Single Cell

单细胞
  • 文章类型: Journal Article
    扩大单细胞组学在肺动脉高压(PH)研究中带来的重要进步,这篇综述深入研究了这些技术是如何在理解这种复杂疾病的新时代进行试验的。通过利用单细胞转录组学(scRNA-seq)的功能,研究人员现在可以解剖复杂的肺部细胞生态系统,检查关键参与者,如内皮细胞,平滑肌细胞,周细胞,和免疫细胞,及其在PH发病机制中的独特作用。这种更细粒度的观点超出了传统批量分析的限制,允许识别先前在汇总数据中模糊的新治疗靶标。基于涉及病理变化的细胞的单细胞组学的连接体分析可以更清晰地揭示细胞亚型中的细胞相互作用和转变。此外,审查承认未来的挑战,包括需要增强scRNA-seq的分辨率以捕获细胞变化的更精细的细节,克服处理人体组织样本的后勤障碍,以及整合各种组学方法以充分理解PH的分子基础的必要性。这些单细胞技术的前景是巨大的,为靶向药物开发和发现用于早期诊断和疾病监测的生物标志物提供了潜力。通过这些进步,该领域越来越接近实现PH患者的精准医疗目标。
    Expanding upon the critical advancements brought forth by single-cell omics in pulmonary hypertension (PH) research, this review delves deep into how these technologies have been piloted in a new era of understanding this complex disease. By leveraging the power of single cell transcriptomics (scRNA-seq), researchers can now dissect the complicated cellular ecosystem of the lungs, examining the key players such as endothelial cells, smooth muscle cells, pericytes, and immune cells, and their unique roles in the pathogenesis of PH. This more granular view is beyond the limitations of traditional bulk analysis, allowing for the identification of novel therapeutic targets previously obscured in the aggregated data. Connectome analysis based on single-cell omics of the cells involved in pathological changes can reveal a clearer picture of the cellular interactions and transitions in the cellular subtypes. Furthermore, the review acknowledges the challenges that lie ahead, including the need for enhancing the resolution of scRNA-seq to capture even finer details of cellular changes, overcoming logistical barriers in processing human tissue samples, and the necessity of integrating diverse omics approaches to fully comprehend the molecular underpinnings of PH. The promise of these single-cell technologies is immense, offering the potential for targeted drug development and the discovery of biomarkers for early diagnosis and disease monitoring. Through these advancements, the field moves closer to realizing the goal of precision medicine for patients with PH.
    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
    胶质母细胞瘤(GBM)是一种高度异质性的疾病,临床预后较差。为了全面剖析GBM的分子景观和GBM进展中的异质巨噬细胞簇,这项研究整合了单细胞和批量转录组数据,以识别与GBM预后显著相关的独特的前肿瘤巨噬细胞簇,并开发GBM预后标志以促进以前的亚型.利用神经胶质瘤单细胞测序数据,我们确定了一个新的促肿瘤巨噬细胞亚群,以S100A9为标志,可能与内皮细胞相互作用,通过血管生成促进肿瘤进展。进一步有利于临床应用,利用与肿瘤前巨噬细胞相关的基因建立了预后特征.属于高危人群的患者,其特征是与肿瘤进展相关的功能富集,包括上皮-间质转化和缺氧,在TERT启动子区域显示升高的突变,MGMT启动子区域的甲基化减少,较差的预后,对替莫唑胺治疗的反应减弱,从而有效区分GBM患者的预后结果。我们的研究揭示了神经胶质瘤复杂的微环境,并确定了开发新治疗方法的潜在分子靶标。
    Glioblastoma (GBM) is a highly heterogeneous disease with poor clinical outcomes. To comprehensively dissect the molecular landscape of GBM and heterogeneous macrophage clusters in the progression of GBM, this study integrates single-cell and bulk transcriptome data to recognize a distinct pro-tumor macrophage cluster significantly associated with the prognosis of GBM and develop a GBM prognostic signature to facilitate prior subtypes. Leveraging glioma single-cell sequencing data, we identified a novel pro-tumor macrophage subgroup, marked by S100A9, which might interact with endothelial cells to facilitate tumor progression via angiogenesis. To further benefit clinical application, a prognostic signature was established with the genes associated with pro-tumor macrophages. Patients classified within the high-risk group characterized with enrichment in functions related to tumor progression, including epithelial-mesenchymal transition and hypoxia, displays elevated mutations in the TERT promoter region, reduced methylation in the MGMT promoter region, poorer prognoses, and diminished responses to temozolomide therapy, thus effectively discriminating between the prognostic outcomes of GBM patients. Our research sheds light on the intricate microenvironment of gliomas and identifies potential molecular targets for the development of novel therapeutic approaches.
