Pluripotent Stem Cells

多能干细胞
  • 文章类型: Journal Article
    原理:转录因子(TFs)的调节过程塑造心脏发育并影响成年心脏对压力的反应,导致心脏疾病。尽管意义重大,支持TF介导调节的精确机制仍然难以捉摸。这里,我们确定EBF1,作为TF,在人类心脏组织中高度表达。据报道,EBF1与人类心血管疾病有关,但是它的角色在心里并不清楚。在这项研究中,我们研究了EBF1在心脏系统中的功能。方法:利用RNA-seq对EBF1表达模式进行分析。CRISPR/Cas9用于敲除EBF1以研究其作用。分化为心脏谱系的人多能干细胞(hPSC)用于模拟心脏发育。通过使用诸如超声心动图的技术在具有Ebf1敲除的小鼠模型上评估心脏功能。进行RNA-seq以分析转录扰动。ChIP-seq用于阐明EBF1结合的基因和潜在的调节机制。结果:EBF1在部分人和小鼠心肌细胞中均有表达。EBF1基因敲除抑制心脏发育。ChIP-seq表明EBF1对心脏发育至关重要的心源性TFs启动子的结合,促进其转录表达和促进心脏发育。在老鼠身上,Ebf1耗竭触发了基因的转录扰动,导致心脏重塑。机械上,我们发现EBF1直接与心脏肥大诱导基因的上游染色质区域结合,导致心脏肥大.结论:我们揭示了EBF1介导的调节过程的潜在机制,照亮心脏发育,和心脏重塑的发病机制。这些发现强调了EBF1在协调心脏过程的各个方面的关键作用,并为心肌病提供了有希望的治疗干预。
    Rationale: Regulatory processes of transcription factors (TFs) shape heart development and influence the adult heart\'s response to stress, contributing to cardiac disorders. Despite their significance, the precise mechanisms underpinning TF-mediated regulation remain elusive. Here, we identify that EBF1, as a TF, is highly expressed in human heart tissues. EBF1 is reported to be associated with human cardiovascular disease, but its roles are unclear in heart. In this study, we investigated EBF1 function in cardiac system. Methods: RNA-seq was utilized to profile EBF1 expression patterns. CRISPR/Cas9 was utilized to knock out EBF1 to investigate its effects. Human pluripotent stem cells (hPSCs) differentiated into cardiac lineages were used to mimic cardiac development. Cardiac function was evaluated on mouse model with Ebf1 knockout by using techniques such as echocardiography. RNA-seq was conducted to analyze transcriptional perturbations. ChIP-seq was employed to elucidate EBF1-bound genes and the underlying regulatory mechanisms. Results: EBF1 was expressed in some human and mouse cardiomyocyte. Knockout of EBF1 inhibited cardiac development. ChIP-seq indicated EBF1\'s binding on promoters of cardiogenic TFs pivotal to cardiac development, facilitating their transcriptional expression and promoting cardiac development. In mouse, Ebf1 depletion triggered transcriptional perturbations of genes, resulting in cardiac remodeling. Mechanistically, we found that EBF1 directly bound to upstream chromatin regions of cardiac hypertrophy-inducing genes, contributing to cardiac hypertrophy. Conclusions: We uncover the mechanisms underlying EBF1-mediated regulatory processes, shedding light on cardiac development, and the pathogenesis of cardiac remodeling. These findings emphasize EBF1\'s critical role in orchestrating diverse aspects of cardiac processes and provide a promising therapeutic intervention for cardiomyopathy.
