How far is single-cell sequencing from clinical application?
Jiaqiang Zhang,William Wang,Jianan Huang,Xiangdong Wang,Yiming Zeng
DOI: https://doi.org/10.1002/ctm2.117
IF: 8.554
2020-01-01
Clinical and Translational Medicine
Abstract:Single-cell sequencing (SC-seq) is rapidly becoming one of the most important approaches to single-cell biomedicine and is centered around single-cell RNA sequencing (scRNA-seq) and single-cell DNA sequencing (scDNA-seq). Growing evidence suggests that SC-seq can significantly speed up the identification of new cell types, intercellular heterogeneity, function-based sub-categories, intelligent cell and human cell altars, disease biomarkers, therapeutic targets, and it can evaluate intra- and intercellular communication.1-3 Molecular characteristics of single cells are uncovered through transcriptome to chromatin accessibility, DNA mutation to histone modification, genome sequences to multi-dimensions, phenomes to functions, and from dynamics to spatial alternations. SC-seq is an approach that contributes to the understanding of interactions and communications between RNA-DNA-proteins, proteins-proteins, cells-cells, and pathogens-host cells from which molecular mechanisms of disease pathogeneses and progression can be explored. Tumor heterogeneity is a critical factor that influences the response of tumor cells to chemotherapy and target-based individualized therapy. New clinical strategies for precision medicine are designed according to tumor heterogeneity and dynamic mutations to improve the prognosis of patients with multiple metastases and reoccurrences.4 The value of SC-seq in clinical and translational medicine lies in its ability to develop our understanding in tumor heterogeneity, genesis, evolution, and metastasis in various diseases.5-8 SC-seq also plays an important role in monitoring on/off-targets and the post-operation of gene editing for gene therapy.9 The aim of this editorial is to provide a brief overview of the recent development of scRNA-seq, scDNA-seq, single cell multi-omics (scMO), trans-omics (scTO), and to highlight the challenges that need to be faced during the clinical application of SC-seq. scRNA-seq is no longer a foreign word for clinicians and is almost ready for clinical application. Single-cell transcriptome profiles have been widely mapped in many human tissues/organs and diseases, due to the application of scRNA-seq. Most studies on single cells from patients are designed to explore molecular mechanisms of diseases, map transcriptomic phenomes of disorders, uncover rare cell types and cellular subpopulations developed during disease occurrence, define new categories of disease and patient stratification, and to monitor therapeutic responses. Differences in single-cell transcriptomic profiles are usually observed by comparing healthy patients to patients with disease. However, there is usually a lack in comparison based on clinical considerations, for example, among disease subtypes, stages, severities, durations, and dynamic responses to drugs. A key challenge is to develop a definition for scRNA-seq platforming that can be used for clinical application, translation of single-cell transcriptome phenomes to clinical precision medicine, and the dynamic monitoring of target cell-specific responses to drugs. The integration of scRNA-seq profiles with scDNA-seq and genomic or proteomic phenomes has recently been proposed as it can aid in understanding transcriptome-associated regulations and functions, heterogeneity-dependent tumorigenesis, and the interaction among intracellular signal pathways, although comprehensive analyses needs to be standardized and uncomplicated. Copy numbers, mutations, and modifications at a single cell resolution can demonstrate intercellular variations, metastatic lineages, and clonal lineages in cancer. scDNA-seq is an approach that can help clinicians in understanding the existence and formation of aneuploidy evolution, phylogenetic trees, heterogeneity, and can prospectively predict the occurrence of metastasis while improving the quality of clinical precision therapy. Target gene panels have been developed for both single-cell and bulk tissue DNA sequencing to improve on efficiency. Leung et al. modeled the clonal evolution and metastatic dissemination of colonic cancer in patients using scDNA-seq, exome sequencing, and targeted deep-sequencing and found the association between cancer cell evolution, acquired mutations numbers, and copy numbers with distant organ sites of metastasis.10 In contrast to tumor bulk sequencing, scDNA-seq analyses could define the self-seeding of tumor cells from the early-dissemination of metastasis and discover complex clonal evolution within tumors. With the improvement of high-throughput capacity, scDNA-seq can be routinely and efficiently performed to detect tumor heterogeneity and evolution. In addition, scDNA-seq is a critical approach to measure epigenetic modifications, for example, aberrant DNA and histone, DNA methylation, acetylation, phosphorylation, aberrant gene expression, imprinting loss, and chromosomal and microsatellite instability. Bohrson et al recently developed a new method named “linked-read analysis” that can precisely profile somatic single nucleotide variants using read-level phasing with nearby germline heterozygous polymorphisms and characterize mutational profiles while predicting somatic mutation rates in single cell level.11 This analysis resulted in more accurate sensitivity for single-cell somatic heterozygous single nucleotide variants, especially for singletons and in the condition with high false discovery rates, while only can detect the part of the single-cell genome that rounds polymorphic germline heterozygous single nucleotide variants. This method provides a new approach that can precisely link the number of mutations with age, mutational processes, and predict mutagenetic rates and intercellular heterogeneity. scMO is a new alternative in clinical and translational medicine that provides multi-dimensional insights into intracellular signal networks and functions and can help to define new function categories using comprehensive analyses. Wang