Human embryonic stem cells

人胚胎干细胞
  • 文章类型: Journal Article
    生物活性脂质如鞘氨醇-1-磷酸(S1P)和溶血磷脂酸作为在干细胞增殖和分化中具有调节作用的信号分子已获得显著关注。新型化学合成的鞘氨醇代谢物O-环状植物鞘氨醇-1-磷酸(cP1P)源自植物鞘氨醇-1-磷酸(P1P),并与S1P具有结构相似性。以前,cP1P在人胚胎干细胞(hESC)心肌细胞分化过程中调节ALK3/BMPR信号传导中的作用被证明。在这项研究中,研究了cP1P从hESCs分化为内皮细胞(ECs)的适用性,并对数代获得高产量功能性ECs的有效方法进行了标准化.源自hESC的EC显示出与天然EC相似的细胞和分子特征。因此,这项研究的结果为进一步研究基于cP1P的干细胞分化用于再生疗法开辟了途径.
    Bioactive lipids like sphingosine-1-phosphate (S1P) and lysophosphatidic acid have gained significant attention as signaling molecules with regulatory roles in stem cell proliferation and differentiation. The novel chemically synthesized sphingosine metabolite O-cyclic phytosphingosine-1-phosphate (cP1P) is derived from phytosphingosine-1-phosphate (P1P) and shares structural similarities with S1P. Previously, the role of cP1P in regulating ALK3/BMPR signaling during cardiomyocyte differentiation from human embryonic stem cells (hESCs) was demonstrated. In this study, the applicability of cP1P for endothelial cells (ECs) differentiation from hESCs was investigated an efficient method to obtain a high yield of functional ECs over several passages was standardized. The ECs derived from hESCs showed cellular and molecular characteristics similar to the native ECs. Thus, the results of this study open avenues for further research into cP1P-based stem cell differentiation for regenerative 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
    在开发的第一周,人类胚胎形成由内部细胞团和滋养外胚层(TE)细胞组成的胚泡,后者是胎盘滋养层的祖细胞。这里,我们研究了从胚泡早期到晚期的人TE中转录本的表达。我们鉴定了转录因子GATA2,GATA3,TFAP2C和KLF5的富集,并表征了它们在TE发育过程中的蛋白质表达动力学。通过诱导型过表达和mRNA转染,我们确定这些因素,和MYC一起,足以从引发的人胚胎干细胞建立诱导的滋养层干细胞(iTSC)。这些iTSCs自我更新并概括了形态学特征,基因表达谱,和定向分化潜力,与现有的人类TSC相似。每个系统的遗漏,或多种因素的组合,揭示了GATA2和GATA3对iTSC转分化的重要性。总之,这些发现提供了对可能在人类TE中起作用的转录因子网络的见解,并拓宽了建立早期人类胎盘祖细胞细胞模型的方法,这可能在未来对胎盘相关疾病的模型是有用的。
    During the first week of development, human embryos form a blastocyst composed of an inner cell mass and trophectoderm (TE) cells, the latter of which are progenitors of placental trophoblast. Here, we investigated the expression of transcripts in the human TE from early to late blastocyst stages. We identified enrichment of the transcription factors GATA2, GATA3, TFAP2C and KLF5 and characterised their protein expression dynamics across TE development. By inducible overexpression and mRNA transfection, we determined that these factors, together with MYC, are sufficient to establish induced trophoblast stem cells (iTSCs) from primed human embryonic stem cells. These iTSCs self-renew and recapitulate morphological characteristics, gene expression profiles, and directed differentiation potential, similar to existing human TSCs. Systematic omission of each, or combinations of factors, revealed the crucial importance of GATA2 and GATA3 for iTSC transdifferentiation. Altogether, these findings provide insights into the transcription factor network that may be operational in the human TE and broaden the methods for establishing cellular models of early human placental progenitor cells, which may be useful in the future to model placental-associated diseases.
