1
|
Alanzi AR. Exploring Microbial Dark Matter for the Discovery of Novel Natural Products: Characteristics, Abundance Challenges and Methods. J Microbiol Biotechnol 2024; 35:e2407064. [PMID: 39639495 PMCID: PMC11813339 DOI: 10.4014/jmb.2407.07064] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2024] [Revised: 10/22/2024] [Accepted: 10/30/2024] [Indexed: 12/07/2024]
Abstract
The objective of this review is to investigate microbial dark matter (MDM) with a focus on its potential for discovering novel natural products (NPs). This first part will examine the characteristics and abundance of these previously unexplored microbial communities, as well as the challenges faced in identifying and harnessing their unique biochemical properties and novel methods in this field. MDMs are thought to hold great potential for the discovery of novel NPs, which could have significant applications in medicine, agriculture, and industry. In recent years, there has been a growing interest in exploring MDM to unlock its potential. In fact, developments in genome-sequencing technologies and sophisticated phylogenetic procedures and metagenomic techniques have contributed to drastically make important changes in our sights on the diversity of microbial life, including the very outline of the tree of life. This has led to the development of novel technologies and methodologies for studying these elusive microorganisms, such as single-cell genomics, metagenomics, and culturomics. These approaches enable researchers to isolate and analyze individual microbial cells, as well as entire communities, providing insights into their genetic and metabolic potential. By delving into the MDM, scientists hope to uncover new compounds and biotechnological advancements that could have far-reaching impacts on various fields.
Collapse
Affiliation(s)
- Abdullah R Alanzi
- Department of Pharmacognosy, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia
| |
Collapse
|
2
|
Chen J, Larsson L, Swarbrick A, Lundeberg J. Spatial landscapes of cancers: insights and opportunities. Nat Rev Clin Oncol 2024; 21:660-674. [PMID: 39043872 DOI: 10.1038/s41571-024-00926-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/28/2024] [Indexed: 07/25/2024]
Abstract
Solid tumours comprise many different cell types organized in spatially structured arrangements, with substantial intratumour and intertumour heterogeneity. Advances in spatial profiling technologies over the past decade hold promise to capture the complexity of these cellular architectures to build a holistic view of the intricate molecular mechanisms that shape the tumour ecosystem. Some of these mechanisms act at the cellular scale and are controlled by cell-autonomous programmes or communication between nearby cells, whereas other mechanisms result from coordinated efforts between large networks of cells and extracellular molecules organized into tissues and organs. In this Review we provide insights into the application of single-cell and spatial profiling tools, with a focus on spatially resolved transcriptomic tools developed to understand the cellular architecture of the tumour microenvironment and identify opportunities to use them to improve clinical management of cancers.
Collapse
Affiliation(s)
- Julia Chen
- Cancer Ecosystems Program, Garvan Institute of Medical Research, Sydney, New South Wales, Australia
- School of Clinical Medicine, Faculty of Medicine and Health, University of New South Wales, Sydney, New South Wales, Australia
- Department of Medical Oncology, St George Hospital, Sydney, New South Wales, Australia
| | - Ludvig Larsson
- Department of Gene Technology, KTH Royal Institute of Technology, Science for Life Laboratory, Stockholm, Sweden
| | - Alexander Swarbrick
- Cancer Ecosystems Program, Garvan Institute of Medical Research, Sydney, New South Wales, Australia.
- School of Clinical Medicine, Faculty of Medicine and Health, University of New South Wales, Sydney, New South Wales, Australia.
| | - Joakim Lundeberg
- Department of Gene Technology, KTH Royal Institute of Technology, Science for Life Laboratory, Stockholm, Sweden.
| |
Collapse
|
3
|
Chen X, Yu Y, Zheng H, Yang M, Wang C, Cai Q, Zhang W, Jiang F, Zhu Y, Yang H, Zhang T, Zhou Z. Single-cell transcriptome analysis reveals dynamic changes of the preclinical A549 cancer models, and the mechanism of dacomitinib. Eur J Pharmacol 2023; 960:176046. [PMID: 37708985 DOI: 10.1016/j.ejphar.2023.176046] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2023] [Revised: 09/06/2023] [Accepted: 09/11/2023] [Indexed: 09/16/2023]
Abstract
The in vitro A549 cells, and A549 xenografts in nude mouse, were two commonly used models for anti-cancer drug discovery. However, the biological and molecular characteristics of these two classic models, and also the dynamic transcriptome changes after dacomitinib exposure remains elusive. We performed single-cell RNA sequencing to define the transcriptome profile at single-cell resolution, and processed tumor samples for bulk RNA and protein analysis to validate the differently expressed genes. Transcriptome profiling revealed that the in vitro A549 cells are heterogeneous. The minimal subpopulation of the in vitro A549 cells, which were characterized by the signature of response to unfolded protein, became the overriding subpopulation of the xenografts. The EGFR non-activating A549 cells were resistant to dacomitinib in vitro, while A549 xenografts were comparatively sensitive as EGFR-activating HCC827 xenografts. Dacomitinib inhibited MAPK signaling pathway, and increased the immune response in the A549 xenografts. A phagocytosis checkpoint stanniocalcin-1 (STC1) was significantly inhibited in dacomitinib-treated xenografts. So here our study gives the first insight of the heterogeneity of the two classic models, and the translational potential of dacomitinib being used into a broader patient population rather than EGFR common activating mutation.
Collapse
Affiliation(s)
- Xiaoyan Chen
- Jiading District Central Hospital Affiliated Shanghai University of Medicine and Health Sciences, Shanghai University of Medicine and Health Sciences, Shanghai, 201318, China; The College of Medical Technology, Shanghai University of Medicine and Health Sciences, Shanghai, 201318, China; Collaborative Research Center, Shanghai University of Medicine and Health Sciences, Shanghai, 201318, China
| | - Yangziwei Yu
- Jiading District Central Hospital Affiliated Shanghai University of Medicine and Health Sciences, Shanghai University of Medicine and Health Sciences, Shanghai, 201318, China; School of Health Science and Engineering, University of Shanghai for Science and Technology, Shanghai, 200093, China; Collaborative Research Center, Shanghai University of Medicine and Health Sciences, Shanghai, 201318, China
| | - Haoyang Zheng
- Jiading District Central Hospital Affiliated Shanghai University of Medicine and Health Sciences, Shanghai University of Medicine and Health Sciences, Shanghai, 201318, China; School of Pharmacy, Shanghai University of Medicine and Health Sciences, Shanghai, 201318, China; Collaborative Research Center, Shanghai University of Medicine and Health Sciences, Shanghai, 201318, China
| | - Mengjing Yang
- Jiading District Central Hospital Affiliated Shanghai University of Medicine and Health Sciences, Shanghai University of Medicine and Health Sciences, Shanghai, 201318, China; Shanghai Key Laboratory of Molecular Imaging, Shanghai University of Medicine and Health Sciences, Shanghai, 201318, China; Collaborative Research Center, Shanghai University of Medicine and Health Sciences, Shanghai, 201318, China
| | - Chuqiao Wang
- Department of Pharmaceutical Toxicology, School of Pharmacy, China Medical University, Shenyang, 110122, China
| | - Qianqian Cai
- Jiading District Central Hospital Affiliated Shanghai University of Medicine and Health Sciences, Shanghai University of Medicine and Health Sciences, Shanghai, 201318, China; Shanghai Key Laboratory of Molecular Imaging, Shanghai University of Medicine and Health Sciences, Shanghai, 201318, China; Collaborative Research Center, Shanghai University of Medicine and Health Sciences, Shanghai, 201318, China
| | - Weiguo Zhang
- Jiading District Central Hospital Affiliated Shanghai University of Medicine and Health Sciences, Shanghai University of Medicine and Health Sciences, Shanghai, 201318, China; Shanghai Key Laboratory of Molecular Imaging, Shanghai University of Medicine and Health Sciences, Shanghai, 201318, China; Collaborative Research Center, Shanghai University of Medicine and Health Sciences, Shanghai, 201318, China
| | - Feixiang Jiang
- Jiading District Central Hospital Affiliated Shanghai University of Medicine and Health Sciences, Shanghai University of Medicine and Health Sciences, Shanghai, 201318, China; School of Pharmacy, Shanghai University of Medicine and Health Sciences, Shanghai, 201318, China; Collaborative Research Center, Shanghai University of Medicine and Health Sciences, Shanghai, 201318, China
| | - Yanmei Zhu
- Department of Pathology, Affiliated Cancer Hospital of Dalian University of Technology, Shenyang, 110042, China; Liaoning Cancer Hospital and Institute, Shenyang, 110042, China; Cancer Hospital of China Medical University, Shenyang, 110042, China
| | - Hedi Yang
- Collaborative Research Center, Shanghai University of Medicine and Health Sciences, Shanghai, 201318, China
| | - Tianbiao Zhang
- Department of Biochemistry and Molecular Biology, China Medical University, Shenyang, 110122, China
| | - Zhaoli Zhou
- Jiading District Central Hospital Affiliated Shanghai University of Medicine and Health Sciences, Shanghai University of Medicine and Health Sciences, Shanghai, 201318, China; School of Pharmacy, Shanghai University of Medicine and Health Sciences, Shanghai, 201318, China; Collaborative Research Center, Shanghai University of Medicine and Health Sciences, Shanghai, 201318, China.
| |
Collapse
|
4
|
Xu C, Wang K, Huang P, Liu D, Guan Y. Single-Cell Isolation Microfluidic Chip Based on Thermal Bubble Micropump Technology. SENSORS (BASEL, SWITZERLAND) 2023; 23:s23073623. [PMID: 37050683 PMCID: PMC10099219 DOI: 10.3390/s23073623] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/09/2023] [Revised: 03/25/2023] [Accepted: 03/29/2023] [Indexed: 05/31/2023]
Abstract
The isolation of single cells is essential for the development of single cell analysis methods, such as single-cell sequencing, monoclonal antibodies, and drug development. Traditional single-cell isolation techniques include flow cytometry (FACS), laser capture microdissection (LCM), micromanipulation, etc., but their operations are complex and have low throughput. Here, we present a microfluidic chip that can isolate individual cells from cell suspension and release them onto a well plate. It uses thermal bubble micropump technology to drive the fluid flow, and single-cell isolation is achieved by matching the flow resistance of the flow channel. Therefore, injection pumps and peristaltic pumps are not required for cell loading. Because of its small size, we can integrate hundreds of single-cell functional modules, which makes high-throughput single-cell isolation possible. For polystyrene beads, the capture rate of the single bead is close to 100%. Finally, the method has been applied to cells, and the capture rate of the single cell is also about 75%. This is a promising method for single-cell isolation.
Collapse
Affiliation(s)
- Chao Xu
- School of Microelectronics, Shanghai University, Shanghai 201800, China
| | - Kun Wang
- Shanghai Aure Technology Limited Company, Shanghai 201800, China
| | - Peng Huang
- Shanghai Aure Technology Limited Company, Shanghai 201800, China
| | - Demeng Liu
- School of Microelectronics, Shanghai University, Shanghai 201800, China
- Shanghai Aure Technology Limited Company, Shanghai 201800, China
| | - Yimin Guan
- School of Microelectronics, Shanghai University, Shanghai 201800, China
- Shanghai Aure Technology Limited Company, Shanghai 201800, China
| |
Collapse
|
5
|
Xu T, Li Y, Han X, Kan L, Ren J, Sun L, Diao Z, Ji Y, Zhu P, Xu J, Ma B. Versatile, facile and low-cost single-cell isolation, culture and sequencing by optical tweezer-assisted pool-screening. LAB ON A CHIP 2022; 23:125-135. [PMID: 36477690 DOI: 10.1039/d2lc00888b] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/17/2023]
Abstract
Real-time image-based sorting of target cells in a precisely indexed manner is desirable for sequencing or cultivating individual human or microbial cells directly from clinical or environmental samples; however, the versatility of existing methods is limited as they are usually not broadly applicable to all cell sizes. Here, an optical tweezer-assisted pool-screening and single-cell isolation (OPSI) system is established for precise, indexed isolation of individual bacterial, yeast or human-cancer cells. A controllable static flow field that acts as a cell pool is achieved in a microfluidics chip, to enable precise and ready screening of cells of 1 to 40 μm in size by bright-field, fluorescence, or Raman imaging. The target cell is then captured by a 1064 nm optical tweezer and deposited as one-cell-harboring nanoliter microdroplets in a one-cell-one-tube manner. For bacterial, yeast and human cells, OPSI achieves a >99.7% target-cell sorting purity and a 10-fold elevated speed of 10-20 cells per min. Moreover, OPSI-based one-cell RNA-seq of human cancer cells yields high quality and reproducible single-cell transcriptome profiles. The versatility, facileness, flexibility, modularized design, and low cost of OPSI suggest its broad applications for image-based sorting of target cells.
Collapse
Affiliation(s)
- Teng Xu
- Single-Cell Center, CAS Key Laboratory of Biofuels, Shandong Key Laboratory of Energy Genetics, Qingdao Institute of Bioenergy and Bioprocess Technology, Chinese Academy of Sciences, Qingdao, China.
