首页 | 本学科首页   官方微博 | 高级检索  
相似文献
 共查询到20条相似文献,搜索用时 171 毫秒
1.
单细胞转录组测序是一种在单细胞水平上研究基因表达的技术.多孔板法和液滴法是目前应用于植物研究的两类主要的单细胞转录组技术.首先概述了植物单细胞转录组测序的技术原理和数据分析流程,然后介绍了植物单细胞转录组的研究进展,重点阐述了单细胞转录组测序技术在鉴定植物细胞类型、揭示细胞演化轨迹和构建细胞间调控网络中的应用.单细胞转...  相似文献   

2.
细胞是机体最基本的结构组成及功能单位,细胞类型和功能由其整个转录表达谱决定,通过单细胞转录组测序可以获得单个细胞转录表达谱,由此以高精度分辨率鉴定细胞类型、细胞状态以及稀有类型细胞,从而可以在单细胞水平分析细胞动态变化及细胞间的关系,深入解析驱动细胞变化及细胞异常背后的分子细胞机制。随着单细胞测序技术稳定性和测序通量的提高,以及测序成本的降低,其在发育生物学、肿瘤、免疫及疾病等领域被广泛应用,研究对象主要涉及人及模式生物,在动物上的应用研究相对较少。本文主要介绍单细胞转录组测序技术及其生物学应用并综述目前其在动物上的一些开创性研究,以期为今后更好的在动物上应用单细胞转录组测序提供方法参考。  相似文献   

3.
单细胞测序技术凭借其能全面反映细胞群体异质性这一优势,近年来发展迅速.其中,单细胞转录组测序技术提供了在分子水平上对细胞作分类或表征的替代方法,在发育生物学、神经科学、血液学、免疫及癌症等研究领域均展示出了广泛的应用前景.本文总结了近年来单细胞转录组测序技术的主要发展趋势,并列举了该技术在造血系统中的应用.  相似文献   

4.
近年来,高通量测序技术(Next-generation sequencing,NGS)快速发展,已广泛应用于生命科学各个领域,但传统的混合细胞测序(Bulk cell sequencing)检测的是细胞群体的总平均反应,无法反应每个细胞的真实情况,这会影响研究者对细胞功能认知的准确性。单细胞测序技术(Single cell sequencing,sc-Seq)的出现,从一定程度上解决了传统测序固有的缺陷。单细胞测序是针对单个细胞的RNA或DNA进行测序,能够准确测出单个细胞的基因结构和表达状态,从而分析相同表型细胞的异质性。本文首先介绍单细胞测序的原理、测序类型和测序平台,有助于理解单细胞测序和在进行科研项目时设计合适的项目方案。进一步介绍单细胞转录组测序的分析流程和各种常用的分析工具或软件,并重点阐述单细胞转录组测序分析中的细胞聚类和拟时序分析的原理和研究进展,为进行单细胞转录组测序数据分析提供参考。最后,本文简述了单细胞测序研究热度、单细胞测序的应用、挑战和展望等,有助于更全面地认识单细胞测序。  相似文献   

5.
单细胞转录组技术在单细胞水平上进行转录组测序,提供了单个细胞的基因表达差异信息,使在单细胞尺度下研究个体细胞、相关环境细胞及其相互作用的机理成为可能.近年来,单细胞转录组技术在c DNA扩增原理上经历了从末端加尾、体外逆转录到模板置换的方法发展,大大提高了基因检测的数量、基因表达的准确性等.同时,在单细胞选取方式上进行了从96/384孔板到油包水液滴以及纳米微孔的创新,在提高通量和重复性的同时降低了整体实验成本.单细胞转录组技术广泛应用于细胞群体分类和异质性研究,推动了从发育生物学到正常、病态组织细胞图谱的构建.本文对单细胞转录组技术近年的技术进展以及在人类细胞图谱构建中的应用进行了综述.  相似文献   

6.
随着现代生物医学的发展,基因测序技术已经在肿瘤学研究的多个领域得到广泛应用,但对肿瘤整体进行测序往往会掩盖细胞之间的异质性。为了探索单个肿瘤细胞的行为特性,肿瘤细胞异质性已成为从事肿瘤方面研究的科研和临床专家目前共同关注的焦点。单细胞转录组测序能从单个细胞的水平揭示同一肿瘤内不同细胞之间的变异程度与相互联系。随着单细胞分离、二代测序技术的发展,单细胞转录组测序日趋成熟。未来在临床中,单细胞转录组测序技术将会在肿瘤的早期诊断、监测、临床预后以及个体化用药等方面得到越来越广泛的应用。  相似文献   