    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
    在一些眼部疾病中,视网膜神经元变性可导致永久性失明。干细胞(SC)衍生的RGC的移植已被提出作为RGC损失的潜在疗法。尽管有SC衍生的RGC移植成功病例的报道,实现远距离再生和功能连接仍然是一个挑战。为了解决这些障碍,视网膜类器官被用于研究干细胞移植的调控机制。在这里,我们提出了一种改良的方案,用于将人类胚胎干细胞(ESC)分化为视网膜类器官,并将类器官衍生的RGC移植到鼠眼中。
    In several ocular diseases, degeneration of retinal neurons can lead to permanent blindness. Transplantation of stem cell (SC)-derived RGCs has been proposed as a potential therapy for RGC loss. Although there are reports of successful cases of SC-derived RGC transplantation, achieving long-distance regeneration and functional connectivity remains a challenge. To address these hurdles, retinal organoids are being used to study the regulatory mechanism of stem cell transplantation. Here we present a modified protocol for differentiating human embryonic stem cells (ESCs) into retinal organoids and transplanting organoid-derived RGCs into the murine eyes.
    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
    基因组印记涉及同源父系和母系基因座之间的差异DNA甲基化和基因表达。目前还不清楚,然而,DNA复制是否在印迹或其他基因组区域也显示出亲本起源特异性模式。这里,我们利用单亲人类胚胎干细胞研究了全基因组异步DNA复制,所述单亲人类胚胎干细胞含有仅母本(孤雌生殖)或仅父本(雄激素)DNA.四个印迹基因簇表现出基于亲本起源的差异复制时间,而基因组的其余部分,99.82%,亲本起源之间没有明显的复制不同步。印迹基因簇中的活性等位基因比非活性等位基因更早复制。在Prader-Willi综合征基因座,异步复制几乎跨越了整个S阶段。通过以与基因表达一致的方式分化成神经元前体细胞来进行复制异步。这项研究将异步DNA复制确立为大型印迹基因簇的标志。
    Genomic imprinting involves differential DNA methylation and gene expression between homologous paternal and maternal loci. It remains unclear, however, whether DNA replication also shows parent-of-origin-specific patterns at imprinted or other genomic regions. Here, we investigate genome-wide asynchronous DNA replication utilizing uniparental human embryonic stem cells containing either maternal-only (parthenogenetic) or paternal-only (androgenetic) DNA. Four clusters of imprinted genes exhibited differential replication timing based on parent of origin, while the remainder of the genome, 99.82%, showed no significant replication asynchrony between parental origins. Active alleles in imprinted gene clusters replicated earlier than their inactive counterparts. At the Prader-Willi syndrome locus, replication asynchrony spanned virtually the entirety of S phase. Replication asynchrony was carried through differentiation to neuronal precursor cells in a manner consistent with gene expression. This study establishes asynchronous DNA replication as a hallmark of large imprinted gene clusters.
    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
    关于MECP2功能及其在Rett综合征(RTT)中的意义的研究传统上集中在神经元上。这里,使用人类胚胎干细胞(hESC)系,我们对MECP2功能丧失进行建模,以探讨其对星形胶质细胞(AST)发育和脑功能障碍的影响.RTThESC衍生的大脑类器官的超微结构分析显示,与对照组(CTRs)相比,线粒体明显较小,在神经胶质和神经元中尤其明显。采用多元组学方法,我们观察到,与神经元相比,ASTs中MECP2突变后,核编码线粒体基因亚组的基因表达和可及性增加.对hESC衍生的AST的分析显示,线粒体呼吸减少,RTT与CTR中的三羧酸循环和电子传递链中的关键蛋白发生了变化。此外,RTTAST在基础条件下表现出增加的细胞溶质氨基酸,随着能源需求的增加而耗尽。值得注意的是,从RTTAST中分离出的线粒体在转移到皮质神经元时表现出增加的活性氧并影响神经元活性。这些发现强调了MECP2突变对ASTs和神经元中线粒体和代谢途径的不同影响,提示AST线粒体功能失调可能通过影响神经元健康而导致RTT病理生理学。
    Studies on MECP2 function and its implications in Rett Syndrome (RTT) have traditionally centered on neurons. Here, using human embryonic stem cell (hESC) lines, we modeled MECP2 loss-of-function to explore its effects on astrocyte (AST) development and dysfunction in the brain. Ultrastructural analysis of RTT hESC-derived cerebral organoids revealed significantly smaller mitochondria compared to controls (CTRs), particularly pronounced in glia versus neurons. Employing a multiomics approach, we observed increased gene expression and accessibility of a subset of nuclear-encoded mitochondrial genes upon mutation of MECP2 in ASTs compared to neurons. Analysis of hESC-derived ASTs showed reduced mitochondrial respiration and altered key proteins in the tricarboxylic acid cycle and electron transport chain in RTT versus CTRs. Additionally, RTT ASTs exhibited increased cytosolic amino acids under basal conditions, which were depleted upon increased energy demands. Notably, mitochondria isolated from RTT ASTs exhibited increased reactive oxygen species and influenced neuronal activity when transferred to cortical neurons. These findings underscore MECP2 mutation\'s differential impact on mitochondrial and metabolic pathways in ASTs versus neurons, suggesting that dysfunctional AST mitochondria may contribute to RTT pathophysiology by affecting neuronal health.