- Shandong Energy Institute, Qingdao, China
- Qingdao New Energy Shandong Laboratory, Qingdao, China
| | - Yuandong Li
- Single-Cell Center, CAS Key Laboratory of Biofuels, Shandong Key Laboratory of Energy Genetics, Qingdao Institute of Bioenergy and Bioprocess Technology, Chinese Academy of Sciences, Qingdao, China.
- Shandong Energy Institute, Qingdao, China
- Qingdao New Energy Shandong Laboratory, Qingdao, China
| | - Xiao Han
- Single-Cell Center, CAS Key Laboratory of Biofuels, Shandong Key Laboratory of Energy Genetics, Qingdao Institute of Bioenergy and Bioprocess Technology, Chinese Academy of Sciences, Qingdao, China.
- School of Chemistry and Chemical Engineering, University of Jinan, Jinan, China
| | - Lingyan Kan
- Single-Cell Center, CAS Key Laboratory of Biofuels, Shandong Key Laboratory of Energy Genetics, Qingdao Institute of Bioenergy and Bioprocess Technology, Chinese Academy of Sciences, Qingdao, China.
- Shandong Energy Institute, Qingdao, China
- Qingdao New Energy Shandong Laboratory, Qingdao, China
| | - Jing Ren
- Single-Cell Center, CAS Key Laboratory of Biofuels, Shandong Key Laboratory of Energy Genetics, Qingdao Institute of Bioenergy and Bioprocess Technology, Chinese Academy of Sciences, Qingdao, China.
- Shandong Energy Institute, Qingdao, China
- Qingdao New Energy Shandong Laboratory, Qingdao, China
| | - Luyang Sun
- Single-Cell Center, CAS Key Laboratory of Biofuels, Shandong Key Laboratory of Energy Genetics, Qingdao Institute of Bioenergy and Bioprocess Technology, Chinese Academy of Sciences, Qingdao, China.
- Shandong Energy Institute, Qingdao, China
- Qingdao New Energy Shandong Laboratory, Qingdao, China
| | - Zhidian Diao
- Single-Cell Center, CAS Key Laboratory of Biofuels, Shandong Key Laboratory of Energy Genetics, Qingdao Institute of Bioenergy and Bioprocess Technology, Chinese Academy of Sciences, Qingdao, China.
- Shandong Energy Institute, Qingdao, China
- Qingdao New Energy Shandong Laboratory, Qingdao, China
| | - Yuetong Ji
- Single-Cell Center, CAS Key Laboratory of Biofuels, Shandong Key Laboratory of Energy Genetics, Qingdao Institute of Bioenergy and Bioprocess Technology, Chinese Academy of Sciences, Qingdao, China.
- Single-Cell Biotechnology Ltd., Qingdao, Shandong, China
| | - Pengfei Zhu
- Single-Cell Center, CAS Key Laboratory of Biofuels, Shandong Key Laboratory of Energy Genetics, Qingdao Institute of Bioenergy and Bioprocess Technology, Chinese Academy of Sciences, Qingdao, China.
- Single-Cell Biotechnology Ltd., Qingdao, Shandong, China
| | - Jian Xu
- Single-Cell Center, CAS Key Laboratory of Biofuels, Shandong Key Laboratory of Energy Genetics, Qingdao Institute of Bioenergy and Bioprocess Technology, Chinese Academy of Sciences, Qingdao, China.
- Shandong Energy Institute, Qingdao, China
- Qingdao New Energy Shandong Laboratory, Qingdao, China
- Laboratory of Marine Biology and Biotechnology, Qingdao National Laboratory for Marine Science and Technology, Qingdao, China
| | - Bo Ma
- Single-Cell Center, CAS Key Laboratory of Biofuels, Shandong Key Laboratory of Energy Genetics, Qingdao Institute of Bioenergy and Bioprocess Technology, Chinese Academy of Sciences, Qingdao, China.
- Shandong Energy Institute, Qingdao, China
- Qingdao New Energy Shandong Laboratory, Qingdao, China
- Laboratory of Marine Biology and Biotechnology, Qingdao National Laboratory for Marine Science and Technology, Qingdao, China
| |
Collapse
|
6
|
Caputo V, Megalizzi D, Fabrizio C, Termine A, Colantoni L, Caltagirone C, Giardina E, Cascella R, Strafella C. Update on the Molecular Aspects and Methods Underlying the Complex Architecture of FSHD. Cells 2022; 11:cells11172687. [PMID: 36078093 PMCID: PMC9454908 DOI: 10.3390/cells11172687] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2022] [Revised: 08/24/2022] [Accepted: 08/26/2022] [Indexed: 11/16/2022] Open
Abstract
Despite the knowledge of the main mechanisms involved in facioscapulohumeral muscular dystrophy (FSHD), the high heterogeneity and variable penetrance of the disease complicate the diagnosis, characterization and genotype–phenotype correlation of patients and families, raising the need for further research and data. Thus, the present review provides an update of the main molecular aspects underlying the complex architecture of FSHD, including the genetic factors (related to D4Z4 repeated units and FSHD-associated genes), epigenetic elements (D4Z4 methylation status, non-coding RNAs and high-order chromatin interactions) and gene expression profiles (FSHD transcriptome signatures both at bulk tissue and single-cell level). In addition, the review will also describe the methods currently available for investigating the above-mentioned features and how the resulting data may be combined with artificial-intelligence-based pipelines, with the purpose of developing a multifunctional tool tailored to enhancing the knowledge of disease pathophysiology and progression and fostering the research for novel treatment strategies, as well as clinically useful biomarkers. In conclusion, the present review highlights how FSHD should be regarded as a disease characterized by a molecular spectrum of genetic and epigenetic factors, whose alteration plays a differential role in DUX4 repression and, subsequently, contributes to determining the FSHD phenotype.
Collapse
Affiliation(s)
- Valerio Caputo
- Genomic Medicine Laboratory-UILDM, Santa Lucia Foundation IRCCS, 00179 Rome, Italy
- Department of Biomedicine and Prevention, Tor Vergata University, 00133 Rome, Italy
| | - Domenica Megalizzi
- Genomic Medicine Laboratory-UILDM, Santa Lucia Foundation IRCCS, 00179 Rome, Italy
- Department of Biomedicine and Prevention, Tor Vergata University, 00133 Rome, Italy
| | - Carlo Fabrizio
- Data Science Unit, Santa Lucia Foundation IRCCS, 00179 Rome, Italy
| | - Andrea Termine
- Data Science Unit, Santa Lucia Foundation IRCCS, 00179 Rome, Italy
| | - Luca Colantoni
- Genomic Medicine Laboratory-UILDM, Santa Lucia Foundation IRCCS, 00179 Rome, Italy
| | - Carlo Caltagirone
- Department of Clinical and Behavorial Neurology, Santa Lucia Foundation IRCCS, 00179 Rome, Italy
| | - Emiliano Giardina
- Genomic Medicine Laboratory-UILDM, Santa Lucia Foundation IRCCS, 00179 Rome, Italy
- Department of Biomedicine and Prevention, Tor Vergata University, 00133 Rome, Italy
- Correspondence: ; Tel.: +39-0651501550
| | - Raffaella Cascella
- Genomic Medicine Laboratory-UILDM, Santa Lucia Foundation IRCCS, 00179 Rome, Italy
- Department of Biomedicine and Prevention, Tor Vergata University, 00133 Rome, Italy
| | - Claudia Strafella
- Genomic Medicine Laboratory-UILDM, Santa Lucia Foundation IRCCS, 00179 Rome, Italy
- Department of Biomedicine and Prevention, Tor Vergata University, 00133 Rome, Italy
| |
Collapse
|
7
|
Salwan R, Sharma V. Genomics of Prokaryotic Extremophiles to Unfold the Mystery of Survival in Extreme Environments. Microbiol Res 2022; 264:127156. [DOI: 10.1016/j.micres.2022.127156] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2022] [Revised: 07/30/2022] [Accepted: 07/31/2022] [Indexed: 11/26/2022]
|
8
|
Gonzalez A, Quintela F, Leon DA, Bringas-Vega ML, Valdes-Sosa PA. Estimating the number of available states for normal and tumor tissues in gene expression space. BIOPHYSICAL REPORTS 2022; 2:100053. [PMID: 36425772 PMCID: PMC9680729 DOI: 10.1016/j.bpr.2022.100053] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/21/2021] [Accepted: 03/25/2022] [Indexed: 05/15/2023]
Abstract
The topology of gene expression space for a set of 12 cancer types is studied by means of an entropy-like magnitude, which measures the volumes of the regions occupied by tumor and normal samples, i.e., the number of available states (genotypes) that can be classified as tumor-like or normal-like, respectively. Computations show that the number of available states is much greater for tumors than for normal tissues, suggesting the irreversibility of the progression to the tumor phase. The entropy is nearly constant for tumors, whereas it exhibits a higher variability in normal tissues, probably due to tissue differentiation. In addition, we show an interesting correlation between the fraction (tumor/normal) of available states and the overlap between the tumor and normal sample clouds, interpreted as a way of reducing the decay rate to the tumor phase in more ordered or structured tissues.
Collapse
Affiliation(s)
- Augusto Gonzalez
- The Clinical Hospital Chengdu Brain Sciences Institute, University of Electronic Sciences and Technology of China (UESTC), Chengdu, People Republic of China
- Institute of Cybernetics, Mathematics and Physics, Havana, Cuba
| | - Frank Quintela
- Institute of Cybernetics, Mathematics and Physics, Havana, Cuba
- University of Modena & Reggio Emilia, Modena, Italy
| | - Dario A. Leon
- Institute of Cybernetics, Mathematics and Physics, Havana, Cuba
- S3 Centre, Istituto Nanoscienze, CNR, Modena, Italy
| | - Maria Luisa Bringas-Vega
- The Clinical Hospital Chengdu Brain Sciences Institute, University of Electronic Sciences and Technology of China (UESTC), Chengdu, People Republic of China
- Cuban Neurosciences Center, Havana, Cuba
| | - Pedro A. Valdes-Sosa
- The Clinical Hospital Chengdu Brain Sciences Institute, University of Electronic Sciences and Technology of China (UESTC), Chengdu, People Republic of China
- Cuban Neurosciences Center, Havana, Cuba
| |
Collapse
|
9
|
Rai P, Sengupta D, Majumdar A. SelfE: Gene Selection via Self-Expression for Single-Cell Data. IEEE/ACM TRANSACTIONS ON COMPUTATIONAL BIOLOGY AND BIOINFORMATICS 2022; 19:624-632. [PMID: 32750851 DOI: 10.1109/tcbb.2020.2997326] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
Single-cell RNA sequencing has been proved to be advantageous in discerning molecular heterogeneity in seemingly similar cells in a tissue. Due to the paucity of starting RNA, a large fraction of transcripts fail to amplify during the polymerase chain reaction cycle. This gets compounded by trivial biological noise such as variability in the cell cycle specific genes. As a result expression matrix obtained from a single-cell study is highly sparse with a large number of missing values. This hinders downstream analysis of single-cell expression data. It has been observed that feature engineering significantly improves the analysis outcomes. Feature extraction methods such as principal component analysis and zero-inflated factor analysis have been shown to be useful for subsequent steps of data analysis including clustering. However, too little or no visible efforts have been observed for developing feature selection techniques, which offer transparency for the analyst's consumption. We propose SelfE, a novel l2,0 -minimization algorithm that determines an optimal subset of feature vectors that preserves sub-space structures as observed in the data. We compared SelfE with the commonly used feature selection methods for single-cell expression data analysis.
Collapse
|
10
|
Zhao ZH, Wang XY, Schatten H, Sun QY. Single cell RNA sequencing techniques and applications in research of ovary development and related diseases. Reprod Toxicol 2021; 107:97-103. [PMID: 34896591 DOI: 10.1016/j.reprotox.2021.12.003] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2021] [Revised: 11/21/2021] [Accepted: 12/08/2021] [Indexed: 11/17/2022]
Abstract
The ovary is a highly organized composite of germ cells and various types of somatic cells, whose communications dictate ovary development to generate functional oocytes. The differences between individual cells might have profound effects on ovary functions. Single cell RNA sequencing techniques are promising approaches to explore the cell type composition of organisms, the dynamics of transcriptomes, the regulatory network between genes and the signaling pathways between cell types at the single cell resolution. In this review, we provide an overview of the currently available single cell RNA sequencing techniques including Smart-seq2 and Drop-seq, as well as their applications in biological and clinical research to provide a better understanding on the molecular mechanisms underlying ovary development and associated diseases.
