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
    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:染色体稳定性对于多能干细胞(PSC)的稳态和早期胚胎发育至关重要。当使用PSC作为原始材料时,染色体缺陷可能会增加再生医学中的致癌风险。然而,关于PSC染色体稳定性维持的详细机制尚不完全清楚。
    方法:在标准条件下培养小鼠胚胎干细胞(细胞系D3)和人胚胎干细胞(细胞系H9)。为了证实RetSat蛋白在PSC有丝分裂染色体上的负载,在小鼠胚胎成纤维细胞(MEFs)的PSC自发分化试验和iPSC重编程试验中进行免疫染色,分别。此外,qPCR,免疫沉淀,LC-MS/MS和免疫印迹法研究RetSat的表达,以及RetSat与共粘素/凝缩素组分的相互作用。进行RNA测序和畸胎瘤形成测定以评估具有RetSat缺失的小鼠胚胎干细胞的致癌风险。
    结果:我们报道了一个PSC高表达基因,RetSat,在染色体稳定中起着关键作用。我们鉴定了位于有丝分裂染色体上的RetSat蛋白,特别是在干细胞阳性细胞中,例如胚胎干细胞(ESC)和诱导多能干细胞(iPSC)。我们发现染色体不稳定,例如染色体桥接,当下调RetSat时,小鼠和人ESCs中的滞后和相间微核。RetSat敲除小鼠ESCs上调癌症相关基因通路,并在畸胎瘤形成试验中显示出较高的致瘤能力。机械上,我们证实RetSat与粘附素/凝缩素成分Smc1a和Nudcd2相互作用。RetSat缺失损害了Smc1a的染色体负荷剂量,Smc3和Nudcd2。
    结论:总之,我们报道RetSat是多能干细胞染色体凝聚的关键稳定剂.这突出了RetSat在早期胚胎发育中的关键作用,以及RetSat作为评估多能干细胞质量的有效生物标志物的潜在价值。
    BACKGROUND: Chromosome stability is crucial for homeostasis of pluripotent stem cells (PSCs) and early-stage embryonic development. Chromosomal defects may raise carcinogenic risks in regenerative medicine when using PSCs as original materials. However, the detailed mechanism regarding PSCs chromosome stability maintenance is not fully understood.
    METHODS: Mouse embryonic stem cells (line D3) and human embryonic stem cells (line H9) were cultured under standard conditions. To confirm the loading of RetSat protein on mitotic chromosomes of PSCs, immunostaining was performed in PSCs spontaneous differentiation assay and iPSC reprogramming assay from mouse embryonic fibroblasts (MEFs), respectively. In addition, qPCR, immunoprecipitation, LC-MS/MS and immunoblotting were used to study the expression of RetSat, and interactions of RetSat with cohesin/condensin components. RNA sequencing and teratoma formation assay was conducted to evaluate the carcinogenic risk of mouse embryonic stem cells with RetSat deletion.
    RESULTS: We reported a PSC high-expressing gene, RetSat, plays key roles in chromosome stabilization. We identified RetSat protein localizing onto mitotic chromosomes specifically in stemness positive cells such as embryonic stem cells (ESCs) and induced pluripotent stem cells (iPSCs). We found dramatic chromosome instability, e.g. chromosome bridging, lagging and interphase micronuclei in mouse and human ESCs when down regulating RetSat. RetSat knock-out mouse ESCs upregulated cancer associated gene pathways, and displayed higher tumorigenic capacities in teratoma formation assay. Mechanistically, we confirmed that RetSat interacts with cohesin/condensin components Smc1a and Nudcd2. RetSat deletion impaired the chromosome loading dosage of Smc1a, Smc3 and Nudcd2.
    CONCLUSIONS: In summary, we reported RetSat to be a key stabilizer of chromosome condensation in pluripotent stem cells. This highlights the crucial roles of RetSat in early-stage embryonic development, and potential value of RetSat as an effective biomarker for assessing the quality of pluripotent stem cells.
    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
    我们对人类生物学的认识主要来源于使用动物模型的研究。然而,人类和模型生物之间的种间差异可能导致从模型生物获得的结果的不精确外推。类器官是源自多能或成体干细胞的三维细胞簇,其自组织成类似于同源器官的器官结构。人类类器官的建立使得研究人类特有的器官或组织病理生理学成为可能。然而,大多数类器官没有器官特异性脉管系统,神经元,和免疫细胞,因此限制了它们在模拟复杂病理生理表型方面的效用。在解决这些限制的各种方法中,异种移植是一种有希望的“捷径”。我们将讨论在类器官中构建组织复杂性的最新进展,特别关注异种移植。
    Our knowledge of human biology is mainly originated from studies using animal models. However, interspecies differences between human and model organisms may lead to imprecise extrapolation of results obtained from model organisms. Organoids are three-dimensional cell clusters derived from pluripotent or adult stem cells that self-organize into organ-like structures reminiscent of the cognate organ. The establishment of human organoids makes it possible to study organ or tissue pathophysiology that is specific to human beings. However, most organoids do not have organ-specific vasculature, neurons, and immune cells, hence limiting their utility in emulating complex pathophysiological phenotypes. Among the various approaches to address these limitations, xenotransplantation represents a promising \'shortcut\'. We will discuss recent advance in constructing tissue complexity in organoids, with a special focus on xenotransplantation.
