351
|
Chen J, Rénia L, Ginhoux F. Constructing cell lineages from single-cell transcriptomes. Mol Aspects Med 2017; 59:95-113. [PMID: 29107741 DOI: 10.1016/j.mam.2017.10.004] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2017] [Revised: 10/23/2017] [Accepted: 10/25/2017] [Indexed: 12/25/2022]
Abstract
Advances in single-cell RNA-sequencing have helped reveal the previously underappreciated level of cellular heterogeneity present during cellular differentiation. A static snapshot of single-cell transcriptomes provides a good representation of the various stages of differentiation as differentiation is rarely synchronized between cells. Data from numerous single-cell analyses has suggested that cellular differentiation and development can be conceptualized as continuous processes. Consequently, computational algorithms have been developed to infer lineage relationships between cell types and construct developmental trajectories along which cells are re-ordered such that similarity between successive cell pairs is maximized. Here, we compare and contrast the existing computational methods, and illustrate how they may be applied to build mouse myeloid progenitor lineages from massively parallel RNA single-cell sequencing data.
Collapse
Affiliation(s)
- Jinmiao Chen
- Singapore Immunology Network (SIgN), A*STAR, 8A Biomedical Grove, Immunos Building, Level 4, Singapore 138648, Singapore.
| | - Laurent Rénia
- Singapore Immunology Network (SIgN), A*STAR, 8A Biomedical Grove, Immunos Building, Level 4, Singapore 138648, Singapore
| | - Florent Ginhoux
- Singapore Immunology Network (SIgN), A*STAR, 8A Biomedical Grove, Immunos Building, Level 4, Singapore 138648, Singapore
| |
Collapse
|
352
|
van Unen V, Höllt T, Pezzotti N, Li N, Reinders MJT, Eisemann E, Koning F, Vilanova A, Lelieveldt BPF. Visual analysis of mass cytometry data by hierarchical stochastic neighbour embedding reveals rare cell types. Nat Commun 2017; 8:1740. [PMID: 29170529 PMCID: PMC5700955 DOI: 10.1038/s41467-017-01689-9] [Citation(s) in RCA: 149] [Impact Index Per Article: 18.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2017] [Accepted: 09/25/2017] [Indexed: 12/17/2022] Open
Abstract
Mass cytometry allows high-resolution dissection of the cellular composition of the immune system. However, the high-dimensionality, large size, and non-linear structure of the data poses considerable challenges for the data analysis. In particular, dimensionality reduction-based techniques like t-SNE offer single-cell resolution but are limited in the number of cells that can be analyzed. Here we introduce Hierarchical Stochastic Neighbor Embedding (HSNE) for the analysis of mass cytometry data sets. HSNE constructs a hierarchy of non-linear similarities that can be interactively explored with a stepwise increase in detail up to the single-cell level. We apply HSNE to a study on gastrointestinal disorders and three other available mass cytometry data sets. We find that HSNE efficiently replicates previous observations and identifies rare cell populations that were previously missed due to downsampling. Thus, HSNE removes the scalability limit of conventional t-SNE analysis, a feature that makes it highly suitable for the analysis of massive high-dimensional data sets.
Collapse
Affiliation(s)
- Vincent van Unen
- Department of Immunohematology and Blood Transfusion, Leiden University Medical Center, Albinusdreef 2, 2333, ZA, Leiden, The Netherlands.
| | - Thomas Höllt
- Computer Graphics and Visualization Group, Delft University of Technology, Mekelweg 4, 2628, CD, Delft, The Netherlands
- Computational Biology Center, Leiden University Medical Center, Albinusdreef 2, 2333, ZA, Leiden, The Netherlands
| | - Nicola Pezzotti
- Computer Graphics and Visualization Group, Delft University of Technology, Mekelweg 4, 2628, CD, Delft, The Netherlands
| | - Na Li
- Department of Immunohematology and Blood Transfusion, Leiden University Medical Center, Albinusdreef 2, 2333, ZA, Leiden, The Netherlands
| | - Marcel J T Reinders
- Pattern Recognition and Bioinformatics Group, Delft University of Technology, Mekelweg 4, 2628, CD, Delft, The Netherlands
| | - Elmar Eisemann
- Computer Graphics and Visualization Group, Delft University of Technology, Mekelweg 4, 2628, CD, Delft, The Netherlands
| | - Frits Koning
- Department of Immunohematology and Blood Transfusion, Leiden University Medical Center, Albinusdreef 2, 2333, ZA, Leiden, The Netherlands
| | - Anna Vilanova
- Computer Graphics and Visualization Group, Delft University of Technology, Mekelweg 4, 2628, CD, Delft, The Netherlands
| | - Boudewijn P F Lelieveldt
- Pattern Recognition and Bioinformatics Group, Delft University of Technology, Mekelweg 4, 2628, CD, Delft, The Netherlands.
- Division of Image Processing, Department of Radiology, Leiden University Medical Center, Albinusdreef 2, 2333, ZA, Leiden, The Netherlands.
| |
Collapse
|
353
|
Haber AL, Biton M, Rogel N, Herbst RH, Shekhar K, Smillie C, Burgin G, Delorey TM, Howitt MR, Katz Y, Tirosh I, Beyaz S, Dionne D, Zhang M, Raychowdhury R, Garrett WS, Rozenblatt-Rosen O, Shi HN, Yilmaz O, Xavier RJ, Regev A. A single-cell survey of the small intestinal epithelium. Nature 2017; 551:333-339. [PMID: 29144463 PMCID: PMC6022292 DOI: 10.1038/nature24489] [Citation(s) in RCA: 1161] [Impact Index Per Article: 145.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2016] [Accepted: 10/03/2017] [Indexed: 12/17/2022]
Abstract
Intestinal epithelial cells absorb nutrients, respond to microbes, function as a barrier and help to coordinate immune responses. Here we report profiling of 53,193 individual epithelial cells from the small intestine and organoids of mice, which enabled the identification and characterization of previously unknown subtypes of intestinal epithelial cell and their gene signatures. We found unexpected diversity in hormone-secreting enteroendocrine cells and constructed the taxonomy of newly identified subtypes, and distinguished between two subtypes of tuft cell, one of which expresses the epithelial cytokine Tslp and the pan-immune marker CD45, which was not previously associated with non-haematopoietic cells. We also characterized the ways in which cell-intrinsic states and the proportions of different cell types respond to bacterial and helminth infections: Salmonella infection caused an increase in the abundance of Paneth cells and enterocytes, and broad activation of an antimicrobial program; Heligmosomoides polygyrus caused an increase in the abundance of goblet and tuft cells. Our survey highlights previously unidentified markers and programs, associates sensory molecules with cell types, and uncovers principles of gut homeostasis and response to pathogens.
Collapse
Affiliation(s)
- Adam L Haber
- Broad Institute of Harvard and Massachusetts Institute of Technology, Cambridge, Massachusetts 02142, USA
| | - Moshe Biton
- Broad Institute of Harvard and Massachusetts Institute of Technology, Cambridge, Massachusetts 02142, USA
- Center for Computational and Integrative Biology, Massachusetts General Hospital, Boston, Massachusetts 02114, USA
| | - Noga Rogel
- Broad Institute of Harvard and Massachusetts Institute of Technology, Cambridge, Massachusetts 02142, USA
| | - Rebecca H Herbst
- Broad Institute of Harvard and Massachusetts Institute of Technology, Cambridge, Massachusetts 02142, USA
- Department of Systems Biology, Harvard Medical School, Boston, Massachusetts 02114, USA
| | - Karthik Shekhar
- Broad Institute of Harvard and Massachusetts Institute of Technology, Cambridge, Massachusetts 02142, USA
| | - Christopher Smillie
- Broad Institute of Harvard and Massachusetts Institute of Technology, Cambridge, Massachusetts 02142, USA
| | - Grace Burgin
- Broad Institute of Harvard and Massachusetts Institute of Technology, Cambridge, Massachusetts 02142, USA
| | - Toni M Delorey
- Broad Institute of Harvard and Massachusetts Institute of Technology, Cambridge, Massachusetts 02142, USA
- Department of Biology and Biotechnology, Worcester Polytechnic Institute, Worcester, Massachusetts 01609, USA
| | - Michael R Howitt
- Departments of Immunology and Infectious Diseases and Genetics and Complex Diseases, Harvard T. H. Chan School of Public Health, Boston, Massachusetts 02115, USA
| | - Yarden Katz
- Department of Systems Biology, Harvard Medical School, Boston, Massachusetts 02114, USA
| | - Itay Tirosh
- Broad Institute of Harvard and Massachusetts Institute of Technology, Cambridge, Massachusetts 02142, USA
| | - Semir Beyaz
- The David H. Koch Institute for Integrative Cancer Research at Massachusetts Institute of Technology, Department of Biology, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, USA
- Division of Hematology/Oncology, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts 02115, USA
- Department of Pediatric Oncology, Dana-Farber Cancer Institute, Howard Hughes Medical Institute, Harvard Stem Cell Institute, Harvard Medical School, Boston, Massachusetts 02115, USA
| | - Danielle Dionne
- Broad Institute of Harvard and Massachusetts Institute of Technology, Cambridge, Massachusetts 02142, USA
| | - Mei Zhang
- Mucosal Immunology and Biology Research Center, Massachusetts General Hospital and Harvard Medical School, Charlestown, Massachusetts 02129, USA
| | - Raktima Raychowdhury
- Broad Institute of Harvard and Massachusetts Institute of Technology, Cambridge, Massachusetts 02142, USA
| | - Wendy S Garrett
- Broad Institute of Harvard and Massachusetts Institute of Technology, Cambridge, Massachusetts 02142, USA
- Departments of Immunology and Infectious Diseases and Genetics and Complex Diseases, Harvard T. H. Chan School of Public Health, Boston, Massachusetts 02115, USA
| | - Orit Rozenblatt-Rosen
- Broad Institute of Harvard and Massachusetts Institute of Technology, Cambridge, Massachusetts 02142, USA
| | - Hai Ning Shi
- Mucosal Immunology and Biology Research Center, Massachusetts General Hospital and Harvard Medical School, Charlestown, Massachusetts 02129, USA
| | - Omer Yilmaz
- Broad Institute of Harvard and Massachusetts Institute of Technology, Cambridge, Massachusetts 02142, USA
- The David H. Koch Institute for Integrative Cancer Research at Massachusetts Institute of Technology, Department of Biology, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, USA
- Departments of Pathology, Gastroenterology, and Surgery, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts 02114, USA
| | - Ramnik J Xavier
- Broad Institute of Harvard and Massachusetts Institute of Technology, Cambridge, Massachusetts 02142, USA
- Center for Computational and Integrative Biology, Massachusetts General Hospital, Boston, Massachusetts 02114, USA
- Gastrointestinal Unit and Center for the Study of Inflammatory Bowel Disease, Massachusetts General Hospital, Boston, Massachusetts 02114, USA
| | - Aviv Regev
- Broad Institute of Harvard and Massachusetts Institute of Technology, Cambridge, Massachusetts 02142, USA
- Department of Biology, Howard Hughes Medical Institute, Massachusetts Institute of Technology, Cambridge, Massachusetts 02140, USA
| |
Collapse
|
354
|
Herring CA, Banerjee A, McKinley ET, Simmons AJ, Ping J, Roland JT, Franklin JL, Liu Q, Gerdes MJ, Coffey RJ, Lau KS. Unsupervised Trajectory Analysis of Single-Cell RNA-Seq and Imaging Data Reveals Alternative Tuft Cell Origins in the Gut. Cell Syst 2017; 6:37-51.e9. [PMID: 29153838 DOI: 10.1016/j.cels.2017.10.012] [Citation(s) in RCA: 135] [Impact Index Per Article: 16.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2017] [Revised: 08/17/2017] [Accepted: 10/13/2017] [Indexed: 12/19/2022]
Abstract
Modern single-cell technologies allow multiplexed sampling of cellular states within a tissue. However, computational tools that can infer developmental cell-state transitions reproducibly from such single-cell data are lacking. Here, we introduce p-Creode, an unsupervised algorithm that produces multi-branching graphs from single-cell data, compares graphs with differing topologies, and infers a statistically robust hierarchy of cell-state transitions that define developmental trajectories. We have applied p-Creode to mass cytometry, multiplex immunofluorescence, and single-cell RNA-seq data. As a test case, we validate cell-state-transition trajectories predicted by p-Creode for intestinal tuft cells, a rare, chemosensory cell type. We clarify that tuft cells are specified outside of the Atoh1-dependent secretory lineage in the small intestine. However, p-Creode also predicts, and we confirm, that tuft cells arise from an alternative, Atoh1-driven developmental program in the colon. These studies introduce p-Creode as a reliable method for analyzing large datasets that depict branching transition trajectories.