recently described multi-omics as the parallel association and correction among multiple molecular omic profiles.12 SC-seq of genomes, DNA mutations, methylomes, and transcriptomes can demonstrate accurate mechanisms and how multiple network layers regulate and correlate among themselves. For example, some genomic copy-number variations altered RNA expression of genes within the gained or lost genomic regions, rather than DNA methylation to address the association among genetic, epigenetic, and transcriptomic heterogeneities.13 The simultaneous and comprehensive analyses of single cell transcriptome, genome, epigenome, and proteome profiles can demonstrate genotypic and phenotypic characteristics of single cells at multiple layers and integrate regulatory mechanisms of cell evolution with their function. A new method of simultaneous DNA methylome and RNA transcriptome profiles in cells could detect the DNA methylation status of 0.5-1 million CpG sites, mRNA expression of 10 000 genes, and both promoter DNA methylation and RNA transcription of one-third genes.14 It is possible that the numbers of measured genes and methylation sites increase with the improvement of methodology. scMO-seq could play a crucial role in deepening our understanding of molecular mechanisms and the balance between heterogeneity and homogeneity of epigenomic reprogramming in reproduction and development. For example, the reprogramming of genome-scale chromatin statuses and DNA methylation dynamics at the single-cell stage of early embryos was evidenced by chromatin state/nucleosome positioning. DNA methylation, copy number variation, and ploidy were achieved using single-cell multi-omics sequencing technology. Single-cell chromatins overall omic-scale landscape sequencing was used to map DNA methylation, chromatin accessibility, and to find feedback mechanisms between transcription and open chromatin maintenance. Epigenetic reprogramming during human preimplantation development, that is, the inhibition of RNA transcriptional profiles leads to the closure of more than one-third of the open chromatin regions in promoters of human early embryos.15 Early cell-fate choice and the coordination among the three primary germ layers can be regulated by epigenetic reprogramming accompanied by alternations of transcriptional profiles during the onset gastrulation of embryos.16 The ten-eleven translocation-mediated demethylation and elevated chromatin accessibility could drive epigenome rearrangements, contributing to epigenetic prime or the remodeling of regulatory elements associated with each germ layer before cell-fate decisions. There is no doubt that scRNA-seq, scDNA-seq, and scMO/TO are important approaches to exploring the molecular mechanisms of transcriptional function, heterogeneity, epigenetic modification, chromatin inaccessibility, multi-dimensional regulation, and control at single-cell level. Single-cell biomedicine is a new emerging discipline that will improve the understanding of molecular pathogenesis and pathophysiology, facilitate the discovery and validation of biomarkers and targets, strategize target-based precision medicine, and positively impact the life quality of patients. The questions are whether single-cell measurements are ready for clinical application and how far SC-seq is from disease diagnosis, dynamical monitoring, therapeutic efficacy, and prognostic prediction. Luecken and Theis recommended a best-practice of scRNA-seq workflow in data generation and analysis, for example, how to preprocess and visualize raw data, perform quality control, normalize and standardize single cell gene expression and epigenetic modification, correct and integrate transcriptomic profiles with other omics, select and carry out multiple analyses and annotations, and interrupt results to clinicians and patients.17 With the maturation and development of SC-seq methodologies, it seems that the concept of single-cell biomedicine is becoming more accepted by clinicians and patients and the clinical translation of scRNA-seq into real life practice needs to be further considered. It also needs to be further standardized and consortiums of clinical SC-seq usage are urgently required. Lähnemann et al highlighted major challenges that existed in the translational process from single-cell biology to clinical utilities, including sparsity, flexible statistical frameworks, single cell alters, trajectories, patterns in spatially resolved measurements, errors, and missing data, phylogenetic models, data and types of measurement, population genetic parameters, and analysis tools.18 For clinical practice, it is important to standardize the processing of tissue sampling from operations (eg, human sample resection, selection, transfer, and isolation from tissue to single cells), establishing criteria for scRNA-seq, scDNA-seq, and scMO/TO for each disease (eg, disease nature, severity, stage, duration, location, complexity, control pair, genetics, and history), and to simplify and minimize the procedures for measurements and analyses (eg, protocols for each disease and tissue, methodologies of data analyses, and clinical descriptions of result reports to clinicians). Of those factors, the most important issue is to clarify what scRNA-seq, scDNA-seq, and scMO/TO can provide for decision-making, disease-monitoring, and therapy-assistance in clinical practice. In conclusion, the fast development of scRNA-seq, scDNA-seq, and scMO/TO provides a new insight into intra- and intercellular communications and network interactions, and is a new emerging area that can uncover disease-specific biomarkers and target-based precision therapies. The integration of transcriptomic profiles, epigenetic modifications, and multi-dimensional networks at single-cell levels with clinical phenomes has a significant impact on the development of single-cell biomedicine and will be a new strategy to discover new generations of diagnosis and treatment. Clinical translation of single cell measurements into routine practice is highly dependent upon the development of individual protocols and consortiums for each disease, tissue, and cell, with many challenges remaining.
