human pluripotent stem cells

人多能干细胞
  • 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
    视网膜变性疾病,如年龄相关性黄斑变性(AMD)和视网膜色素变性(RP),最初表现为视网膜色素上皮(RPE)的功能障碍或死亡。人类多能干细胞(hPSC)衍生的RPE细胞的视网膜下移植已成为视网膜变性的潜在疗法。然而,使用当前方案从hPSC分化的RPE细胞是异种的,并且很少在临床试验中应用。临床应用迫切需要开发使用无异种生物材料的hPSC衍生的RPE细胞分化方案。在这项研究中,选择两种方案(活化素A和NIC84方案)进行修饰并用于将hiPSCs分化为RPE细胞;逐渐增加切托明浓度以实现RPE细胞的高分化效率。无异种细胞外基质(ECM)蛋白,层粘连蛋白-511、层粘连蛋白-521和重组玻连蛋白,被选为板材涂层基材,使用Matrigel(含异种ECM)涂覆的表面作为阳性对照。健康,将成熟的hPSC衍生的RPE细胞移植到21天大的皇家外科医学院(RCS)大鼠中,视网膜变性疾病的模型.移植hPSC衍生的RPE细胞后,通过视运动反应(qOMR)和视网膜电图评估RCS大鼠的视觉功能。我们的研究表明,使用NIC84方案,hPSC可以在LN521包被的培养皿上有效分化为RPE细胞。视网膜下移植细胞悬液可以延缓RCS大鼠视力丧失的进展。
    Retinal degeneration diseases, such as age-related macular degeneration (AMD) and retinitis pigmentosa (RP), initially manifest as dysfunction or death of the retinal pigment epithelium (RPE). Subretinal transplantation of human pluripotent stem cell (hPSC)-derived RPE cells has emerged as a potential therapy for retinal degeneration. However, RPE cells differentiated from hPSCs using current protocols are xeno-containing and are rarely applied in clinical trials. The development of hPSC-derived RPE cell differentiation protocols using xeno-free biomaterials is urgently needed for clinical applications. In this study, two protocols (the activin A and NIC84 protocols) were selected for modification and use in the differentiation of hiPSCs into RPE cells; the chetomin concentration was gradually increased to achieve high differentiation efficiency of RPE cells. The xeno-free extracellular matrix (ECM) proteins, laminin-511, laminin-521 and recombinant vitronectin, were selected as plate-coating substrates, and a Matrigel (xeno-containing ECM)-coated surface was used as a positive control. Healthy, mature hPSC-derived RPE cells were transplanted into 21-day-old Royal College of Surgeons (RCS) rats, a model of retinal degeneration disease. The visual function of RCS rats was evaluated by optomotor response (qOMR) and electroretinography after transplantation of hPSC-derived RPE cells. Our study demonstrated that hPSCs can be efficiently differentiated into RPE cells on LN521-coated dishes using the NIC84 protocol, and that subretinal transplantation of the cell suspensions can delay the progression of vision loss in RCS rats.
    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
    研究免疫介导的宿主损伤的人类模型很少。这里,我们利用GeoMx空间多组学平台分析了COVID-19胰腺尸检样本中的免疫细胞变化,显示促炎巨噬细胞的积累。暴露于严重急性呼吸综合征冠状病毒2(SARS-CoV-2)或柯萨奇病毒B4(CVB4)病毒的人胰岛的单细胞RNA测序(scRNA-seq)分析确定了促炎性巨噬细胞的激活和β细胞焦亡。为了区分病毒和促炎巨噬细胞介导的β细胞焦亡,我们开发了人类多能干细胞(hPSC)衍生的血管化巨噬细胞胰岛(VMI)类器官。与单独培养的细胞相比,VMI类器官在β细胞和内皮细胞中均表现出增强的标志物表达和功能。值得注意的是,VMI类器官内的促炎巨噬细胞诱导β细胞焦亡。机制研究强调了TNFSF12-TNFRSF12A参与促炎巨噬细胞介导的β细胞焦亡。这项研究确立了hPSC衍生的VMI类器官作为研究免疫细胞介导的宿主损伤的有价值的工具,并揭示了病毒暴露期间β细胞损伤的机制。
    There is a paucity of human models to study immune-mediated host damage. Here, we utilized the GeoMx spatial multi-omics platform to analyze immune cell changes in COVID-19 pancreatic autopsy samples, revealing an accumulation of proinflammatory macrophages. Single-cell RNA sequencing (scRNA-seq) analysis of human islets exposed to severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) or coxsackievirus B4 (CVB4) viruses identified activation of proinflammatory macrophages and β cell pyroptosis. To distinguish viral versus proinflammatory-macrophage-mediated β cell pyroptosis, we developed human pluripotent stem cell (hPSC)-derived vascularized macrophage-islet (VMI) organoids. VMI organoids exhibited enhanced marker expression and function in both β cells and endothelial cells compared with separately cultured cells. Notably, proinflammatory macrophages within VMI organoids induced β cell pyroptosis. Mechanistic investigations highlighted TNFSF12-TNFRSF12A involvement in proinflammatory-macrophage-mediated β cell pyroptosis. This study established hPSC-derived VMI organoids as a valuable tool for studying immune-cell-mediated host damage and uncovered the mechanism of β cell damage during viral exposure.