Collapse
Affiliation(s)
- Charles A Herring
- Epithelial Biology Center, Vanderbilt University Medical Center, 2213 Garland Avenue, 10475 MRB IV, Nashville, TN 37232, USA; Program in Chemical and Physical Biology, Vanderbilt University School of Medicine, Nashville, TN 37232, USA
| | - Amrita Banerjee
- Epithelial Biology Center, Vanderbilt University Medical Center, 2213 Garland Avenue, 10475 MRB IV, Nashville, TN 37232, USA; Department of Cell and Developmental Biology, Vanderbilt University School of Medicine, Nashville, TN 37232, USA
| | - Eliot T McKinley
- Epithelial Biology Center, Vanderbilt University Medical Center, 2213 Garland Avenue, 10475 MRB IV, Nashville, TN 37232, USA; Department of Medicine, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Alan J Simmons
- Epithelial Biology Center, Vanderbilt University Medical Center, 2213 Garland Avenue, 10475 MRB IV, Nashville, TN 37232, USA; Department of Cell and Developmental Biology, Vanderbilt University School of Medicine, Nashville, TN 37232, USA
| | - Jie Ping
- Department of Biomedical Informatics, Vanderbilt University School of Medicine, Nashville, TN 37232, USA; Center for Quantitative Sciences, Vanderbilt University School of Medicine, Nashville, TN 37232, USA
| | - Joseph T Roland
- Epithelial Biology Center, Vanderbilt University Medical Center, 2213 Garland Avenue, 10475 MRB IV, Nashville, TN 37232, USA
| | - Jeffrey L Franklin
- Epithelial Biology Center, Vanderbilt University Medical Center, 2213 Garland Avenue, 10475 MRB IV, Nashville, TN 37232, USA; Department of Cell and Developmental Biology, Vanderbilt University School of Medicine, Nashville, TN 37232, USA
| | - Qi Liu
- Department of Biomedical Informatics, Vanderbilt University School of Medicine, Nashville, TN 37232, USA; Center for Quantitative Sciences, Vanderbilt University School of Medicine, Nashville, TN 37232, USA
| | - Michael J Gerdes
- Life Sciences Division, GE Global Research, Niskayuna, NY 12309, USA
| | - Robert J Coffey
- Epithelial Biology Center, Vanderbilt University Medical Center, 2213 Garland Avenue, 10475 MRB IV, Nashville, TN 37232, USA; Department of Cell and Developmental Biology, Vanderbilt University School of Medicine, Nashville, TN 37232, USA; Department of Medicine, Vanderbilt University Medical Center, Nashville, TN 37232, USA; Veterans Affairs Medical Center, Tennessee Valley Healthcare System, Nashville, TN 37232, USA
| | - Ken S Lau
- Epithelial Biology Center, Vanderbilt University Medical Center, 2213 Garland Avenue, 10475 MRB IV, Nashville, TN 37232, USA; Program in Chemical and Physical Biology, Vanderbilt University School of Medicine, Nashville, TN 37232, USA; Department of Cell and Developmental Biology, Vanderbilt University School of Medicine, Nashville, TN 37232, USA; Center for Quantitative Sciences, Vanderbilt University School of Medicine, Nashville, TN 37232, USA.
| |
Collapse
|
355
|
Abstract
Purpose of review Hematopoietic stem cell (HSC) transplantation has yielded tremendous information on experimental properties of HSCs. Yet, it remains unclear whether transplantation reflects the physiology of hematopoiesis. A limitation is the difficulty in accessing HSC functions without isolation, in-vitro manipulation and readout for potential. New genetic fate mapping and clonal marking techniques now shed light on hematopoiesis under physiological conditions. Recent findings Transposon-based genetic marks were introduced across the entire hematopoietic system to follow the clonal dynamics of these tags over time. A polyclonal source downstream from stem cells was found responsible for the production of at least granulocytes. In independent experiments, HSCs were genetically marked in adult mice, and the kinetics of label emergence throughout the system was followed over time. These experiments uncovered that during physiological steady-state hematopoiesis large numbers of HSCs yield differentiated progeny. Individual HSCs were active only rarely, indicating their very slow periodicity of differentiation rather than quiescence. Summary Noninvasive genetic experiments in mice have identified a major role of stem and progenitor cells downstream from HSCs as drivers of adult hematopoiesis, and revealed that post-transplantation hematopoiesis differs quantitatively from normal steady-state hematopoiesis.
Collapse
|
356
|
Chlis NK, Wolf FA, Theis FJ. Model-based branching point detection in single-cell data by K-branches clustering. Bioinformatics 2017; 33:3211-3219. [PMID: 28582478 PMCID: PMC5860029 DOI: 10.1093/bioinformatics/btx325] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2016] [Revised: 04/11/2017] [Accepted: 05/31/2017] [Indexed: 11/14/2022] Open
Abstract
MOTIVATION The identification of heterogeneities in cell populations by utilizing single-cell technologies such as single-cell RNA-Seq, enables inference of cellular development and lineage trees. Several methods have been proposed for such inference from high-dimensional single-cell data. They typically assign each cell to a branch in a differentiation trajectory. However, they commonly assume specific geometries such as tree-like developmental hierarchies and lack statistically sound methods to decide on the number of branching events. RESULTS We present K-Branches, a solution to the above problem by locally fitting half-lines to single-cell data, introducing a clustering algorithm similar to K-Means. These halflines are proxies for branches in the differentiation trajectory of cells. We propose a modified version of the GAP statistic for model selection, in order to decide on the number of lines that best describe the data locally. In this manner, we identify the location and number of subgroups of cells that are associated with branching events and full differentiation, respectively. We evaluate the performance of our method on single-cell RNA-Seq data describing the differentiation of myeloid progenitors during hematopoiesis, single-cell qPCR data of mouse blastocyst development, single-cell qPCR data of human myeloid monocytic leukemia and artificial data. AVAILABILITY AND IMPLEMENTATION An R implementation of K-Branches is freely available at https://github.com/theislab/kbranches. CONTACT fabian.theis@helmholtz-muenchen.de. SUPPLEMENTARY INFORMATION Supplementary data are available at Bioinformatics online.
Collapse
Affiliation(s)
- Nikolaos K Chlis
- Institute of Computational Biology, Helmholtz Zentrum München, Neuherberg, Germany
- School of Life Sciences Weihenstephan, Technical University of Munich, Freising, Germany
| | - F Alexander Wolf
- Institute of Computational Biology, Helmholtz Zentrum München, Neuherberg, Germany
| | - Fabian J Theis
- Institute of Computational Biology, Helmholtz Zentrum München, Neuherberg, Germany
- School of Life Sciences Weihenstephan, Technical University of Munich, Freising, Germany
- Department of Mathematics, Technical University of Munich, Garching, Germany
| |
Collapse
|
357
|
Su Y, Shi Q, Wei W. Single cell proteomics in biomedicine: High-dimensional data acquisition, visualization, and analysis. Proteomics 2017; 17. [PMID: 28128880 DOI: 10.1002/pmic.201600267] [Citation(s) in RCA: 65] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2016] [Revised: 01/20/2017] [Accepted: 01/23/2017] [Indexed: 11/11/2022]
Abstract
New insights on cellular heterogeneity in the last decade provoke the development of a variety of single cell omics tools at a lightning pace. The resultant high-dimensional single cell data generated by these tools require new theoretical approaches and analytical algorithms for effective visualization and interpretation. In this review, we briefly survey the state-of-the-art single cell proteomic tools with a particular focus on data acquisition and quantification, followed by an elaboration of a number of statistical and computational approaches developed to date for dissecting the high-dimensional single cell data. The underlying assumptions, unique features, and limitations of the analytical methods with the designated biological questions they seek to answer will be discussed. Particular attention will be given to those information theoretical approaches that are anchored in a set of first principles of physics and can yield detailed (and often surprising) predictions.
Collapse
Affiliation(s)
- Yapeng Su
- NanoSystems Biology Cancer Center, Division of Chemistry and Chemical Engineering, California Institute of Technology, Pasadena, CA, USA
| | - Qihui Shi
- Key Laboratory of Systems Biomedicine (Ministry of Education), School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, China
| | - Wei Wei
- NanoSystems Biology Cancer Center, Division of Chemistry and Chemical Engineering, California Institute of Technology, Pasadena, CA, USA.,Department of Molecular and Medical Pharmacology, David Geffen School of Medicine, University of California - Los Angeles, Los Angeles, CA, USA
| |
Collapse
|
358
|
Loughran SJ, Comoglio F, Hamey FK, Giustacchini A, Errami Y, Earp E, Göttgens B, Jacobsen SEW, Mead AJ, Hendrich B, Green AR. Mbd3/NuRD controls lymphoid cell fate and inhibits tumorigenesis by repressing a B cell transcriptional program. J Exp Med 2017; 214:3085-3104. [PMID: 28899870 PMCID: PMC5626393 DOI: 10.1084/jem.20161827] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2016] [Revised: 07/04/2017] [Accepted: 07/25/2017] [Indexed: 02/02/2023] Open
Abstract
Differentiation of lineage-committed cells from multipotent progenitors requires the establishment of accessible chromatin at lineage-specific transcriptional enhancers and promoters, which is mediated by pioneer transcription factors that recruit activating chromatin remodeling complexes. Here we show that the Mbd3/nucleosome remodeling and deacetylation (NuRD) chromatin remodeling complex opposes this transcriptional pioneering during B cell programming of multipotent lymphoid progenitors by restricting chromatin accessibility at B cell enhancers and promoters. Mbd3/NuRD-deficient lymphoid progenitors therefore prematurely activate a B cell transcriptional program and are biased toward overproduction of pro-B cells at the expense of T cell progenitors. The striking reduction in early thymic T cell progenitors results in compensatory hyperproliferation of immature thymocytes and development of T cell lymphoma. Our results reveal that Mbd3/NuRD can regulate multilineage differentiation by constraining the activation of dormant lineage-specific enhancers and promoters. In this way, Mbd3/NuRD protects the multipotency of lymphoid progenitors, preventing B cell-programming transcription factors from prematurely enacting lineage commitment. Mbd3/NuRD therefore controls the fate of lymphoid progenitors, ensuring appropriate production of lineage-committed progeny and suppressing tumor formation.
Collapse
Affiliation(s)
- Stephen J Loughran
- Department of Haematology, Cambridge Institute for Medical Research, University of Cambridge, Cambridge, England, UK
- Wellcome Trust/MRC Stem Cell Institute, University of Cambridge, Cambridge, England, UK
| | - Federico Comoglio
- Department of Haematology, Cambridge Institute for Medical Research, University of Cambridge, Cambridge, England, UK
- Wellcome Trust/MRC Stem Cell Institute, University of Cambridge, Cambridge, England, UK
| | - Fiona K Hamey
- Department of Haematology, Cambridge Institute for Medical Research, University of Cambridge, Cambridge, England, UK
- Wellcome Trust/MRC Stem Cell Institute, University of Cambridge, Cambridge, England, UK
| | - Alice Giustacchini
- MRC Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, England, UK
| | - Youssef Errami
- Department of Haematology, Cambridge Institute for Medical Research, University of Cambridge, Cambridge, England, UK
- Wellcome Trust/MRC Stem Cell Institute, University of Cambridge, Cambridge, England, UK
| | - Eleanor Earp
- Department of Haematology, Cambridge Institute for Medical Research, University of Cambridge, Cambridge, England, UK
- Wellcome Trust/MRC Stem Cell Institute, University of Cambridge, Cambridge, England, UK
| | - Berthold Göttgens
- Department of Haematology, Cambridge Institute for Medical Research, University of Cambridge, Cambridge, England, UK
- Wellcome Trust/MRC Stem Cell Institute, University of Cambridge, Cambridge, England, UK
| | - Sten Eirik W Jacobsen
- MRC Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, England, UK
- Wallenberg Institute for Regenerative Medicine, Department of Cell and Molecular Biology and Department of Medicine Huddinge, Karolinska Institutet and Center for Hematology and Regenerative Medicine, Karolinska University Hospital Huddinge, Stockholm, Sweden
| | - Adam J Mead
- MRC Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, England, UK
| | - Brian Hendrich
- Wellcome Trust/MRC Stem Cell Institute, University of Cambridge, Cambridge, England, UK
- Department of Biochemistry, University of Cambridge, Cambridge, England, UK
| | - Anthony R Green
- Department of Haematology, Cambridge Institute for Medical Research, University of Cambridge, Cambridge, England, UK
- Wellcome Trust/MRC Stem Cell Institute, University of Cambridge, Cambridge, England, UK
| |
Collapse
|
359
|
|
360
|
Kumar P, Tan Y, Cahan P. Understanding development and stem cells using single cell-based analyses of gene expression. Development 2017; 144:17-32. [PMID: 28049689 DOI: 10.1242/dev.133058] [Citation(s) in RCA: 76] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
In recent years, genome-wide profiling approaches have begun to uncover the molecular programs that drive developmental processes. In particular, technical advances that enable genome-wide profiling of thousands of individual cells have provided the tantalizing prospect of cataloging cell type diversity and developmental dynamics in a quantitative and comprehensive manner. Here, we review how single-cell RNA sequencing has provided key insights into mammalian developmental and stem cell biology, emphasizing the analytical approaches that are specific to studying gene expression in single cells.