7.
药物成瘾是复杂的中枢神经系统疾病,相关基础与临床研究均证实药物成瘾的神经机制及神经环路在成瘾行为形成的不同阶段逐渐发生改变。利用全基因组关联研究、全基因组测序、全外显子测序或高通量转录组测序等技术的组学研究对包括药物成瘾在内的精神疾病遗传的脆弱性进行了深入研究。上述单核苷酸多态性检测技术或测序技术主要预测疾病的遗传风险位点。然而,许多中枢神经系统疾病的发生与环境因素密切相关,而且在疾病发展的不同阶段,相关基因的表达存在脑区特异性的细胞异质性信息。因此,传统研究对发病机制的解释存在一定的局限性。单细胞转录组测序技术是针对单个细胞进行转录水平的测定,规避了传统测序对细胞群体平均转录水平检测的缺点,可以定量描述细胞异质性。近年来,单细胞转录测序技术在神经精神科学研究中的应用逐渐受到关注,本文总结了该技术在神经科学研究中的重要应用,并以药物成瘾为例,重点阐述说明其在中枢神经系统疾病中的应用价值。  相似文献   

8.
同一组织中的细胞往往被认为是具有相同状态的功能单位,传统的检测手段分析的是细胞群体的总体平均反应。然而通过对单个细胞的DNA或RNA进行测序,表明组织系统层面的功能是由异质性细胞构成的。单细胞测序以单个细胞为单位,通过全基因组或转录组扩增,进行高通量测序,能够揭示单个细胞的基因结构和基因表达状态,反映细胞间的异质性,在肿瘤、发育生物学、微生物学、神经科学等领域发挥重要作用,正成为生命科学研究的焦点。单细胞测序的难点是单个细胞的分离、单细胞基因组和转录组的扩增。本文主要介绍和分析了单细胞测序技术中常用的单细胞分离技术、单细胞基因组扩增技术和转录组扩增技术及其优缺点,并对当前已经取得成果的应用领域进行了阐述,为单细胞测序技术的研究与应用提供参考。  相似文献   

9.
单细胞测序技术是在单个细胞水平上对基因组或者转录组进行测序,从而分析相同表型细胞的遗传异质性,或获得难以培养微生物的遗传信息。单细胞测序技术的应用已经深刻地改变了我们对一系列生物学现象的理解,包括基因转录、胚胎发育、癌变。对单细胞测序技术的方法和应用进行了概述,并对其在传染病研究中的应用进行了详细介绍。  相似文献   

10.
细胞的转录组决定其生理状态,每个细胞的转录组都是唯一的。借助单细胞转录组测序可分析单个干细胞的转录组特征,通过进一步的运算方法可以根据转录组特征对细胞进行细胞状态测定以及系谱分化特征的重建,在干细胞及组织发育研究中发挥了强大的作用,推动了其快速发展,加速了对干细胞分化及组织发育的相关过程及调控路径的认识。尤其是在干细胞领域的应用,得益于新算法的发展,单细胞转录组测序分析可用来阐述干细胞的起源、异质性,尤其是对干细胞的分化过程进行连续观察。本文主要对应用于干细胞分化相关研究的单细胞转录组测序数据新的算法及其应用进行了综述。  相似文献   

11.
多细胞有机体的细胞类型多且复杂,细胞间普遍存在异质性。目前,单细胞转录组测序(single-cell RNA sequencing,scRNA-seq)技术是一项新兴的研究单个细胞转录水平的技术,其从数千个平行的细胞中生成转录谱,揭示个体细胞基因组的差异性表达,反映细胞间的异质性,从而鉴定出不同细胞类型,形成组织或器官的细胞图谱,在生物和临床医学等领域发挥重要作用。该文在对scRNA-seq测序平台进行阐述和比较的基础上,着重介绍其在神经系统和免疫系统细胞类型探索中的应用,并且总结scRNA-seq与空间转录组技术相结合的研究成果。  相似文献   

12.
屈亮  李素  仇华吉 《遗传》2020,(3):269-277
单细胞RNA测序(single-cell RNA sequencing, scRNA-seq)技术已经成为不同领域中研究细胞异质性的有效工具。在病毒研究领域中,利用该技术分析病毒和细胞的转录组,可以在单细胞水平上检测病毒感染的动态变化,了解病毒与细胞间复杂的相互作用。本文简述了scRNA-seq技术,着重介绍病毒感染宿主细胞后scRNA-seq研究的最新进展,同时也描述了细胞周期、基因表达、细胞状态等细胞异质性对病毒感染过程的影响,以及病毒变异对其本身感染过程的影响。此外,本文还分析了scRNA-seq在研究病毒–宿主互作动态变化方面具有的独特优势,及其在病毒研究领域中广阔的应用前景,为揭示病毒的感染与致病机制、抗病毒靶标的开发等提供参考。  相似文献   

13.
Accurate identification of cell types from single-cell RNA sequencing(scRNA-seq) data plays a critical role in a variety of scRNA-seq analysis studies. This task corresponds to solving an unsupervised clustering problem, in which the similarity measurement between cells affects the result significantly. Although many approaches for cell type identification have been proposed,the accuracy still needs to be improved. In this study, we proposed a novel single-cell clustering framework based on similarity learning, called SSRE. SSRE models the relationships between cells based on subspace assumption, and generates a sparse representation of the cell-to-cell similarity.The sparse representation retains the most similar neighbors for each cell. Besides, three classical pairwise similarities are incorporated with a gene selection and enhancement strategy to further improve the effectiveness of SSRE. Tested on ten real scRNA-seq datasets and five simulated datasets, SSRE achieved the superior performance in most cases compared to several state-of-the-art single-cell clustering methods. In addition, SSRE can be extended to visualization of scRNA-seq data and identification of differentially expressed genes. The matlab and python implementations of SSRE are available at https://github.com/CSUBioGroup/SSRE.  相似文献   