    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
    心脏生物起搏(BP)是缓慢性心律失常的未来干预方向之一。目前,用于心脏BP的心脏起搏器细胞(PC)主要来自多能干细胞(PSC)。然而,从PSC生产高质量的心脏PC仍然是一个挑战。这里,我们通过采用双重PC标记并模拟PC的发育路线,开发了一种心脏PC分化策略。首先,两个PC标记,选择Shox2和Hcn4以建立Shox2:EGFP;Hcn4:mCherry小鼠PSC报道系。然后,通过逐步引导幼稚PSC到心脏PC,在幼稚到形成多能性转变和在心脏PC分化过程中操纵信号通路,我们设计了FSK方法,提高了SHOX2+的产量;HCN4+细胞具有典型的PC特性,分别比拟胚体(EB)和单层M10方法高12倍和42倍。此外,通过单细胞RNA测序(scRNA-seq)绘制体外心脏PCs分化轨迹,这类似于体内PCs的发展,ZFP503被证实是心脏PCs分化的关键调节因子。这些PSC衍生的心脏PC具有推动心脏BP技术进步的潜力,帮助了解PC(病理)生理学,并有益于PC相关疾病的药物发现。
    Cardiac biological pacing (BP) is one of the future directions for bradyarrhythmias intervention. Currently, cardiac pacemaker cells (PCs) used for cardiac BP are mainly derived from pluripotent stem cells (PSCs). However, the production of high-quality cardiac PCs from PSCs remains a challenge. Here, we developed a cardiac PC differentiation strategy by adopting dual PC markers and simulating the developmental route of PCs. First, two PC markers, Shox2 and Hcn4, were selected to establish Shox2:EGFP; Hcn4:mCherry mouse PSC reporter line. Then, by stepwise guiding naïve PSCs to cardiac PCs following naïve to formative pluripotency transition and manipulating signaling pathways during cardiac PCs differentiation, we designed the FSK method that increased the yield of SHOX2+; HCN4+ cells with typical PC characteristics, which was 12 and 42 folds higher than that of the embryoid body (EB) and the monolayer M10 methods respectively. In addition, the in vitro cardiac PCs differentiation trajectory was mapped by single-cell RNA sequencing (scRNA-seq), which resembled in vivo PCs development, and ZFP503 was verified as a key regulator of cardiac PCs differentiation. These PSC-derived cardiac PCs have the potential to drive advances in cardiac BP technology, help with the understanding of PCs (patho)physiology, and benefit drug discovery for PC-related diseases as well.
    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
    原始生殖细胞(PGCs)是配子的前体,也是动物跨代传递遗传信息的唯一机制。在小鼠胚胎中,PGC规范的转录和表观遗传调控已被广泛表征。然而,引发soma-种系分离的初始事件仍然知之甚少.这里,我们揭示了基底膜在调节种系进入中的关键作用。我们发现PGCs出现在小鼠胚胎缺乏与基底膜接触的区域,外源性细胞外基质(ECM)的添加抑制小鼠胚胎和干细胞模型中的PGC和PGC样细胞(PGCLC)规格,分别。机械上,我们证明了β1整合素与层粘连蛋白的结合通过阻止PGC转录阻遏物的Wnt信号依赖性下调来阻断PGCLC的特异性,Otx2。这样,细胞远离基底膜的物理分离充当形态发生命运开关,控制体细胞-种系分叉。
    Primordial germ cells (PGCs) are the precursors of gametes and the sole mechanism by which animals transmit genetic information across generations. In the mouse embryo, the transcriptional and epigenetic regulation of PGC specification has been extensively characterized. However, the initial event that triggers the soma-germline segregation remains poorly understood. Here, we uncover a critical role for the basement membrane in regulating germline entry. We show that PGCs arise in a region of the mouse embryo that lacks contact with the basement membrane, and the addition of exogenous extracellular matrix (ECM) inhibits both PGC and PGC-like cell (PGCLC) specification in mouse embryos and stem cell models, respectively. Mechanistically, we demonstrate that the engagement of β1 integrin with laminin blocks PGCLC specification by preventing the Wnt signaling-dependent down-regulation of the PGC transcriptional repressor, Otx2. In this way, the physical segregation of cells away from the basement membrane acts as a morphogenetic fate switch that controls the soma-germline bifurcation.