What problem does this paper attempt to address?
-
Simultaneous Profiling of Mrna Transcriptome and DNA Methylome from a Single Cell.
Youjin Hu,Qin An,Ying Guo,Jiawei Zhong,Shuxin Fan,Pinhong Rao,Xialin Liu,Yizhi Liu,Guoping Fan
DOI: https://doi.org/10.1007/978-1-4939-9240-9_21
2019-01-01
Abstract:Single-cell transcriptome and single-cell methylome analysis have successfully revealed the heterogeneity in transcriptome and DNA methylome between single cells, and have become powerful tools to understand the dynamics of transcriptome and DNA methylome during the complicated biological processes, such as differentiation and carcinogenesis. Inspired by the success of using these single-cell -omics methods to understand the regulation of a particular "-ome," more interests have been put on elucidating the regulatory relationship among multiple-omics at single-cell resolution. The simultaneous profiling of multiple-omics from the same single cell would provide us the ultimate power to understand the relationship among different "-omes," but this idea is not materialized for decades due to difficulties to assay extremely tiny amount of DNA or RNA in a single cell. To address this technical challenge, we have recently developed a novel method named scMT-seq that can simultaneously profile both DNA methylome and RNA transcriptome from the same cell. This method enabled us to measure, from a single cell, the DNA methylation status of the most informative 0.5-1 million CpG sites and mRNA level of 10,000 genes, of which 3200 genes can be further analyzed with both promoter DNA methylation and RNA transcription. Using the scMT-seq data, we have successfully shown the regulatory relationship between DNA methylation and transcriptional level in a single dorsal root ganglion neuron (Hu et al., Genome Biol 17:88, 2016). We believe the scMT-seq would be a powerful technique to uncover the regulatory mechanism between transcription and DNA methylation, and would be of wide interest beyond the epigenetics community.
-
Single cell RNA‐sequencing: A powerful yet still challenging technology to study cellular heterogeneity
May Ke,Badran Elshenawy,Helen Sheldon,Anjali Arora,Francesca M Buffa
DOI: https://doi.org/10.1002/bies.202200084
2022-09-08
BioEssays
Abstract:Single‐cell RNA sequencing compares the transcriptomes of individual cells to study their function, evolution and interactions. We review emerging technologies and their applications, and highlight the limitations and the need for reproducible experimental protocols and robust computational pipelines. Almost all biomedical research to date has relied upon mean measurements from cell populations, however it is well established that what it is observed at this macroscopic level can be the result of many interactions of several different single cells. Thus, the observable macroscopic 'average' cannot outright be used as representative of the 'average cell'. Rather, it is the resulting emerging behaviour of the actions and interactions of many different cells. Single‐cell RNA sequencing (scRNA‐Seq) enables the comparison of the transcriptomes of individual cells. This provides high‐resolution maps of the dynamic cellular programmes allowing us to answer fundamental biological questions on their function and evolution. It also allows to address medical questions such as the role of rare cell populations contributing to disease progression and therapeutic resistance. Furthermore, it provides an understanding of context‐specific dependencies, namely the behaviour and function that a cell has in a specific context, which can be crucial to understand some complex diseases, such as diabetes, cardiovascular disease and cancer. Here, we provide an overview of scRNA‐Seq, including a comparative review of emerging technologies and computational pipelines. We discuss the current and emerging applications and focus on tumour heterogeneity a clear example of how scRNA‐Seq can provide new understanding of a complex disease. Additionally, we review the limitations and highlight the need of powerful computational pipelines and reproducible protocols for the broader acceptance of this technique in basic and clinical research.
biochemistry & molecular biology,biology
-
Single-cell sequencing to multi-omics: technologies and applications
Xiangyu Wu,Xin Yang,Yunhan Dai,Zihan Zhao,Junmeng Zhu,Hongqian Guo,Rong Yang
DOI: https://doi.org/10.1186/s40364-024-00643-4
2024-09-27
Abstract:Cells, as the fundamental units of life, contain multidimensional spatiotemporal information. Single-cell RNA sequencing (scRNA-seq) is revolutionizing biomedical science by analyzing cellular state and intercellular heterogeneity. Undoubtedly, single-cell transcriptomics has emerged as one of the most vibrant research fields today. With the optimization and innovation of single-cell sequencing technologies, the intricate multidimensional details concealed within cells are gradually unveiled. The combination of scRNA-seq and other multi-omics is at the forefront of the single-cell field. This involves simultaneously measuring various omics data within individual cells, expanding our understanding across a broader spectrum of dimensions. Single-cell multi-omics precisely captures the multidimensional aspects of single-cell transcriptomes, immune repertoire, spatial information, temporal information, epitopes, and other omics in diverse spatiotemporal contexts. In addition to depicting the cell atlas of normal or diseased tissues, it also provides a cornerstone for studying cell differentiation and development patterns, disease heterogeneity, drug resistance mechanisms, and treatment strategies. Herein, we review traditional single-cell sequencing technologies and outline the latest advancements in single-cell multi-omics. We summarize the current status and challenges of applying single-cell multi-omics technologies to biological research and clinical applications. Finally, we discuss the limitations and challenges of single-cell multi-omics and potential strategies to address them.