Collapse
Affiliation(s)
- Pavithra Kumar
- Department of Biomedical Engineering, Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Yuqi Tan
- Department of Biomedical Engineering, Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Patrick Cahan
- Department of Biomedical Engineering, Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| |
Collapse
|
361
|
Sanchez-Castillo M, Blanco D, Tienda-Luna IM, Carrion MC, Huang Y. A Bayesian framework for the inference of gene regulatory networks from time and pseudo-time series data. Bioinformatics 2017; 34:964-970. [DOI: 10.1093/bioinformatics/btx605] [Citation(s) in RCA: 62] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2016] [Accepted: 09/22/2017] [Indexed: 11/12/2022] Open
Affiliation(s)
| | - D Blanco
- Department of Applied Physics, University of Granada, Granada, Spain
| | - I M Tienda-Luna
- Department of Electrical Engineering, University of Granada, Granada, Spain
| | - M C Carrion
- Department of Applied Physics, University of Granada, Granada, Spain
| | - Yufei Huang
- Department of Electrical Engineering, University of Texas at San Antonio, San Antonio, TX, USA
| |
Collapse
|
362
|
Data-analysis strategies for image-based cell profiling. Nat Methods 2017; 14:849-863. [PMID: 28858338 PMCID: PMC6871000 DOI: 10.1038/nmeth.4397] [Citation(s) in RCA: 435] [Impact Index Per Article: 54.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2016] [Accepted: 07/28/2017] [Indexed: 12/16/2022]
Abstract
Image-based cell profiling is a high-throughput strategy for the quantification of phenotypic differences among a variety of cell populations. It paves the way to studying biological systems on a large scale by using chemical and genetic perturbations. The general workflow for this technology involves image acquisition with high-throughput microscopy systems and subsequent image processing and analysis. Here, we introduce the steps required to create high-quality image-based (i.e., morphological) profiles from a collection of microscopy images. We recommend techniques that have proven useful in each stage of the data analysis process, on the basis of the experience of 20 laboratories worldwide that are refining their image-based cell-profiling methodologies in pursuit of biological discovery. The recommended techniques cover alternatives that may suit various biological goals, experimental designs, and laboratories' preferences.
Collapse
|
363
|
Papili Gao N, Ud-Dean SMM, Gandrillon O, Gunawan R. SINCERITIES: inferring gene regulatory networks from time-stamped single cell transcriptional expression profiles. Bioinformatics 2017; 34:258-266. [PMID: 28968704 PMCID: PMC5860204 DOI: 10.1093/bioinformatics/btx575] [Citation(s) in RCA: 126] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2016] [Revised: 06/12/2017] [Accepted: 09/13/2017] [Indexed: 11/13/2022] Open
Abstract
Motivation Single cell transcriptional profiling opens up a new avenue in studying the functional role of cell-to-cell variability in physiological processes. The analysis of single cell expression profiles creates new challenges due to the distributive nature of the data and the stochastic dynamics of gene transcription process. The reconstruction of gene regulatory networks (GRNs) using single cell transcriptional profiles is particularly challenging, especially when directed gene-gene relationships are desired. Results We developed SINCERITIES (SINgle CEll Regularized Inference using TIme-stamped Expression profileS) for the inference of GRNs from single cell transcriptional profiles. We focused on time-stamped cross-sectional expression data, commonly generated from transcriptional profiling of single cells collected at multiple time points after cell stimulation. SINCERITIES recovers directed regulatory relationships among genes by employing regularized linear regression (ridge regression), using temporal changes in the distributions of gene expressions. Meanwhile, the modes of the gene regulations (activation and repression) come from partial correlation analyses between pairs of genes. We demonstrated the efficacy of SINCERITIES in inferring GRNs using in silico time-stamped single cell expression data and single cell transcriptional profiles of THP-1 monocytic human leukemia cells. The case studies showed that SINCERITIES could provide accurate GRN predictions, significantly better than other GRN inference algorithms such as TSNI, GENIE3 and JUMP3. Moreover, SINCERITIES has a low computational complexity and is amenable to problems of extremely large dimensionality. Finally, an application of SINCERITIES to single cell expression data of T2EC chicken erythrocytes pointed to BATF as a candidate novel regulator of erythroid development. Availability and implementation MATLAB and R version of SINCERITIES are freely available from the following websites: http://www.cabsel.ethz.ch/tools/sincerities.html and https://github.com/CABSEL/SINCERITIES. The single cell THP-1 and T2EC transcriptional profiles are available from the original publications (Kouno et al., 2013; Richard et al., 2016). The in silico single cell data are available on SINCERITIES websites. Supplementary information Supplementary data are available at Bioinformatics online.
Collapse
Affiliation(s)
- Nan Papili Gao
- Institute for Chemical and Bioengineering, ETH Zurich, Zurich, Switzerland.,Swiss Institute of Bioinformatics, Lausanne, Switzerland
| | - S M Minhaz Ud-Dean
- Department of Environmental Health Sciences, Mailman School of Public Health, Columbia University, New York, NY, USA
| | - Olivier Gandrillon
- Laboratory of Biology and Modelling of the Cell, Univ Lyon, ENS de Lyon, Univ Claude Bernard, CNRS UMR, INSERM Lyon, France.,Inria Team Dracula, Inria Center Grenoble Rhône-Alpes, Rhône-Alpes, France
| | - Rudiyanto Gunawan
- Institute for Chemical and Bioengineering, ETH Zurich, Zurich, Switzerland.,Swiss Institute of Bioinformatics, Lausanne, Switzerland
| |
Collapse
|
364
|
Haque A, Engel J, Teichmann SA, Lönnberg T. A practical guide to single-cell RNA-sequencing for biomedical research and clinical applications. Genome Med 2017; 9:75. [PMID: 28821273 PMCID: PMC5561556 DOI: 10.1186/s13073-017-0467-4] [Citation(s) in RCA: 626] [Impact Index Per Article: 78.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
RNA sequencing (RNA-seq) is a genomic approach for the detection and quantitative analysis of messenger RNA molecules in a biological sample and is useful for studying cellular responses. RNA-seq has fueled much discovery and innovation in medicine over recent years. For practical reasons, the technique is usually conducted on samples comprising thousands to millions of cells. However, this has hindered direct assessment of the fundamental unit of biology-the cell. Since the first single-cell RNA-sequencing (scRNA-seq) study was published in 2009, many more have been conducted, mostly by specialist laboratories with unique skills in wet-lab single-cell genomics, bioinformatics, and computation. However, with the increasing commercial availability of scRNA-seq platforms, and the rapid ongoing maturation of bioinformatics approaches, a point has been reached where any biomedical researcher or clinician can use scRNA-seq to make exciting discoveries. In this review, we present a practical guide to help researchers design their first scRNA-seq studies, including introductory information on experimental hardware, protocol choice, quality control, data analysis and biological interpretation.
Collapse
Affiliation(s)
- Ashraful Haque
- QIMR Berghofer Medical Research Institute, Herston, Brisbane, Queensland, 4006, Australia.
| | - Jessica Engel
- QIMR Berghofer Medical Research Institute, Herston, Brisbane, Queensland, 4006, Australia
| | - Sarah A Teichmann
- Wellcome Trust Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridge, CB10 1SA, UK
| | - Tapio Lönnberg
- Turku Centre for Biotechnology, University of Turku and Åbo Akademi University, FI-20520, Turku, Finland.
| |
Collapse
|
365
|
Functional Virtual Flow Cytometry: A Visual Analytic Approach for Characterizing Single-Cell Gene Expression Patterns. BIOMED RESEARCH INTERNATIONAL 2017; 2017:3035481. [PMID: 28798928 PMCID: PMC5536134 DOI: 10.1155/2017/3035481] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/03/2017] [Accepted: 05/22/2017] [Indexed: 01/13/2023]
Abstract
We presented a novel workflow for detecting distribution patterns in cell populations based on single-cell transcriptome study. With the fast adoption of single-cell analysis, a challenge to researchers is how to effectively extract gene features to meaningfully separate the cell population. Considering that coexpressed genes are often functionally or structurally related and the number of coexpressed modules is much smaller than the number of genes, our workflow uses gene coexpression modules as features instead of individual genes. Thus, when the coexpressed modules are summarized into eigengenes, not only can we interactively explore the distribution of cells but also we can promptly interpret the gene features. The interactive visualization is aided by a novel application of spatial statistical analysis to the scatter plots using a clustering index parameter. This parameter helps to highlight interesting 2D patterns in the scatter plot matrix (SPLOM). We demonstrated the effectiveness of the workflow using two large single-cell studies. In the Allen Brain scRNA-seq dataset, the visual analytics suggested a new hypothesis such as the involvement of glutamate metabolism in the separation of the brain cells. In a large glioblastoma study, a sample with a unique cell migration related signature was identified.
Collapse
|
366
|
An Immune Atlas of Clear Cell Renal Cell Carcinoma. Cell 2017; 169:736-749.e18. [PMID: 28475899 PMCID: PMC5422211 DOI: 10.1016/j.cell.2017.04.016] [Citation(s) in RCA: 722] [Impact Index Per Article: 90.3] [Reference Citation Analysis] [Abstract] [Key Words] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2017] [Revised: 03/19/2017] [Accepted: 04/12/2017] [Indexed: 01/07/2023]
Abstract
Immune cells in the tumor microenvironment modulate cancer progression and are attractive therapeutic targets. Macrophages and T cells are key components of the microenvironment, yet their phenotypes and relationships in this ecosystem and to clinical outcomes are ill defined. We used mass cytometry with extensive antibody panels to perform in-depth immune profiling of samples from 73 clear cell renal cell carcinoma (ccRCC) patients and five healthy controls. In 3.5 million measured cells, we identified 17 tumor-associated macrophage phenotypes, 22 T cell phenotypes, and a distinct immune composition correlated with progression-free survival, thereby presenting an in-depth human atlas of the immune tumor microenvironment in this disease. This study revealed potential biomarkers and targets for immunotherapy development and validated tools that can be used for immune profiling of other tumor types. Mass cytometry reveals the immune cell diversity of the ccRCC tumor ecosystem PD-1+ cells display heterogeneous combinations of inhibitory receptors CD38+CD204+CD206− tumor-associated macrophages correlate with immunosuppression A specific immune signature is linked to shorter progression-free survival
Collapse
|
367
|
Pouyan MB, Nourani M. Identifying Cell Populations in Flow Cytometry Data Using Phenotypic Signatures. IEEE/ACM TRANSACTIONS ON COMPUTATIONAL BIOLOGY AND BIOINFORMATICS 2017; 14:880-891. [PMID: 27076456 DOI: 10.1109/tcbb.2016.2550428] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/05/2023]
Abstract
Single-cell flow cytometry is a technology that measures the expression of several cellular markers simultaneously for a large number of cells. Identification of homogeneous cell populations, currently done by manual biaxial gating, is highly subjective and time consuming. To overcome the shortcomings of manual gating, automatic algorithms have been proposed. However, the performance of these methods highly depends on the shape of populations and the dimension of the data. In this paper, we have developed a time-efficient method that accurately identifies cellular populations. This is done based on a novel technique that estimates the initial number of clusters in high dimension and identifies the final clusters by merging clusters using their phenotypic signatures in low dimension. The proposed method is called SigClust. We have applied SigClust to four public datasets and compared it with five well known methods in the field. The results are promising and indicate higher performance and accuracy compared to similar approaches reported in literature.
Collapse
|
368
|
Hamey FK, Nestorowa S, Kinston SJ, Kent DG, Wilson NK, Göttgens B. Reconstructing blood stem cell regulatory network models from single-cell molecular profiles. Proc Natl Acad Sci U S A 2017; 114:5822-5829. [PMID: 28584094 PMCID: PMC5468644 DOI: 10.1073/pnas.1610609114] [Citation(s) in RCA: 70] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023] Open
Abstract
Adult blood contains a mixture of mature cell types, each with specialized functions. Single hematopoietic stem cells (HSCs) have been functionally shown to generate all mature cell types for the lifetime of the organism. Differentiation of HSCs toward alternative lineages must be balanced at the population level by the fate decisions made by individual cells. Transcription factors play a key role in regulating these decisions and operate within organized regulatory programs that can be modeled as transcriptional regulatory networks. As dysregulation of single HSC fate decisions is linked to fatal malignancies such as leukemia, it is important to understand how these decisions are controlled on a cell-by-cell basis. Here we developed and applied a network inference method, exploiting the ability to infer dynamic information from single-cell snapshot expression data based on expression profiles of 48 genes in 2,167 blood stem and progenitor cells. This approach allowed us to infer transcriptional regulatory network models that recapitulated differentiation of HSCs into progenitor cell types, focusing on trajectories toward megakaryocyte-erythrocyte progenitors and lymphoid-primed multipotent progenitors. By comparing these two models, we identified and subsequently experimentally validated a difference in the regulation of nuclear factor, erythroid 2 (Nfe2) and core-binding factor, runt domain, alpha subunit 2, translocated to, 3 homolog (Cbfa2t3h) by the transcription factor Gata2. Our approach confirms known aspects of hematopoiesis, provides hypotheses about regulation of HSC differentiation, and is widely applicable to other hierarchical biological systems to uncover regulatory relationships.
