],京都基因和基因组百科全书[KEGG],HeatMap,和主成分分析[PCA])。通过组合TWIST1过表达(表示为T)和所示的小分子混合物(表示为S),hESCs高效分化成MSCs(表示为TS-MSCs,由T和S联合诱导)在2周内。TS-MSC满足MSC定义的标准,并显示出相当的三谱系分化潜力和对ALI小鼠的改善功效。根据RNA测序(SEQ)分析,我们最初阐明了基因表达模式的逐渐变化和程序性hESC-MSCs伴随的生物功能。总的来说,我们的数据表明通过TWIST1和基于混合物的编程高效生成hESC-MSCs的可行性.产生的hESC-MSCs在体内和体外表现出多方面的生物功能,作为成年BM-MSCs,这共同暗示了未来ALI管理的前景。
    Human embryonic stem cells (hESCs) are advantaged sources for large-scale and homogeneous mesenchymal stem/stromal cells (MSCs) generation. However, due to the limitations in high-efficiency procedures for hESC-MSCs induction, the systematic and detailed information of mesengenesis and early MSC development are largely obscure. In this study, we took advantage of the well-established twist-related protein 1 (TWIST1)-overexpressing hESCs and two small molecular cocktails (CHIR99021, decitabine) for high-efficient MSC induction. To assess the multidimensional biological and transcriptomic characteristics, we turned to cellular and molecular methods, such as flow cytometry (FCM), quantitative reverse transcription-polymerase chain reaction (qRT-PCR), in vitro tri-lineage differentiation, cytokine secretion analysis, in vivo transplantation for acute liver injury (ALI) management, and bioinformatics analyses (eg, gene ontology-biological processes [GO-BP], Kyoto Encyclopedia of Genes and Genomes [KEGG], HeatMap, and principal component analysis [PCA]). By combining TWIST1 overexpression (denoted as T) and the indicated small molecular cocktails (denoted as S), hESCs high-efficiently differentiated into MSCs (denoted as TS-MSCs, induced by T and S combination) within 2 weeks. TS-MSCs satisfied the criteria for MSC definition and revealed comparable tri-lineage differentiation potential and ameliorative efficacy upon ALI mice. According to RNA-sequencing (SEQ) analysis, we originally illuminated the gradual variations in gene expression pattern and the concomitant biofunctions of the programmed hESC-MSCs. Overall, our data indicated the feasibility of high-efficient generation of hESC-MSCs by TWIST1 and cocktail-based programming. The generated hESC-MSCs revealed multifaceted in vivo and in vitro biofunctions as adult BM-MSCs, which collectively suggested promising prospects in ALI management in future.
    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
    Polycomb抑制复合物(PRCs)通过介导组蛋白修饰(如H3K27me3和H2AK119ub)在正常发育和疾病进展期间在细胞命运决定中发挥关键作用。如何将PRC招募到染色质中仍有待充分阐明。这里,我们报道了YTHDF1,N6-甲基腺嘌呤(m6A)RNA阅读器,以前已知主要是细胞质,与RNF2相关,RNF2是一种在人胚胎干细胞(hESC)中介导H2AK119ub的PRC1蛋白。YTHDF1的一部分位于细胞核中,并在与神经发育功能相关的基因位点子集上与RNF2/H2AK119ub相关联。敲低YTHDF1减弱对这些基因的H2AK119ub修饰并促进hESC中的神经分化。我们的发现提供了YTHDF1参与hESC中PRC1功能的非规范机制。