    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
    动脉内皮细胞(AECs)是胚胎内造血的基础细胞。这里,我们报道了一种从多能干细胞(PSC)高效生成人血源性DLL4+AECs的方法.时间序列单细胞RNA测序揭示了造血和淋巴细胞生成的动态进化,产生细胞类型与早期人类胚胎中存在的对应物,包括以造血前干细胞基因MECOM/EVI1,MLLT3和SPINK2标记的阶段。DLL4+AECs强烈支持淋巴样分化,不需要外源性NOTCH配体。使用这个系统,我们发现IL7作为一种形态发生因子,决定了T和先天淋巴谱系之间的命运选择,并且在调节RAG1的相对表达水平中也起着作用。此外,我们记录了人类RAG1淋巴样前体产生自然杀伤细胞谱系的发育途径。我们的研究描述了一种产生淋巴祖细胞的有效方法,提供对它们的内皮和造血个体发育的见解,建立研究人类血液系统发展的平台。
    Arterial endothelial cells (AECs) are the founder cells for intraembryonic haematopoiesis. Here, we report a method for the efficient generation of human haemogenic DLL4+ AECs from pluripotent stem cells (PSC). Time-series single-cell RNA-sequencing reveals the dynamic evolution of haematopoiesis and lymphopoiesis, generating cell types with counterparts present in early human embryos, including stages marked by the pre-haematopoietic stem cell genes MECOM/EVI1, MLLT3 and SPINK2. DLL4+ AECs robustly support lymphoid differentiation, without the requirement for exogenous NOTCH ligands. Using this system, we find IL7 acts as a morphogenic factor determining the fate choice between the T and innate lymphoid lineages and also plays a role in regulating the relative expression level of RAG1. Moreover, we document a developmental pathway by which human RAG1+ lymphoid precursors give rise to the natural killer cell lineage. Our study describes an efficient method for producing lymphoid progenitors, providing insights into their endothelial and haematopoietic ontogeny, and establishing a platform to investigate the development of the human blood system.
    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:早产儿慢性肺病,称为支气管肺发育不良(BPD),缺乏有效的治疗方法,强调需要反映人类病理学的临床前测试系统来识别因果途径和测试新化合物。源自人类多能干细胞(hPSC)的肺泡类器官是研究BPD等远端气道疾病的有前途的测试平台。但是目前的方案不能准确复制天然肺的远端小生境环境。在这里,我们研究了肺泡的细胞成分和胎儿呼吸运动对hPSC衍生的肺泡类器官形成的贡献。
    方法:人PSC在2D培养物中分化成肺祖细胞(LPC),然后在去除共发育的中胚层细胞之前和之后进一步分化成肺泡类器官。LPC也在有和没有人胎儿肺成纤维细胞的Transwell®共培养物中分化。使用Flexcell®系统使形成的类器官经受阶段性机械应变。通过流式细胞术评估类器官和Transwell®培养物中的分化,免疫荧光,和qPCR的肺上皮和肺泡分化标志物,包括GATA结合蛋白6(GATA6),E-钙黏着蛋白(CDH1),NK2Homeobox1(NKX2-1),HT2-280、表面活性蛋白B(SFTPB)和C(SFTPC)。
    结果:我们观察到共同发育的间充质祖细胞促进hPSC衍生的肺类器官内的肺泡2型上皮细胞(AEC2)分化。通过共培养hPSC-NKX2-1+肺祖细胞与人胚肺成纤维细胞证实了这种对AEC2分化的间质效应。刺激作用不需要成纤维细胞和NKX2-1+肺祖细胞之间的直接接触。此外,我们证明了hPSC衍生的肺类器官的阵发性机械变形,模仿原位胎儿呼吸运动,AEC2分化增加而不影响近端上皮分化。
    结论:我们的数据表明,生物物理和间充质成分在体外促进hPSC衍生的远端类器官内的AEC2分化。
    BACKGROUND: Chronic lung disease of prematurity, called bronchopulmonary dysplasia (BPD), lacks effective therapies, stressing the need for preclinical testing systems that reflect human pathology for identifying causal pathways and testing novel compounds. Alveolar organoids derived from human pluripotent stem cells (hPSC) are promising test platforms for studying distal airway diseases like BPD, but current protocols do not accurately replicate the distal niche environment of the native lung. Herein, we investigated the contributions of cellular constituents of the alveolus and fetal respiratory movements on hPSC-derived alveolar organoid formation.