Collapse
Affiliation(s)
- Fiona K Hamey
- Department of Haematology, Wellcome Trust-Medical Research Council Cambridge Stem Cell Institute, University of Cambridge, Cambridge Institute for Medical Research, Cambridge CB2 0XY, United Kingdom
| | - Sonia Nestorowa
- Department of Haematology, Wellcome Trust-Medical Research Council Cambridge Stem Cell Institute, University of Cambridge, Cambridge Institute for Medical Research, Cambridge CB2 0XY, United Kingdom
| | - Sarah J Kinston
- Department of Haematology, Wellcome Trust-Medical Research Council Cambridge Stem Cell Institute, University of Cambridge, Cambridge Institute for Medical Research, Cambridge CB2 0XY, United Kingdom
| | - David G Kent
- Department of Haematology, Wellcome Trust-Medical Research Council Cambridge Stem Cell Institute, University of Cambridge, Cambridge Institute for Medical Research, Cambridge CB2 0XY, United Kingdom
| | - Nicola K Wilson
- Department of Haematology, Wellcome Trust-Medical Research Council Cambridge Stem Cell Institute, University of Cambridge, Cambridge Institute for Medical Research, Cambridge CB2 0XY, United Kingdom
| | - Berthold Göttgens
- Department of Haematology, Wellcome Trust-Medical Research Council Cambridge Stem Cell Institute, University of Cambridge, Cambridge Institute for Medical Research, Cambridge CB2 0XY, United Kingdom
| |
Collapse
|
369
|
Wagner A, Regev A, Yosef N. Revealing the vectors of cellular identity with single-cell genomics. Nat Biotechnol 2017; 34:1145-1160. [PMID: 27824854 DOI: 10.1038/nbt.3711] [Citation(s) in RCA: 401] [Impact Index Per Article: 50.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Single-cell genomics has now made it possible to create a comprehensive atlas of human cells. At the same time, it has reopened definitions of a cell's identity and of the ways in which identity is regulated by the cell's molecular circuitry. Emerging computational analysis methods, especially in single-cell RNA sequencing (scRNA-seq), have already begun to reveal, in a data-driven way, the diverse simultaneous facets of a cell's identity, from discrete cell types to continuous dynamic transitions and spatial locations. These developments will eventually allow a cell to be represented as a superposition of 'basis vectors', each determining a different (but possibly dependent) aspect of cellular organization and function. However, computational methods must also overcome considerable challenges-from handling technical noise and data scale to forming new abstractions of biology. As the scale of single-cell experiments continues to increase, new computational approaches will be essential for constructing and characterizing a reference map of cell identities.
Collapse
Affiliation(s)
- Allon Wagner
- Department of Electrical Engineering and Computer Science and the Center for Computational Biology, University of California, Berkeley, California, USA
| | - Aviv Regev
- Howard Hughes Medical Institute, Department of Biology, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA.,Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
| | - Nir Yosef
- Department of Electrical Engineering and Computer Science and the Center for Computational Biology, University of California, Berkeley, California, USA.,Ragon Institute of Massachusetts General Hospital, Massachusetts Institute of Technology, and Harvard University, Boston, Massachusetts, USA
| |
Collapse
|
370
|
Abstract
Immunologists are being compelled to develop new high-dimensional perspectives of cellular heterogeneity and to determine which applications best exploit the power of mass cytometry and associated multiplex approaches.
Collapse
Affiliation(s)
- Evan W Newell
- Singapore Immunology Network, Agency for Science Technology and Research, Singapore, and the Nanyang Technological University School of Biological Sciences, Singapore
| | - Yang Cheng
- Singapore Immunology Network, Agency for Science Technology and Research, Singapore, and the Nanyang Technological University School of Biological Sciences, Singapore
| |
Collapse
|
371
|
McCarthy DJ, Campbell KR, Lun ATL, Wills QF. Scater: pre-processing, quality control, normalization and visualization of single-cell RNA-seq data in R. Bioinformatics 2017; 33:1179-1186. [PMID: 28088763 PMCID: PMC5408845 DOI: 10.1093/bioinformatics/btw777] [Citation(s) in RCA: 912] [Impact Index Per Article: 114.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2016] [Accepted: 12/07/2016] [Indexed: 12/26/2022] Open
Abstract
Motivation Single-cell RNA sequencing (scRNA-seq) is increasingly used to study gene expression at the level of individual cells. However, preparing raw sequence data for further analysis is not a straightforward process. Biases, artifacts and other sources of unwanted variation are present in the data, requiring substantial time and effort to be spent on pre-processing, quality control (QC) and normalization. Results We have developed the R/Bioconductor package scater to facilitate rigorous pre-processing, quality control, normalization and visualization of scRNA-seq data. The package provides a convenient, flexible workflow to process raw sequencing reads into a high-quality expression dataset ready for downstream analysis. scater provides a rich suite of plotting tools for single-cell data and a flexible data structure that is compatible with existing tools and can be used as infrastructure for future software development. Availability and Implementation The open-source code, along with installation instructions, vignettes and case studies, is available through Bioconductor at http://bioconductor.org/packages/scater. Supplementary information Supplementary data are available at Bioinformatics online.
Collapse
Affiliation(s)
- Davis J McCarthy
- European Molecular Biology Laboratory, European Bioinformatics Institute, Wellcome Genome Campus, CB10 1SD Hinxton, Cambridge, UK.,Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford OX3 7BN, UK.,St Vincent's Institute of Medical Research, Fitzroy, Victoria 3065, Australia
| | - Kieran R Campbell
- Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford OX3 7BN, UK.,Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford OX1 3QX, UK
| | - Aaron T L Lun
- CRUK Cambridge Institute, University of Cambridge, Cambridge CB2 0RE, UK
| | - Quin F Wills
- Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford OX3 7BN, UK.,Weatherall Institute for Molecular Medicine, University of Oxford, John Radcliffe Hospital, Oxford OX3 9DS, UK
| |
Collapse
|
372
|
Nowicka M, Krieg C, Crowell HL, Weber LM, Hartmann FJ, Guglietta S, Becher B, Levesque MP, Robinson MD. CyTOF workflow: differential discovery in high-throughput high-dimensional cytometry datasets. F1000Res 2017; 6:748. [PMID: 28663787 PMCID: PMC5473464 DOI: 10.12688/f1000research.11622.1] [Citation(s) in RCA: 227] [Impact Index Per Article: 28.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 05/24/2017] [Indexed: 01/02/2023] Open
Abstract
High dimensional mass and flow cytometry (HDCyto) experiments have become a method of choice for high throughput interrogation and characterization of cell populations.Here, we present an R-based pipeline for differential analyses of HDCyto data, largely based on Bioconductor packages. We computationally define cell populations using FlowSOM clustering, and facilitate an optional but reproducible strategy for manual merging of algorithm-generated clusters. Our workflow offers different analysis paths, including association of cell type abundance with a phenotype or changes in signaling markers within specific subpopulations, or differential analyses of aggregated signals. Importantly, the differential analyses we show are based on regression frameworks where the HDCyto data is the response; thus, we are able to model arbitrary experimental designs, such as those with batch effects, paired designs and so on. In particular, we apply generalized linear mixed models to analyses of cell population abundance or cell-population-specific analyses of signaling markers, allowing overdispersion in cell count or aggregated signals across samples to be appropriately modeled. To support the formal statistical analyses, we encourage exploratory data analysis at every step, including quality control (e.g. multi-dimensional scaling plots), reporting of clustering results (dimensionality reduction, heatmaps with dendrograms) and differential analyses (e.g. plots of aggregated signals).
Collapse
Affiliation(s)
- Malgorzata Nowicka
- Institute for Molecular Life Sciences, University of Zurich, Zurich, 8057, Switzerland
- SIB Swiss Institute of Bioinformatics, University of Zurich, Zurich, 8057, Switzerland
| | - Carsten Krieg
- Institute of Experimental Immunology, University of Zurich, Zurich, 8057, Switzerland
| | - Helena L. Crowell
- Institute for Molecular Life Sciences, University of Zurich, Zurich, 8057, Switzerland
- SIB Swiss Institute of Bioinformatics, University of Zurich, Zurich, 8057, Switzerland
| | - Lukas M. Weber
- Institute for Molecular Life Sciences, University of Zurich, Zurich, 8057, Switzerland
- SIB Swiss Institute of Bioinformatics, University of Zurich, Zurich, 8057, Switzerland
| | - Felix J. Hartmann
- Institute of Experimental Immunology, University of Zurich, Zurich, 8057, Switzerland
| | - Silvia Guglietta
- Department of Experimental Oncology, European Institute of Oncology, Via Adamello 16, Milan, I-20139, Italy
| | - Burkhard Becher
- Institute of Experimental Immunology, University of Zurich, Zurich, 8057, Switzerland
| | - Mitchell P. Levesque
- Department of Dermatology, University Hospital Zurich, Zurich, CH-8091, Switzerland
| | - Mark D. Robinson
- Institute for Molecular Life Sciences, University of Zurich, Zurich, 8057, Switzerland
- SIB Swiss Institute of Bioinformatics, University of Zurich, Zurich, 8057, Switzerland
| |
Collapse
|
373
|
Nowicka M, Krieg C, Crowell HL, Weber LM, Hartmann FJ, Guglietta S, Becher B, Levesque MP, Robinson MD. CyTOF workflow: differential discovery in high-throughput high-dimensional cytometry datasets. F1000Res 2017; 6:748. [PMID: 28663787 PMCID: PMC5473464 DOI: 10.12688/f1000research.11622.3] [Citation(s) in RCA: 209] [Impact Index Per Article: 26.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 04/15/2019] [Indexed: 12/15/2022] Open
Abstract
High-dimensional mass and flow cytometry (HDCyto) experiments have become a method of choice for high-throughput interrogation and characterization of cell populations. Here, we present an updated R-based pipeline for differential analyses of HDCyto data, largely based on Bioconductor packages. We computationally define cell populations using FlowSOM clustering, and facilitate an optional but reproducible strategy for manual merging of algorithm-generated clusters. Our workflow offers different analysis paths, including association of cell type abundance with a phenotype or changes in signalling markers within specific subpopulations, or differential analyses of aggregated signals. Importantly, the differential analyses we show are based on regression frameworks where the HDCyto data is the response; thus, we are able to model arbitrary experimental designs, such as those with batch effects, paired designs and so on. In particular, we apply generalized linear mixed models or linear mixed models to analyses of cell population abundance or cell-population-specific analyses of signaling markers, allowing overdispersion in cell count or aggregated signals across samples to be appropriately modeled. To support the formal statistical analyses, we encourage exploratory data analysis at every step, including quality control (e.g., multi-dimensional scaling plots), reporting of clustering results (dimensionality reduction, heatmaps with dendrograms) and differential analyses (e.g., plots of aggregated signals).
Collapse
Affiliation(s)
- Malgorzata Nowicka
- Institute for Molecular Life Sciences, University of Zurich, Zurich, 8057, Switzerland
- SIB Swiss Institute of Bioinformatics, University of Zurich, Zurich, 8057, Switzerland
| | - Carsten Krieg
- Institute of Experimental Immunology, University of Zurich, Zurich, 8057, Switzerland
| | - Helena L. Crowell
- Institute for Molecular Life Sciences, University of Zurich, Zurich, 8057, Switzerland
- SIB Swiss Institute of Bioinformatics, University of Zurich, Zurich, 8057, Switzerland
| | - Lukas M. Weber
- Institute for Molecular Life Sciences, University of Zurich, Zurich, 8057, Switzerland
- SIB Swiss Institute of Bioinformatics, University of Zurich, Zurich, 8057, Switzerland
| | - Felix J. Hartmann
- Institute of Experimental Immunology, University of Zurich, Zurich, 8057, Switzerland
| | - Silvia Guglietta
- Department of Experimental Oncology, European Institute of Oncology, Via Adamello 16, Milan, I-20139, Italy
| | - Burkhard Becher
- Institute of Experimental Immunology, University of Zurich, Zurich, 8057, Switzerland
| | - Mitchell P. Levesque
- Department of Dermatology, University Hospital Zurich, Zurich, CH-8091, Switzerland
| | - Mark D. Robinson
- Institute for Molecular Life Sciences, University of Zurich, Zurich, 8057, Switzerland
- SIB Swiss Institute of Bioinformatics, University of Zurich, Zurich, 8057, Switzerland
| |
Collapse
|
374
|
Nowicka M, Krieg C, Crowell HL, Weber LM, Hartmann FJ, Guglietta S, Becher B, Levesque MP, Robinson MD. CyTOF workflow: differential discovery in high-throughput high-dimensional cytometry datasets. F1000Res 2017; 6:748. [PMID: 28663787 PMCID: PMC5473464 DOI: 10.12688/f1000research.11622.2] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 11/15/2017] [Indexed: 12/19/2022] Open
Abstract
High dimensional mass and flow cytometry (HDCyto) experiments have become a method of choice for high throughput interrogation and characterization of cell populations.Here, we present an R-based pipeline for differential analyses of HDCyto data, largely based on Bioconductor packages. We computationally define cell populations using FlowSOM clustering, and facilitate an optional but reproducible strategy for manual merging of algorithm-generated clusters. Our workflow offers different analysis paths, including association of cell type abundance with a phenotype or changes in signaling markers within specific subpopulations, or differential analyses of aggregated signals. Importantly, the differential analyses we show are based on regression frameworks where the HDCyto data is the response; thus, we are able to model arbitrary experimental designs, such as those with batch effects, paired designs and so on. In particular, we apply generalized linear mixed models to analyses of cell population abundance or cell-population-specific analyses of signaling markers, allowing overdispersion in cell count or aggregated signals across samples to be appropriately modeled. To support the formal statistical analyses, we encourage exploratory data analysis at every step, including quality control (e.g. multi-dimensional scaling plots), reporting of clustering results (dimensionality reduction, heatmaps with dendrograms) and differential analyses (e.g. plots of aggregated signals).