-
Exploiting Single-Cell Tools in Gene and Cell Therapy
Daniel Bode,Alyssa H. Cull,Juan A. Rubio-Lara,David G. Kent
DOI: https://doi.org/10.3389/fimmu.2021.702636
IF: 7.3
2021-07-12
Frontiers in Immunology
Abstract:Single-cell molecular tools have been developed at an incredible pace over the last five years as sequencing costs continue to drop and numerous molecular assays have been coupled to sequencing readouts. This rapid period of technological development has facilitated the delineation of individual molecular characteristics including the genome, transcriptome, epigenome, and proteome of individual cells, leading to an unprecedented resolution of the molecular networks governing complex biological systems. The immense power of single-cell molecular screens has been particularly highlighted through work in systems where cellular heterogeneity is a key feature, such as stem cell biology, immunology, and tumor cell biology. Single-cell-omics technologies have already contributed to the identification of novel disease biomarkers, cellular subsets, therapeutic targets and diagnostics, many of which would have been undetectable by bulk sequencing approaches. More recently, efforts to integrate single-cell multi-omics with single cell functional output and/or physical location have been challenging but have led to substantial advances. Perhaps most excitingly, there are emerging opportunities to reach beyond the description of static cellular states with recent advances in modulation of cells through CRISPR technology, in particular with the development of base editors which greatly raises the prospect of cell and gene therapies. In this review, we provide a brief overview of emerging single-cell technologies and discuss current developments in integrating single-cell molecular screens and performing single-cell multi-omics for clinical applications. We also discuss how single-cell molecular assays can be usefully combined with functional data to unpick the mechanism of cellular decision-making. Finally, we reflect upon the introduction of spatial transcriptomics and proteomics, its complementary role with single-cell RNA sequencing (scRNA-seq) and potential application in cellular and gene therapy.
immunology
-
Single-cell RNA-sequencing: The future of genome biology is now
Simone Picelli
DOI: https://doi.org/10.1080/15476286.2016.1201618
2016-07-21
RNA Biology
Abstract:Genome-wide single-cell analysis represents the ultimate frontier of genomics research. In particular, single-cell RNA-sequencing (scRNA-seq) studies have been boosted in the last few years by an explosion of new technologies enabling the study of the transcriptomic landscape of thousands of single cells in complex multicellular organisms. More sensitive and automated methods are being continuously developed and promise to deliver better data quality and higher throughput with less hands-on time. The outstanding amount of knowledge that is going to be gained from present and future studies will have a profound impact in many aspects of our society, from the introduction of truly tailored cancer treatments, to a better understanding of antibiotic resistance and host-pathogen interactions; from the discovery of the mechanisms regulating stem cell differentiation to the characterization of the early event of human embryogenesis.
biochemistry & molecular biology
-
The new technologies of high-throughput single-cell RNA sequencing
E. A. Vodiasova,E. S. Chelebieva,O. N. Kuleshova
DOI: https://doi.org/10.18699/vj19.520
2019-08-24
Vavilov Journal of Genetics and Breeding
Abstract:A wealth of genome and transcriptome data obtained using new generation sequencing (NGS) technologies for whole organisms could not answer many questions in oncology, immunology, physiology, neurobiology, zoology and other fields of science and medicine. Since the cell is the basis for the living of all unicellular and multicellular organisms, it is necessary to study the biological processes at its level. This understanding gave impetus to the development of a new direction – the creation of technologies that allow working with individual cells (single-cell technology). The rapid development of not only instruments, but also various advanced protocols for working with single cells is due to the relevance of these studies in many fields of science and medicine. Studying the features of various stages of ontogenesis, identifying patterns of cell differentiation and subsequent tissue development, conducting genomic and transcriptome analyses in various areas of medicine (especially in demand in immunology and oncology), identifying cell types and states, patterns of biochemical and physiological processes using single cell technologies, allows the comprehensive research to be conducted at a new level. The first RNA-sequencing technologies of individual cell transcriptomes (scRNA-seq) captured no more than one hundred cells at a time, which was insufficient due to the detection of high cell heterogeneity, existence of the minor cell types (which were not detected by morphology) and complex regulatory pathways. The unique techniques for isolating, capturing and sequencing transcripts of tens of thousands of cells at a time are evolving now. However, new technologies have certain differences both at the sample preparation stage and during the bioinformatics analysis. In the paper we consider the most effective methods of multiple parallel scRNA-seq using the example of 10XGenomics, as well as the specifics of such an experiment, further bioinformatics analysis of the data, future outlook and applications of new high-performance technologies.