    METHODS: Human PSCs were differentiated in 2D culture into lung progenitor cells (LPC) which were then further differentiated into alveolar organoids before and after removal of co-developing mesodermal cells. LPCs were also differentiated in Transwell® co-cultures with and without human fetal lung fibroblast. Forming organoids were subjected to phasic mechanical strain using a Flexcell® system. Differentiation within organoids and Transwell® cultures was assessed by flow cytometry, immunofluorescence, and qPCR for lung epithelial and alveolar markers of differentiation including GATA binding protein 6 (GATA 6), E-cadherin (CDH1), NK2 Homeobox 1 (NKX2-1), HT2-280, surfactant proteins B (SFTPB) and C (SFTPC).
    RESULTS: We observed that co-developing mesenchymal progenitors promote alveolar epithelial type 2 cell (AEC2) differentiation within hPSC-derived lung organoids. This mesenchymal effect on AEC2 differentiation was corroborated by co-culturing hPSC-NKX2-1+ lung progenitors with human embryonic lung fibroblasts. The stimulatory effect did not require direct contact between fibroblasts and NKX2-1+ lung progenitors. Additionally, we demonstrate that episodic mechanical deformation of hPSC-derived lung organoids, mimicking in situ fetal respiratory movements, increased AEC2 differentiation without affecting proximal epithelial differentiation.
    CONCLUSIONS: Our data suggest that biophysical and mesenchymal components promote AEC2 differentiation within hPSC-derived distal organoids in vitro.
    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
    茎尖分生组织(SAM)包含产生植物的所有地上部分的多能干细胞。干细胞经历不对称细胞分裂以自我更新并产生分化细胞。我们的研究集中在揭示干细胞分裂后这两种不同细胞命运规范的机制。为此,我们使用了模型生物Physcomitriumpatens,其特征是称为配子体顶端细胞的单一多能干细胞。我们证明细胞分裂素的活性,关键的干细胞调节剂,由于PpLOG的特定定位,仅限于配子体顶端细胞,负责细胞分裂素激活的酶。反过来,PpTAW,这促进了植物的细胞身份的分化,通过细胞分裂素的作用从配子体顶端细胞中排除。我们提出了一种基于细胞分裂素的模型,用于建立多能干细胞分裂中的不对称性。
    The shoot apical meristem (SAM) contains pluripotent stem cells that produce all the aerial parts of the plant. Stem cells undergo asymmetric cell divisions to self-renew and to produce differentiating cells. Our research focused on unraveling the mechanisms governing the specification of these two distinct cell fates following the stem cell division. For this purpose, we used the model organism Physcomitrium patens, which features a singular pluripotent stem cell known as the gametophore apical cell. We show that the activity of cytokinins, critical stem cell regulators, is restricted to the gametophore apical cell due to the specific localization of PpLOG, the enzyme responsible for cytokinin activation. In turn, PpTAW, which promotes differentiating cell identity of the merophyte, is excluded from the gametophore apical cell by the action of cytokinins. We propose a cytokinin-based model for the establishment of asymmetry in the pluripotent stem cell division.