Collapse
Affiliation(s)
- Malgorzata Nowicka
- Institute for Molecular Life Sciences, University of Zurich, Zurich, 8057, Switzerland
- SIB Swiss Institute of Bioinformatics, University of Zurich, Zurich, 8057, Switzerland
| | - Carsten Krieg
- Institute of Experimental Immunology, University of Zurich, Zurich, 8057, Switzerland
| | - Helena L. Crowell
- Institute for Molecular Life Sciences, University of Zurich, Zurich, 8057, Switzerland
- SIB Swiss Institute of Bioinformatics, University of Zurich, Zurich, 8057, Switzerland
| | - Lukas M. Weber
- Institute for Molecular Life Sciences, University of Zurich, Zurich, 8057, Switzerland
- SIB Swiss Institute of Bioinformatics, University of Zurich, Zurich, 8057, Switzerland
| | - Felix J. Hartmann
- Institute of Experimental Immunology, University of Zurich, Zurich, 8057, Switzerland
| | - Silvia Guglietta
- Department of Experimental Oncology, European Institute of Oncology, Via Adamello 16, Milan, I-20139, Italy
| | - Burkhard Becher
- Institute of Experimental Immunology, University of Zurich, Zurich, 8057, Switzerland
| | - Mitchell P. Levesque
- Department of Dermatology, University Hospital Zurich, Zurich, CH-8091, Switzerland
| | - Mark D. Robinson
- Institute for Molecular Life Sciences, University of Zurich, Zurich, 8057, Switzerland
- SIB Swiss Institute of Bioinformatics, University of Zurich, Zurich, 8057, Switzerland
| |
Collapse
|
375
|
Cabezas-Wallscheid N, Buettner F, Sommerkamp P, Klimmeck D, Ladel L, Thalheimer FB, Pastor-Flores D, Roma LP, Renders S, Zeisberger P, Przybylla A, Schönberger K, Scognamiglio R, Altamura S, Florian CM, Fawaz M, Vonficht D, Tesio M, Collier P, Pavlinic D, Geiger H, Schroeder T, Benes V, Dick TP, Rieger MA, Stegle O, Trumpp A. Vitamin A-Retinoic Acid Signaling Regulates Hematopoietic Stem Cell Dormancy. Cell 2017; 169:807-823.e19. [PMID: 28479188 DOI: 10.1016/j.cell.2017.04.018] [Citation(s) in RCA: 325] [Impact Index Per Article: 40.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2017] [Revised: 04/06/2017] [Accepted: 04/12/2017] [Indexed: 02/07/2023]
Abstract
Dormant hematopoietic stem cells (dHSCs) are atop the hematopoietic hierarchy. The molecular identity of dHSCs and the mechanisms regulating their maintenance or exit from dormancy remain uncertain. Here, we use single-cell RNA sequencing (RNA-seq) analysis to show that the transition from dormancy toward cell-cycle entry is a continuous developmental path associated with upregulation of biosynthetic processes rather than a stepwise progression. In addition, low Myc levels and high expression of a retinoic acid program are characteristic for dHSCs. To follow the behavior of dHSCs in situ, a Gprc5c-controlled reporter mouse was established. Treatment with all-trans retinoic acid antagonizes stress-induced activation of dHSCs by restricting protein translation and levels of reactive oxygen species (ROS) and Myc. Mice maintained on a vitamin A-free diet lose HSCs and show a disrupted re-entry into dormancy after exposure to inflammatory stress stimuli. Our results highlight the impact of dietary vitamin A on the regulation of cell-cycle-mediated stem cell plasticity. VIDEO ABSTRACT.
Collapse
Affiliation(s)
- Nina Cabezas-Wallscheid
- Division of Stem Cells and Cancer, German Cancer Research Center (DKFZ) and DKFZ-ZMBH Alliance, 69120 Heidelberg, Germany; Heidelberg Institute for Stem Cell Technology and Experimental Medicine (HI-STEM gGmbH), 69120 Heidelberg, Germany.
| | - Florian Buettner
- European Molecular Biology Laboratory, European Bioinformatics Institute (EBI), Wellcome Genome Campus, Hinxton, Cambridge CB10 1SD, UK
| | - Pia Sommerkamp
- Division of Stem Cells and Cancer, German Cancer Research Center (DKFZ) and DKFZ-ZMBH Alliance, 69120 Heidelberg, Germany; Heidelberg Institute for Stem Cell Technology and Experimental Medicine (HI-STEM gGmbH), 69120 Heidelberg, Germany
| | - Daniel Klimmeck
- Division of Stem Cells and Cancer, German Cancer Research Center (DKFZ) and DKFZ-ZMBH Alliance, 69120 Heidelberg, Germany; Heidelberg Institute for Stem Cell Technology and Experimental Medicine (HI-STEM gGmbH), 69120 Heidelberg, Germany
| | - Luisa Ladel
- Division of Stem Cells and Cancer, German Cancer Research Center (DKFZ) and DKFZ-ZMBH Alliance, 69120 Heidelberg, Germany; Heidelberg Institute for Stem Cell Technology and Experimental Medicine (HI-STEM gGmbH), 69120 Heidelberg, Germany
| | - Frederic B Thalheimer
- LOEWE Center for Cell and Gene Therapy and Department of Medicine, Hematology/Oncology, Goethe University Frankfurt, 60590 Frankfurt am Main, Germany
| | - Daniel Pastor-Flores
- Division of Redox Regulation, DKFZ-ZMBH Alliance, German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
| | - Leticia P Roma
- Division of Redox Regulation, DKFZ-ZMBH Alliance, German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
| | - Simon Renders
- Division of Stem Cells and Cancer, German Cancer Research Center (DKFZ) and DKFZ-ZMBH Alliance, 69120 Heidelberg, Germany; Heidelberg Institute for Stem Cell Technology and Experimental Medicine (HI-STEM gGmbH), 69120 Heidelberg, Germany
| | - Petra Zeisberger
- Division of Stem Cells and Cancer, German Cancer Research Center (DKFZ) and DKFZ-ZMBH Alliance, 69120 Heidelberg, Germany; Heidelberg Institute for Stem Cell Technology and Experimental Medicine (HI-STEM gGmbH), 69120 Heidelberg, Germany
| | - Adriana Przybylla
- Division of Stem Cells and Cancer, German Cancer Research Center (DKFZ) and DKFZ-ZMBH Alliance, 69120 Heidelberg, Germany; Heidelberg Institute for Stem Cell Technology and Experimental Medicine (HI-STEM gGmbH), 69120 Heidelberg, Germany
| | - Katharina Schönberger
- Division of Stem Cells and Cancer, German Cancer Research Center (DKFZ) and DKFZ-ZMBH Alliance, 69120 Heidelberg, Germany; Heidelberg Institute for Stem Cell Technology and Experimental Medicine (HI-STEM gGmbH), 69120 Heidelberg, Germany
| | - Roberta Scognamiglio
- Division of Stem Cells and Cancer, German Cancer Research Center (DKFZ) and DKFZ-ZMBH Alliance, 69120 Heidelberg, Germany; Heidelberg Institute for Stem Cell Technology and Experimental Medicine (HI-STEM gGmbH), 69120 Heidelberg, Germany
| | - Sandro Altamura
- Department of Pediatric Hematology, Oncology and Immunology, University of Heidelberg, 69120 Heidelberg, Germany
| | - Carolina M Florian
- Institute for Molecular Medicine, Stem Cells and Aging, Ulm University, 89081 Ulm, Germany
| | - Malak Fawaz
- LOEWE Center for Cell and Gene Therapy and Department of Medicine, Hematology/Oncology, Goethe University Frankfurt, 60590 Frankfurt am Main, Germany
| | - Dominik Vonficht
- Division of Stem Cells and Cancer, German Cancer Research Center (DKFZ) and DKFZ-ZMBH Alliance, 69120 Heidelberg, Germany; Heidelberg Institute for Stem Cell Technology and Experimental Medicine (HI-STEM gGmbH), 69120 Heidelberg, Germany
| | - Melania Tesio
- Division of Stem Cells and Cancer, German Cancer Research Center (DKFZ) and DKFZ-ZMBH Alliance, 69120 Heidelberg, Germany; Heidelberg Institute for Stem Cell Technology and Experimental Medicine (HI-STEM gGmbH), 69120 Heidelberg, Germany
| | - Paul Collier
- Genomics Core Facility, European Molecular Biology Laboratory (EMBL), 69117 Heidelberg, Germany
| | - Dinko Pavlinic
- Genomics Core Facility, European Molecular Biology Laboratory (EMBL), 69117 Heidelberg, Germany
| | - Hartmut Geiger
- Institute for Molecular Medicine, Stem Cells and Aging, Ulm University, 89081 Ulm, Germany
| | - Timm Schroeder
- Department of Biosystems Science and Engineering (D-BSSE), ETH Zurich, 4058 Basel, Switzerland
| | - Vladimir Benes
- Genomics Core Facility, European Molecular Biology Laboratory (EMBL), 69117 Heidelberg, Germany
| | - Tobias P Dick
- Division of Redox Regulation, DKFZ-ZMBH Alliance, German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
| | - Michael A Rieger
- LOEWE Center for Cell and Gene Therapy and Department of Medicine, Hematology/Oncology, Goethe University Frankfurt, 60590 Frankfurt am Main, Germany
| | - Oliver Stegle
- European Molecular Biology Laboratory, European Bioinformatics Institute (EBI), Wellcome Genome Campus, Hinxton, Cambridge CB10 1SD, UK
| | - Andreas Trumpp
- Division of Stem Cells and Cancer, German Cancer Research Center (DKFZ) and DKFZ-ZMBH Alliance, 69120 Heidelberg, Germany; Heidelberg Institute for Stem Cell Technology and Experimental Medicine (HI-STEM gGmbH), 69120 Heidelberg, Germany; German Cancer Consortium (DKTK), 69120 Heidelberg, Germany.
| |
Collapse
|
376
|
Mildner A, Schönheit J, Giladi A, David E, Lara-Astiaso D, Lorenzo-Vivas E, Paul F, Chappell-Maor L, Priller J, Leutz A, Amit I, Jung S. Genomic Characterization of Murine Monocytes Reveals C/EBPβ Transcription Factor Dependence of Ly6C − Cells. Immunity 2017; 46:849-862.e7. [DOI: 10.1016/j.immuni.2017.04.018] [Citation(s) in RCA: 217] [Impact Index Per Article: 27.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2016] [Revised: 02/22/2017] [Accepted: 04/26/2017] [Indexed: 12/11/2022]
|
377
|
Scaling single-cell genomics from phenomenology to mechanism. Nature 2017; 541:331-338. [PMID: 28102262 DOI: 10.1038/nature21350] [Citation(s) in RCA: 521] [Impact Index Per Article: 65.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2016] [Accepted: 11/14/2016] [Indexed: 02/08/2023]
Abstract
Three of the most fundamental questions in biology are how individual cells differentiate to form tissues, how tissues function in a coordinated and flexible fashion and which gene regulatory mechanisms support these processes. Single-cell genomics is opening up new ways to tackle these questions by combining the comprehensive nature of genomics with the microscopic resolution that is required to describe complex multicellular systems. Initial single-cell genomic studies provided a remarkably rich phenomenology of heterogeneous cellular states, but transforming observational studies into models of dynamics and causal mechanisms in tissues poses fresh challenges and requires stronger integration of theoretical, computational and experimental frameworks.
Collapse
|
378
|
Pouyan MB, Birjandtalab J, Nourani M. Distance metric learning using random forest for cytometry data. ANNUAL INTERNATIONAL CONFERENCE OF THE IEEE ENGINEERING IN MEDICINE AND BIOLOGY SOCIETY. IEEE ENGINEERING IN MEDICINE AND BIOLOGY SOCIETY. ANNUAL INTERNATIONAL CONFERENCE 2017; 2016:2590. [PMID: 28268852 DOI: 10.1109/embc.2016.7591260] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Visualization and clustering of single-cell mass cytometry (CyTOF) data are analytic techniques to identify different cell types. Most of such techniques, such as Euclidean norm, lose their effectiveness when the data dimension increases due to the curse of dimensionality. In this paper, we propose a new cell distance (called CytoRFD) that works based on Random Forest (RF) concept. The experimental results show that the proposed distance can achieve a much higher quality and effectiveness in large data analysis than traditional metrics specially for CyTOF data.