    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
    人胚胎干细胞和人诱导多能干细胞可用于创建称为脑类器官的3D组织。它们在结构和功能两方面更忠实地复制了人脑组织的生理和病理特征,它们更精确地类似于人类胚胎大脑的形态和细胞结构。这使它们成为有价值的模型,用于药物筛选和关于人脑和相关疾病发展的体外研究。脑类器官实现的技术突破对不同脑区的研究产生了重大影响,大脑发育和疾病,大脑与其他组织和器官之间的联系,和大脑进化。本文讨论了脑类器官的发育,它们在糖尿病研究中的应用,和他们的进步。
    Human embryonic stem cells and human induced pluripotent stem cells may be used to create 3D tissues called brain organoids. They duplicate the physiological and pathological characteristics of human brain tissue more faithfully in terms of both structure and function, and they more precisely resemble the morphology and cellular structure of the human embryonic brain. This makes them valuable models for both drug screening and in vitro studies on the development of the human brain and associated disorders. The technical breakthroughs enabled by brain organoids have a significant impact on the research of different brain regions, brain development and sickness, the connections between the brain and other tissues and organs, and brain evolution. This article discusses the development of brain organoids, their use in diabetes research, and their progress.
    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
    细胞大小是显著影响细胞生理学和功能的关键物理性质。然而,细胞大小对干细胞规格的影响在很大程度上仍然未知.这里,我们研究了人多能干细胞分化为定形内胚层(DE)过程中细胞大小的动态变化.有趣的是,随着DE分化的发展,细胞大小表现出逐渐减小的刚度较高。此外,应用高渗压力或化学物质来加速细胞大小的显著减小和特异性地增强DE分化。通过在功能上干预机械敏感元件,我们已经确定肌动球蛋白活性是DE分化和细胞大小减小的关键介质。机械上,细胞大小的减少诱导肌动球蛋白依赖性血管动蛋白(AMOT)核易位,抑制Yes相关蛋白(YAP)活性,从而促进DE分化。一起,我们的研究在细胞大小缩小和DE分化之间建立了一种新的联系,由AMOT核易位介导。此外,我们的发现表明,渗透压的应用可以有效地促进人的内胚层谱系分化。
    Cell size is a crucial physical property that significantly impacts cellular physiology and function. However, the influence of cell size on stem cell specification remains largely unknown. Here, we investigated the dynamic changes in cell size during the differentiation of human pluripotent stem cells into definitive endoderm (DE). Interestingly, cell size exhibited a gradual decrease as DE differentiation progressed with higher stiffness. Furthermore, the application of hypertonic pressure or chemical to accelerate the reduction in cell size significantly and specifically enhanced DE differentiation. By functionally intervening in mechanosensitive elements, we have identified actomyosin activity as a crucial mediator of both DE differentiation and cell size reduction. Mechanistically, the reduction in cell size induces actomyosin-dependent angiomotin (AMOT) nuclear translocation, which suppresses Yes-associated protein (YAP) activity and thus facilitates DE differentiation. Together, our study has established a novel connection between cell size diminution and DE differentiation, which is mediated by AMOT nuclear translocation. Additionally, our findings suggest that the application of osmotic pressure can effectively promote human endodermal lineage 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
    从基因操纵的多能干细胞中衍生低免疫原性人细胞对于未来的移植医学和过继性免疫疗法具有巨大的希望。破坏多能干细胞中的β-2-微球蛋白(B2M),然后分化为专门的细胞类型是一种有希望的方法来获得低免疫原性细胞。鉴于基于CRISPR/Cas9的基因编辑工具和杆状病毒递送系统的有吸引力的特征,杆状病毒可以提供CRISPR/Cas9组件用于B2M的位点特异性基因编辑。在这里,我们报道了一种杆状病毒CRISPR/Cas9载体系统的开发,用于人细胞中B2M基因座的破坏。在人类胚胎干细胞(hESCs)中进行测试时,成功实现了B2M基因敲除/敲除,导致人白细胞抗原I类在细胞表面表达的稳定下调。然后将源自B2M基因破坏的hESC的成纤维细胞用作与人外周血单核细胞共培养的刺激细胞。如通过敏感的Elispot测定所评估的,这些成纤维细胞触发的同种免疫应答显著降低。B2M阴性hESC在体外和体内保持多能性和分化成三个胚芽谱系的能力。这些发现证明了使用杆状病毒-CRISPR/Cas9系统建立B2M破坏的多能干细胞的可行性。B2M敲低/敲除足以导致低免疫原性条件,从而支持B2M阴性细胞作为同种异体细胞治疗的通用供体细胞的潜在用途。
    Derivation of hypoimmunogenic human cells from genetically manipulated pluripotent stem cells holds great promise for future transplantation medicine and adoptive immunotherapy. Disruption of beta-2-microglobulin (B2M) in pluripotent stem cells followed by differentiation into specialized cell types is a promising approach to derive hypoimmunogenic cells. Given the attractive features of CRISPR/Cas9-based gene editing tool and baculoviral delivery system, baculovirus can deliver CRISPR/Cas9 components for site-specific gene editing of B2M. Herein, we report the development of a baculoviral CRISPR/Cas9 vector system for the B2M locus disruption in human cells. When tested in human embryonic stem cells (hESCs), the B2M gene knockdown/out was successfully achieved, leading to the stable down-regulation of human leukocyte antigen class I expression on the cell surface. Fibroblasts derived from the B2M gene-disrupted hESCs were then used as stimulator cells in the co-cultures with human peripheral blood mononuclear cells. These fibroblasts triggered significantly reduced alloimmune responses as assessed by sensitive Elispot assays. The B2M-negative hESCs maintained the pluripotency and the ability to differentiate into three germ lineages in vitro and in vivo. These findings demonstrated the feasibility of using the baculoviral-CRISPR/Cas9 system to establish B2M-disrupted pluripotent stem cells. B2M knockdown/out sufficiently leads to hypoimmunogenic conditions, thereby supporting the potential use of B2M-negative cells as universal donor cells for allogeneic cell therapy.