    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
    临床前数据已经证实,人类多能干细胞衍生的心肌细胞(PSC-CM)可以使受伤或患病的心脏重新肌化,几项临床试验目前处于计划或招募阶段。然而,因为室性心律失常是心肌内注射PSC-CM植入后的并发症,有必要提供治疗策略来控制或预防植入心律失常(EA)。这里,我们在猪心肌梗死和PSC-CM移植模型中显示,EA与输入PSC-CM和所得移植物中的细胞异质性有机械联系。具体来说,我们确定心房和起搏器样心肌细胞是导致心律失常的罪犯亚群.两个独特的表面标记签名,信号调节蛋白α(SIRPA)+CD90-CD200+和SIRPA+CD90-CD200-,识别心律失常性和非致心律失常性心肌细胞,分别。我们的数据表明,对当前PSC-CM生产和/或PSC-CM选择协议的修改可能会阻止EA。我们进一步表明,药理学和介入性抗心律失常策略可以控制并可能消除这些心律失常。
    Preclinical data have confirmed that human pluripotent stem cell-derived cardiomyocytes (PSC-CMs) can remuscularize the injured or diseased heart, with several clinical trials now in planning or recruitment stages. However, because ventricular arrhythmias represent a complication following engraftment of intramyocardially injected PSC-CMs, it is necessary to provide treatment strategies to control or prevent engraftment arrhythmias (EAs). Here, we show in a porcine model of myocardial infarction and PSC-CM transplantation that EAs are mechanistically linked to cellular heterogeneity in the input PSC-CM and resultant graft. Specifically, we identify atrial and pacemaker-like cardiomyocytes as culprit arrhythmogenic subpopulations. Two unique surface marker signatures, signal regulatory protein α (SIRPA)+CD90-CD200+ and SIRPA+CD90-CD200-, identify arrhythmogenic and non-arrhythmogenic cardiomyocytes, respectively. Our data suggest that modifications to current PSC-CM-production and/or PSC-CM-selection protocols could potentially prevent EAs. We further show that pharmacologic and interventional anti-arrhythmic strategies can control and potentially abolish these arrhythmias.
    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)在再生医学中至关重要,然而它们的体外扩增往往会导致遗传异常,引起人们对其在临床应用中的安全性的担忧。这项研究分析了多个传代的十种人类胚胎干细胞系,以阐明380个癌症相关基因中染色体异常和单核苷酸变异(SNV)的动力学。延长体外培养导致80%的品系获得染色体20q或1q的增益,两者都以赋予体外生长优势而闻名。70%的品系还获得了轮回集外的其他拷贝数变体(CNV)。此外,我们在88个基因中检测到122个SNV,所有品系在培养期间获得至少一个从头SNV。我们的研究结果表明,在以后的传代中,CNV和SNV的负荷都较高,这是由于在较长的培养时间内累积获得突变,而不是随着时间的推移诱变率增加。重要的是,我们观察到SNVs和稀有CNVs在1q和20q获得染色体增益后,而大多数低传代和遗传平衡的样本没有癌症相关突变。这表明复发性染色体异常是获得其他突变的潜在驱动因素。
    Human pluripotent stem cells (hPSCs) are pivotal in regenerative medicine, yet their in vitro expansion often leads to genetic abnormalities, raising concerns about their safety in clinical applications. This study analyzed ten human embryonic stem cell lines across multiple passages to elucidate the dynamics of chromosomal abnormalities and single-nucleotide variants (SNVs) in 380 cancer-related genes. Prolonged in vitro culture resulted in 80% of the lines acquiring gains of chromosome 20q or 1q, both known for conferring an in vitro growth advantage. 70% of lines also acquired other copy number variants (CNVs) outside the recurrent set. Additionally, we detected 122 SNVs in 88 genes, with all lines acquiring at least one de novo SNV during culture. Our findings showed higher loads of both CNVs and SNVs at later passages, which were due to the cumulative acquisition of mutations over a longer time in culture, and not to an increased rate of mutagenesis over time. Importantly, we observed that SNVs and rare CNVs followed the acquisition of chromosomal gains in 1q and 20q, while most of the low-passage and genetically balanced samples were devoid of cancer-associated mutations. This suggests that recurrent chromosomal abnormalities are potential drivers for the acquisition of other mutations.