Collapse
|
379
|
Lönnberg T, Svensson V, James KR, Fernandez-Ruiz D, Sebina I, Montandon R, Soon MSF, Fogg LG, Nair AS, Liligeto U, Stubbington MJT, Ly LH, Bagger FO, Zwiessele M, Lawrence ND, Souza-Fonseca-Guimaraes F, Bunn PT, Engwerda CR, Heath WR, Billker O, Stegle O, Haque A, Teichmann SA. Single-cell RNA-seq and computational analysis using temporal mixture modelling resolves Th1/Tfh fate bifurcation in malaria. Sci Immunol 2017; 2:eaal2192. [PMID: 28345074 PMCID: PMC5365145 DOI: 10.1126/sciimmunol.aal2192] [Citation(s) in RCA: 206] [Impact Index Per Article: 25.8] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Differentiation of naïve CD4+ T cells into functionally distinct T helper subsets is crucial for the orchestration of immune responses. Due to extensive heterogeneity and multiple overlapping transcriptional programs in differentiating T cell populations, this process has remained a challenge for systematic dissection in vivo. By using single-cell transcriptomics and computational analysis using a temporal mixtures of Gaussian processes model, termed GPfates, we reconstructed the developmental trajectories of Th1 and Tfh cells during blood-stage Plasmodium infection in mice. By tracking clonality using endogenous TCR sequences, we first demonstrated that Th1/Tfh bifurcation had occurred at both population and single-clone levels. Next, we identified genes whose expression was associated with Th1 or Tfh fates, and demonstrated a T-cell intrinsic role for Galectin-1 in supporting a Th1 differentiation. We also revealed the close molecular relationship between Th1 and IL-10-producing Tr1 cells in this infection. Th1 and Tfh fates emerged from a highly proliferative precursor that upregulated aerobic glycolysis and accelerated cell cycling as cytokine expression began. Dynamic gene expression of chemokine receptors around bifurcation predicted roles for cell-cell in driving Th1/Tfh fates. In particular, we found that precursor Th cells were coached towards a Th1 but not a Tfh fate by inflammatory monocytes. Thus, by integrating genomic and computational approaches, our study has provided two unique resources, a database www.PlasmoTH.org, which facilitates discovery of novel factors controlling Th1/Tfh fate commitment, and more generally, GPfates, a modelling framework for characterizing cell differentiation towards multiple fates.
Collapse
Affiliation(s)
- Tapio Lönnberg
- European Molecular Biology Laboratory, European Bioinformatics Institute (EMBL-EBI), Wellcome Genome Campus, Hinxton, Cambridge, UK
- Wellcome Trust Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridge, UK
| | - Valentine Svensson
- European Molecular Biology Laboratory, European Bioinformatics Institute (EMBL-EBI), Wellcome Genome Campus, Hinxton, Cambridge, UK
| | - Kylie R. James
- QIMR Berghofer Medical Research Institute, Herston, Brisbane, Queensland, Australia
| | - Daniel Fernandez-Ruiz
- Department of Microbiology and Immunology, The Peter Doherty Institute, University of Melbourne, Parkville, Victoria, Australia
| | - Ismail Sebina
- QIMR Berghofer Medical Research Institute, Herston, Brisbane, Queensland, Australia
| | - Ruddy Montandon
- Wellcome Trust Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridge, UK
| | - Megan S. F. Soon
- QIMR Berghofer Medical Research Institute, Herston, Brisbane, Queensland, Australia
| | - Lily G. Fogg
- QIMR Berghofer Medical Research Institute, Herston, Brisbane, Queensland, Australia
| | - Arya Sheela Nair
- QIMR Berghofer Medical Research Institute, Herston, Brisbane, Queensland, Australia
| | - Urijah Liligeto
- QIMR Berghofer Medical Research Institute, Herston, Brisbane, Queensland, Australia
| | - Michael J. T. Stubbington
- European Molecular Biology Laboratory, European Bioinformatics Institute (EMBL-EBI), Wellcome Genome Campus, Hinxton, Cambridge, UK
- Wellcome Trust Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridge, UK
| | - Lam-Ha Ly
- Wellcome Trust Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridge, UK
| | - Frederik Otzen Bagger
- European Molecular Biology Laboratory, European Bioinformatics Institute (EMBL-EBI), Wellcome Genome Campus, Hinxton, Cambridge, UK
- Wellcome Trust Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridge, UK
- Department of Haematology, University of Cambridge, Cambridge Biomedical Campus, Cambridge, UK
- National Health Service (NHS) Blood and Transplant, Cambridge Biomedical Campus, Long Road, Cambridge, UK
| | - Max Zwiessele
- Department of Computer Science, University of Sheffield, Sheffield, UK
| | - Neil D. Lawrence
- Department of Computer Science, University of Sheffield, Sheffield, UK
| | | | - Patrick T. Bunn
- QIMR Berghofer Medical Research Institute, Herston, Brisbane, Queensland, Australia
| | | | - William R. Heath
- Department of Microbiology and Immunology, The Peter Doherty Institute, University of Melbourne, Parkville, Victoria, Australia
- The Australian Research Council Centre of Excellence in Advanced Molecular Imaging, The University of Melbourne, Parkville, Victoria, Australia
| | - Oliver Billker
- Wellcome Trust Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridge, UK
| | - Oliver Stegle
- European Molecular Biology Laboratory, European Bioinformatics Institute (EMBL-EBI), Wellcome Genome Campus, Hinxton, Cambridge, UK
| | - Ashraful Haque
- QIMR Berghofer Medical Research Institute, Herston, Brisbane, Queensland, Australia
| | - Sarah A. Teichmann
- European Molecular Biology Laboratory, European Bioinformatics Institute (EMBL-EBI), Wellcome Genome Campus, Hinxton, Cambridge, UK
- Wellcome Trust Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridge, UK
| |
Collapse
|
380
|
Dpath software reveals hierarchical haemato-endothelial lineages of Etv2 progenitors based on single-cell transcriptome analysis. Nat Commun 2017; 8:14362. [PMID: 28181481 PMCID: PMC5309826 DOI: 10.1038/ncomms14362] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2016] [Accepted: 12/20/2016] [Indexed: 01/04/2023] Open
Abstract
Developmental, stem cell and cancer biologists are interested in the molecular definition of cellular differentiation. Although single-cell RNA sequencing represents a transformational advance for global gene analyses, novel obstacles have emerged, including the computational management of dropout events, the reconstruction of biological pathways and the isolation of target cell populations. We develop an algorithm named dpath that applies the concept of metagene entropy and allows the ranking of cells based on their differentiation potential. We also develop self-organizing map (SOM) and random walk with restart (RWR) algorithms to separate the progenitors from the differentiated cells and reconstruct the lineage hierarchies in an unbiased manner. We test these algorithms using single cells from Etv2-EYFP transgenic mouse embryos and reveal specific molecular pathways that direct differentiation programmes involving the haemato-endothelial lineages. This software program quantitatively assesses the progenitor and committed states in single-cell RNA-seq data sets in a non-biased manner. Single-cell RNA sequencing has enabled great advances in understanding developmental biology but reconstructing cellular lineages from this data remains challenging. Here the authors develop an algorithm, dpath, which models the lineage relationships of underlying single cells based on single cell RNA seq data and apply it to study lineage progression of Etv2 expressing progenitors.
Collapse
|
381
|
Dulken BW, Leeman DS, Boutet SC, Hebestreit K, Brunet A. Single-Cell Transcriptomic Analysis Defines Heterogeneity and Transcriptional Dynamics in the Adult Neural Stem Cell Lineage. Cell Rep 2017; 18:777-790. [PMID: 28099854 PMCID: PMC5269583 DOI: 10.1016/j.celrep.2016.12.060] [Citation(s) in RCA: 283] [Impact Index Per Article: 35.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2016] [Revised: 10/04/2016] [Accepted: 12/19/2016] [Indexed: 12/31/2022] Open
Abstract
Neural stem cells (NSCs) in the adult mammalian brain serve as a reservoir for the generation of new neurons, oligodendrocytes, and astrocytes. Here, we use single-cell RNA sequencing to characterize adult NSC populations and examine the molecular identities and heterogeneity of in vivo NSC populations. We find that cells in the NSC lineage exist on a continuum through the processes of activation and differentiation. Interestingly, rare intermediate states with distinct molecular profiles can be identified and experimentally validated, and our analysis identifies putative surface markers and key intracellular regulators for these subpopulations of NSCs. Finally, using the power of single-cell profiling, we conduct a meta-analysis to compare in vivo NSCs and in vitro cultures, distinct fluorescence-activated cell sorting strategies, and different neurogenic niches. These data provide a resource for the field and contribute to an integrative understanding of the adult NSC lineage.
Collapse
Affiliation(s)
- Ben W Dulken
- Department of Genetics, Stanford University, Stanford, CA 94305, USA; Stanford Medical Scientist Training Program, Stanford University, Stanford, CA 94305, USA; Institute for Stem Cell Biology and Regenerative Medicine, Stanford University, Stanford, CA 94305, USA
| | - Dena S Leeman
- Department of Genetics, Stanford University, Stanford, CA 94305, USA; Cancer Biology Program, Stanford University, Stanford, CA 94305, USA
| | | | - Katja Hebestreit
- Department of Genetics, Stanford University, Stanford, CA 94305, USA
| | - Anne Brunet
- Department of Genetics, Stanford University, Stanford, CA 94305, USA; Glenn Laboratories for the Biology of Aging at Stanford University, Stanford University, Stanford, CA 94305, USA.
| |
Collapse
|
382
|
Richard A, Boullu L, Herbach U, Bonnafoux A, Morin V, Vallin E, Guillemin A, Papili Gao N, Gunawan R, Cosette J, Arnaud O, Kupiec JJ, Espinasse T, Gonin-Giraud S, Gandrillon O. Single-Cell-Based Analysis Highlights a Surge in Cell-to-Cell Molecular Variability Preceding Irreversible Commitment in a Differentiation Process. PLoS Biol 2016; 14:e1002585. [PMID: 28027290 PMCID: PMC5191835 DOI: 10.1371/journal.pbio.1002585] [Citation(s) in RCA: 138] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2016] [Accepted: 09/22/2016] [Indexed: 12/31/2022] Open
Abstract
In some recent studies, a view emerged that stochastic dynamics governing the switching of cells from one differentiation state to another could be characterized by a peak in gene expression variability at the point of fate commitment. We have tested this hypothesis at the single-cell level by analyzing primary chicken erythroid progenitors through their differentiation process and measuring the expression of selected genes at six sequential time-points after induction of differentiation. In contrast to population-based expression data, single-cell gene expression data revealed a high cell-to-cell variability, which was masked by averaging. We were able to show that the correlation network was a very dynamical entity and that a subgroup of genes tend to follow the predictions from the dynamical network biomarker (DNB) theory. In addition, we also identified a small group of functionally related genes encoding proteins involved in sterol synthesis that could act as the initial drivers of the differentiation. In order to assess quantitatively the cell-to-cell variability in gene expression and its evolution in time, we used Shannon entropy as a measure of the heterogeneity. Entropy values showed a significant increase in the first 8 h of the differentiation process, reaching a peak between 8 and 24 h, before decreasing to significantly lower values. Moreover, we observed that the previous point of maximum entropy precedes two paramount key points: an irreversible commitment to differentiation between 24 and 48 h followed by a significant increase in cell size variability at 48 h. In conclusion, when analyzed at the single cell level, the differentiation process looks very different from its classical population average view. New observables (like entropy) can be computed, the behavior of which is fully compatible with the idea that differentiation is not a “simple” program that all cells execute identically but results from the dynamical behavior of the underlying molecular network. A single-cell transcriptomics analysis offers a new dynamical view of the differentiation process, involving an increase in between-cell variability prior to commitment. The differentiation process has classically been seen as a stereotyped program leading from one progenitor toward a functional cell. This vision was based upon cell population-based analyses averaged over millions of cells. However, new methods have recently emerged that allow interrogation of the molecular content at the single-cell level, challenging this view with a new model suggesting that cell-to-cell gene expression stochasticity could play a key role in differentiation. We took advantage of a physiologically relevant avian cellular model to analyze the expression level of 92 genes in individual cells collected at several time-points during differentiation. We first observed that the process analyzed at the single-cell level is very different and much less well ordered than the population-based average view. Furthermore, we showed that cell-to-cell variability in gene expression peaks transiently before strongly decreasing. This rise in variability precedes two key events: an irreversible commitment to differentiation, followed by a significant increase in cell size variability. Altogether, our results support the idea that differentiation is not a “simple” series of well-ordered molecular events executed identically by all cells in a population but likely results from dynamical behavior of the underlying molecular network.