    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
    目前可用于研究丁型肝炎病毒(HDV)的培养系统是次优的。在这项研究中,我们证明,源自人多能干细胞(hPSC)的肝细胞样细胞(HLC)在各种测试基因型中完全允许HDV感染。当共感染辅助乙型肝炎病毒(HBV)或转导表达HBV包膜蛋白HBsAg时,HLC有效地释放感染性子代病毒粒子。我们还表明,表达HBsAg的HLC支持HDV的细胞外传播,从而为测试可用的抗HDV方案提供了有价值的平台。通过挑战细胞沿着分化与HDV感染,我们已经确定CD63是一种潜在的HDV共进入因子,它是未成熟肝细胞中HDV感染的限速因子.鉴于其可再生来源和从个体患者中获得hPSC的潜力,我们建议将HLC作为研究HDV生物学的有前途的模型。我们的发现为HDV感染提供了新的见解,并扩展了可用于开发治疗干预措施的研究工具库。
    Current culture systems available for studying hepatitis D virus (HDV) are suboptimal. In this study, we demonstrate that hepatocyte-like cells (HLCs) derived from human pluripotent stem cells (hPSCs) are fully permissive to HDV infection across various tested genotypes. When co-infected with the helper hepatitis B virus (HBV) or transduced to express the HBV envelope protein HBsAg, HLCs effectively release infectious progeny virions. We also show that HBsAg-expressing HLCs support the extracellular spread of HDV, thus providing a valuable platform for testing available anti-HDV regimens. By challenging the cells along the differentiation with HDV infection, we have identified CD63 as a potential HDV co-entry factor that was rate-limiting for HDV infection in immature hepatocytes. Given their renewable source and the potential to derive hPSCs from individual patients, we propose HLCs as a promising model for investigating HDV biology. Our findings offer new insights into HDV infection and expand the repertoire of research tools available for the development of therapeutic interventions.
    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
    人类多能干细胞衍生的心肌细胞(hPSC-CM)正在推进心血管发育和疾病建模,药物测试,和再生疗法。然而,hPSC-CM的产生受到分化过程中的显著可变性的阻碍。建立早期质量标记以监测谱系进展和预测终末分化结果将解决hPSC-CM生产中的这种稳健性和可重复性障碍。整合的转录组和表观基因组分析评估心脏祖细胞(CPC)的属性如何影响CM分化结果。结果分析确定了产生高纯度CM批次的CPC的预测性标志物,包括TTN,TRIM55,DGKI,MEF2C,MAB21L2、MYL7、LDB3、SLC7A11、MAB21L2和CALD1。从这些基因开发的预测模型在确定CPC阶段的最终CM纯度方面提供了高准确性。Further,阐明了对批次失败机制和在失败批次中产生的主要非CM细胞类型的见解。即EMT,MAPK,和WNT信号成为批次差异的重要驱动因素,产生成纤维细胞/壁细胞的脱靶群体,骨骼肌细胞,心外膜细胞,和非CPCSLC7A11亚群。这项研究证明了祖细胞的综合多组分析如何识别祖细胞的质量属性并预测分化结果。从而改进了区分方案并增加了过程的鲁棒性。
    Human pluripotent stem cell-derived cardiomyocytes (hPSC-CMs) are advancing cardiovascular development and disease modeling, drug testing, and regenerative therapies. However, hPSC-CM production is hindered by significant variability in the differentiation process. Establishment of early quality markers to monitor lineage progression and predict terminal differentiation outcomes would address this robustness and reproducibility roadblock in hPSC-CM production. An integrated transcriptomic and epigenomic analysis assesses how attributes of the cardiac progenitor cell (CPC) affect CM differentiation outcome. Resulting analysis identifies predictive markers of CPCs that give rise to high purity CM batches, including TTN, TRIM55, DGKI, MEF2C, MAB21L2, MYL7, LDB3, SLC7A11, MAB21L2, and CALD1. Predictive models developed from these genes provide high accuracy in determining terminal CM purities at the CPC stage. Further, insights into mechanisms of batch failure and dominant non-CM cell types generated in failed batches are elucidated. Namely EMT, MAPK, and WNT signaling emerge as significant drivers of batch divergence, giving rise to off-target populations of fibroblasts/mural cells, skeletal myocytes, epicardial cells, and a non-CPC SLC7A11+ subpopulation. This study demonstrates how integrated multi-omic analysis of progenitor cells can identify quality attributes of that progenitor and predict differentiation outcomes, thereby improving differentiation protocols and increasing process robustness.