    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
    FOXR2激活的中枢神经系统(CNS)神经母细胞瘤(CNSNB-FOXR2)是最近鉴定的脑肿瘤亚型,其特征在于转录因子FOXR2的表达升高主要是由于基因组重排。然而,精确的致病机制,包括起源的细胞类型,仍然难以捉摸。
    进行患者肿瘤的基因表达分析以鉴定推定的起源细胞类型。基于这一预测,我们开发了一种新的基于人类胚胎干细胞的模型,以验证CNSNB-FOXR2形成的起源和分子和细胞机制.
    我们的数据显示,CNSNB-FOXR2肿瘤表达高水平的与内侧神经节隆起(MGE)相关的谱系标记基因,位于发育中的腹侧前脑的一过性结构。我们的模型证实了FOXR2对增殖和体内致瘤性的细胞类型特异性作用。此外,我们发现FOXR2过表达通过抑制内源性RAS抑制剂DIRAS3激活MEK/ERK信号通路.MEK抑制剂曲美替尼比非表达细胞更能抑制表达FOXR2的MGE祖细胞的增殖。
    我们的研究共同表明,MGE祖细胞是CNSNB-FOXR2的起源细胞,FOXR2激活MEK/ERK信号通路,提供潜在的治疗靶点。
    UNASSIGNED: FOXR2-activated central nervous system (CNS) neuroblastoma (CNS NB-FOXR2) is a recently identified subtype of brain tumor characterized by the elevated expression of the transcription factor FOXR2 mainly due to genomic rearrangements. However, the precise pathogenic mechanisms, including the cell type of origin, remain elusive.
    UNASSIGNED: A gene expression analysis of patient tumors was performed to identify putative cell types of origin. Based on this prediction, a new human embryonic stem cell-based model was developed to validate the origin and to examine the molecular and cellular mechanisms underlying the formation of CNS NB-FOXR2.
    UNASSIGNED: Our data showed that CNS NB-FOXR2 tumors express a high level of lineage marker genes associated with the medial ganglionic eminence (MGE), a transient structure located in the developing ventral forebrain. Our model confirmed the cell-type-specific effect of FOXR2 on the proliferation and in vivo tumorigenicity. Additionally, we found that FOXR2 overexpression activated the MEK/ERK signaling pathway through a suppression of the endogenous RAS inhibitor DIRAS3. The MEK inhibitor trametinib suppressed the proliferation of FOXR2-expressing MGE progenitors more than nonexpressing cells.
    UNASSIGNED: Our study collectively demonstrates that MGE progenitors are the cell of origin of CNS NB-FOXR2 and that FOXR2 activates the MEK/ERK signaling pathway, providing a potential therapeutic target.