Collapse
Affiliation(s)
- Zheng-Hui Zhao
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China
| | - Xiao-Yu Wang
- School of Social Development and Public Policy, Beijing Normal University, Beijing, 100875, China
| | - Heide Schatten
- Department of Veterinary Pathobiology, University of Missouri, Columbia, MO, 65211, United States
| | - Qing-Yuan Sun
- Fertility Preservation Lab, Guangdong-Hong Kong Metabolism & Reproduction Joint Laboratory, Reproductive Medicine Center, Guangdong Second Provincial General Hospital, Guangzhou, 510317, China.
| |
Collapse
|
11
|
Zhou WM, Yan YY, Guo QR, Ji H, Wang H, Xu TT, Makabel B, Pilarsky C, He G, Yu XY, Zhang JY. Microfluidics applications for high-throughput single cell sequencing. J Nanobiotechnology 2021; 19:312. [PMID: 34635104 PMCID: PMC8507141 DOI: 10.1186/s12951-021-01045-6] [Citation(s) in RCA: 60] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2021] [Accepted: 09/16/2021] [Indexed: 12/22/2022] Open
Abstract
The inherent heterogeneity of individual cells in cell populations plays significant roles in disease development and progression, which is critical for disease diagnosis and treatment. Substantial evidences show that the majority of traditional gene profiling methods mask the difference of individual cells. Single cell sequencing can provide data to characterize the inherent heterogeneity of individual cells, and reveal complex and rare cell populations. Different microfluidic technologies have emerged for single cell researches and become the frontiers and hot topics over the past decade. In this review article, we introduce the processes of single cell sequencing, and review the principles of microfluidics for single cell analysis. Also, we discuss the common high-throughput single cell sequencing technologies along with their advantages and disadvantages. Lastly, microfluidics applications in single cell sequencing technology for the diagnosis of cancers and immune system diseases are briefly illustrated.
Collapse
Affiliation(s)
- Wen-Min Zhou
- Key Laboratory of Molecular Target & Clinical Pharmacology , The State & NMPA Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences & the Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, 511436, People's Republic of China
| | - Yan-Yan Yan
- School of Medicine, Shanxi Datong University, Datong, 037009, People's Republic of China
| | - Qiao-Ru Guo
- Key Laboratory of Molecular Target & Clinical Pharmacology , The State & NMPA Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences & the Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, 511436, People's Republic of China
| | - Hong Ji
- Key Laboratory of Molecular Target & Clinical Pharmacology , The State & NMPA Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences & the Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, 511436, People's Republic of China
| | - Hui Wang
- Guangzhou Institute of Pediatrics/Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, 510623, People's Republic of China
| | - Tian-Tian Xu
- Guangzhou Institute of Pediatrics/Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, 510623, People's Republic of China
| | - Bolat Makabel
- Xinjiang Institute of Materia Medica, Urumqi, 830004, People's Republic of China
| | - Christian Pilarsky
- Department of Surgery, Friedrich-Alexander University of Erlangen-Nuremberg (FAU), University Hospital of Erlangen, Erlangen, Germany
| | - Gen He
- Key Laboratory of Molecular Target & Clinical Pharmacology , The State & NMPA Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences & the Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, 511436, People's Republic of China.
| | - Xi-Yong Yu
- Key Laboratory of Molecular Target & Clinical Pharmacology , The State & NMPA Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences & the Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, 511436, People's Republic of China.
| | - Jian-Ye Zhang
- Key Laboratory of Molecular Target & Clinical Pharmacology , The State & NMPA Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences & the Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, 511436, People's Republic of China.
| |
Collapse
|
12
|
A modular single-cell pipette microfluidic chip coupling to ETAAS and ICP-MS for single cell analysis. CHINESE CHEM LETT 2021. [DOI: 10.1016/j.cclet.2021.08.024] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
13
|
Prokaryotic and eukaryotic diversity in hydrothermal continental systems. Arch Microbiol 2021; 203:3751-3766. [PMID: 34143270 DOI: 10.1007/s00203-021-02416-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2020] [Revised: 05/28/2021] [Accepted: 05/31/2021] [Indexed: 02/07/2023]
Abstract
The term extremophile was suggested more than 30 years ago and represents microorganisms that are capable of developing and living under extreme conditions, these conditions being particularly hostile to other types of microorganisms and to humankind. In terrestrial hydrothermal sites, like hot springs, "mud pools", solfataras, and geysers, the dominant extreme conditions are high temperature, low or high pH, and high levels of salinity. The diversity of microorganisms inhabiting these sites is determined by the conditions of the environment. Organisms belonging to the domains Archaea and Bacteria are more represented than the one belonging to Eukarya. Eukarya members tend to be less present because of their lower tolerance to higher temperatures, however, they perform important ecosystem processes when present. Both prokaryotes and eukaryotes have morphological and physical adaptations that allow them to colonize extreme environments. Microbial mats are complex associations of microorganisms that help the colonization of more extreme systems. In this review, a characterization of prokaryotic and eukaryotic organisms that populate terrestrial hydrothermal systems are made.
Collapse
|
14
|
Strategies for Natural Products Discovery from Uncultured Microorganisms. Molecules 2021; 26:molecules26102977. [PMID: 34067778 PMCID: PMC8156983 DOI: 10.3390/molecules26102977] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2021] [Revised: 05/13/2021] [Accepted: 05/13/2021] [Indexed: 12/12/2022] Open
Abstract
Microorganisms are highly regarded as a prominent source of natural products that have significant importance in many fields such as medicine, farming, environmental safety, and material production. Due to this, only tiny amounts of microorganisms can be cultivated under standard laboratory conditions, and the bulk of microorganisms in the ecosystems are still unidentified, which restricts our knowledge of uncultured microbial metabolism. However, they could hypothetically provide a large collection of innovative natural products. Culture-independent metagenomics study has the ability to address core questions in the potential of NP production by cloning and analysis of microbial DNA derived directly from environmental samples. Latest advancements in next generation sequencing and genetic engineering tools for genome assembly have broadened the scope of metagenomics to offer perspectives into the life of uncultured microorganisms. In this review, we cover the methods of metagenomic library construction, and heterologous expression for the exploration and development of the environmental metabolome and focus on the function-based metagenomics, sequencing-based metagenomics, and single-cell metagenomics of uncultured microorganisms.
Collapse
|
15
|
Gonzalez A, Nieves J, Leon DA, Bringas Vega ML, Sosa PV. Gene expression rearrangements denoting changes in the biological state. Sci Rep 2021; 11:8470. [PMID: 33875699 PMCID: PMC8055689 DOI: 10.1038/s41598-021-87764-0] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2020] [Accepted: 03/30/2021] [Indexed: 11/30/2022] Open
Abstract
In many situations, the gene expression signature is a unique marker of the biological state. We study the modification of the gene expression distribution function when the biological state of a system experiences a change. This change may be the result of a selective pressure, as in the Long Term Evolution Experiment with E. Coli populations, or the progression to Alzheimer disease in aged brains, or the progression from a normal tissue to the cancer state. The first two cases seem to belong to a class of transitions, where the initial and final states are relatively close to each other, and the distribution function for the differential expressions is short ranged, with a tail of only a few dozens of strongly varying genes. In the latter case, cancer, the initial and final states are far apart and separated by a low-fitness barrier. The distribution function shows a very heavy tail, with thousands of silenced and over-expressed genes. We characterize the biological states by means of their principal component representations, and the expression distribution functions by their maximal and minimal differential expression values and the exponents of the Pareto laws describing the tails.
Collapse
Affiliation(s)
- Augusto Gonzalez
- University of Electronic Science and Technology, 610051, Chengdu, People's Republic of China
- Institute of Cybernetics, Mathematics and Physics, 10400, Havana, Cuba
| | - Joan Nieves
- Faculty of Physics, University of Havana, 10400, Havana, Cuba
| | - Dario A Leon
- Institute of Cybernetics, Mathematics and Physics, 10400, Havana, Cuba
- University of Modena and Reggio Emilia, 41125, Modena, Italy
| | - Maria Luisa Bringas Vega
- University of Electronic Science and Technology, 610051, Chengdu, People's Republic of China
- Cuban Neurosciences Center, 11600, Havana, Cuba
| | - Pedro Valdes Sosa
- University of Electronic Science and Technology, 610051, Chengdu, People's Republic of China.
- Cuban Neurosciences Center, 11600, Havana, Cuba.
| |
Collapse
|
16
|
Mao Y, Zeineldin M, Usmani M, Uprety S, Shisler JL, Jutla A, Unnikrishnan A, Nguyen TH. Distribution and Antibiotic Resistance Profiles of Salmonella enterica in Rural Areas of North Carolina After Hurricane Florence in 2018. GEOHEALTH 2021; 5:e2020GH000294. [PMID: 33709047 PMCID: PMC7892206 DOI: 10.1029/2020gh000294] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/01/2020] [Revised: 12/10/2020] [Accepted: 12/15/2020] [Indexed: 05/07/2023]
Abstract
In this study, water samples were analyzed from a rural area of North Carolina after Hurricane Florence in 2018 and the distribution of the ttrC virulence gene of Salmonella enterica were investigated. We also examined the distribution of culturable S. enterica and determined their antibiotic resistance profiles. Antibiotic resistance genes (ARGs) in the classes of aminoglycoside, beta-lactam, and macrolide-lincosamide-streptogramin B (MLSB) were targeted in this study. The ttrC gene was detected in 23 out of 25 locations. There was a wider and higher range of the ttrC gene in flooded water versus unflooded water samples (0-2.12 × 105 copies/L vs. 0-4.86 × 104 copies/L). Culturable S. enterica was isolated from 10 of 25 sampling locations, which was less prevalent than the distribution of the ttrC gene. The antibiotic resistance profiles were not distinct among the S. enterica isolates. The aminoglycoside resistance gene aac(6')-Iy had the highest relative abundance (around 0.05 copies/16S rRNA gene copy in all isolates) among all ARGs. These findings suggested that the 2018 flooding event led to higher copy numbers of the ttrC genes of S. enterica in some flooded water bodies compared to those in unflooded water bodies. The high ARG level and similar ARG profiles were observed in all S. enterica isolates from both flooded and unflooded samples, suggesting that the antibiotic resistance was prevalent in S. enterica within this region, regardless of flooding.
Collapse
Affiliation(s)
- Yuqing Mao
- Department of Civil and Environmental EngineeringUniversity of Illinois at Urbana‐ChampaignUrbanaILUSA
| | - Mohamed Zeineldin
- Institute for Genomic BiologyUniversity of Illinois at Urbana‐ChampaignUrbanaILUSA
- Animal Medicine DepartmentCollege of Veterinary MedicineBenha UniversityBenhaEgypt
| | - Moiz Usmani
- Environmental Engineering SciencesUniversity of FloridaGainesvilleFLUSA
| | - Sital Uprety
- Department of Civil and Environmental EngineeringUniversity of Illinois at Urbana‐ChampaignUrbanaILUSA
| | - Joanna L. Shisler
- Institute for Genomic BiologyUniversity of Illinois at Urbana‐ChampaignUrbanaILUSA
- Department of MicrobiologyUniversity of Illinois at Urbana‐ChampaignUrbanaILUSA
| | - Antarpreet Jutla
- Environmental Engineering SciencesUniversity of FloridaGainesvilleFLUSA
| | | | - Thanh H. Nguyen
- Department of Civil and Environmental EngineeringUniversity of Illinois at Urbana‐ChampaignUrbanaILUSA
- Institute for Genomic BiologyUniversity of Illinois at Urbana‐ChampaignUrbanaILUSA
| |
Collapse
|
17
|
Gupta S, Kumar P, Das BC. HPV +ve/-ve oral-tongue cancer stem cells: A potential target for relapse-free therapy. Transl Oncol 2021; 14:100919. [PMID: 33129107 PMCID: PMC7590584 DOI: 10.1016/j.tranon.2020.100919] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2020] [Revised: 09/27/2020] [Accepted: 10/12/2020] [Indexed: 12/12/2022] Open
Abstract
The tongue squamous cell carcinoma (TSCC) is a highly prevalent head and neck cancer often associated with tobacco and/or alcohol abuse or high-risk human papillomavirus (HR-HPV) infection. HPV positive TSCCs present a unique mechanism of tumorigenesis as compared to tobacco and alcohol-induced TSCCs and show a better prognosis when treated. The poor prognosis and/or recurrence of TSCC is due to presence of a small subpopulation of tumor-initiating tongue cancer stem cells (TCSCs) that are intrinsically resistant to conventional chemoradio-therapies enabling cancer to relapse. Therefore, targeting TCSCs may provide efficient therapeutic strategy for relapse-free survival of TSCC patients. Indeed, the development of new TCSC targeting therapeutic approaches for the successful elimination of HPV+ve/-ve TCSCs could be achieved either by targeting the self-renewal pathways, epithelial mesenchymal transition, vascular niche, nanoparticles-based therapy, induction of differentiation, chemoradio-sensitization of TCSCs or TCSC-derived exosome-based drug delivery and inhibition of HPV oncogenes or by regulating epigenetic pathways. In this review, we have discussed all these potential approaches and highlighted several important signaling pathways/networks involved in the formation and maintenance of TCSCs, which are targetable as novel therapeutic targets to sensitize/eliminate TCSCs and to improve survival of TSCC patients.