    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
    胰腺δ细胞的功能障碍是糖尿病的病因。尽管发挥了重要作用,人类δ细胞很少,限制了针对δ细胞的生理学研究和药物发现。迄今为止,没有建立直接的δ细胞分化方法。这里,我们证明成纤维细胞生长因子(FGF)7促进胰腺内胚层/祖细胞分化,而FGF2通过FGF受体1将细胞偏向胰腺δ细胞谱系。我们开发了一种分化方法,通过将FGF2与FGF7组合来从人干细胞中产生δ细胞,该方法在内胚层/内分泌前体诱导过程中协同指导胰腺谱系分化并调节转录因子和SST激活剂的表达。这些δ细胞表现出成熟的RNA谱和细小的分泌颗粒,分泌生长抑素以响应各种刺激,并抑制移植后体外共培养的β细胞和小鼠β细胞的胰岛素分泌。体外从干细胞产生人δ胰腺细胞将为糖尿病中的药物发现和细胞移植研究提供前所未有的细胞来源。
    Dysfunction of pancreatic δ cells contributes to the etiology of diabetes. Despite their important role, human δ cells are scarce, limiting physiological studies and drug discovery targeting δ cells. To date, no directed δ-cell differentiation method has been established. Here, we demonstrate that fibroblast growth factor (FGF) 7 promotes pancreatic endoderm/progenitor differentiation, whereas FGF2 biases cells towards the pancreatic δ-cell lineage via FGF receptor 1. We develop a differentiation method to generate δ cells from human stem cells by combining FGF2 with FGF7, which synergistically directs pancreatic lineage differentiation and modulates the expression of transcription factors and SST activators during endoderm/endocrine precursor induction. These δ cells display mature RNA profiles and fine secretory granules, secrete somatostatin in response to various stimuli, and suppress insulin secretion from in vitro co-cultured β cells and mouse β cells upon transplantation. The generation of human pancreatic δ cells from stem cells in vitro would provide an unprecedented cell source for drug discovery and cell transplantation studies in diabetes.
    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
    从人类多能干细胞(hPSC)制造胰岛素分泌胰岛的方案开发已经取得了显著进展。与目前的方法不同,我们设计了一种可调节的策略,通过将PDX1细胞出芽形态发生整合到分期分化中来生成富含主要胰岛细胞类型的胰岛球体。在这个模拟正常胰岛形态发生的过程中,分化的胰岛球状体与内分泌细胞相互混合或排列在核心-地幔结构中,伴随着功能异质性。通过对人类胰腺发育的体外建模,我们说明了PDX1的重要性和EphB3/4信号在引发细胞出芽形态发生中的需求。使用这种新方法,我们用RFX6敲除hPSC对Mitchell-Riley综合征进行建模,说明在向胰岛细胞分化过程中存在意想不到的形态发生缺陷。这项工作中描述的可调分化系统和干细胞衍生的胰岛模型可能有助于解决胰岛生物学和人类胰腺疾病探测中的基本问题。
    Remarkable advances in protocol development have been achieved to manufacture insulin-secreting islets from human pluripotent stem cells (hPSCs). Distinct from current approaches, we devised a tunable strategy to generate islet spheroids enriched for major islet cell types by incorporating PDX1+ cell budding morphogenesis into staged differentiation. In this process that appears to mimic normal islet morphogenesis, the differentiating islet spheroids organize with endocrine cells that are intermingled or arranged in a core-mantle architecture, accompanied with functional heterogeneity. Through in vitro modelling of human pancreas development, we illustrate the importance of PDX1 and the requirement for EphB3/4 signaling in eliciting cell budding morphogenesis. Using this new approach, we model Mitchell-Riley syndrome with RFX6 knockout hPSCs illustrating unexpected morphogenesis defects in the differentiation towards islet cells. The tunable differentiation system and stem cell-derived islet models described in this work may facilitate addressing fundamental questions in islet biology and probing human pancreas diseases.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

公众号