    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
    人类多能干细胞(hPS)分化为胰腺内分泌细胞的重大进展,包括功能性β细胞,基于对潜在发展机制的详细了解。然而,最后的分化步骤,从内分泌祖细胞到单激素和成熟的胰腺内分泌细胞,仍然有待充分理解,这反映在hPS细胞衍生的胰岛细胞(SC-胰岛细胞)的剩余缺点中,其中包括缺乏β细胞成熟和不同细胞系之间的变异性。最终分化步骤的附加信号和修改将必须以组合方式评估以解决剩余的问题,并且适当的报告线在本任务中将是有用的。在这里,我们报告了hPS细胞报告系的生成和功能验证,该报告系可以监测INS和GCG细胞的生成及其在单激素细胞中的分辨率(INSeGFP,INSeGFP/GCGmCHERRY)以及β细胞成熟(INSeGFP/MAFAmCHERRY)和功能(INSGCaMP6)。报告hPS细胞系维持了多能性标记物的强大和广泛表达,并有效分化为定形内胚层和胰腺祖细胞(PP)。来自所有细胞系的PP细胞有效分化为胰岛细胞簇,这些细胞簇强烈表达相应的报告基因并含有葡萄糖反应性,产生胰岛素的细胞。为了证明这些hPS细胞报告系在高含量实时成像方法中的适用性,用于鉴定最佳分化条件,我们调整了我们的分化程序以在微孔中产生SC-胰岛簇。这允许在单个条件下对多个SC胰岛进行实时共聚焦成像,使用这种方法,我们发现在分化的最后阶段使用N21补充剂可以增加单激素β细胞的数量,而不会影响SC胰岛中α细胞的数量。本文描述的hPS细胞报告系和高含量实时成像方法将能够有效评估SC-胰岛的最佳分化和成熟的多种条件。
    The significant advances in the differentiation of human pluripotent stem (hPS) cells into pancreatic endocrine cells, including functional β-cells, have been based on a detailed understanding of the underlying developmental mechanisms. However, the final differentiation steps, leading from endocrine progenitors to mono-hormonal and mature pancreatic endocrine cells, remain to be fully understood and this is reflected in the remaining shortcomings of the hPS cell-derived islet cells (SC-islet cells), which include a lack of β-cell maturation and variability among different cell lines. Additional signals and modifications of the final differentiation steps will have to be assessed in a combinatorial manner to address the remaining issues and appropriate reporter lines would be useful in this undertaking. Here we report the generation and functional validation of hPS cell reporter lines that can monitor the generation of INS+ and GCG+ cells and their resolution into mono-hormonal cells (INSeGFP, INSeGFP/GCGmCHERRY) as well as β-cell maturation (INSeGFP/MAFAmCHERRY) and function (INSGCaMP6). The reporter hPS cell lines maintained strong and widespread expression of pluripotency markers and differentiated efficiently into definitive endoderm and pancreatic progenitor (PP) cells. PP cells from all lines differentiated efficiently into islet cell clusters that robustly expressed the corresponding reporters and contained glucose-responsive, insulin-producing cells. To demonstrate the applicability of these hPS cell reporter lines in a high-content live imaging approach for the identification of optimal differentiation conditions, we adapted our differentiation procedure to generate SC-islet clusters in microwells. This allowed the live confocal imaging of multiple SC-islets for a single condition and, using this approach, we found that the use of the N21 supplement in the last stage of the differentiation increased the number of monohormonal β-cells without affecting the number of α-cells in the SC-islets. The hPS cell reporter lines and the high-content live imaging approach described here will enable the efficient assessment of multiple conditions for the optimal differentiation and maturation of SC-islets.
    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(hPSC)技术的最新进展促使人类神经元和脑类器官的新研究领域和应用的出现。脑类器官作为一种概括高级结构的体外模型系统而受到关注,细胞多样性和大脑的功能来探索大脑发育,疾病建模,药物筛选,和再生医学。脑类器官技术与各种研究领域的丰富相互作用加速了这一进展。与人脑类器官技术的跨学科方法为更准确地理解人脑提供了更有序的进步。在这次审查中,我们总结了hPSCs在二维和三维培养系统中的神经诱导状态以及使用脑类器官对神经退行性疾病的建模。我们还强调了用于组装空间高阶神经组织的最新生物工程技术,以及脑类器官技术对理解人脑潜力和能力的前景。
    Recent advances in human pluripotent stem cell (hPSC) technologies have prompted the emergence of new research fields and applications for human neurons and brain organoids. Brain organoids have gained attention as an in vitro model system that recapitulates the higher structure, cellular diversity and function of the brain to explore brain development, disease modeling, drug screening, and regenerative medicine. This progress has been accelerated by abundant interactions of brain organoid technology with various research fields. A cross-disciplinary approach with human brain organoid technology offers a higher-ordered advance for more accurately understanding the human brain. In this review, we summarize the status of neural induction in two- and three-dimensional culture systems from hPSCs and the modeling of neurodegenerative diseases using brain organoids. We also highlight the latest bioengineered technologies for the assembly of spatially higher-ordered neural tissues and prospects of brain organoid technology toward the understanding of the potential and abilities of the human brain.