Collapse
Affiliation(s)
- Shilpi Gupta
- Stem Cell and Cancer Research Lab, Amity Institute of Molecular Medicine & Stem Cell Research (AIMMSCR), Amity University Uttar Pradesh, Sector-125, Noida 201313, India; National Institute of Cancer Prevention and Research (NICPR), I-7, Sector-39, Noida 201301, India
| | - Prabhat Kumar
- Stem Cell and Cancer Research Lab, Amity Institute of Molecular Medicine & Stem Cell Research (AIMMSCR), Amity University Uttar Pradesh, Sector-125, Noida 201313, India
| | - Bhudev C Das
- Stem Cell and Cancer Research Lab, Amity Institute of Molecular Medicine & Stem Cell Research (AIMMSCR), Amity University Uttar Pradesh, Sector-125, Noida 201313, India.
| |
Collapse
|
18
|
Abstract
Over the last several years, next-generation sequencing and its recent push toward single-cell resolution have transformed the landscape of immunology research by revealing novel complexities about all components of the immune system. With the vast amounts of diverse data currently being generated, and with the methods of analyzing and combining diverse data improving as well, integrative systems approaches are becoming more powerful. Previous integrative approaches have combined multiple data types and revealed ways that the immune system, both as a whole and as individual parts, is affected by genetics, the microbiome, and other factors. In this review, we explore the data types that are available for studying immunology with an integrative systems approach, as well as the current strategies and challenges for conducting such analyses.
Collapse
Affiliation(s)
- Silvia Pineda
- Bakar Computational Health Sciences Institute, University of California, San Francisco, California 94158, USA
- Genetic and Molecular Epidemiology Group, Spanish National Cancer Research Centre, 28029 Madrid, Spain
| | - Daniel G. Bunis
- Bakar Computational Health Sciences Institute, University of California, San Francisco, California 94158, USA
| | - Idit Kosti
- Bakar Computational Health Sciences Institute, University of California, San Francisco, California 94158, USA
- Department of Pediatrics, University of California, San Francisco, California 94143, USA
| | - Marina Sirota
- Bakar Computational Health Sciences Institute, University of California, San Francisco, California 94158, USA
- Department of Pediatrics, University of California, San Francisco, California 94143, USA
| |
Collapse
|
19
|
Sesen M, Whyte G. Image-Based Single Cell Sorting Automation in Droplet Microfluidics. Sci Rep 2020; 10:8736. [PMID: 32457421 PMCID: PMC7250914 DOI: 10.1038/s41598-020-65483-2] [Citation(s) in RCA: 58] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2019] [Accepted: 05/06/2020] [Indexed: 12/13/2022] Open
Abstract
The recent boom in single-cell omics has brought researchers one step closer to understanding the biological mechanisms associated with cell heterogeneity. Rare cells that have historically been obscured by bulk measurement techniques are being studied by single cell analysis and providing valuable insight into cell function. To support this progress, novel upstream capabilities are required for single cell preparation for analysis. Presented here is a droplet microfluidic, image-based single-cell sorting technique that is flexible and programmable. The automated system performs real-time dual-camera imaging (brightfield & fluorescent), processing, decision making and sorting verification. To demonstrate capabilities, the system was used to overcome the Poisson loading problem by sorting for droplets containing a single red blood cell with 85% purity. Furthermore, fluorescent imaging and machine learning was used to load single K562 cells amongst clusters based on their instantaneous size and circularity. The presented system aspires to replace manual cell handling techniques by translating expert knowledge into cell sorting automation via machine learning algorithms. This powerful technique finds application in the enrichment of single cells based on their micrographs for further downstream processing and analysis.
Collapse
Affiliation(s)
- Muhsincan Sesen
- Heriot-Watt University, Institute of Biological Chemistry, Biophysics and Bioengineering, Edinburgh, EH14 4AS, United Kingdom
- Imperial College London, Department of Bioengineering, London, SW7 2AZ, United Kingdom
| | - Graeme Whyte
- Heriot-Watt University, Institute of Biological Chemistry, Biophysics and Bioengineering, Edinburgh, EH14 4AS, United Kingdom.
| |
Collapse
|
20
|
Lim W, Lee S, Park S, Baac HW. Differential detachment of intact and viable cells of different sizes using laser-induced microbubbles. BIOMEDICAL OPTICS EXPRESS 2019; 10:4919-4930. [PMID: 31646019 PMCID: PMC6788613 DOI: 10.1364/boe.10.004919] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/02/2019] [Revised: 06/04/2019] [Accepted: 06/25/2019] [Indexed: 06/10/2023]
Abstract
Single cell isolation is a prerequisite for the analysis of rare or small cell subtypes. Here, we selectively detach single cells in a heterogeneous population comprised of different morphological subtypes whose sizes vary in body and extension. Such a cellular environment is first accommodated for by a photomechanical method in which pulsed laser irradiation produces microbubbles from a polymer substrate, thus pushing out and detaching cultured cells in an intact, viable, and spatially tailored way. While this has previously only bene used at a very low cell density with lack of quantitative characterization, we determine optimal detachment conditions for different cell sizes in terms of an optical fluence and the number of laser pulses. Importantly, our approach is employed to isolate cancer cells with inherent size variation and elucidate cellular heterogeneity in drug sensitivity: i.e., higher resistance for larger cell size. For cells detached by laser-induced microbubbles, morphology, proliferation, and viability are compared with those of conventional trypsin-treated cells detached without any spatial selectivity. These results support the suitability of our photomechanical method for biochemical screen and secondary analysis of cells with unusual responses.
Collapse
Affiliation(s)
- Wanyoung Lim
- Department of Biomedical Engineering, Sungkyunkwan University, Suwon 16419, South Korea
| | - Seungjin Lee
- Department of Electrical and Computer Engineering, Sungkyunkwan University, Suwon 16419, South Korea
| | - Sungsu Park
- Department of Biomedical Engineering, Sungkyunkwan University, Suwon 16419, South Korea
- School of Mechanical Engineering, Sungkyunkwan University, Suwon 16419, South Korea
| | - Hyoung Won Baac
- Department of Electrical and Computer Engineering, Sungkyunkwan University, Suwon 16419, South Korea
| |
Collapse
|
21
|
Szubert B, Cole JE, Monaco C, Drozdov I. Structure-preserving visualisation of high dimensional single-cell datasets. Sci Rep 2019; 9:8914. [PMID: 31222035 PMCID: PMC6586841 DOI: 10.1038/s41598-019-45301-0] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2018] [Accepted: 05/24/2019] [Indexed: 11/08/2022] Open
Abstract
Single-cell technologies offer an unprecedented opportunity to effectively characterize cellular heterogeneity in health and disease. Nevertheless, visualisation and interpretation of these multi-dimensional datasets remains a challenge. We present a novel framework, ivis, for dimensionality reduction of single-cell expression data. ivis utilizes a siamese neural network architecture that is trained using a novel triplet loss function. Results on simulated and real datasets demonstrate that ivis preserves global data structures in a low-dimensional space, adds new data points to existing embeddings using a parametric mapping function, and scales linearly to hundreds of thousands of cells. ivis is made publicly available through Python and R interfaces on https://github.com/beringresearch/ivis .
Collapse
Affiliation(s)
| | - Jennifer E Cole
- Kennedy Institute of Rheumatology, Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, University of Oxford, Oxford, OX3 7FY, UK
| | - Claudia Monaco
- Kennedy Institute of Rheumatology, Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, University of Oxford, Oxford, OX3 7FY, UK
| | | |
Collapse
|
22
|
O'Cathail SM, Buffa FM. Science in Focus: Bioinformatics Part 1 - Lost in Translation. Clin Oncol (R Coll Radiol) 2019; 31:337-340. [PMID: 30975523 DOI: 10.1016/j.clon.2019.03.043] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2019] [Revised: 03/02/2019] [Accepted: 03/04/2019] [Indexed: 02/07/2023]
Affiliation(s)
- S M O'Cathail
- CRUK/MRC Oxford Institute of Radiation Oncology, University of Oxford, Oxford, UK.
| | - F M Buffa
- Department of Oncology, University of Oxford, Oxford, UK
| |
Collapse
|
23
|
Nadappuram BP, Cadinu P, Barik A, Ainscough AJ, Devine MJ, Kang M, Gonzalez-Garcia J, Kittler JT, Willison KR, Vilar R, Actis P, Wojciak-Stothard B, Oh SH, Ivanov AP, Edel JB. Nanoscale tweezers for single-cell biopsies. NATURE NANOTECHNOLOGY 2019; 14:80-88. [PMID: 30510280 DOI: 10.1038/s41565-018-0315-8] [Citation(s) in RCA: 116] [Impact Index Per Article: 19.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/18/2018] [Accepted: 10/19/2018] [Indexed: 05/19/2023]
Abstract
Much of the functionality of multicellular systems arises from the spatial organization and dynamic behaviours within and between cells. Current single-cell genomic methods only provide a transcriptional 'snapshot' of individual cells. The real-time analysis and perturbation of living cells would generate a step change in single-cell analysis. Here we describe minimally invasive nanotweezers that can be spatially controlled to extract samples from living cells with single-molecule precision. They consist of two closely spaced electrodes with gaps as small as 10-20 nm, which can be used for the dielectrophoretic trapping of DNA and proteins. Aside from trapping single molecules, we also extract nucleic acids for gene expression analysis from living cells without affecting their viability. Finally, we report on the trapping and extraction of a single mitochondrion. This work bridges the gap between single-molecule/organelle manipulation and cell biology and can ultimately enable a better understanding of living cells.
Collapse
Affiliation(s)
| | - Paolo Cadinu
- Department of Chemistry, Imperial College London, London, UK
| | - Avijit Barik
- Department of Electrical and Computer Engineering, University of Minnesota, Minneapolis, MN, USA
| | - Alexander J Ainscough
- Department of Chemistry, Imperial College London, London, UK
- Department of Experimental Medicine and Toxicology, Imperial College London, London, UK
| | - Michael J Devine
- Department of Neuroscience, Physiology and Pharmacology, University College London, London, UK
| | - Minkyung Kang
- Department of Chemistry, Imperial College London, London, UK
| | | | - Josef T Kittler
- Department of Neuroscience, Physiology and Pharmacology, University College London, London, UK
| | | | - Ramon Vilar
- Department of Chemistry, Imperial College London, London, UK
| | - Paolo Actis
- School of Electronic and Electrical Engineering, Pollard Institute, University of Leeds, Leeds, UK
| | | | - Sang-Hyun Oh
- Department of Electrical and Computer Engineering, University of Minnesota, Minneapolis, MN, USA
| | | | - Joshua B Edel
- Department of Chemistry, Imperial College London, London, UK.
| |
Collapse
|
24
|
Abstract
![]()
The compartmentalization of reactions
in monodispersed droplets
is valuable for applications across biology. However, the requirement
of microfluidics to partition the sample into monodispersed droplets
is a significant barrier that impedes implementation. Here, we introduce
particle-templated emulsification, a method to encapsulate samples
in monodispersed emulsions without microfluidics. By vortexing a mixture
of hydrogel particles and sample solution, we encapsulate the sample
in monodispersed emulsions that are useful for most droplet applications.
We illustrate the method with ddPCR and single cell culture. The ability
to encapsulate samples in monodispersed droplets without microfluidics
should facilitate the implementation of compartmentalized reactions
in biology.
Collapse
Affiliation(s)
- Makiko N Hatori
- Department of Bioengineering and Therapeutic Sciences, California Institute for Quantitative Biosciences , University of California , San Francisco , California 94158 , United States
| | - Samuel C Kim
- Department of Bioengineering and Therapeutic Sciences, California Institute for Quantitative Biosciences , University of California , San Francisco , California 94158 , United States
| | - Adam R Abate
- Department of Bioengineering and Therapeutic Sciences, California Institute for Quantitative Biosciences , University of California , San Francisco , California 94158 , United States.,Chan Zuckerberg Biohub , San Francisco , California 94158 , United States
| |
Collapse
|
25
|
Developing novel methods to image and visualize 3D genomes. Cell Biol Toxicol 2018; 34:367-380. [PMID: 29577183 PMCID: PMC6133007 DOI: 10.1007/s10565-018-9427-z] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2017] [Accepted: 03/11/2018] [Indexed: 02/07/2023]
Abstract
To investigate three-dimensional (3D) genome organization in prokaryotic and eukaryotic cells, three main strategies are employed, namely nuclear proximity ligation-based methods, imaging tools (such as fluorescence in situ hybridization (FISH) and its derivatives), and computational/visualization methods. Proximity ligation-based methods are based on digestion and re-ligation of physically proximal cross-linked chromatin fragments accompanied by massively parallel DNA sequencing to measure the relative spatial proximity between genomic loci. Imaging tools enable direct visualization and quantification of spatial distances between genomic loci, and advanced implementation of (super-resolution) microscopy helps to significantly improve the resolution of images. Computational methods are used to map global 3D genome structures at various scales driven by experimental data, and visualization methods are used to visualize genome 3D structures in virtual 3D space-based on algorithms. In this review, we focus on the introduction of novel imaging and visualization methods to study 3D genomes. First, we introduce the progress made recently in 3D genome imaging in both fixed cell and live cells based on long-probe labeling, short-probe labeling, RNA FISH, and the CRISPR system. As the fluorescence-capturing capability of a particular microscope is very important for the sensitivity of bioimaging experiments, we also introduce two novel super-resolution microscopy methods, SDOM and low-power super-resolution STED, which have potential for time-lapse super-resolution live-cell imaging of chromatin. Finally, we review some software tools developed recently to visualize proximity ligation-based data. The imaging and visualization methods are complementary to each other, and all three strategies are not mutually exclusive. These methods provide powerful tools to explore the mechanisms of gene regulation and transcription in cell nuclei.