-
The Advancement and Application of the Single-Cell Transcriptome in Biological and Medical Research
Kongwei Huang,Yixue Xu,Tong Feng,Hong Lan,Fei Ling,Hai Xiang,Qingyou Liu
DOI: https://doi.org/10.3390/biology13060451
2024-06-20
Biology
Abstract:Single-cell RNA sequencing technology (scRNA-seq) has been steadily developing since its inception in 2009. Unlike bulk RNA-seq, scRNA-seq identifies the heterogeneity of tissue cells and reveals gene expression changes in individual cells at the microscopic level. Here, we review the development of scRNA-seq, which has gone through iterations of reverse transcription, in vitro transcription, smart-seq, drop-seq, 10 × Genomics, and spatial single-cell transcriptome technologies. The technology of 10 × Genomics has been widely applied in medicine and biology, producing rich research results. Furthermore, this review presents a summary of the analytical process for single-cell transcriptome data and its integration with other omics analyses, including genomes, epigenomes, proteomes, and metabolomics. The single-cell transcriptome has a wide range of applications in biology and medicine. This review analyzes the applications of scRNA-seq in cancer, stem cell research, developmental biology, microbiology, and other fields. In essence, scRNA-seq provides a means of elucidating gene expression patterns in single cells, thereby offering a valuable tool for scientific research. Nevertheless, the current single-cell transcriptome technology is still imperfect, and this review identifies its shortcomings and anticipates future developments. The objective of this review is to facilitate a deeper comprehension of scRNA-seq technology and its applications in biological and medical research, as well as to identify avenues for its future development in alignment with practical needs.
biology
-
Single-Cell Sequencing Methodologies: From Transcriptome To Multi-Dimensional Measurement
Yingwen Chen,Jia Song,Qingyu Ruan,Xi Zeng,Lingling Wu,Linfeng Cai,Xuanqun Wang,Chaoyong Yang
DOI: https://doi.org/10.1002/smtd.202100111
IF: 12.4
2021-01-01
Small Methods
Abstract:Cells are the basic building blocks of biological systems, with inherent unique molecular features and development trajectories. The study of single cells facilitates in-depth understanding of cellular diversity, disease processes, and organization of multicellular organisms. Single-cell RNA sequencing (scRNA-seq) technologies have become essential tools for the interrogation of gene expression patterns and the dynamics of single cells, allowing cellular heterogeneity to be dissected at unprecedented resolution. Nevertheless, measuring at only transcriptome level or 1D is incomplete; the cellular heterogeneity reflects in multiple dimensions, including the genome, epigenome, transcriptome, spatial, and even temporal dimensions. Hence, integrative single cell analysis is highly desired. In addition, the way to interpret sequencing data by virtue of bioinformatic tools also exerts critical roles in revealing differential gene expression. Here, a comprehensive review that summarizes the cutting-edge single-cell transcriptome sequencing methodologies, including scRNA-seq, spatial and temporal transcriptome profiling, multi-omics sequencing and computational methods developed for scRNA-seq data analysis is provided. Finally, the challenges and perspectives of this field are discussed.
-
Editorial: Bioinformatics Analysis of Single Cell Sequencing Data and Applications in Precision Medicine
Jialiang Yang,Bo Liao,Tuo Zhang,Yifei Xu
DOI: https://doi.org/10.3389/fgene.2019.01358
IF: 3.7
2020-01-01
Frontiers in Genetics
Abstract:Next-generation sequencing (NGS) technology has been successfully applied in disease diagnostics, oncological immunotherapy, and drug repurposing, especially for precision medicine where optimized medication is tailored to individual patients. Recently, the development of single cell techniques makes it possible to examine gene expression and mutation at individual cell resolution, which provides an unprecedented opportunity to study cell development and differentiation, and reveal cell-to-cell heterogeneity during disease development, treatment, and drug response for individual patients. With the exponential increase of single cell sequencing data, it is critical to develop appropriate bioinformatics and machine learning tools to mine the rules behind them. However, due to the technical barriers in single cell sequencing and the noisy nature of raw sequencing data, this task is challenging especially in the context of disease diagnosis and drug development. To promote the translation and efficient usage of single cell sequencing data to precision medicine, it is necessary to develop new analysis tools for analyzing and integrating multi-level single cell data including DNA, RNA, protein, and so on, comparing existing methods and results derived from different studies, and enhancing disease diagnostics and drug development. For example, the quality control, normalization, differential gene calling, and clustering methods are quite different between single cell sequencing and traditional bulk cell sequencing. Thus, it is critical to develop a best practice specifically for dealing with single cell sequencing data. For disease treatment, it is also important to identify disease driver genes common to all cell types as well as those specific to a particular cell type or subgroup as revealed by single cell techniques, based on existing or novel network and machine learning-based methods. Finally, more translational work should be done to bridge the bioinformatics analyses and clinical applications for single cell researchers. To provide a platform bridging single cell analysis and translational studies, we organized this special issue, in which 11 manuscripts have been accepted for publication. Firstly, Zen and Dai presented a comprehensive review on scRNA-seq associated biological experiments as well as computational methods for evaluating disease heterogeneity. They described the early impact of such technologies as well as a variety of common methods applicable to upstream and downstream processes. Upstream processes include several computational methods related to the detection and removal of technical noise given commonly assumed statistical distributions. In addition, the
-
Advancing precision medicine through single‐cell sequencing: Insights and implications
Xu Zhang,Rongrong Gao,Liuke Yang,Youwei Zhu,Tiancheng Zhang,Xiaorong Shen,Wenwen Gu,Long Yang,Shenjie Peng
DOI: https://doi.org/10.1002/ctd2.293
2024-04-19
Clinical and Translational Discovery
Abstract:Background Single‐cell sequencing (SCS) marks the advent of a transformative period in biomedical studies, enabling unprecedented insight into the cellular intricacies of health and disease. Methods By dissecting the genetic, epigenetic and proteomic landscapes at the single‐cell level, SCS transcends traditional bulk sequencing methodologies, illuminating the heterogeneity and dynamics of individual cells. Results This analytical leap facilitates a deeper understanding of disease mechanisms, offers novel diagnostic and therapeutic targets and underpins the development of precision medicine across diverse fields such as neurology, oncology and immunology. Conclusions Despite its profound potential, SCS encounters challenges, including complex sample preparation, sophisticated data analysis and cost considerations. Nevertheless, ongoing advancements promise to overcome these barriers, integrating SCS with other omics data and leveraging machine learning to enhance biological understanding and clinical application. With the advancement of SCS technologies, personalised healthcare might be fundamentally altered, facilitating tailored and efficacious treatment strategies.