    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
    小胶质细胞是中枢神经系统(CNS)的常驻巨噬细胞。它们的吞噬活动在大脑发育和体内平衡以及大量的大脑病理中是中心的。然而,对构图知之甚少,动力学,以及在稳态和病理条件下人类小胶质细胞吞噬体的功能。这里,我们开发了一种在各种体外条件下从人多能干细胞衍生的小胶质细胞中快速分离纯的和完整的吞噬体的方法,从人脑活检中,用于无偏的多维分析。吞噬体谱分析表明,小胶质细胞吞噬体可以感知其环境的微小变化,并且具有高度动态性。我们检测到参与突触体内平衡的蛋白质,或者与脑部病理有关,并将吞噬体鉴定为喹啉酸在细胞质中从头产生烟酰胺腺嘌呤二核苷酸(NAD)的储存和代谢位点。我们的发现强调了吞噬体在健康和患病大脑的小胶质细胞功能中的核心作用。
    Microglia are the resident macrophages of the central nervous system (CNS). Their phagocytic activity is central during brain development and homeostasis-and in a plethora of brain pathologies. However, little is known about the composition, dynamics, and function of human microglial phagosomes under homeostatic and pathological conditions. Here, we developed a method for rapid isolation of pure and intact phagosomes from human pluripotent stem cell-derived microglia under various in vitro conditions, and from human brain biopsies, for unbiased multiomic analysis. Phagosome profiling revealed that microglial phagosomes were equipped to sense minute changes in their environment and were highly dynamic. We detected proteins involved in synapse homeostasis, or implicated in brain pathologies, and identified the phagosome as the site where quinolinic acid was stored and metabolized for de novo nicotinamide adenine dinucleotide (NAD+) generation in the cytoplasm. Our findings highlight the central role of phagosomes in microglial functioning in the healthy and diseased brain.
    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
    与传统的二维(2D)模型相比,三维(3D)组织模型的模拟天然细胞微环境的能力得到了认可。这一进展是由3D生物打印等组织工程技术的进步推动的。一种制造仿生活组织的有前途的方法。迄今为止,生物打印已经成功地产生了各种组织,创建神经组织模型仍然具有挑战性。在这种情况下,我们提出了一种加速的方法来制造3D感觉神经元(SN)结构使用转基因人类多能干细胞(hPSC)-系,其中包含可诱导的神经原-2(NGN2)表达盒。NGN2hPSC系首先分化为神经c细胞(NCC)祖细胞,然后整合到基于细胞相容性GelMA的生物墨水中进行3D生物打印。在生物打印的NCC中上调的NGN2表达导致诱导的SN(iSN)群体表现出特定的细胞标志物,3D分析揭示了通过支架体积广泛的神经突生长。钙成像显示iSNs的功能活性,包括膜兴奋性和电压门控钠通道(NaV)活性。这种生成3D生物打印iSN结构的有效方法简化了神经组织模型的发展,可用于研究神经发育和疾病状态,并提供翻译潜力。
    Three-dimensional (3D) tissue models have gained recognition for their improved ability to mimic the native cell microenvironment compared to traditional two-dimensional models. This progress has been driven by advances in tissue-engineering technologies such as 3D bioprinting, a promising method for fabricating biomimetic living tissues. While bioprinting has succeeded in generating various tissues to date, creating neural tissue models remains challenging. In this context, we present an accelerated approach to fabricate 3D sensory neuron (SN) structures using a transgenic human pluripotent stem cell (hPSC)-line that contains an inducible Neurogenin-2 (NGN2) expression cassette. The NGN2 hPSC line was first differentiated to neural crest cell (NCC) progenitors, then incorporated into a cytocompatible gelatin methacryloyl-based bioink for 3D bioprinting. Upregulated NGN2 expression in the bioprinted NCCs resulted in induced SN (iSN) populations that exhibited specific cell markers, with 3D analysis revealing widespread neurite outgrowth through the scaffold volume. Calcium imaging demonstrated functional activity of iSNs, including membrane excitability properties and voltage-gated sodium channel (NaV) activity. This efficient approach to generate 3D bioprinted iSN structures streamlines the development of neural tissue models, useful for the study of neurodevelopment and disease states and offering translational potential.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

    求助全文

公众号