14.
15.
16.
The single-cell RNA sequencing (scRNA-seq) technologies obtain gene expression at single-cell resolution and provide a tool for exploring cell heterogeneity and cell types. As the low amount of extracted mRNA copies per cell, scRNA-seq data exhibit a large number of dropouts, which hinders the downstream analysis of the scRNA-seq data. We propose a statistical method, SDImpute (Single-cell RNA-seq Dropout Imputation), to implement block imputation for dropout events in scRNA-seq data. SDImpute automatically identifies the dropout events based on the gene expression levels and the variations of gene expression across similar cells and similar genes, and it implements block imputation for dropouts by utilizing gene expression unaffected by dropouts from similar cells. In the experiments, the results of the simulated datasets and real datasets suggest that SDImpute is an effective tool to recover the data and preserve the heterogeneity of gene expression across cells. Compared with the state-of-the-art imputation methods, SDImpute improves the accuracy of the downstream analysis including clustering, visualization, and differential expression analysis.  相似文献   

17.
During early embryonic development, cell fate commitment represents a critical transition or"tipping point"of embryonic differentiation, at which there is a drastic and qualitative shift of the cell populations. In this study, we presented a computational approach, scGET, to explore the gene–gene associations based on single-cell RNA sequencing (scRNA-seq) data for critical transition prediction. Specifically, by transforming the gene expression data to the local network entropy, the single-cell graph entropy (SGE) value quantitatively characterizes the stability and criticality of gene regu-latory networks among cell populations and thus can be employed to detect the critical signal of cell fate or lineage commitment at the single-cell level. Being applied to five scRNA-seq datasets of embryonic differentiation, scGET accurately predicts all the impending cell fate transitions. After identifying the"dark genes"that are non-differentially expressed genes but sensitive to the SGE value, the underlying signaling mechanisms were revealed, suggesting that the synergy of dark genes and their downstream targets may play a key role in various cell development processes. The application in all five datasets demonstrates the effectiveness of scGET in analyzing scRNA-seq data from a network perspective and its potential to track the dynamics of cell differentiation. The source code of scGET is accessible at https://github.com/zhongjiayuna/scGET_Project.  相似文献   

18.
High-throughput single-cell RNA sequencing (scRNA-seq) has advantages over traditional RNA-seq to explore spatiotemporal information on gene dynamic expressions in heterogenous tissues. We performed Drop-seq, a method for the dropwise sequestration of single cells for sequencing, on protoplasts from the differentiating xylem of Populus alba×Populus glandulosa. The scRNA-seq profiled 9,798 cells, which were grouped into 12 clusters. Through characterization of differentially expressed genes in each cluster and RNA in situ hybridizations, we identified vessel cells, fiber cells, ray parenchyma cells and xylem precursor cells. Diffusion pseudotime analyses revealed the differentiating trajectory of vessels, fiber cells and ray parenchyma cells and indicated a different differentiation process between vessels and fiber cells, and a similar differentiation process between fiber cells and ray parenchyma cells. We identified marker genes for each cell type (cluster) and key candidate regulators during developmental stages of xylem cell differentiation. Our study generates a high-resolution expression atlas of wood formation at the single cell level and provides valuable information on wood formation.  相似文献   

19.
With the tremendous increase of publicly available single-cell RNA-sequencing (scRNA-seq) datasets, bioinformatics methods based on gene co-expression network are becoming efficient tools for analyzing scRNA-seq data, improving cell type prediction accuracy and in turn facilitating biological discovery. However, the current methods are mainly based on overall co-expression correlation and overlook co-expression that exists in only a subset of cells, thus fail to discover certain rare cell types and sensitive to batch effect. Here, we developed independent component analysis-based gene co-expression network inference (ICAnet) that decomposed scRNA-seq data into a series of independent gene expression components and inferred co-expression modules, which improved cell clustering and rare cell-type discovery. ICAnet showed efficient performance for cell clustering and batch integration using scRNA-seq datasets spanning multiple cells/tissues/donors/library types. It works stably on datasets produced by different library construction strategies and with different sequencing depths and cell numbers. We demonstrated the capability of ICAnet to discover rare cell types in multiple independent scRNA-seq datasets from different sources. Importantly, the identified modules activated in acute myeloid leukemia scRNA-seq datasets have the potential to serve as new diagnostic markers. Thus, ICAnet is a competitive tool for cell clustering and biological interpretations of single-cell RNA-seq data analysis.  相似文献   

20.
设为首页 | 免责声明 | 关于勤云 | 加入收藏

Copyright©北京勤云科技发展有限公司  京ICP备09084417号