    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:了解人类前列腺的谱系分化不仅对人类发育生物学的基础研究至关重要,而且对前列腺相关疾病的治疗也有重要贡献。当前的知识主要依赖于对啮齿动物模型的研究,尽管临床样本缺乏人类来源的替代品,但可能会在某些阶段提供快照。人类胚胎干细胞可以产生包括前列腺在内的所有胚胎谱系,事实上,一些研究证明了这种可能性,基于共培养或与泌尿生殖道间质共同移植到小鼠肾包膜中。
    方法:为了逐步建立从人胚胎干细胞体外获得前列腺类器官的方案,我们通过模拟转录因子和信号转导通路的调节网络来应用化学物质和生长因子,并构建携带诱导型NKX3-1表达盒的细胞系,以及三维文化系统。非配对t检验用于统计分析。
    结果:我们首先成功生成定形内胚层,后肠,和泌尿生殖道窦细胞.胚胎干细胞来源的泌尿生殖窦细胞表达前列腺关键转录因子AR和FOXA1,但不能表达NKX3-1。因此,我们通过同源重组构建NKX3-1诱导细胞系,最终能够产生AR,FOXA1和NKX3-1三阳性泌尿生殖前列腺谱系细胞通过逐步分化。最后,结合3D培养,我们成功地获得了具有某些结构和前列腺细胞群的前列腺样类器官。
    结论:这项研究揭示了NKX3-1在前列腺分化中的关键作用,并提供了可诱导的NKX3-1细胞系,以及提供逐步分化方案以生成人类前列腺样器官,这将有助于前列腺发育和疾病发病机制的研究。
    BACKGROUND: Understanding the lineage differentiation of human prostate not only is crucial for basic research on human developmental biology but also significantly contributes to the management of prostate-related disorders. Current knowledge mainly relies on studies on rodent models, lacking human-derived alternatives despite clinical samples may provide a snapshot at certain stage. Human embryonic stem cells can generate all the embryonic lineages including the prostate, and indeed a few studies demonstrate such possibility based on co-culture or co-transplantation with urogenital mesenchyme into mouse renal capsule.
    METHODS: To establish a stepwise protocol to obtain prostatic organoids in vitro from human embryonic stem cells, we apply chemicals and growth factors by mimicking the regulation network of transcription factors and signal transduction pathways, and construct cell lines carrying an inducible NKX3-1 expressing cassette, together with three-dimensional culture system. Unpaired t test was applied for statistical analyses.
    RESULTS: We first successfully generate the definitive endoderm, hindgut, and urogenital sinus cells. The embryonic stem cell-derived urogenital sinus cells express prostatic key transcription factors AR and FOXA1, but fail to express NKX3-1. Therefore, we construct NKX3-1-inducible cell line by homologous recombination, which is eventually able to yield AR, FOXA1, and NKX3-1 triple-positive urogenital prostatic lineage cells through stepwise differentiation. Finally, combined with 3D culture we successfully derive prostate-like organoids with certain structures and prostatic cell populations.
    CONCLUSIONS: This study reveals the crucial role of NKX3-1 in prostatic differentiation and offers the inducible NKX3-1 cell line, as well as provides a stepwise differentiation protocol to generate human prostate-like organoids, which should facilitate the studies on prostate development and disease pathogenesis.
    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
    严重的结膜损伤可导致广泛的眼部瘢痕,穹窿缩短,甚至眼表故障,导致严重的视力损害。结膜重建是这些临床结膜疾病的主要治疗策略。然而,关于干细胞来源的结膜上皮细胞诱导分化的研究有限。在这项研究中,我们建立了从人胚胎干细胞(hESCs)向结膜上皮细胞分化的化学方法.hES细胞系H1用于分化,和RT-qPCR,免疫荧光染色周期性酸性希夫染色(PAS),和转录组分析用于鉴定分化的细胞。这里,模仿脊椎动物结膜的发育,使用三步法诱导hESCs,包括第一切托明用于诱导眼表外胚层,然后用烟酰胺诱导眼表上皮祖细胞,最后是表皮生长因子,角质细胞生长因子和其他因子用于分化成熟的结膜上皮细胞。hESC来源的结膜上皮样细胞表达成熟的结膜上皮谱系标记(包括PAX6、P63、K13)。通过阳性PAS证实杯状细胞的存在。转录组分析显示,hESC衍生的结膜上皮样细胞具有更幼稚的表型,与成熟的人结膜上皮细胞相比,表现出更大的增殖能力,表明它们作为结膜重建替代种子细胞的潜力。