-
Single-cell Transcriptome in the Identification of Disease Biomarkers: Opportunities and Challenges
Zhitu Zhu,Diane C. Wang,Laurentiu M. Popescu,Xiangdong Wang
DOI: https://doi.org/10.1186/s12967-014-0212-3
IF: 8.44
2014-01-01
Journal of Translational Medicine
Abstract:Single cell transcriptome defined as the entire RNA or polyadenylated products of RNA polymerase II on a cell can describe the gene regulation networks responsible for physiological functions, behaviours, and phenotypes in response to signals and microenvironmental changes. Single cell transcriptome/sequencing has the special power to investigate small groups of differentiating cells, circulating tumour cells, or tissue stem cells. A large number of factors may influence the extent of single-cell heterogeneity within a system. It is the opportunity that the single-cell sequencing can be used for the identification of genetic changes in rare cells, e.g. cancer and tissue stem cells, in clinical samples. The methodologies of single-cell sequencing have been improved and developed with the increase of the understanding and attention. The clinical research and application of the single cell sequencing analysis are expected to identify and validate disease-specific biomarkers, network biomarkers, dynamic network biomarkers. The single cell research and value will be also dependent upon the understanding of genomic heterogeneity, planning and design of study protocol, representative of selected and targeted cells, and sensitivity and repeatability of the methodology. The single cell sequencing can be used to develop new diagnostics, monitor disease progresses, measure responses to therapies, and predict the prognosis of patients, although there are still a large number of challenges and difficulties to be faced. It would be more values and specificities of the single cell sequencing to integrate with the function of cells, organs, and systems of the body, the clinical phenotypes of patients, and the description of clinical bioinformatics.
-
Single‐cell profiling for advancing birth defects research and prevention
Thomas B. Knudsen,Malte Spielmann,Sean G. Megason,Elaine M. Faustman
DOI: https://doi.org/10.1002/bdr2.1870
2021-01-25
Birth Defects Research
Abstract:<p>Cellular analysis of developmental processes and toxicities has traditionally entailed bulk methods (e.g., transcriptomics) that lack single cell resolution or tissue localization methods (e.g., immunostaining) that allow only a few genes to be monitored in each experiment. Recent technological advances have enabled interrogation of genomic function at the single‐cell level, providing new opportunities to unravel developmental pathways and processes with unprecedented resolution. Here, we review emerging technologies of single‐cell RNA‐sequencing (scRNA‐seq) to globally characterize the gene expression sets of different cell types and how different cell types emerge from earlier cell states in development. Cell atlases of experimental embryology and human embryogenesis at single‐cell resolution will provide an encyclopedia of genes that define key stages from gastrulation to organogenesis. This technology, combined with computational models to discover key organizational principles, was recognized by Science magazine as the "Breakthrough of the year" for 2018 due to transformative potential on the way we study how human cells mature over a lifetime, how tissues regenerate, and how cells change in diseases (e.g., patient‐derived organoids to screen disease‐specific targets and design precision therapy). Profiling transcriptomes at the single‐cell level can fulfill the need for greater detail in the molecular progression of all cell lineages, from pluripotency to adulthood and how cell–cell signaling pathways control progression at every step. Translational opportunities emerge for elucidating pathogenesis of genetic birth defects with cellular precision and improvements for predictive toxicology of chemical teratogenesis.</p>
toxicology,developmental biology
-
Deciphering cell-cell interactions and communication in the tumor microenvironment and unraveling intratumoral genetic heterogeneity via single-cell genomic sequencing
Ya-Hong Cao,Jie Ding,Qing-Hai Tang,Jie Zhang,Zhong-Yan Huang,Xiao-Mei Tang,Ji-Bin Liu,Yu-Shui Ma,Da Fu
DOI: https://doi.org/10.1080/21655979.2023.2185434
Bioengineered
Abstract:A tumor's heterogeneity has important implications in terms of its clonal origin, progression, stemness, and drug resistance. Therefore, because of its significance in treatment, it is important to understand the gene expression pattern of a single cell, track gene expression or mutation in heterogeneous cells, evaluate the clonal origin of cancer cells, and determine the selective evolution of different subpopulations of cancer cells. Researchers are able to trace a cell's mutation and identify different types of tumor cells by measuring the whole transcriptome with single-cell sequencing (scRNA-seq). This technology provides a better understanding of the molecular mechanisms driving tumor growth than that offered by traditional RNA sequencing methods. In addition, it has revealed changes in the mutations and functions of somatic cells as a tumor evolves; it has also clarified immune cell infiltration and activation. Research on scRNA-seq technology has recently advanced significantly, suggesting new strategies for the treatment of cancer. In short, cancer researchers have become increasingly dependent on scRNA-seq. This paper reviews the development, detection principles, and processes of scRNA-seq technology and their application in tumor research. It also considers potential clinical applications.