    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
    胚胎干细胞(ESCs)具有高活性的泛素-蛋白酶体系统-细胞中蛋白质降解的分子机制。包含不同亚基和相互作用调节剂的各种形式的蛋白酶体复合物负责底物选择性和降解。免疫蛋白酶体是这些在抗原呈递中起重要作用的形式之一;然而,最近的大量证据表明它们在多能干细胞中的功能。以前的研究已经建立了三个连续的多能性阶段,以上胚细胞及其培养的对应物为特征:幼稚,形成性,和准备阶段。在这项工作中,我们报告说,免疫蛋白酶体及其伴侣共调节因子在幼稚状态下被抑制,但在多能性连续体的形成阶段容易上调,以上皮样细胞(EpiLCs)为特征。我们的数据为进一步研究免疫蛋白酶体在哺乳动物早期发育过程中调节蛋白质停滞的生物学功能奠定了基础。
    Embryonic stem cells (ESCs) are remarkable for the high activity level of ubiquitin-proteasome system-the molecular machinery of protein degradation in the cell. Various forms of the proteasome complexes comprising different subunits and interacting regulators are responsible for the substrate selectivity and degradation. Immunoproteasomes are amongst these forms which play an important role in antigen presentation; however, a body of recent evidence suggests their functions in pluripotent stem cells. Previous studies have established three consecutive phases of pluripotency, featured by epiblast cells and their cultured counterparts: naïve, formative, and primed phase. In this work, we report that immunoproteasomes and their chaperone co-regulators are suppressed in the naïve state but are readily upregulated in the formative phase of the pluripotency continuum, featured by epiblast-like cells (EpiLCs). Our data lay ground for the further investigation of the biological functions of immunoproteasome in the regulation of proteostasis during early mammalian development.
    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
    人类多能干细胞衍生的心肌细胞(hPSC-CM)构成了一个吸引人的药物发现工具,疾病建模,和心脏毒性筛查。然而,他们的生理不成熟,类似于胎儿晚期的CMs,限制了它的效用。在这里,我们开发了一部小说,可扩展的细胞培养基旨在增强hPSC-CM的成熟。该培养基通过用特定的小分子抑制剂靶向乙酰辅酶A羧化酶2(ACC2)来促进向脂肪酸利用的代谢转变,并增强线粒体功能。我们的研究结果表明,这种成熟方案显着促进了新陈代谢,结构,hPSC-CM在不同分化阶段的分子和功能成熟度。此外,它能够创建具有优越结构完整性和收缩特性的心脏微组织。值得注意的是,在这种优化的成熟培养基中培养的hPSC-CM在模拟急性内皮素-1诱导后的肥大性心脏表型方面显示出更高的准确性,并且在药物筛选工作中的体外和体内靶标参与之间显示出很强的相关性。这种方法有望改善hPSC-CM在心脏病建模和药物发现中的实用性和可翻译性。
    Human pluripotent stem cell-derived cardiomyocytes (hPSC-CMs) constitute an appealing tool for drug discovery, disease modeling, and cardiotoxicity screening. However, their physiological immaturity, resembling CMs in the late fetal stage, limits their utility. Herein, we have developed a novel, scalable cell culture medium designed to enhance the maturation of hPSC-CMs. This medium facilitates a metabolic shift towards fatty acid utilization and augments mitochondrial function by targeting Acetyl-CoA carboxylase 2 (ACC2) with a specific small molecule inhibitor. Our findings demonstrate that this maturation protocol significantly advances the metabolic, structural, molecular and functional maturity of hPSC-CMs at various stages of differentiation. Furthermore, it enables the creation of cardiac microtissues with superior structural integrity and contractile properties. Notably, hPSC-CMs cultured in this optimized maturation medium display increased accuracy in modeling a hypertrophic cardiac phenotype following acute endothelin-1 induction and show a strong correlation between in vitro and in vivo target engagement in drug screening efforts. This approach holds promise for improving the utility and translatability of hPSC-CMs in cardiac disease modeling and drug discovery.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

公众号