    Polycomb repressive complexes (PRCs) play critical roles in cell fate decisions during normal development as well as disease progression through mediating histone modifications such as H3K27me3 and H2AK119ub. How exactly PRCs recruited to chromatin remains to be fully illuminated. Here, we report that YTHDF1, the N6-methyladenine (m6 A) RNA reader that was previously known to be mainly cytoplasmic, associates with RNF2, a PRC1 protein that mediates H2AK119ub in human embryonic stem cells (hESCs). A portion of YTHDF1 localizes in the nuclei and associates with RNF2/H2AK119ub on a subset of gene loci related to neural development functions. Knock-down YTHDF1 attenuates H2AK119ub modification on these genes and promotes neural differentiation in hESCs. Our findings provide a noncanonical mechanism that YTHDF1 participates in PRC1 functions in 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
    METTL3,一种负责N6-甲基腺苷(m6A)修饰的甲基转移酶,在哺乳动物中枢神经系统(CNS)发育中起着关键的调节作用。然而,控制人类中枢神经系统发育的特定表观遗传机制仍未阐明。这里,我们产生了小分子辅助关闭(SMASh)标记的hESC系,以降低METTL3蛋白水平,并发现METTL3不是人类神经祖细胞(hNPC)形成和神经元分化所必需的。然而,METTL3缺乏通过降低SLIT2表达抑制hNPC增殖。机制研究表明,METTL3在hNPCs中的降解显着降低了SLIT2mRNA中m6A的富集,因此减少了它的表达。我们的发现揭示了hNPC中METTL3的新功能靶标(SLIT2),并有助于更好地理解hNPC增殖中m6A依赖性机制。
    METTL3, a methyltransferase responsible for N6-methyladenosine (m6A) modification, plays key regulatory roles in mammal central neural system (CNS) development. However, the specific epigenetic mechanisms governing human CNS development remain poorly elucidated. Here, we generated small-molecule-assisted shut-off (SMASh)-tagged hESC lines to reduce METTL3 protein levels, and found that METTL3 is not required for human neural progenitor cell (hNPC) formation and neuron differentiation. However, METTL3 deficiency inhibited hNPC proliferation by reducing SLIT2 expression. Mechanistic studies revealed that METTL3 degradation in hNPCs significantly decreased the enrichment of m6A in SLIT2 mRNA, consequently reducing its expression. Our findings reveal a novel functional target (SLIT2) for METTL3 in hNPCs and contribute to a better understanding of m6A-dependent mechanisms in hNPC proliferation.
    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
    人类干细胞的实验室培养寻求捕获细胞状态作为生物系统的体外替代。为了使这项研究的结果和输出准确,有意义的,耐用,应采用确保数据再现性和可靠性的标准。尽管以前已经为存储库和分发中心提出了此类标准,对于人类多能干细胞和组织干细胞的实验室研究,尚无广泛接受的最佳实践。为了填补这个空白,国际干细胞研究协会提出了一套建议,包括报告标准,为基础研究实验室的科学家。这些标准的设计在技术上和财务上是可行的,当实施时,增强干细胞研究的可重复性和严谨性。
    The laboratory culture of human stem cells seeks to capture a cellular state as an in vitro surrogate of a biological system. For the results and outputs from this research to be accurate, meaningful, and durable, standards that ensure reproducibility and reliability of the data should be applied. Although such standards have been previously proposed for repositories and distribution centers, no widely accepted best practices exist for laboratory research with human pluripotent and tissue stem cells. To fill that void, the International Society for Stem Cell Research has developed a set of recommendations, including reporting criteria, for scientists in basic research laboratories. These criteria are designed to be technically and financially feasible and, when implemented, enhance the reproducibility and rigor of stem cell research.