-
Time-resolved Single-cell Transcriptomic Sequencing
Xing Xu,Qianxi Wen,Tianchen Lan,Liuqing Zeng,Yonghao Zeng,Shiyan Lin,Minghao Qiu,Xing Na,Chaoyong Yang
DOI: https://doi.org/10.1039/d4sc05700g
IF: 8.4
2024-11-02
Chemical Science
Abstract:Cells experience continuous transformation under both physiological and pathological circumstances. Single-cell RNA sequencing (scRNA-seq) is competent in disclosing the disparities of cells; nevertheless, it poses challenges in linking the individual cell state at distinct time points. Although computational approaches based on scRNA-seq data have been put forward for trajectory analysis, the result is based on assumptions and fails to reflect the actual states. Consequently, it is necessary to incorporate "time anchor" into the scRNA-seq library for the temporal documentation of the dynamic expression pattern. This review comprehensively overviews the time-resolved single-cell transcriptomic sequencing methodologies and applications. As scRNA-seq functions as the basis for profiling single-cell expression patterns, the review initially introduces various scRNA-seq approaches. Subsequently, the review focuses on the different experimental strategies for introducing a "time anchor" to scRNA-seq, highlighting their principles, strengths, weaknesses, and comparing their adaptation in various scenarios. Next, it provides a brief summary of applications in immunity response, cancer progression, and embryo development. Finally, the review concludes with a forward-looking perspective on future advancements in time-resolved single-cell transcriptomic sequencing.
chemistry, multidisciplinary
-
Application of Multi-Omics in Single Cells
Xiaolong Kang,Andrew Liu,George E Liu
DOI: https://doi.org/10.33582/2637-4927/1007
2018-01-01
Annals of Biotechnology
Abstract:In recent years, single cell assays have made exciting progresses, overcoming the issue of heterogeneity associated with bulk populations. The fast-developing sequencing methods now enable unbiased, high-throughput and highresolution view of the heterogeneity from individual cell within a population, in terms of its fate decisions, identity and function. The cell’s state is regulated at different levels, such as DNA, RNA and protein, by complex interplay of intrinsic molecules existing in the organism and extrinsic stimuli such as local environment. Comprehensive profiling of single cell requires a simultaneously dissection from different levels (multi-omics) to avoid incomplete information generated from single cell. In this short review, we first examine the whole genome amplification methods, and then survey the features of the single cell approaches for genome, epigenome, transcriptome, proteome and metabolome profiling. Finally, we briefly analyze advantages of multi-omics measurement from single cells as compared to separate measurement of each molecular type, and discuss opportunities and challenges of combining single cell multiomics information on resolving phenotype variants.
-
Exploring Additional Valuable Information from Single-Cell RNA-Seq Data.
Yunjin Li,Qiyue Xu,Duojiao Wu,Geng Chen
DOI: https://doi.org/10.3389/fcell.2020.593007
IF: 5.5
2020-01-01
Frontiers in Cell and Developmental Biology
Abstract:Single-cell RNA-seq (scRNA-seq) technologies are broadly applied to dissect the cellular heterogeneity and expression dynamics, providing unprecedented insights into single-cell biology. Most of the scRNA-seq studies mainly focused on the dissection of cell types/states, developmental trajectory, gene regulatory network, and alternative splicing. However, besides these routine analyses, many other valuable scRNA-seq investigations can be conducted. Here, we first review cell-to-cell communication exploration, RNA velocity inference, identification of large-scale copy number variations and single nucleotide changes, and chromatin accessibility prediction based on single-cell transcriptomics data. Next, we discuss the identification of novel genes/transcripts through transcriptome reconstruction approaches, as well as the profiling of long non-coding RNAs and circular RNAs. Additionally, we survey the integration of single-cell and bulk RNA-seq datasets for deconvoluting the cell composition of large-scale bulk samples and linking single-cell signatures to patient outcomes. These additional analyses could largely facilitate corresponding basic science and clinical applications.