    Severe conjunctival damage can lead to extensive ocular cicatrisation, fornix shortening, and even ocular surface failure, resulting in significant vision impairment. Conjunctival reconstruction is the primary therapeutic strategy for these clinical conjunctival diseases. However, there have been limited studies on induced differentiation of conjunctival epithelial cells derived from stem cells. In this study, we established a chemical defined differentiation protocol from human embryonic stem cells (hESCs) into conjunctival epithelial cells. hES cell line H1 was used for differentiation, and RT-qPCR, immunofluorescence staining, Periodic-acid-Schiff staining (PAS), and transcriptome analysis were employed to identify the differentiated cells. Here, to imitate the development of the vertebrate conjunctiva, hESCs were induced using a three-step process involving first chetomin was used to induce ocular surface ectoderm, then nicotinamide was used to induce ocular surface epithelial progenitor cells, and finally epidermal growth factor, keratinocyte growth factor and other factors were used to differentiate mature conjunctival epithelial cells. hESC-derived conjunctival epithelial cells expressed mature conjunctival epithelial lineage markers (including PAX6, P63, K13). The presence of goblet cells was confirmed by positive PAS. Transcriptome analysis revealed that hESC-derived conjunctival epithelial cells possessed a more naïve phenotype, and exhibited greater proliferation capacity compared to mature human conjunctival epithelial cells, suggesting their potential as alternative seed cells for conjunctival reconstruction.
    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
    自闭症易感性候选2(AUTS2)基因中断的个体表现出智力障碍等症状,小头畸形,生长迟缓,和明显的骨骼和面部差异。已经使用动物和胚胎干细胞模型研究了AUTS2在神经发育中的作用。然而,AUTS2如何影响神经发育的精确分子机制,特别是在人类中,没有被彻底理解。我们的研究采用了3D人脑类器官培养系统,结合遗传,基因组,细胞,和分子方法,研究AUTS2如何通过细胞信号通路影响神经发育。我们使用CRISPR/Cas9技术来创建缺乏AUTS2的人类胚胎干细胞,然后用这些细胞生成大脑类器官。我们的转录组学分析显示,脑类器官中AUTS2的缺失减少了致力于神经元谱系的细胞群,导致转录谱类似于脉络丛(ChP)细胞的细胞过多。有趣的是,我们发现AUTS2负调节WNT/β-catenin信号通路,其在缺乏AUTS2的脑类器官和缺乏AUTS2的荧光素酶报告细胞中的过度激活证明。重要的是,用WNT抑制剂治疗缺乏AUTS2的脑类器官逆转了ChP基因的过表达,并增加了下调的神经元基因表达。这项研究为AUTS2在神经发育中的作用提供了新的见解,并提出了神经发育障碍的潜在靶向疗法。
    Individuals with the Autism Susceptibility Candidate 2 (AUTS2) gene disruptions exhibit symptoms such as intellectual disability, microcephaly, growth retardation, and distinct skeletal and facial differences. The role of AUTS2 in neurodevelopment has been investigated using animal and embryonic stem cell models. However, the precise molecular mechanisms of how AUTS2 influences neurodevelopment, particularly in humans, are not thoroughly understood. Our study employed a 3D human cerebral organoid culture system, in combination with genetic, genomic, cellular, and molecular approaches, to investigate how AUTS2 impacts neurodevelopment through cellular signaling pathways. We used CRISPR/Cas9 technology to create AUTS2-deficient human embryonic stem cells and then generated cerebral organoids with these cells. Our transcriptomic analyses revealed that the absence of AUTS2 in cerebral organoids reduces the populations of cells committed to the neuronal lineage, resulting in an overabundance of cells with a transcription profile resembling that of choroid plexus (ChP) cells. Intriguingly, we found that AUTS2 negatively regulates the WNT/β-catenin signaling pathway, evidenced by its overactivation in AUTS2-deficient cerebral organoids and in luciferase reporter cells lacking AUTS2. Importantly, treating the AUTS2-deficient cerebral organoids with a WNT inhibitor reversed the overexpression of ChP genes and increased the downregulated neuronal gene expression. This study offers new insights into the role of AUTS2 in neurodevelopment and suggests potential targeted therapies for neurodevelopmental disorders.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

公众号