    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
    具有双标记的马赛克分析(MADM)是一种强大的遗传方法,通常用于谱系追踪和解开具有单细胞分辨率的候选基因的细胞自主和组织范围的作用。鉴于标签相对稀疏,根据一个人选择的19条MADM染色体中的哪一条,MADM方法代表了细胞形态分析的绝佳机会。各种MADM研究包括中枢神经系统(CNS)形态异常和表型的报告。任何候选基因的MADM都可以轻松地将形态学分析纳入实验工作流程。这里,我们描述了我们在最近的各种MADM研究过程中开发的形态细胞分析方法。本章将特别关注量化中枢神经系统内神经元和星形胶质细胞形态方面的方法,但是这些方法可以广泛应用于整个生物体中的任何MADM标记的细胞。我们将涵盖两个分析-体细胞体积和树突表征-体感皮层中锥体神经元的物理特征,和两个分析-体积和Sholl分析-星形胶质细胞形态。
    Mosaic Analysis with Double Markers (MADM) is a powerful genetic method typically used for lineage tracing and to disentangle cell autonomous and tissue-wide roles of candidate genes with single cell resolution. Given the relatively sparse labeling, depending on which of the 19 MADM chromosomes one chooses, the MADM approach represents the perfect opportunity for cell morphology analysis. Various MADM studies include reports of morphological anomalies and phenotypes in the central nervous system (CNS). MADM for any candidate gene can easily incorporate morphological analysis within the experimental workflow. Here, we describe the methods of morphological cell analysis which we developed in the course of diverse recent MADM studies. This chapter will specifically focus on methods to quantify aspects of the morphology of neurons and astrocytes within the CNS, but these methods can broadly be applied to any MADM-labeled cells throughout the entire organism. We will cover two analyses-soma volume and dendrite characterization-of physical characteristics of pyramidal neurons in the somatosensory cortex, and two analyses-volume and Sholl analysis-of astrocyte morphology.
    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
    了解胃肠道(GI)免疫细胞的多样性,尤其是固有肌层,对于了解它们在维持肠神经元和平滑肌中的作用以及它们对胃肠道运动的贡献至关重要。这里,我们提出了从人胃固有肌层分离单个免疫细胞的详细方案。我们描述了组织保存的步骤,解剖,和固有肌层的分离。然后,我们详细介绍了CD45+细胞的磁性分选和单细胞RNA测序(scRNA-seq)分析的程序。有关此协议的使用和执行的完整详细信息,请参考Chikkamenahalli等1。
    Understanding the diversity of gastrointestinal (GI) immune cells, especially in the muscularis propria, is crucial for understanding their role in the maintenance of enteric neurons and smooth muscle and their contribution to GI motility. Here, we present a detailed protocol for isolating single immune cells from the human gastric muscularis propria. We describe steps for tissue preservation, dissection, and dissociation of the muscularis propria. We then detail procedures for magnetic sorting of CD45+ cells and single-cell RNA sequencing (scRNA-seq) analysis. For complete details on the use and execution of this protocol, please refer to Chikkamenahalli et al.1.
    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
    细胞类型特异性结构域是空间分辨转录组(SRT)组织中的解剖结构域,其中特定细胞类型同时富集。使用现有的计算方法来检测具有低比例细胞类型的特定域是具有挑战性的,与其他细胞类型特异性结构域部分重叠或甚至在内部。这里,我们建议去现场,它将分割和反卷积合成为一个集合来生成细胞类型的模式,检测低比例的细胞类型特异性结构域,并直观地显示这些领域。实验评估表明,De-spot使我们能够发现癌症相关成纤维细胞和免疫相关细胞之间的共定位,这表明给定切片中潜在的肿瘤微环境(TME)域,被以前的计算方法掩盖了。我们进一步阐明了鉴定的结构域,发现Srgn可能是SRT切片中的关键TME标记。通过破译乳腺癌组织中的T细胞特异性结构域,De-spot还显示,耗竭T细胞的比例在侵袭性与侵袭性之间显着增加。导管癌.