Collapse
Affiliation(s)
- Angélique Richard
- Univ Lyon, ENS de Lyon, Univ Claude Bernard, CNRS UMR 5239, INSERM U1210, Laboratory of Biology and Modelling of the Cell, 46 allée d’Italie Site Jacques Monod, F-69007, Lyon, France
| | - Loïs Boullu
- Inria Team Dracula, Inria Center Grenoble Rhône-Alpes, France
- Université de Lyon, Université Lyon 1, CNRS UMR 5208, Institut Camille Jordan 43 blvd du 11 novembre 1918, F-69622 Villeurbanne-Cedex, France
- Département de Mathématiques et de statistiques de l’Université de Montréal, Pavillon André-Aisenstadt, 2920, chemin de la Tour, Montréal (Québec) H3T 1J4 Canada
| | - Ulysse Herbach
- Univ Lyon, ENS de Lyon, Univ Claude Bernard, CNRS UMR 5239, INSERM U1210, Laboratory of Biology and Modelling of the Cell, 46 allée d’Italie Site Jacques Monod, F-69007, Lyon, France
- Inria Team Dracula, Inria Center Grenoble Rhône-Alpes, France
- Université de Lyon, Université Lyon 1, CNRS UMR 5208, Institut Camille Jordan 43 blvd du 11 novembre 1918, F-69622 Villeurbanne-Cedex, France
| | - Arnaud Bonnafoux
- Univ Lyon, ENS de Lyon, Univ Claude Bernard, CNRS UMR 5239, INSERM U1210, Laboratory of Biology and Modelling of the Cell, 46 allée d’Italie Site Jacques Monod, F-69007, Lyon, France
- Inria Team Dracula, Inria Center Grenoble Rhône-Alpes, France
- The CoSMo company. 5 passage du Vercors – 69007 LYON – France
| | - Valérie Morin
- Univ Lyon, Univ Claude Bernard, CNRS UMR 5310 - INSERM U1217, Institut NeuroMyoGène, F-69622 Villeurbanne-Cedex, France
| | - Elodie Vallin
- Univ Lyon, ENS de Lyon, Univ Claude Bernard, CNRS UMR 5239, INSERM U1210, Laboratory of Biology and Modelling of the Cell, 46 allée d’Italie Site Jacques Monod, F-69007, Lyon, France
| | - Anissa Guillemin
- Univ Lyon, ENS de Lyon, Univ Claude Bernard, CNRS UMR 5239, INSERM U1210, Laboratory of Biology and Modelling of the Cell, 46 allée d’Italie Site Jacques Monod, F-69007, Lyon, France
| | - Nan Papili Gao
- Institute for Chemical and Bioengineering, ETH Zurich, Zurich, Switzerland
- Swiss Institute of Bioinformatics, Quartier Sorge - Batiment Genopode, 1015 Lausanne Switzerland
| | - Rudiyanto Gunawan
- Institute for Chemical and Bioengineering, ETH Zurich, Zurich, Switzerland
- Swiss Institute of Bioinformatics, Quartier Sorge - Batiment Genopode, 1015 Lausanne Switzerland
| | - Jérémie Cosette
- Genethon – Institut National de la Santé et de la Recherche Médicale – INSERM, Université d’Evry-Val-d’Essone – 1 rue de l’internationale 91000 Evry, France
| | - Ophélie Arnaud
- RIKEN - Center for Life Science Technologies (Division of Genomic Technologies)—CLST (DGT), 1-7-22 Suehiro-cho, Tsurumi-ku, Yokohama, Kanagawa 230-0045, Japan
| | | | - Thibault Espinasse
- Université de Lyon, Université Lyon 1, CNRS UMR 5208, Institut Camille Jordan 43 blvd du 11 novembre 1918, F-69622 Villeurbanne-Cedex, France
| | - Sandrine Gonin-Giraud
- Univ Lyon, ENS de Lyon, Univ Claude Bernard, CNRS UMR 5239, INSERM U1210, Laboratory of Biology and Modelling of the Cell, 46 allée d’Italie Site Jacques Monod, F-69007, Lyon, France
| | - Olivier Gandrillon
- Univ Lyon, ENS de Lyon, Univ Claude Bernard, CNRS UMR 5239, INSERM U1210, Laboratory of Biology and Modelling of the Cell, 46 allée d’Italie Site Jacques Monod, F-69007, Lyon, France
- Inria Team Dracula, Inria Center Grenoble Rhône-Alpes, France
- * E-mail:
| |
Collapse
|
383
|
Velasco S, Ibrahim MM, Kakumanu A, Garipler G, Aydin B, Al-Sayegh MA, Hirsekorn A, Abdul-Rahman F, Satija R, Ohler U, Mahony S, Mazzoni EO. A Multi-step Transcriptional and Chromatin State Cascade Underlies Motor Neuron Programming from Embryonic Stem Cells. Cell Stem Cell 2016; 20:205-217.e8. [PMID: 27939218 DOI: 10.1016/j.stem.2016.11.006] [Citation(s) in RCA: 69] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2016] [Revised: 09/04/2016] [Accepted: 11/04/2016] [Indexed: 11/28/2022]
Abstract
Direct cell programming via overexpression of transcription factors (TFs) aims to control cell fate with the degree of precision needed for clinical applications. However, the regulatory steps involved in successful terminal cell fate programming remain obscure. We have investigated the underlying mechanisms by looking at gene expression, chromatin states, and TF binding during the uniquely efficient Ngn2, Isl1, and Lhx3 motor neuron programming pathway. Our analysis reveals a highly dynamic process in which Ngn2 and the Isl1/Lhx3 pair initially engage distinct regulatory regions. Subsequently, Isl1/Lhx3 binding shifts from one set of targets to another, controlling regulatory region activity and gene expression as cell differentiation progresses. Binding of Isl1/Lhx3 to later motor neuron enhancers depends on the Ebf and Onecut TFs, which are induced by Ngn2 during the programming process. Thus, motor neuron programming is the product of two initially independent transcriptional modules that converge with a feedforward transcriptional logic.
Collapse
Affiliation(s)
- Silvia Velasco
- Department of Biology, New York University, 100 Washington Square East, New York, NY 10003, USA
| | - Mahmoud M Ibrahim
- Department of Biology, Humboldt Universität zu Berlin, Unter den Linden 6, Berlin 10117, Germany.,Berlin Institute for Medical Systems Biology, Max-Delbrück-Center for Molecular Medicine, Robert-Rössle-Str. 10, Berlin 13125, Germany
| | - Akshay Kakumanu
- Center for Eukaryotic Gene Regulation, Department of Biochemistry & Molecular Biology, Penn State University, Pennsylvania, USA
| | - Görkem Garipler
- Department of Biology, New York University, 100 Washington Square East, New York, NY 10003, USA
| | - Begüm Aydin
- Department of Biology, New York University, 100 Washington Square East, New York, NY 10003, USA
| | - Mohamed Ahmed Al-Sayegh
- Department of Biology, New York University, 100 Washington Square East, New York, NY 10003, USA.,Division of Science and Math, New York University, Abu-Dhabi, UAE
| | - Antje Hirsekorn
- Berlin Institute for Medical Systems Biology, Max-Delbrück-Center for Molecular Medicine, Robert-Rössle-Str. 10, Berlin 13125, Germany
| | - Farah Abdul-Rahman
- Department of Biology, New York University, 100 Washington Square East, New York, NY 10003, USA
| | - Rahul Satija
- New York Genome Center, New York University, New York, USA
| | - Uwe Ohler
- Department of Biology, Humboldt Universität zu Berlin, Unter den Linden 6, Berlin 10117, Germany.,Berlin Institute for Medical Systems Biology, Max-Delbrück-Center for Molecular Medicine, Robert-Rössle-Str. 10, Berlin 13125, Germany
| | - Shaun Mahony
- Center for Eukaryotic Gene Regulation, Department of Biochemistry & Molecular Biology, Penn State University, Pennsylvania, USA
| | - Esteban O Mazzoni
- Department of Biology, New York University, 100 Washington Square East, New York, NY 10003, USA
| |
Collapse
|
384
|
Campbell KR, Yau C. Order Under Uncertainty: Robust Differential Expression Analysis Using Probabilistic Models for Pseudotime Inference. PLoS Comput Biol 2016; 12:e1005212. [PMID: 27870852 PMCID: PMC5117567 DOI: 10.1371/journal.pcbi.1005212] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2016] [Accepted: 10/13/2016] [Indexed: 11/18/2022] Open
Abstract
Single cell gene expression profiling can be used to quantify transcriptional dynamics in temporal processes, such as cell differentiation, using computational methods to label each cell with a ‘pseudotime’ where true time series experimentation is too difficult to perform. However, owing to the high variability in gene expression between individual cells, there is an inherent uncertainty in the precise temporal ordering of the cells. Pre-existing methods for pseudotime estimation have predominantly given point estimates precluding a rigorous analysis of the implications of uncertainty. We use probabilistic modelling techniques to quantify pseudotime uncertainty and propagate this into downstream differential expression analysis. We demonstrate that reliance on a point estimate of pseudotime can lead to inflated false discovery rates and that probabilistic approaches provide greater robustness and measures of the temporal resolution that can be obtained from pseudotime inference. Understanding the “cellular programming” that controls fundamental, dynamic biological processes is important for determining normal cellular function and potential perturbations that might give rise to physiological disorders. Ideally, investigations would employ time series experiments to periodically measure the properties of each cell. This would allow us to understand the sequence of gene (in)activations that constitute the program being followed. In practice, such experiments can be difficult to perform as cellular activity may be asynchronous with each cell occupying a different phase of the process of interested. Furthermore, the unbiased measurement of all transcripts or proteins requires the cells to be captured and lysed precluding the continued monitoring of that cell. In the absence of the ability to conduct true time series experiments, pseudotime algorithms exploit the asynchronous cellular nature of these systems to mathematically assign a “pseudotime” to each cell based on its molecular profile allowing the cells to be aligned and the sequence of gene activation events retrospectively inferred. Existing approaches predominantly use deterministic methods that ignore the statistical uncertainties associated with the problem. This paper demonstrates that this statistical uncertainty limits the temporal resolution that can be extracted from static snapshots of cell expression profiles and can also detrimentally affect downstream analysis.
Collapse
Affiliation(s)
- Kieran R. Campbell
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, United Kingdom
- Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford, United Kingdom
| | - Christopher Yau
- Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford, United Kingdom
- Department of Statistics, University of Oxford, Oxford, United Kingdom
- * E-mail:
| |
Collapse
|
385
|
Cannoodt R, Saelens W, Saeys Y. Computational methods for trajectory inference from single-cell transcriptomics. Eur J Immunol 2016; 46:2496-2506. [DOI: 10.1002/eji.201646347] [Citation(s) in RCA: 125] [Impact Index Per Article: 13.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2016] [Revised: 08/30/2016] [Accepted: 09/26/2016] [Indexed: 12/22/2022]
Affiliation(s)
- Robrecht Cannoodt
- Data Mining and Modelling for Biomedicine group; VIB Inflammation Research Center; Ghent Belgium
- Department of Internal Medicine; Ghent University; Ghent Belgium
- Center for Medical Genetics; Ghent University; Ghent Belgium
- Cancer Research Institute Ghent (CRIG); Ghent Belgium
| | - Wouter Saelens
- Data Mining and Modelling for Biomedicine group; VIB Inflammation Research Center; Ghent Belgium
- Department of Internal Medicine; Ghent University; Ghent Belgium
| | - Yvan Saeys
- Data Mining and Modelling for Biomedicine group; VIB Inflammation Research Center; Ghent Belgium
- Department of Internal Medicine; Ghent University; Ghent Belgium
| |
Collapse
|
386
|
Shao C, Höfer T. Robust classification of single-cell transcriptome data by nonnegative matrix factorization. Bioinformatics 2016; 33:235-242. [DOI: 10.1093/bioinformatics/btw607] [Citation(s) in RCA: 76] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2016] [Revised: 09/15/2016] [Accepted: 09/16/2016] [Indexed: 11/14/2022] Open
|
387
|
Chen H, Lau MC, Wong MT, Newell EW, Poidinger M, Chen J. Cytofkit: A Bioconductor Package for an Integrated Mass Cytometry Data Analysis Pipeline. PLoS Comput Biol 2016; 12:e1005112. [PMID: 27662185 PMCID: PMC5035035 DOI: 10.1371/journal.pcbi.1005112] [Citation(s) in RCA: 263] [Impact Index Per Article: 29.2] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2016] [Accepted: 08/22/2016] [Indexed: 11/18/2022] Open
Abstract
Single-cell mass cytometry significantly increases the dimensionality of cytometry analysis as compared to fluorescence flow cytometry, providing unprecedented resolution of cellular diversity in tissues. However, analysis and interpretation of these high-dimensional data poses a significant technical challenge. Here, we present cytofkit, a new Bioconductor package, which integrates both state-of-the-art bioinformatics methods and in-house novel algorithms to offer a comprehensive toolset for mass cytometry data analysis. Cytofkit provides functions for data pre-processing, data visualization through linear or non-linear dimensionality reduction, automatic identification of cell subsets, and inference of the relatedness between cell subsets. This pipeline also provides a graphical user interface (GUI) for ease of use, as well as a shiny application (APP) for interactive visualization of cell subpopulations and progression profiles of key markers. Applied to a CD14−CD19− PBMCs dataset, cytofkit accurately identified different subsets of lymphocytes; applied to a human CD4+ T cell dataset, cytofkit uncovered multiple subtypes of TFH cells spanning blood and tonsils. Cytofkit is implemented in R, licensed under the Artistic license 2.0, and freely available from the Bioconductor website, https://bioconductor.org/packages/cytofkit/. Cytofkit is also applicable for flow cytometry data analysis.
Collapse
Affiliation(s)
- Hao Chen
- Agency for Science, Technology and Research (A*STAR), Singapore Immunology Network (SIgN), Singapore
| | - Mai Chan Lau
- Agency for Science, Technology and Research (A*STAR), Singapore Immunology Network (SIgN), Singapore
| | - Michael Thomas Wong
- Agency for Science, Technology and Research (A*STAR), Singapore Immunology Network (SIgN), Singapore
| | - Evan W. Newell
- Agency for Science, Technology and Research (A*STAR), Singapore Immunology Network (SIgN), Singapore
| | - Michael Poidinger
- Agency for Science, Technology and Research (A*STAR), Singapore Immunology Network (SIgN), Singapore
| | - Jinmiao Chen
- Agency for Science, Technology and Research (A*STAR), Singapore Immunology Network (SIgN), Singapore
- * E-mail:
| |
Collapse
|
388
|
Moignard V, Göttgens B. Dissecting stem cell differentiation using single cell expression profiling. Curr Opin Cell Biol 2016; 43:78-86. [PMID: 27665068 DOI: 10.1016/j.ceb.2016.08.005] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2016] [Revised: 06/15/2016] [Accepted: 08/19/2016] [Indexed: 01/08/2023]
Abstract
Many assumptions about the way cells behave are based on analyses of populations. However, it is now widely recognized that even apparently pure populations can display a remarkable level of heterogeneity. This is particularly true in stem cell biology where it hinders our understanding of normal development and the development of strategies for regenerative medicine. Over the past decade technologies facilitating gene expression analysis at the single cell level have become widespread, providing access to rare cell populations and insights into population structure and function. Here we review the contributions of single cell biology to understanding stem cell differentiation so far, both as a new methodology for defining cell types and a tool for understanding the complexities of cellular decision-making.