-
Forward single‐cell sequencing into clinical application: Understanding of cancer microenvironment at single‐cell solution
Xuanqi Liu,Charles A Powell,Xiangdong Wang
DOI: https://doi.org/10.1002/ctm2.782
IF: 8.554
2022-04-01
Clinical and Translational Medicine
Abstract:Single‐cell RNA sequencing (scRNA‐seq) is considered an important approach to understand the molecular mechanisms of cancer microenvironmental functions and has the potential for clinical and translational discovery and development. The recent concerns on the impact of scRNA‐seq for clinical practice are whether scRNA can be applied as a routine measurement of clinical biochemistry to assist in clinical decision‐making for diagnosis and therapy. Pushing single‐cell sequencing into clinical application is one of the important missions for clinical and translational medicine (CTM), although there still are a large number of challenges to be overcome. The present Editorial as one of serials aims at overviewing the history of scRNA‐seq publications in CTM, sharing the understanding and consideration of the cancer microenvironment at the single‐cell solution and emphasising the objective of translating scRNA‐seq into clinical application. The dynamic characteristics and patterns of single‐cell identity, regulatory networks, and intercellular communication play decisive roles in the properties of the microenvironment, malignancy and migrative capacity of cancer cells, and defensive capacity of immune cells. The microenvironmental single‐cell transcriptomic profiles and cell clusters defined by scRNA‐seq have great value for exploring the molecular mechanisms of diseases and predicting cell sensitivities to therapy and patient prognosis.
oncology,medicine, research & experimental
-
Single‐cell RNA sequencing technologies and applications: A brief overview
Dragomirka Jovic,Xue Liang,Hua Zeng,Lin Lin,Fengping Xu,Yonglun Luo
DOI: https://doi.org/10.1002/ctm2.694
IF: 8.554
2022-03-01
Clinical and Translational Medicine
Abstract:Single-cell RNA sequencing (scRNA-seq) technology has become the state-of-the-art approach for unravelling the heterogeneity and complexity of RNA transcripts within individual cells, as well as revealing the composition of different cell types and functions within highly organized tissues/organs/organisms. Since its first discovery in 2009, studies based on scRNA-seq provide massive information across different fields making exciting new discoveries in better understanding the composition and interaction of cells within humans, model animals and plants. In this review, we provide a concise overview about the scRNA-seq technology, experimental and computational procedures for transforming the biological and molecular processes into computational and statistical data. We also provide an explanation of the key technological steps in implementing the technology. We highlight a few examples on how scRNA-seq can provide unique information for better understanding health and diseases. One important application of the scRNA-seq technology is to build a better and high-resolution catalogue of cells in all living organism, commonly known as atlas, which is key resource to better understand and provide a solution in treating diseases. While great promises have been demonstrated with the technology in all areas, we further highlight a few remaining challenges to be overcome and its great potentials in transforming current protocols in disease diagnosis and treatment.
oncology,medicine, research & experimental
-
Single-Cell RNA Sequencing: Technological Progress and Biomedical Application in Cancer Research
Xu Chang,Yunxi Zheng,Kai Xu
DOI: https://doi.org/10.1007/s12033-023-00777-0
2023-06-16
Molecular Biotechnology
Abstract:Single-cell RNA-seq (scRNA-seq) is a revolutionary technology that allows for the genomic investigation of individual cells in a population, allowing for the discovery of unusual cells associated with cancer and metastasis. ScRNA-seq has been used to discover different types of cancers with poor prognosis and medication resistance such as lung cancer, breast cancer, ovarian cancer, and gastric cancer. Besides, scRNA-seq is a promising method that helps us comprehend the biological features and dynamics of cell development, as well as other disorders. This review gives a concise summary of current scRNA-seq technology. We also explain the main technological steps involved in implementing the technology. We highlight the present applications of scRNA-seq in cancer research, including tumor heterogeneity analysis in lung cancer, breast cancer, and ovarian cancer. In addition, this review elucidates potential applications of scRNA-seq in lineage tracing, personalized medicine, illness prediction, and disease diagnosis, which reveals that scRNA-seq facilitates these events by producing genetic variations on the single-cell level.
biochemistry & molecular biology,biotechnology & applied microbiology
-
Single-cell Multiomics: a New Frontier in Drug Research and Development
Jiaxiu Ma,Chao Dong,Aibin He,Haiqing Xiong
DOI: https://doi.org/10.3389/fddsv.2024.1474331
2024-01-01
Frontiers in Drug Discovery
Abstract:Single-cell multiomics (sc-multiomics) is a burgeoning field that simultaneously integrates multiple layers of molecular information, enabling the characterization of dynamic cell states and activities in development and disease as well as treatment response. Studying drug actions and responses using sc-multiomics technologies has revolutionized our understanding of how small molecules intervene for specific cell types in cancer treatment and how they are linked with disease etiology and progression. Here, we summarize recent advances in sc-multiomics technologies that have been adapted and improved in drug research and development, with a focus on genome-wide examination of drug-chromatin engagement and the applications in drug response and the mechanisms of drug resistance. Furthermore, we discuss how state-of-the-art technologies can be taken forward to devise innovative personalized treatment modalities in biomedical research.