    Cell-type-specific domains are the anatomical domains in spatially resolved transcriptome (SRT) tissues where particular cell types are enriched coincidentally. It is challenging to use existing computational methods to detect specific domains with low-proportion cell types, which are partly overlapped with or even inside other cell-type-specific domains. Here, we propose De-spot, which synthesizes segmentation and deconvolution as an ensemble to generate cell-type patterns, detect low-proportion cell-type-specific domains, and display these domains intuitively. Experimental evaluation showed that De-spot enabled us to discover the co-localizations between cancer-associated fibroblasts and immune-related cells that indicate potential tumor microenvironment (TME) domains in given slices, which were obscured by previous computational methods. We further elucidated the identified domains and found that Srgn may be a critical TME marker in SRT slices. By deciphering T cell-specific domains in breast cancer tissues, De-spot also revealed that the proportions of exhausted T cells were significantly increased in invasive vs. ductal carcinoma.
    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
    脂肪组织免疫细胞具有异质性和动态性,改变新陈代谢,驱动免疫反应。这里,我们提出了一种使用基于荧光的流式细胞术和分选成纯群体的小鼠脂肪组织免疫细胞的评估和表征方案。我们描述了分离基质血管部分的步骤,抗体染色,并通过流式细胞术收集数据。我们还将讨论常见问题和故障排除步骤。有关此协议的使用和执行的完整详细信息,PleaserefertoCareyetal.1.
    Adipose tissue immune cells are heterogeneous and dynamic, alter metabolism, and drive immune responses. Here, we present a protocol for assessment and characterization of murine adipose tissue immune cells using fluorescence-based flow cytometry and sorting into pure populations. We describe steps for isolation of the stromovascular fraction, antibody staining, and data collection by flow cytometry. We also discuss common issues and troubleshooting steps. For complete details on the use and execution of this protocol, please refer to Carey et al.1.
    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
    哺乳动物肾脏通过从肾单位和输尿管祖细胞产生的多种上皮细胞类型维持流体稳态。为了扩展对肾脏上皮网络的发展理解,我们比较了发展中的人类(10.6-17.6周;n=10)和小鼠(出生后第0天[P]0;n=10)肾脏的染色质组织(转座酶可接近的染色质测序[ATAC-seq];112,864个细胞核)和基因表达(单细胞/细胞核RNA测序[RNA-seq];109,477个细胞/细胞核),补充分析与早期发布的小鼠数据集。在物种水平上分析单细胞/核数据集,然后肾单位和输尿管细胞谱系被提取并整合到一个共同的,跨物种,多模态数据集。比较计算分析确定了染色质组织和相关基因活性的保守和不同特征,确定特定物种和特定细胞类型的调控计划。人类富集基因活性的原位验证指向肾脏发育中人类特异性信号相互作用。Further,通过全基因组关联研究(GWAS),人类特异性增强子区域与肾脏疾病相关,突出了从发育建模中获得临床洞察力的潜力。
    The mammalian kidney maintains fluid homeostasis through diverse epithelial cell types generated from nephron and ureteric progenitor cells. To extend a developmental understanding of the kidney\'s epithelial networks, we compared chromatin organization (single-nuclear assay for transposase-accessible chromatin sequencing [ATAC-seq]; 112,864 nuclei) and gene expression (single-cell/nuclear RNA sequencing [RNA-seq]; 109,477 cells/nuclei) in the developing human (10.6-17.6 weeks; n = 10) and mouse (post-natal day [P]0; n = 10) kidney, supplementing analysis with published mouse datasets from earlier stages. Single-cell/nuclear datasets were analyzed at a species level, and then nephron and ureteric cellular lineages were extracted and integrated into a common, cross-species, multimodal dataset. Comparative computational analyses identified conserved and divergent features of chromatin organization and linked gene activity, identifying species-specific and cell-type-specific regulatory programs. In situ validation of human-enriched gene activity points to human-specific signaling interactions in kidney development. Further, human-specific enhancer regions were linked to kidney diseases through genome-wide association studies (GWASs), highlighting the potential for clinical insight from developmental modeling.