Collapse
|
26
|
|
27
|
Wu AR, Wang J, Streets AM, Huang Y. Single-Cell Transcriptional Analysis. ANNUAL REVIEW OF ANALYTICAL CHEMISTRY (PALO ALTO, CALIF.) 2017; 10:439-462. [PMID: 28301747 DOI: 10.1146/annurev-anchem-061516-045228] [Citation(s) in RCA: 60] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/06/2023]
Abstract
Despite being a relatively recent technological development, single-cell transcriptional analysis through high-throughput sequencing has already been used in hundreds of fruitful studies to make exciting new biological discoveries that would otherwise be challenging or even impossible. Consequently, this has fueled a virtuous cycle of even greater interest in the field and compelled development of further improved technical methodologies and approaches. Thanks to the combined efforts of the research community, including the fields of biochemistry and molecular biology, technology and instrumentation, data science, computational biology, and bioinformatics, the single-cell RNA-sequencing field is advancing at a pace that is both astounding and unprecedented. In this review, we provide a broad introduction to this revolutionary technology by presenting the state-of-the-art in sample preparation methodologies, technology platforms, and computational analysis methods, while highlighting the key considerations for designing, executing, and interpreting a study using single-cell RNA sequencing.
Collapse
Affiliation(s)
- Angela R Wu
- Division of Life Science and Division of Biomedical Engineering, The Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong, China;
| | - Jianbin Wang
- School of Life Sciences and Center for Life Sciences, Tsinghua University, Beijing 100084, China;
| | - Aaron M Streets
- Department of Bioengineering, University of California, Berkeley, California 94720;
| | - Yanyi Huang
- Biodynamic Optical Imaging Center (BIOPIC), Beijing Advanced Innovation Center for Genomics (ICG), College of Engineering, and Center for Life Sciences, Peking University, Beijing 100871, China;
| |
Collapse
|
28
|
Yuan GC, Cai L, Elowitz M, Enver T, Fan G, Guo G, Irizarry R, Kharchenko P, Kim J, Orkin S, Quackenbush J, Saadatpour A, Schroeder T, Shivdasani R, Tirosh I. Challenges and emerging directions in single-cell analysis. Genome Biol 2017; 18:84. [PMID: 28482897 PMCID: PMC5421338 DOI: 10.1186/s13059-017-1218-y] [Citation(s) in RCA: 199] [Impact Index Per Article: 24.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2016] [Accepted: 04/21/2017] [Indexed: 12/15/2022] Open
Abstract
Single-cell analysis is a rapidly evolving approach to characterize genome-scale molecular information at the individual cell level. Development of single-cell technologies and computational methods has enabled systematic investigation of cellular heterogeneity in a wide range of tissues and cell populations, yielding fresh insights into the composition, dynamics, and regulatory mechanisms of cell states in development and disease. Despite substantial advances, significant challenges remain in the analysis, integration, and interpretation of single-cell omics data. Here, we discuss the state of the field and recent advances and look to future opportunities.
Collapse
Affiliation(s)
- Guo-Cheng Yuan
- Department of Biostatistics and Computational Biology, Dana-Farber Cancer Institute, Boston, MA, USA.
- Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, MA, USA.
| | - Long Cai
- Division of Chemistry and Chemical Engineering, California Institute of Technology, Pasadena, CA, USA
| | - Michael Elowitz
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, USA
| | - Tariq Enver
- Cancer Institute, University College London, London, UK
| | - Guoping Fan
- Department of Human Genetics, University of California, Los Angeles, CA, USA
| | - Guoji Guo
- Center for Stem Cell and Regenerative Medicine, Zhejiang University, Hangzhou, China
| | - Rafael Irizarry
- Department of Biostatistics and Computational Biology, Dana-Farber Cancer Institute, Boston, MA, USA
- Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Peter Kharchenko
- Department of Biomedical Informatics, Harvard Medical School, Boston, MA, USA
| | - Junhyong Kim
- Department of Biology, University of Pennsylvania, Philadelphia, PA, USA
| | - Stuart Orkin
- Department of Pediatric Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Division of Hematology/Oncology, Boston Children's Hospital, Boston, MA, USA
- Howard Hughes Medical Institute, Boston, MA, USA
| | - John Quackenbush
- Department of Biostatistics and Computational Biology, Dana-Farber Cancer Institute, Boston, MA, USA
- Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Assieh Saadatpour
- Department of Biostatistics and Computational Biology, Dana-Farber Cancer Institute, Boston, MA, USA
- Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Timm Schroeder
- Department of Biosystems Science and Engineering, ETH Zurich, Basel, Switzerland
| | - Ramesh Shivdasani
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Itay Tirosh
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| |
Collapse
|
29
|
Acero Sánchez JL, Joda H, Henry OYF, Solnestam BW, Kvastad L, Akan PS, Lundeberg J, Laddach N, Ramakrishnan D, Riley I, Schwind C, Latta D, O'Sullivan CK. Electrochemical Genetic Profiling of Single Cancer Cells. Anal Chem 2017; 89:3378-3385. [PMID: 28211676 DOI: 10.1021/acs.analchem.6b03973] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Recent understandings in the development and spread of cancer have led to the realization of novel single cell analysis platforms focused on circulating tumor cells (CTCs). A simple, rapid, and inexpensive analytical platform capable of providing genetic information on these rare cells is highly desirable to support clinicians and researchers alike to either support the selection or adjustment of therapy or provide fundamental insights into cell function and cancer progression mechanisms. We report on the genetic profiling of single cancer cells, exploiting a combination of multiplex ligation-dependent probe amplification (MLPA) and electrochemical detection. Cells were isolated using laser capture and lysed, and the mRNA was extracted and transcribed into DNA. Seven markers were amplified by MLPA, which allows for the simultaneous amplification of multiple targets with a single primer pair, using MLPA probes containing unique barcode sequences. Capture probes complementary to each of these barcode sequences were immobilized on a printed circuit board (PCB) manufactured electrode array and exposed to single-stranded MLPA products and subsequently to a single stranded DNA reporter probe bearing a HRP molecule, followed by substrate addition and fast electrochemical pulse amperometric detection. We present a simple, rapid, flexible, and inexpensive approach for the simultaneous quantification of multiple breast cancer related mRNA markers, with single tumor cell sensitivity.
Collapse
Affiliation(s)
- Josep Ll Acero Sánchez
- Universitat Rovira i Virgili , Departament de Enginyeria Química, Av. Països Catalans 26, 43007 Tarragona, Spain
| | - Hamdi Joda
- Universitat Rovira i Virgili , Departament de Enginyeria Química, Av. Països Catalans 26, 43007 Tarragona, Spain
| | - Olivier Y F Henry
- Universitat Rovira i Virgili , Departament de Enginyeria Química, Av. Països Catalans 26, 43007 Tarragona, Spain
| | - Beata W Solnestam
- KTH Royal Institute of Technology , Science for Life Laboratory (SciLifeLab Stockholm), School of Biotechnology, Division of Gene Technology, SE-171 65 Solna, Sweden
| | - Linda Kvastad
- KTH Royal Institute of Technology , Science for Life Laboratory (SciLifeLab Stockholm), School of Biotechnology, Division of Gene Technology, SE-171 65 Solna, Sweden
| | - Pelin S Akan
- KTH Royal Institute of Technology , Science for Life Laboratory (SciLifeLab Stockholm), School of Biotechnology, Division of Gene Technology, SE-171 65 Solna, Sweden
| | - Joakim Lundeberg
- KTH Royal Institute of Technology , Science for Life Laboratory (SciLifeLab Stockholm), School of Biotechnology, Division of Gene Technology, SE-171 65 Solna, Sweden
| | - Nadja Laddach
- MRC-Holland , Willem Schoutenstraat 1, 1057 DL Amsterdam, The Netherlands
| | - Dheeraj Ramakrishnan
- Labman Automation Ltd. , Seamer Hill, Seamer, Stokesley, North Yorkshire TS9 5NQ, United Kingdom
| | - Ian Riley
- Labman Automation Ltd. , Seamer Hill, Seamer, Stokesley, North Yorkshire TS9 5NQ, United Kingdom
| | - Carmen Schwind
- Fraunhofer (ICT-IMM) , Carl-Zeiss-Str. 18-20, 55129 Mainz, Germany
| | - Daniel Latta
- Fraunhofer (ICT-IMM) , Carl-Zeiss-Str. 18-20, 55129 Mainz, Germany
| | - Ciara K O'Sullivan
- Universitat Rovira i Virgili , Departament de Enginyeria Química, Av. Països Catalans 26, 43007 Tarragona, Spain.,Institució Catalana de Recerca i Estudis Avançats , Passeig Lluís Companys 23, 08010 Barcelona, Spain
| |
Collapse
|
30
|
Te Pas MFW, Madsen O, Calus MPL, Smits MA. The Importance of Endophenotypes to Evaluate the Relationship between Genotype and External Phenotype. Int J Mol Sci 2017; 18:E472. [PMID: 28241430 PMCID: PMC5344004 DOI: 10.3390/ijms18020472] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2016] [Revised: 02/02/2017] [Accepted: 02/13/2017] [Indexed: 02/06/2023] Open
Abstract
With the exception of a few Mendelian traits, almost all phenotypes (traits) in livestock science are quantitative or complex traits regulated by the expression of many genes. For most of the complex traits, differential expression of genes, rather than genomic variation in the gene coding sequences, is associated with the genotype of a trait. The expression profiles of the animal's transcriptome, proteome and metabolome represent endophenotypes that influence/regulate the externally-observed phenotype. These expression profiles are generated by interactions between the animal's genome and its environment that range from the cellular, up to the husbandry environment. Thus, understanding complex traits requires knowledge about not only genomic variation, but also environmental effects that affect genome expression. Gene products act together in physiological pathways and interaction networks (of pathways). Due to the lack of annotation of the functional genome and ontologies of genes, our knowledge about the various biological systems that contribute to the development of external phenotypes is sparse. Furthermore, interaction with the animals' microbiome, especially in the gut, greatly influences the external phenotype. We conclude that a detailed understanding of complex traits requires not only understanding of variation in the genome, but also its expression at all functional levels.
Collapse
Affiliation(s)
- Marinus F W Te Pas
- Animal Breeding and Genomics Centre, Wageningen UR Livestock Research, 6700AH Wageningen, The Netherlands.
| | - Ole Madsen
- Animal Breeding and Genomics, Wageningen University, 6700AH Wageningen, The Netherlands.
| | - Mario P L Calus
- Animal Breeding and Genomics Centre, Wageningen UR Livestock Research, 6700AH Wageningen, The Netherlands.
| | - Mari A Smits
- Animal Breeding and Genomics Centre, Wageningen UR Livestock Research, 6700AH Wageningen, The Netherlands.
| |
Collapse
|
31
|
Personalized Treatment Through Detection and Monitoring of Genetic Aberrations in Single Circulating Tumor Cells. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2017; 994:255-273. [DOI: 10.1007/978-3-319-55947-6_14] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
|
32
|
Chen Z, Fu Y, Zhang F, Liu L, Zhang N, Zhou D, Yang J, Pang Y, Huang Y. Spinning micropipette liquid emulsion generator for single cell whole genome amplification. LAB ON A CHIP 2016; 16:4512-4516. [PMID: 27775138 DOI: 10.1039/c6lc01084a] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/06/2023]
Abstract
Many on-chip approaches that use flow-focusing to pinch the continuous aqueous phase into droplets have become the most popular methods that provide monodisperse emulsion droplets. However, not every lab can easily adapt a microfluidic workflow into their familiar protocols. We develop an off-chip approach, spinning micro-pipette liquid emulsion (SiMPLE) generator, to produce highly stable monodisperse water-in-oil emulsions using a moving micropipette to disperse the aqueous phase in an oil-filled microcentrifuge tube. This method provides a simple way to produce picoliter-size droplets in situ with no dead volume during emulsification. With SiMPLE, single-cell emulsion whole genome amplification was performed to demonstrate that this novel method can seamlessly be integrated with experimental operations and supplies that most researchers are familiar with. The SiMPLE generator has effectively lowered the technical difficulties in applications relying on emulsion droplets.