    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
    最近的研究发现,lncRNA-MEG3(MEG3)在子宫内膜异位症(EMs)的发生发展过程中发挥着重要作用。但具体机制有待进一步探讨。本研究旨在探讨MEG3对细胞增殖的影响。EMs细胞的侵袭。作者使用RT-qPCR检测MEG3和miR-21-5p在EMs组织和hESCs细胞中的表达,MTT和Transwell检测细胞增殖和侵袭,蛋白质印迹法检测DNMT3B和Twist的表达,MSP检测Twist的甲基化。本研究的检测结果表明MEG3在EMs组织和hESCs细胞中低表达,过表达MEG3可下调miR-21-5p,抑制子宫内膜细胞增殖和侵袭。此外,MEG3的过表达上调DNMT3B的表达并促进TWIST的甲基化。总之,目前的研究结果表明,MEG3在EMs组织中下调,过表达MEG3可通过下调miR-21-5p促进DNA甲基转移酶DNMT3B的活性,从而促进Twist的甲基化,下调Twist水平抑制hESCs细胞增殖和侵袭。
    Recent studies have found that lncRNA-MEG3(MEG3) plays an important role in the development of EMs (Endometriosis), but the specific mechanism needs to be further explored. This study aimed to investigate the effect of MEG3 on the proliferation, invasion of EMs cells. The authors used RT-qPCR to detect the expression of MEG3 and miR-21-5p in EMs tissues and hESCs cells, MTT and Transwell to detect cell proliferation and invasion, western blotting assay to detect the expression of DNMT3B and Twist, MSP to detect the methylation of Twist. The present study\'s detection results showed that MEG3 was lowly expressed in EMs tissues and hESCs cells, and overexpression of MEG3 could down-regulate miR-21-5p and inhibit endometrial cell proliferation and invasion. In addition, overexpression of MEG3 upregulated the expression of DNMT3B and promoted the methylation of TWIST. In conclusion, the present findings suggest that MEG3 is downregulated in EMs tissues, and overexpression of MEG3 can promote the activity of DNA methyltransferase DNMT3B by downregulating miR-21-5p, thereby promoting the methylation of Twist, downregulating Twist level to inhibits hESCs cells proliferation and invasion.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Letter
    将人胚胎干细胞诱导为NEUROG3+胰腺内分泌细胞的效率是糖尿病干细胞治疗的瓶颈。为了了解分化过程中的细胞特性和命运决定,我们使用单细胞转录组分析了改良的诱导方法,发现DAPT结合了四个因素(4FS):烟酰胺,地塞米松,毛喉素和Alk5抑制剂II(DAPT+4FS)将NEUROG3的表达增加到约34.3%。增加的NEUROG3+细胞主要集中在胰岛素+胰高血糖素+(INS+GCG+)和SLAC18A1+嗜铬粒蛋白A+(SLAC18A1+CHGA+)群体中,表明NEUROG3+细胞的增加促进了胰腺内分泌细胞和肠嗜铬细胞的分化。单细胞转录组分析为进一步筛选胰腺内分泌细胞和胰岛细胞分化提供了有价值的线索。基因集富集分析(GSEA)提示我们可以尝试通过上调G蛋白偶联受体(GPCR)和丝裂原活化蛋白激酶信号和下调Wnt来促进INS+GCG+群体的表达,NIK/NF-κB与细胞因子介导的信号通路。我们也可以尝试通过PLCE1调节GPCR信号,从而增加INS+GCG+群体中NEUROG3+细胞的比例。为了排除非胰腺内分泌细胞,ALCAMhighCD9low可用作内分泌人群的标志物,ALCAMhighCD9lowCDH1低可以去除SLC18A1+CHGA+群体。
    The efficiency of inducing human embryonic stem cells into NEUROG3+ pancreatic endocrine cells is a bottleneck in stem cell therapy for diabetes. To understand the cell properties and fate decisions during differentiation, we analyzed the modified induction method using single-cell transcriptome and found that DAPT combined with four factors (4FS): nicotinamide, dexamethasone, forskolin and Alk5 inhibitor II (DAPT + 4FS) increased the expression of NEUROG3 to approximately 34.3%. The increased NEUROG3+ cells were mainly concentrated in Insulin + Glucagon + (INS + GCG+) and SLAC18A1 + Chromogranin A+(SLAC18A1 + CHGA +) populations, indicating that the increased NEUROG3+ cells promoted the differentiation of pancreatic endocrine cells and enterochromaffin-like cells. Single-cell transcriptome analysis provided valuable clues for further screening of pancreatic endocrine cells and differentiation of pancreatic islet cells. The gene set enrichment analysis (GSEA) suggest that we can try to promote the expression of INS + GCG+ population by up-regulating G protein-coupled receptor (GPCR) and mitogen-activated protein kinase signals and down-regulating Wnt, NIK/NF-KappaB and cytokine-mediated signal pathways. We can also try to regulate GPCR signaling through PLCE1, so as to increase the proportion of NEUROG3+ cells in INS+GCG+ populations. To exclude non-pancreatic endocrine cells, ALCAMhigh CD9low could be used as a marker for endocrine populations, and ALCAMhigh CD9lowCDH1low could remove the SLC18A1 + CHGA+ population.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

公众号