    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
    高通量单细胞测序的最新进展对可以解决单细胞多组学数据的高复杂性的计算模型产生了迫切的需求。需要精细的单细胞多组学整合模型,以避免对特定模式的偏见并克服稀疏性。还必须考虑混淆生物信号的批量效应。这里,我们引入了一种新的单细胞多组学整合模型,基于变分专家乘积自动编码器和对抗性学习的单细胞多组体自动编码器集成(scMaui)。scMaui基于专家乘积方法计算多个边际分布的联合表示,这对于模态中的缺失值特别有效。此外,它克服了以前基于VAE的整合方法在批量效应校正和限制性适用测定方面的局限性。它处理多个批量效果,独立接受离散值和连续值,以及提供各种重建损失函数,以涵盖所有可能的分析和预处理管道。我们证明,与其他方法相比,scMaui在许多任务中实现了卓越的性能。进一步的下游分析也证明了其在鉴定测定和发现隐藏亚群之间的关系方面的潜力。
    The recent advances in high-throughput single-cell sequencing have created an urgent demand for computational models which can address the high complexity of single-cell multiomics data. Meticulous single-cell multiomics integration models are required to avoid biases towards a specific modality and overcome sparsity. Batch effects obfuscating biological signals must also be taken into account. Here, we introduce a new single-cell multiomics integration model, Single-cell Multiomics Autoencoder Integration (scMaui) based on variational product-of-experts autoencoders and adversarial learning. scMaui calculates a joint representation of multiple marginal distributions based on a product-of-experts approach which is especially effective for missing values in the modalities. Furthermore, it overcomes limitations seen in previous VAE-based integration methods with regard to batch effect correction and restricted applicable assays. It handles multiple batch effects independently accepting both discrete and continuous values, as well as provides varied reconstruction loss functions to cover all possible assays and preprocessing pipelines. We demonstrate that scMaui achieves superior performance in many tasks compared to other methods. Further downstream analyses also demonstrate its potential in identifying relations between assays and discovering hidden subpopulations.
    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:来自单细胞RNA测序数据的谱系追踪和轨迹推断在揭示驱动发育和疾病的遗传程序方面具有巨大潜力。单细胞数据集被认为提供了关于组织的不同细胞结构的无偏视图。采样偏差,然而,可以使单细胞数据集偏离它们要代表的细胞组成。
    结果:我们展示了一种新形式的抽样偏差,由与不断增长的重复采样有关的统计现象引起的,异质种群。细胞的相对生长速率影响它们将在多个时间点观察到的克隆中取样的概率。我们通过模拟研究和对T细胞发育的实时过程的分析来支持我们的概率推导。我们发现这种偏差会影响命运概率预测,我们探索如何开发对这种偏差具有鲁棒性的轨迹推理方法。
    背景:用于模拟数据集和创建本手稿中的图形的源代码可以在https://github.com/rbonhamcarter/simulate-clones的python中免费获得。LineageOT方法扩展的python实现可在https://github.com/rbonhamcarter/LineageOT/tree/multi-time-clone上免费获得。
    背景:补充数据可在Bioinformation在线获得。
    BACKGROUND: Lineage tracing and trajectory inference from single-cell RNA-sequencing data hold tremendous potential for uncovering the genetic programs driving development and disease. Single cell datasets are thought to provide an unbiased view on the diverse cellular architecture of tissues. Sampling bias, however, can skew single cell datasets away from the cellular composition they are meant to represent.
    RESULTS: We demonstrate a novel form of sampling bias, caused by a statistical phenomenon related to repeated sampling from a growing, heterogeneous population. Relative growth rates of cells influence the probability that they will be sampled in clones observed across multiple time points. We support our probabilistic derivations with a simulation study and an analysis of a real time-course of T-cell development. We find that this bias can impact fate probability predictions, and we explore how to develop trajectory inference methods which are robust to this bias.
    METHODS: Source code for the simulated datasets and to create the figures in this manuscript is freely available in python at https://github.com/rbonhamcarter/simulate-clones. A python implementation of the extension of the LineageOT method is freely available at https://github.com/rbonhamcarter/LineageOT/tree/multi-time-clones.
    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
    单细胞RNA测序(scRNA-seq)仍然是转录组细胞作图的最新技术。这里,我们提供了一种方案来生成罕见心肌细胞群体的高分辨率scRNA-seq(例如,再生/分割,等。)来自小鼠和斑马鱼心脏以及诱导的多能干细胞,及时收集,以实现详细的转录组学洞察。我们描述了活力染色的连续步骤,甲醇固定,storage,和细胞分选以保持适用于scRNA-seq的RNA完整性以及如示例所示的数据的质量评估。有关此协议的使用和执行的完整详细信息,请参考Bak等人1。
    Single-cell RNA sequencing (scRNA-seq) remains state-of-the-art for transcriptomic cell-mapping. Here, we provide a protocol to generate high-resolution scRNA-seq of rare cardiomyocyte populations (e.g., regenerating/dividing, etc.) from mouse and zebrafish hearts as well as induced pluripotent stem cells, collected in time to achieve detailed transcriptomic insight. We describe the serial steps of viability staining, methanol fixation, storage, and cell sorting to preserve RNA integrity suited for scRNA-seq as well as the quality assessment of the data as shown by examples. For complete details on the use and execution of this protocol, please refer to Bak et al.1.
    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