Collapse
Affiliation(s)
- Zitian Chen
- Biodynamic Optical Imaging Center (BIOPIC), School of Life Sciences, and College of Engineering, Peking University, Beijing, China. and Beijing Advanced Innovation Center for Genomics (ICG), Peking University, Beijing, China
| | - Yusi Fu
- Biodynamic Optical Imaging Center (BIOPIC), School of Life Sciences, and College of Engineering, Peking University, Beijing, China. and Beijing Advanced Innovation Center for Genomics (ICG), Peking University, Beijing, China
| | - Fangli Zhang
- Biodynamic Optical Imaging Center (BIOPIC), School of Life Sciences, and College of Engineering, Peking University, Beijing, China. and Beijing Advanced Innovation Center for Genomics (ICG), Peking University, Beijing, China
| | - Lu Liu
- Biodynamic Optical Imaging Center (BIOPIC), School of Life Sciences, and College of Engineering, Peking University, Beijing, China. and Beijing Advanced Innovation Center for Genomics (ICG), Peking University, Beijing, China
| | - Naiqing Zhang
- Biodynamic Optical Imaging Center (BIOPIC), School of Life Sciences, and College of Engineering, Peking University, Beijing, China.
| | - Dong Zhou
- Biodynamic Optical Imaging Center (BIOPIC), School of Life Sciences, and College of Engineering, Peking University, Beijing, China.
| | - Junrui Yang
- School of Electronics Engineering and Computer Science, Peking University, Beijing, China
| | - Yuhong Pang
- Biodynamic Optical Imaging Center (BIOPIC), School of Life Sciences, and College of Engineering, Peking University, Beijing, China. and Beijing Advanced Innovation Center for Genomics (ICG), Peking University, Beijing, China
| | - Yanyi Huang
- Biodynamic Optical Imaging Center (BIOPIC), School of Life Sciences, and College of Engineering, Peking University, Beijing, China. and Beijing Advanced Innovation Center for Genomics (ICG), Peking University, Beijing, China and Peking-Tsinghua Center for Life Sciences, Peking University, Beijing, China
| |
Collapse
|
33
|
Gangavarapu KJ, Miller A, Huss WJ. Gene Expression in Single Cells Isolated from the CWR-R1 Prostate Cancer Cell Line and Human Prostate Tissue Based on the Side Population Phenotype. ACTA ACUST UNITED AC 2016; 5. [PMID: 27785389 PMCID: PMC5076885 DOI: 10.4172/2168-9431.1000150] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Defining biological signals at the single cell level can identify cancer initiating driver mutations. Techniques to isolate single cells such as microfluidics sorting and magnetic capturing systems have limitations such as: high cost, labor intense, and the requirement of a large number of cells. Therefore, the goal of our current study is to identify a cost and labor effective, reliable, and reproducible technique that allows single cell isolation for analysis to promote regular laboratory use, including standard reverse transcription PCR (RT-PCR). In the current study, we utilized single prostate cells isolated from the CWR-R1 prostate cancer cell line and human prostate clinical specimens, based on the ATP binding cassette (ABC) transporter efflux of dye cycle violet (DCV), side population assay. Expression of four genes: ABCG2; Aldehyde dehydrogenase1A1 (ALDH1A1); androgen receptor (AR); and embryonic stem cell marker, Oct-4, were determined. Results from the current study in the CWR-R1 cell line showed ABCG2 and ALDH1A1 gene expression in 67% of single side population cells and in 17% or 100% of non-side population cells respectively. Studies using single cells isolated from clinical specimens showed that the Oct-4 gene is detected in only 22% of single side population cells and in 78% of single non-side population cells. Whereas, AR gene expression is in 100% single side population and non-side population cells isolated from the same human prostate clinical specimen. These studies show that performing RT-PCR on single cells isolated by FACS can be successfully conducted to determine gene expression in single cells from cell lines and enzymatically digested tissue. While these studies provide a simple yes/no expression readout, the more sensitive quantitative RT-PCR would be able to provide even more information if necessary.
Collapse
Affiliation(s)
- Kalyan J Gangavarapu
- Department of Pharmacology and Therapeutics, Roswell Park Cancer Institute, Buffalo, NY-14263, USA
| | - Austin Miller
- Department of Biostatistics, Roswell Park Cancer Institute, Buffalo, NY-14263, USA
| | - Wendy J Huss
- Department of Pharmacology and Therapeutics, Roswell Park Cancer Institute, Buffalo, NY-14263, USA; Department of Urologic Oncology, Roswell Park Cancer Institute, Buffalo, NY-14263, USA
| |
Collapse
|
34
|
Hu P, Zhang W, Xin H, Deng G. Single Cell Isolation and Analysis. Front Cell Dev Biol 2016; 4:116. [PMID: 27826548 PMCID: PMC5078503 DOI: 10.3389/fcell.2016.00116] [Citation(s) in RCA: 216] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2016] [Accepted: 10/07/2016] [Indexed: 02/05/2023] Open
Abstract
Individual cell heterogeneity within a population can be critical to its peculiar function and fate. Subpopulations studies with mixed mutants and wild types may not be as informative regarding which cell responds to which drugs or clinical treatments. Cell to cell differences in RNA transcripts and protein expression can be key to answering questions in cancer, neurobiology, stem cell biology, immunology, and developmental biology. Conventional cell-based assays mainly analyze the average responses from a population of cells, without regarding individual cell phenotypes. To better understand the variations from cell to cell, scientists need to use single cell analyses to provide more detailed information for therapeutic decision making in precision medicine. In this review, we focus on the recent developments in single cell isolation and analysis, which include technologies, analyses and main applications. Here, we summarize the historical background, limitations, applications, and potential of single cell isolation technologies.
Collapse
Affiliation(s)
- Ping Hu
- The Center for Biotechnology and Biopharmaceutics, Institute of Translational Medicine, Nanchang University Nanchang, China
| | - Wenhua Zhang
- Laboratory of Fear and Anxiety Disorders, Institute of Life Science, Nanchang University Nanchang, China
| | - Hongbo Xin
- The Center for Biotechnology and Biopharmaceutics, Institute of Translational Medicine, Nanchang University Nanchang, China
| | - Glenn Deng
- The Center for Biotechnology and Biopharmaceutics, Institute of Translational Medicine, Nanchang UniversityNanchang, China; Yichang Research Center for Biomedical Industry and Central Laboratory of Yichang Central Hospital, Medical School, China Three Gorges UniversityYichang, China; Division of Surgical Oncology, Stanford University School of MedicineStanford, CA, USA
| |
Collapse
|
35
|
Virtual microfluidics for digital quantification and single-cell sequencing. Nat Methods 2016; 13:759-62. [PMID: 27479330 PMCID: PMC5007149 DOI: 10.1038/nmeth.3955] [Citation(s) in RCA: 72] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2016] [Accepted: 06/27/2016] [Indexed: 01/18/2023]
Abstract
Interest in highly parallelized analysis of single molecules and single cells is growing rapidly. Here we develop hydrogel-based virtual microfluidics as a simple alternative to complex engineered microfluidic systems for the compartmentalization of nucleic acid amplification reactions. We applied digital multiple displacement amplification (dMDA) to purified DNA templates, cultured bacterial cells, and human microbiome samples in the virtual microfluidics system and demonstrated recovery and whole-genome sequencing of single-cell MDA products. Our results from control samples showed excellent coverage uniformity and markedly reduced chimerism compared with single-cell data obtained from conventional liquid MDA reactions. We also demonstrate the applicability of the hydrogel method for genomic studies of naturally occurring microbes in human microbiome samples. The virtual microfluidics approach is a simple and robust method that will enable many laboratories to perform single-cell genomic analyses.
Collapse
|
36
|
Troell K, Hallström B, Divne AM, Alsmark C, Arrighi R, Huss M, Beser J, Bertilsson S. Cryptosporidium as a testbed for single cell genome characterization of unicellular eukaryotes. BMC Genomics 2016; 17:471. [PMID: 27338614 PMCID: PMC4917956 DOI: 10.1186/s12864-016-2815-y] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2016] [Accepted: 06/07/2016] [Indexed: 12/11/2022] Open
Abstract
Background Infectious disease involving multiple genetically distinct populations of pathogens is frequently concurrent, but difficult to detect or describe with current routine methodology. Cryptosporidium sp. is a widespread gastrointestinal protozoan of global significance in both animals and humans. It cannot be easily maintained in culture and infections of multiple strains have been reported. To explore the potential use of single cell genomics methodology for revealing genome-level variation in clinical samples from Cryptosporidium-infected hosts, we sorted individual oocysts for subsequent genome amplification and full-genome sequencing. Results Cells were identified with fluorescent antibodies with an 80 % success rate for the entire single cell genomics workflow, demonstrating that the methodology can be applied directly to purified fecal samples. Ten amplified genomes from sorted single cells were selected for genome sequencing and compared both to the original population and a reference genome in order to evaluate the accuracy and performance of the method. Single cell genome coverage was on average 81 % even with a moderate sequencing effort and by combining the 10 single cell genomes, the full genome was accounted for. By a comparison to the original sample, biological variation could be distinguished and separated from noise introduced in the amplification. Conclusions As a proof of principle, we have demonstrated the power of applying single cell genomics to dissect infectious disease caused by closely related parasite species or subtypes. The workflow can easily be expanded and adapted to target other protozoans, and potential applications include mapping genome-encoded traits, virulence, pathogenicity, host specificity and resistance at the level of cells as truly meaningful biological units. Electronic supplementary material The online version of this article (doi:10.1186/s12864-016-2815-y) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Karin Troell
- Department of Microbiology, National Veterinary Institute, Uppsala, Sweden.
| | - Björn Hallström
- Department of Microbiology, Public Health Agency of Sweden, Solna, Sweden
| | - Anna-Maria Divne
- Microbial Single Cell Genomics Facility, Department of Cell and Molecular Biology and Science for Life Laboratory, Uppsala University, Uppsala, Sweden
| | - Cecilia Alsmark
- Department of Microbiology, National Veterinary Institute, Uppsala, Sweden.,Division of Pharmacognosy, Department of Medicinal Chemistry, Biomedical Center, Uppsala University, Uppsala, Sweden
| | - Romanico Arrighi
- Department of Microbiology, Public Health Agency of Sweden, Solna, Sweden
| | - Mikael Huss
- Department of Biochemistry and Biophysics, Science for Life Laboratory, Stockholm University, Solna, Sweden
| | - Jessica Beser
- Department of Microbiology, Public Health Agency of Sweden, Solna, Sweden
| | - Stefan Bertilsson
- Department of Ecology and Genetics, Limnology and Science for Life Laboratory, Uppsala University, Uppsala, Sweden.
| |
Collapse
|
37
|
Microfluidic single-cell transcriptional analysis rationally identifies novel surface marker profiles to enhance cell-based therapies. Nat Commun 2016; 7:11945. [PMID: 27324848 PMCID: PMC5512622 DOI: 10.1038/ncomms11945] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2015] [Accepted: 05/16/2016] [Indexed: 12/17/2022] Open
Abstract
Current progenitor cell therapies have only modest efficacy, which has limited their clinical adoption. This may be the result of a cellular heterogeneity that decreases the number of functional progenitors delivered to diseased tissue, and prevents correction of underlying pathologic cell population disruptions. Here, we develop a high-resolution method of identifying phenotypically distinct progenitor cell subpopulations via single-cell transcriptional analysis and advanced bioinformatics. When combined with high-throughput cell surface marker screening, this approach facilitates the rational selection of surface markers for prospective isolation of cell subpopulations with desired transcriptional profiles. We establish the usefulness of this platform in costly and highly morbid diabetic wounds by identifying a subpopulation of progenitor cells that is dysfunctional in the diabetic state, and normalizes diabetic wound healing rates following allogeneic application. We believe this work presents a logical framework for the development of targeted cell therapies that can be customized to any clinical application. Unrecognized progenitor cell perturbations underlying a disease state may limit the efficacy of cell therapies. Here, the authors use high-throughput, single-cell transcriptional analysis to identify disease-specific cellular alterations and prospectively isolate restorative cell subpopulations.
Collapse
|
38
|
Rennert RC, Achrol AS, Januszyk M, Kahn SA, Liu TT, Liu Y, Sahoo D, Rodrigues M, Maan ZN, Wong VW, Cheshier SH, Chang SD, Steinberg GK, Harsh GR, Gurtner GC. Multiple Subsets of Brain Tumor Initiating Cells Coexist in Glioblastoma. Stem Cells 2016; 34:1702-7. [PMID: 26991945 DOI: 10.1002/stem.2359] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2015] [Accepted: 02/07/2016] [Indexed: 12/31/2022]
Abstract
Brain tumor-initiating cells (BTICs) are self-renewing multipotent cells critical for tumor maintenance and growth. Using single-cell microfluidic profiling, we identified multiple subpopulations of BTICs coexisting in human glioblastoma, characterized by distinct surface marker expression and single-cell molecular profiles relating to divergent bulk tissue molecular subtypes. These data suggest BTIC subpopulation heterogeneity as an underlying source of intra-tumoral bulk tissue molecular heterogeneity, and will support future studies into BTIC subpopulation-specific therapies. Stem Cells 2016;34:1702-1707.
Collapse
Affiliation(s)
- Robert C Rennert
- Department of Surgery, Stanford University School of Medicine, Stanford, California, USA
| | - Achal S Achrol
- Department of Neurosurgery, Stanford University School of Medicine, Stanford, California, USA
| | - Michael Januszyk
- Department of Surgery, Stanford University School of Medicine, Stanford, California, USA.,Stanford Center for Biomedical Informatics, Stanford University, Stanford, California, USA
| | - Suzana A Kahn
- Department of Neurosurgery, Stanford University School of Medicine, Stanford, California, USA
| | - Tiffany T Liu
- Stanford Center for Biomedical Informatics, Stanford University, Stanford, California, USA
| | - Yi Liu
- Stanford Center for Biomedical Informatics, Stanford University, Stanford, California, USA
| | - Debashis Sahoo
- Stanford Center for Biomedical Informatics, Stanford University, Stanford, California, USA
| | - Melanie Rodrigues
- Department of Surgery, Stanford University School of Medicine, Stanford, California, USA
| | - Zeshaan N Maan
- Department of Surgery, Stanford University School of Medicine, Stanford, California, USA
| | - Victor W Wong
- Department of Surgery, Stanford University School of Medicine, Stanford, California, USA
| | - Samuel H Cheshier
- Department of Neurosurgery, Stanford University School of Medicine, Stanford, California, USA
| | - Steven D Chang
- Department of Neurosurgery, Stanford University School of Medicine, Stanford, California, USA
| | - Gary K Steinberg
- Department of Neurosurgery, Stanford University School of Medicine, Stanford, California, USA
| | - Griffith R Harsh
- Department of Neurosurgery, Stanford University School of Medicine, Stanford, California, USA
| | - Geoffrey C Gurtner
- Department of Surgery, Stanford University School of Medicine, Stanford, California, USA
| |
Collapse
|
39
|
Rennert RC, Schäfer R, Bliss T, Januszyk M, Sorkin M, Achrol AS, Rodrigues M, Maan ZN, Kluba T, Steinberg GK, Gurtner GC. High-Resolution Microfluidic Single-Cell Transcriptional Profiling Reveals Clinically Relevant Subtypes among Human Stem Cell Populations Commonly Utilized in Cell-Based Therapies. Front Neurol 2016; 7:41. [PMID: 27047447 PMCID: PMC4801858 DOI: 10.3389/fneur.2016.00041] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2016] [Accepted: 03/10/2016] [Indexed: 12/21/2022] Open
Abstract
Stem cell therapies can promote neural repair and regeneration, yet controversy regarding optimal cell source and mechanism of action has slowed clinical translation, potentially due to undefined cellular heterogeneity. Single-cell resolution is needed to identify clinically relevant subpopulations with the highest therapeutic relevance. We combine single-cell microfluidic analysis with advanced computational modeling to study for the first time two common sources for cell-based therapies, human NSCs and MSCs. This methodology has the potential to logically inform cell source decisions for any clinical application.
Collapse
Affiliation(s)
- Robert C Rennert
- Department of Surgery, Stanford University School of Medicine , Stanford, CA , USA
| | - Richard Schäfer
- Department of Neurosurgery, Stanford University School of Medicine , Stanford, CA , USA
| | - Tonya Bliss
- Department of Neurosurgery, Stanford University School of Medicine , Stanford, CA , USA
| | - Michael Januszyk
- Department of Surgery, Stanford University School of Medicine , Stanford, CA , USA
| | - Michael Sorkin
- Department of Surgery, Stanford University School of Medicine , Stanford, CA , USA
| | - Achal S Achrol
- Department of Neurosurgery, Stanford University School of Medicine , Stanford, CA , USA
| | - Melanie Rodrigues
- Department of Surgery, Stanford University School of Medicine , Stanford, CA , USA
| | - Zeshaan N Maan
- Department of Surgery, Stanford University School of Medicine , Stanford, CA , USA
| | - Torsten Kluba
- Department of Orthopedics, University Hospital Tübingen , Tübingen , Germany
| | - Gary K Steinberg
- Department of Neurosurgery, Stanford University School of Medicine , Stanford, CA , USA
| | - Geoffrey C Gurtner
- Department of Surgery, Stanford University School of Medicine , Stanford, CA , USA
| |
Collapse
|
40
|
Heath JR, Ribas A, Mischel PS. Single-cell analysis tools for drug discovery and development. Nat Rev Drug Discov 2016; 15:204-16. [PMID: 26669673 PMCID: PMC4883669 DOI: 10.1038/nrd.2015.16] [Citation(s) in RCA: 347] [Impact Index Per Article: 38.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
The genetic, functional or compositional heterogeneity of healthy and diseased tissues presents major challenges in drug discovery and development. Such heterogeneity hinders the design of accurate disease models and can confound the interpretation of biomarker levels and of patient responses to specific therapies. The complex nature of virtually all tissues has motivated the development of tools for single-cell genomic, transcriptomic and multiplex proteomic analyses. Here, we review these tools and assess their advantages and limitations. Emerging applications of single cell analysis tools in drug discovery and development, particularly in the field of oncology, are discussed.
Collapse
Affiliation(s)
- James R Heath
- California Institute of Technology Division of Chemistry and Chemical Engineering, MC 127-72, 1200 East California Boulevard, Pasadena, California 91125, USA
| | - Antoni Ribas
- Department of Medicine, University of California, Los Angeles, 10833 Le Conte Avenue, Los Angeles, California 90095, USA
| | - Paul S Mischel
- Ludwig Institute for Cancer Research San Diego, Department of Pathology and Moores Cancer Center, University of California, San Diego, 9500 Gilman Drive, La Jolla, California 92093, USA
| |
Collapse
|
41
|
Yeo T, Tan SJ, Lim CL, Lau DPX, Chua YW, Krisna SS, Iyer G, Tan GS, Lim TKH, Tan DS, Lim WT, Lim CT. Microfluidic enrichment for the single cell analysis of circulating tumor cells. Sci Rep 2016; 6:22076. [PMID: 26924553 PMCID: PMC4770429 DOI: 10.1038/srep22076] [Citation(s) in RCA: 87] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2015] [Accepted: 02/05/2016] [Indexed: 12/18/2022] Open
Abstract
Resistance to drug therapy is a major concern in cancer treatment. To probe clones resistant to chemotherapy, the current approach is to conduct pooled cell analysis. However, this can yield false negative outcomes, especially when we are analyzing a rare number of circulating tumor cells (CTCs) among an abundance of other cell types. Here, we develop a microfluidic device that is able to perform high throughput, selective picking and isolation of single CTC to 100% purity from a larger population of other cells. This microfluidic device can effectively separate the very rare CTCs from blood samples from as few as 1 in 20,000 white blood cells. We first demonstrate isolation of pure tumor cells from a mixed population and track variations of acquired T790M mutations before and after drug treatment using a model PC9 cell line. With clinical CTC samples, we then show that the isolated single CTCs are representative of dominant EGFR mutations such as T790M and L858R found in the primary tumor. With this single cell recovery device, we can potentially implement personalized treatment not only through detecting genetic aberrations at the single cell level, but also through tracking such changes during an anticancer therapy.
Collapse
Affiliation(s)
- Trifanny Yeo
- Clearbridge Accelerator Pte Ltd, 81 Science Park Drive, The Chadwick, #02-03, Singapore Science Park 1, Singapore 118257, Singapore
| | - Swee Jin Tan
- Clearbridge Accelerator Pte Ltd, 81 Science Park Drive, The Chadwick, #02-03, Singapore Science Park 1, Singapore 118257, Singapore
| | - Chew Leng Lim
- School of Biological Science, National Technological University, 60 Nanyang Drive, Singapore 637551, Singapore
| | - Dawn Ping Xi Lau
- Cancer Therapeutics Research Laboratory, National Cancer Centre Singapore, 11 Hospital Drive, Singapore 169610, Singapore
| | - Yong Wei Chua
- Department of Pathology, Singapore General Hospital, Outram Road, Singapore 169608, Singapore
| | - Sai Sakktee Krisna
- Cancer Therapeutics Research Laboratory, National Cancer Centre Singapore, 11 Hospital Drive, Singapore 169610, Singapore
| | - Gopal Iyer
- Cancer Therapeutics Research Laboratory, National Cancer Centre Singapore, 11 Hospital Drive, Singapore 169610, Singapore
| | - Gek San Tan
- Department of Pathology, Singapore General Hospital, Outram Road, Singapore 169608, Singapore
| | - Tony Kiat Hon Lim
- Department of Pathology, Singapore General Hospital, Outram Road, Singapore 169608, Singapore
| | - Daniel S.W. Tan
- Cancer Therapeutics Research Laboratory, National Cancer Centre Singapore, 11 Hospital Drive, Singapore 169610, Singapore
- Cancer Stem Cell Biology, Genome Institute of Singapore, 60 Biopolis St, #02-01, 138672, Singapore
- Division of Medical Oncology, National Cancer Centre Singapore, 11 Hospital Drive, Singapore 169610, Singapore
| | - Wan-Teck Lim
- Division of Medical Oncology, National Cancer Centre Singapore, 11 Hospital Drive, Singapore 169610, Singapore
- Duke-NUS Medical School, 8 College Road, 169857, Singapore
- Institute of Molecular and Cell Biology, A*Star, 61 Biopolis Drive Proteos, 138673, Singapore
| | - Chwee Teck Lim
- Department of Biomedical Engineering, National University of Singapore, 4 Engineering Drive 3, Block E4, #04-08, Singapore 117583, Singapore
- Mechanobiology Institute of Singapore, 5A Engineering Drive 1, Singapore 117411, Singapore
| |
Collapse
|
42
|
High-Throughput Single-Cell Cultivation on Microfluidic Streak Plates. Appl Environ Microbiol 2016; 82:2210-8. [PMID: 26850294 DOI: 10.1128/aem.03588-15] [Citation(s) in RCA: 113] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2015] [Accepted: 01/19/2016] [Indexed: 12/16/2022] Open
Abstract
This paper describes the microfluidic streak plate (MSP), a facile method for high-throughput microbial cell separation and cultivation in nanoliter sessile droplets. The MSP method builds upon the conventional streak plate technique by using microfluidic devices to generate nanoliter droplets that can be streaked manually or robotically onto petri dishes prefilled with carrier oil for cultivation of single cells. In addition, chemical gradients could be encoded in the droplet array for comprehensive dose-response analysis. The MSP method was validated by using single-cell isolation of Escherichia coli and antimicrobial susceptibility testing of Pseudomonas aeruginosa PAO1. The robustness of the MSP work flow was demonstrated by cultivating a soil community that degrades polycyclic aromatic hydrocarbons. Cultivation in droplets enabled detection of the richest species diversity with better coverage of rare species. Moreover, isolation and cultivation of bacterial strains by MSP led to the discovery of several species with high degradation efficiency, including four Mycobacterium isolates and a previously unknown fluoranthene-degrading Blastococcus species.
Collapse
|
43
|
Roberfroid S, Vanderleyden J, Steenackers H. Gene expression variability in clonal populations: Causes and consequences. Crit Rev Microbiol 2016; 42:969-84. [PMID: 26731119 DOI: 10.3109/1040841x.2015.1122571] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
During the last decade it has been shown that among cell variation in gene expression plays an important role within clonal populations. Here, we provide an overview of the different mechanisms contributing to gene expression variability in clonal populations. These are ranging from inherent variations in the biochemical process of gene expression itself, such as intrinsic noise, extrinsic noise and bistability to individual responses to variations in the local micro-environment, a phenomenon called phenotypic plasticity. Also genotypic variations caused by clonal evolution and phase variation can contribute to gene expression variability. Consequently, gene expression studies need to take these fluctuations in expression into account. However, frequently used techniques for expression quantification, such as microarrays, RNA sequencing, quantitative PCR and gene reporter fusions classically determine the population average of gene expression. Here, we discuss how these techniques can be adapted towards single cell analysis by integration with single cell isolation, RNA amplification and microscopy. Alternatively more qualitative selection-based techniques, such as mutant screenings, in vivo expression technology (IVET) and recombination-based IVET (RIVET) can be applied for detection of genes expressed only within a subpopulation. Finally, differential fluorescence induction (DFI), a protocol specially designed for single cell expression is discussed.
Collapse
Affiliation(s)
- Stefanie Roberfroid
- a Department of Microbial and Molecular Systems , Centre of Microbial and Plant Genetics, KU Leuven , Leuven , Belgium
| | - Jos Vanderleyden
- a Department of Microbial and Molecular Systems , Centre of Microbial and Plant Genetics, KU Leuven , Leuven , Belgium
| | - Hans Steenackers
- a Department of Microbial and Molecular Systems , Centre of Microbial and Plant Genetics, KU Leuven , Leuven , Belgium
| |
Collapse
|
44
|
Affiliation(s)
- Sanjin Hosic
- Department of Chemical Engineering, Northeastern University, Boston, MA, USA
| | - Shashi K. Murthy
- Department of Chemical Engineering, Northeastern University, Boston, MA, USA
- Barnett Institute of Chemical and Biological Analysis, Northeastern University, Boston, MA, USA
| | - Abigail N. Koppes
- Department of Chemical Engineering, Northeastern University, Boston, MA, USA
| |
Collapse
|
45
|
ZIFA: Dimensionality reduction for zero-inflated single-cell gene expression analysis. Genome Biol 2015; 16:241. [PMID: 26527291 PMCID: PMC4630968 DOI: 10.1186/s13059-015-0805-z] [Citation(s) in RCA: 363] [Impact Index Per Article: 36.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2015] [Accepted: 10/14/2015] [Indexed: 11/21/2022] Open
Abstract
Single-cell RNA-seq data allows insight into normal cellular function and various disease states through molecular characterization of gene expression on the single cell level. Dimensionality reduction of such high-dimensional data sets is essential for visualization and analysis, but single-cell RNA-seq data are challenging for classical dimensionality-reduction methods because of the prevalence of dropout events, which lead to zero-inflated data. Here, we develop a dimensionality-reduction method, (Z)ero (I)nflated (F)actor (A)nalysis (ZIFA), which explicitly models the dropout characteristics, and show that it improves modeling accuracy on simulated and biological data sets.
Collapse
|
46
|
Hara Y. Tempo and mode of genomic mutations unveil human evolutionary history. Genes Genet Syst 2015; 90:123-31. [PMID: 26510567 DOI: 10.1266/ggs.90.123] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
Mutations that have occurred in human genomes provide insight into various aspects of evolutionary history such as speciation events and degrees of natural selection. Comparing genome sequences between human and great apes or among humans is a feasible approach for inferring human evolutionary history. Recent advances in high-throughput or so-called 'next-generation' DNA sequencing technologies have enabled the sequencing of thousands of individual human genomes, as well as a variety of reference genomes of hominids, many of which are publicly available. These sequence data can help to unveil the detailed demographic history of the lineage leading to humans as well as the explosion of modern human population size in the last several thousand years. In addition, high-throughput sequencing illustrates the tempo and mode of de novo mutations, which are producing human genetic variation at this moment. Pedigree-based human genome sequencing has shown that mutation rates vary significantly across the human genome. These studies have also provided an improved timescale of human evolution, because the mutation rate estimated from pedigree analysis is half that estimated from traditional analyses based on molecular phylogeny. Because of the dramatic reduction in sequencing cost, sequencing on-demand samples designed for specific studies is now also becoming popular. To produce data of sufficient quality to meet the requirements of the study, it is necessary to set an explicit sequencing plan that includes the choice of sample collection methods, sequencing platforms, and number of sequence reads.
Collapse
Affiliation(s)
- Yuichiro Hara
- Phyloinformatics Unit, RIKEN Center for Life Science Technologies
| |
Collapse
|
47
|
Davison M, Hall E, Zare R, Bhaya D. Challenges of metagenomics and single-cell genomics approaches for exploring cyanobacterial diversity. PHOTOSYNTHESIS RESEARCH 2015; 126:135-146. [PMID: 25515769 DOI: 10.1007/s11120-014-0066-9] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/27/2014] [Accepted: 12/10/2014] [Indexed: 06/04/2023]
Abstract
Cyanobacteria have played a crucial role in the history of early earth and continue to be instrumental in shaping our planet, yet applications of cutting edge technology have not yet been widely used to explore cyanobacterial diversity. To provide adequate background, we briefly review current sequencing technologies and their innovative uses in genomics and metagenomics. Next, we focus on current cell capture technologies and the challenges of using them with cyanobacteria. We illustrate the utility in coupling breakthroughs in DNA amplification with cell capture platforms, with an example of microfluidic isolation and subsequent targeted amplicon sequencing from individual terrestrial thermophilic cyanobacteria. Single cells of thermophilic, unicellular Synechococcus sp. JA-2-3-B'a(2-13) (Syn OS-B') were sorted in a microfluidic device, lysed, and subjected to whole genome amplification by multiple displacement amplification. We amplified regions from specific CRISPR spacer arrays, which are known to be highly diverse, contain semi-palindromic repeats which form secondary structure, and can be difficult to amplify. Cell capture, lysis, and genome amplification on a microfluidic device have been optimized, setting a stage for further investigations of individual cyanobacterial cells isolated directly from natural populations.
Collapse
Affiliation(s)
- Michelle Davison
- Department of Plant Biology, Carnegie Institution of Science, 260 Panama Street, Stanford, CA, 94305, USA.
| | - Eric Hall
- SRI International, 333 Ravenswood Ave, Menlo Park, CA, 94025, USA
| | - Richard Zare
- Department of Chemistry, Stanford University, 333 Campus Drive Mudd Building, Room 121, Stanford, CA, 94305-4401, USA
| | - Devaki Bhaya
- Department of Plant Biology, Carnegie Institution of Science, 260 Panama Street, Stanford, CA, 94305, USA
| |
Collapse
|
48
|
Morinishi LS, Blainey P. Simple Bulk Readout of Digital Nucleic Acid Quantification Assays. J Vis Exp 2015. [PMID: 26436576 DOI: 10.3791/52925] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023] Open
Abstract
Digital assays are powerful methods that enable detection of rare cells and counting of individual nucleic acid molecules. However, digital assays are still not routinely applied, due to the cost and specific equipment associated with commercially available methods. Here we present a simplified method for readout of digital droplet assays using a conventional real-time PCR instrument to measure bulk fluorescence of droplet-based digital assays. We characterize the performance of the bulk readout assay using synthetic droplet mixtures and a droplet digital multiple displacement amplification (MDA) assay. Quantitative MDA particularly benefits from a digital reaction format, but our new method applies to any digital assay. For established digital assay protocols such as digital PCR, this method serves to speed up and simplify assay readout. Our bulk readout methodology brings the advantages of partitioned assays without the need for specialized readout instrumentation. The principal limitations of the bulk readout methodology are reduced dynamic range compared with droplet-counting platforms and the need for a standard sample, although the requirements for this standard are less demanding than for a conventional real-time experiment. Quantitative whole genome amplification (WGA) is used to test for contaminants in WGA reactions and is the most sensitive way to detect the presence of DNA fragments with unknown sequences, giving the method great promise in diverse application areas including pharmaceutical quality control and astrobiology.
Collapse
Affiliation(s)
| | - Paul Blainey
- Department of Biological Engineering, Massachusetts Institute of Technology;
| |
Collapse
|
49
|
Gross A, Schoendube J, Zimmermann S, Steeb M, Zengerle R, Koltay P. Technologies for Single-Cell Isolation. Int J Mol Sci 2015; 16:16897-919. [PMID: 26213926 PMCID: PMC4581176 DOI: 10.3390/ijms160816897] [Citation(s) in RCA: 289] [Impact Index Per Article: 28.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2015] [Revised: 07/06/2015] [Accepted: 07/14/2015] [Indexed: 11/16/2022] Open
Abstract
The handling of single cells is of great importance in applications such as cell line development or single-cell analysis, e.g., for cancer research or for emerging diagnostic methods. This review provides an overview of technologies that are currently used or in development to isolate single cells for subsequent single-cell analysis. Data from a dedicated online market survey conducted to identify the most relevant technologies, presented here for the first time, shows that FACS (fluorescence activated cell sorting) respectively Flow cytometry (33% usage), laser microdissection (17%), manual cell picking (17%), random seeding/dilution (15%), and microfluidics/lab-on-a-chip devices (12%) are currently the most frequently used technologies. These most prominent technologies are described in detail and key performance factors are discussed. The survey data indicates a further increasing interest in single-cell isolation tools for the coming years. Additionally, a worldwide patent search was performed to screen for emerging technologies that might become relevant in the future. In total 179 patents were found, out of which 25 were evaluated by screening the title and abstract to be relevant to the field.
Collapse
Affiliation(s)
- Andre Gross
- Laboratory for MEMS Applications, IMTEK-Department of Microsystems Engineering, University of Freiburg, Georges-Koehler-Allee 103, Freiburg 79110, Germany.
- Cytena GmbH, Georges-Koehler-Allee 103, Freiburg 79110, Germany.
| | - Jonas Schoendube
- Laboratory for MEMS Applications, IMTEK-Department of Microsystems Engineering, University of Freiburg, Georges-Koehler-Allee 103, Freiburg 79110, Germany.
- Cytena GmbH, Georges-Koehler-Allee 103, Freiburg 79110, Germany.
| | - Stefan Zimmermann
- Laboratory for MEMS Applications, IMTEK-Department of Microsystems Engineering, University of Freiburg, Georges-Koehler-Allee 103, Freiburg 79110, Germany.
| | - Maximilian Steeb
- Laboratory for MEMS Applications, IMTEK-Department of Microsystems Engineering, University of Freiburg, Georges-Koehler-Allee 103, Freiburg 79110, Germany.
| | - Roland Zengerle
- Laboratory for MEMS Applications, IMTEK-Department of Microsystems Engineering, University of Freiburg, Georges-Koehler-Allee 103, Freiburg 79110, Germany.
- Hahn-Schickard, Georges-Koehler-Allee 103, Freiburg 79110, Germany.
- BIOSS-Centre for Biological Signalling Studies, University of Freiburg, Freiburg 79110, Germany.
| | - Peter Koltay
- Laboratory for MEMS Applications, IMTEK-Department of Microsystems Engineering, University of Freiburg, Georges-Koehler-Allee 103, Freiburg 79110, Germany.
- Cytena GmbH, Georges-Koehler-Allee 103, Freiburg 79110, Germany.
| |
Collapse
|
50
|
Korem Y, Szekely P, Hart Y, Sheftel H, Hausser J, Mayo A, Rothenberg ME, Kalisky T, Alon U. Geometry of the Gene Expression Space of Individual Cells. PLoS Comput Biol 2015; 11:e1004224. [PMID: 26161936 PMCID: PMC4498931 DOI: 10.1371/journal.pcbi.1004224] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2014] [Accepted: 03/04/2015] [Indexed: 12/14/2022] Open
Abstract
There is a revolution in the ability to analyze gene expression of single cells in a tissue. To understand this data we must comprehend how cells are distributed in a high-dimensional gene expression space. One open question is whether cell types form discrete clusters or whether gene expression forms a continuum of states. If such a continuum exists, what is its geometry? Recent theory on evolutionary trade-offs suggests that cells that need to perform multiple tasks are arranged in a polygon or polyhedron (line, triangle, tetrahedron and so on, generally called polytopes) in gene expression space, whose vertices are the expression profiles optimal for each task. Here, we analyze single-cell data from human and mouse tissues profiled using a variety of single-cell technologies. We fit the data to shapes with different numbers of vertices, compute their statistical significance, and infer their tasks. We find cases in which single cells fill out a continuum of expression states within a polyhedron. This occurs in intestinal progenitor cells, which fill out a tetrahedron in gene expression space. The four vertices of this tetrahedron are each enriched with genes for a specific task related to stemness and early differentiation. A polyhedral continuum of states is also found in spleen dendritic cells, known to perform multiple immune tasks: cells fill out a tetrahedron whose vertices correspond to key tasks related to maturation, pathogen sensing and communication with lymphocytes. A mixture of continuum-like distributions and discrete clusters is found in other cell types, including bone marrow and differentiated intestinal crypt cells. This approach can be used to understand the geometry and biological tasks of a wide range of single-cell datasets. The present results suggest that the concept of cell type may be expanded. In addition to discreet clusters in gene-expression space, we suggest a new possibility: a continuum of states within a polyhedron, in which the vertices represent specialists at key tasks. In the past, biological experiments usually pooled together millions of cells, masking the differences between individual cells. Current technology takes a big step forward by measuring gene expression from individual cells. Interpreting this data is challenging because we need to understand how cells are arranged in a high dimensional gene expression space. Here we test recent theory that suggests that cells facing multiple tasks should be arranged in simple low dimensional polygons or polyhedra (generally called polytopes). The vertices of the polytopes are gene expression profiles optimal for each of the tasks. We find evidence for such simplicity in a variety of tissues—spleen, bone marrow, intestine—analyzed by different single-cell technologies. We find that cells are distributed inside polytopes, such as tetrahedrons or four-dimensional simplexes, with cells closest to each vertex responsible for a different key task. For example, intestinal progenitor cells that give rise to the other cell types show a continuous distribution in a tetrahedron whose vertices correspond to several key sub-tasks. Immune dendritic cells likewise are continuously distributed between key immune tasks. This approach of testing whether data falls in polytopes may be useful for interpreting a variety of single-cell datasets in terms of biological tasks.
Collapse
Affiliation(s)
- Yael Korem
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, Israel
| | - Pablo Szekely
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, Israel
| | - Yuval Hart
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, Israel
| | - Hila Sheftel
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, Israel
| | - Jean Hausser
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, Israel
| | - Avi Mayo
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, Israel
| | - Michael E. Rothenberg
- Department of Medicine, Division of Gastroenterology and Hepatology, and Institute for Stem Cell Biology and Regenerative Medicine, Stanford University, Stanford, California, United States of America
| | - Tomer Kalisky
- Faculty of Engineering, Bar Ilan University, Ramat Gan, Israel
| | - Uri Alon
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, Israel
- * E-mail:
| |
Collapse
|