Collapse
Affiliation(s)
- Victoria Moignard
- Department of Haematology and Wellcome Trust - Medical Research Council Cambridge Stem Cell Institute, Cambridge Institute for Medical Research, University of Cambridge, Cambridge, UK
| | - Berthold Göttgens
- Department of Haematology and Wellcome Trust - Medical Research Council Cambridge Stem Cell Institute, Cambridge Institute for Medical Research, University of Cambridge, Cambridge, UK.
| |
Collapse
|
389
|
Poirion OB, Zhu X, Ching T, Garmire L. Single-Cell Transcriptomics Bioinformatics and Computational Challenges. Front Genet 2016; 7:163. [PMID: 27708664 PMCID: PMC5030210 DOI: 10.3389/fgene.2016.00163] [Citation(s) in RCA: 71] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2016] [Accepted: 09/02/2016] [Indexed: 12/21/2022] Open
Abstract
The emerging single-cell RNA-Seq (scRNA-Seq) technology holds the promise to revolutionize our understanding of diseases and associated biological processes at an unprecedented resolution. It opens the door to reveal intercellular heterogeneity and has been employed to a variety of applications, ranging from characterizing cancer cells subpopulations to elucidating tumor resistance mechanisms. Parallel to improving experimental protocols to deal with technological issues, deriving new analytical methods to interpret the complexity in scRNA-Seq data is just as challenging. Here, we review current state-of-the-art bioinformatics tools and methods for scRNA-Seq analysis, as well as addressing some critical analytical challenges that the field faces.
Collapse
Affiliation(s)
- Olivier B Poirion
- Epidemiology Program, University of Hawaii Cancer Center Honolulu, HI, USA
| | - Xun Zhu
- Epidemiology Program, University of Hawaii Cancer CenterHonolulu, HI, USA; Molecular Biosciences and Bioengineering Graduate Program, University of Hawaii at ManoaHonolulu, HI, USA
| | - Travers Ching
- Epidemiology Program, University of Hawaii Cancer CenterHonolulu, HI, USA; Molecular Biosciences and Bioengineering Graduate Program, University of Hawaii at ManoaHonolulu, HI, USA
| | - Lana Garmire
- Epidemiology Program, University of Hawaii Cancer Center Honolulu, HI, USA
| |
Collapse
|
390
|
Haghverdi L, Büttner M, Wolf FA, Buettner F, Theis FJ. Diffusion pseudotime robustly reconstructs lineage branching. Nat Methods 2016; 13:845-8. [PMID: 27571553 DOI: 10.1038/nmeth.3971] [Citation(s) in RCA: 759] [Impact Index Per Article: 84.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2016] [Accepted: 07/20/2016] [Indexed: 12/15/2022]
Abstract
The temporal order of differentiating cells is intrinsically encoded in their single-cell expression profiles. We describe an efficient way to robustly estimate this order according to diffusion pseudotime (DPT), which measures transitions between cells using diffusion-like random walks. Our DPT software implementations make it possible to reconstruct the developmental progression of cells and identify transient or metastable states, branching decisions and differentiation endpoints.
Collapse
Affiliation(s)
- Laleh Haghverdi
- Helmholtz Zentrum München, German Research Center for Environmental Health, Institute of Computational Biology, Neuherberg, Germany.,Department of Mathematics, Technische Universität München, Munich, Germany
| | - Maren Büttner
- Helmholtz Zentrum München, German Research Center for Environmental Health, Institute of Computational Biology, Neuherberg, Germany
| | - F Alexander Wolf
- Helmholtz Zentrum München, German Research Center for Environmental Health, Institute of Computational Biology, Neuherberg, Germany
| | - Florian Buettner
- Helmholtz Zentrum München, German Research Center for Environmental Health, Institute of Computational Biology, Neuherberg, Germany.,European Molecular Biology Laboratory, European Bioinformatics Institute, Wellcome Trust Genome Campus, Hinxton, Cambridge, UK
| | - Fabian J Theis
- Helmholtz Zentrum München, German Research Center for Environmental Health, Institute of Computational Biology, Neuherberg, Germany.,Department of Mathematics, Technische Universität München, Munich, Germany
| |
Collapse
|
391
|
Perié L, Duffy KR. Retracing thein vivohaematopoietic tree using single-cell methods. FEBS Lett 2016; 590:4068-4083. [DOI: 10.1002/1873-3468.12299] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2016] [Revised: 07/08/2016] [Accepted: 07/09/2016] [Indexed: 12/14/2022]
Affiliation(s)
- Leïla Perié
- Institut Curie; PSL Research University; CNRS UMR168; Paris France
- Sorbonne Universités; UPMC Univ Paris 06; France
| | - Ken R. Duffy
- Hamilton Institute; Maynooth University; Co Kildare Ireland
| |
Collapse
|
392
|
Scialdone A, Tanaka Y, Jawaid W, Moignard V, Wilson NK, Macaulay IC, Marioni JC, Göttgens B. Resolving early mesoderm diversification through single-cell expression profiling. Nature 2016; 535:289-293. [PMID: 27383781 PMCID: PMC4947525 DOI: 10.1038/nature18633] [Citation(s) in RCA: 198] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2016] [Accepted: 06/09/2016] [Indexed: 12/21/2022]
Abstract
In mammals, specification of the three major germ layers occurs during gastrulation, when cells ingressing through the primitive streak differentiate into the precursor cells of major organ systems. However, the molecular mechanisms underlying this process remain unclear, as numbers of gastrulating cells are very limited. In the mouse embryo at embryonic day 6.5, cells located at the junction between the extra-embryonic region and the epiblast on the posterior side of the embryo undergo an epithelial-to-mesenchymal transition and ingress through the primitive streak. Subsequently, cells migrate, either surrounding the prospective ectoderm contributing to the embryo proper, or into the extra-embryonic region to form the yolk sac, umbilical cord and placenta. Fate mapping has shown that mature tissues such as blood and heart originate from specific regions of the pre-gastrula epiblast, but the plasticity of cells within the embryo and the function of key cell-type-specific transcription factors remain unclear. Here we analyse 1,205 cells from the epiblast and nascent Flk1(+) mesoderm of gastrulating mouse embryos using single-cell RNA sequencing, representing the first transcriptome-wide in vivo view of early mesoderm formation during mammalian gastrulation. Additionally, using knockout mice, we study the function of Tal1, a key haematopoietic transcription factor, and demonstrate, contrary to previous studies performed using retrospective assays, that Tal1 knockout does not immediately bias precursor cells towards a cardiac fate.
Collapse
Affiliation(s)
- Antonio Scialdone
- EMBL-European Bioinformatics Institute (EMBL-EBI), Wellcome Trust
Genome Campus, Cambridge, UK
- Wellcome Trust Sanger Institute, Hinxton, Cambridge, UK
| | - Yosuke Tanaka
- Department of Haematology, Cambridge Institute for Medical Research,
University of Cambridge, Cambridge, UK
- Wellcome Trust - Medical Research Council Cambridge Stem Cell
Institute, University of Cambridge, Cambridge, UK
| | - Wajid Jawaid
- Department of Haematology, Cambridge Institute for Medical Research,
University of Cambridge, Cambridge, UK
- Wellcome Trust - Medical Research Council Cambridge Stem Cell
Institute, University of Cambridge, Cambridge, UK
| | - Victoria Moignard
- Department of Haematology, Cambridge Institute for Medical Research,
University of Cambridge, Cambridge, UK
- Wellcome Trust - Medical Research Council Cambridge Stem Cell
Institute, University of Cambridge, Cambridge, UK
| | - Nicola K. Wilson
- Department of Haematology, Cambridge Institute for Medical Research,
University of Cambridge, Cambridge, UK
- Wellcome Trust - Medical Research Council Cambridge Stem Cell
Institute, University of Cambridge, Cambridge, UK
| | | | - John C. Marioni
- EMBL-European Bioinformatics Institute (EMBL-EBI), Wellcome Trust
Genome Campus, Cambridge, UK
- Wellcome Trust Sanger Institute, Hinxton, Cambridge, UK
- CRUK Cambridge Institute, University of Cambridge, Cambridge,
UK
| | - Berthold Göttgens
- Department of Haematology, Cambridge Institute for Medical Research,
University of Cambridge, Cambridge, UK
- Wellcome Trust - Medical Research Council Cambridge Stem Cell
Institute, University of Cambridge, Cambridge, UK
| |
Collapse
|
393
|
Single-cell genome-wide studies give new insight into nongenetic cell-to-cell variability in animals. Histochem Cell Biol 2016; 146:239-54. [DOI: 10.1007/s00418-016-1466-z] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/06/2016] [Indexed: 01/21/2023]
|
394
|
Chen J, Schlitzer A, Chakarov S, Ginhoux F, Poidinger M. Mpath maps multi-branching single-cell trajectories revealing progenitor cell progression during development. Nat Commun 2016; 7:11988. [PMID: 27356503 PMCID: PMC4931327 DOI: 10.1038/ncomms11988] [Citation(s) in RCA: 48] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2015] [Accepted: 05/18/2016] [Indexed: 01/07/2023] Open
Abstract
Single-cell RNA-sequencing offers unprecedented resolution of the continuum of state transition during cell differentiation and development. However, tools for constructing multi-branching cell lineages from single-cell data are limited. Here we present Mpath, an algorithm that derives multi-branching developmental trajectories using neighborhood-based cell state transitions. Applied to mouse conventional dendritic cell (cDC) progenitors, Mpath constructs multi-branching trajectories spanning from macrophage/DC progenitors through common DC progenitor to pre-dendritic cells (preDC). The Mpath-generated trajectories detect a branching event at the preDC stage revealing preDC subsets that are exclusively committed to cDC1 or cDC2 lineages. Reordering cells along cDC development reveals sequential waves of gene regulation and temporal coupling between cell cycle and cDC differentiation. Applied to human myoblasts, Mpath recapitulates the time course of myoblast differentiation and isolates a branch of non-muscle cells involved in the differentiation. Our study shows that Mpath is a useful tool for constructing cell lineages from single-cell data. The tools for constructing cell lineages from single cell data are limited. Here, the authors present Mpath: an algorithm to derive multi-branching trajectories using neighbourhood-based cell state transitions, and use this to predict developmental trajectories in dendritic and muscle cells.
Collapse
Affiliation(s)
- Jinmiao Chen
- Singapore Immunology Network (SIgN), Agency for Science, Technology and Research (A*STAR), 8A Biomedical Grove, #03-06, Singapore 138648, Singapore
| | - Andreas Schlitzer
- Singapore Immunology Network (SIgN), Agency for Science, Technology and Research (A*STAR), 8A Biomedical Grove, #03-06, Singapore 138648, Singapore
| | - Svetoslav Chakarov
- Singapore Immunology Network (SIgN), Agency for Science, Technology and Research (A*STAR), 8A Biomedical Grove, #03-06, Singapore 138648, Singapore
| | - Florent Ginhoux
- Singapore Immunology Network (SIgN), Agency for Science, Technology and Research (A*STAR), 8A Biomedical Grove, #03-06, Singapore 138648, Singapore
| | - Michael Poidinger
- Singapore Immunology Network (SIgN), Agency for Science, Technology and Research (A*STAR), 8A Biomedical Grove, #03-06, Singapore 138648, Singapore
| |
Collapse
|
395
|
A single-cell resolution map of mouse hematopoietic stem and progenitor cell differentiation. Blood 2016; 128:e20-31. [PMID: 27365425 DOI: 10.1182/blood-2016-05-716480] [Citation(s) in RCA: 506] [Impact Index Per Article: 56.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2016] [Accepted: 06/28/2016] [Indexed: 02/08/2023] Open
Abstract
Maintenance of the blood system requires balanced cell fate decisions by hematopoietic stem and progenitor cells (HSPCs). Because cell fate choices are executed at the individual cell level, new single-cell profiling technologies offer exciting possibilities for mapping the dynamic molecular changes underlying HSPC differentiation. Here, we have used single-cell RNA sequencing to profile more than 1600 single HSPCs, and deep sequencing has enabled detection of an average of 6558 protein-coding genes per cell. Index sorting, in combination with broad sorting gates, allowed us to retrospectively assign cells to 12 commonly sorted HSPC phenotypes while also capturing intermediate cells typically excluded by conventional gating. We further show that independently generated single-cell data sets can be projected onto the single-cell resolution expression map to directly compare data from multiple groups and to build and refine new hypotheses. Reconstruction of differentiation trajectories reveals dynamic expression changes associated with early lymphoid, erythroid, and granulocyte-macrophage differentiation. The latter two trajectories were characterized by common upregulation of cell cycle and oxidative phosphorylation transcriptional programs. By using external spike-in controls, we estimate absolute messenger RNA (mRNA) levels per cell, showing for the first time that despite a general reduction in total mRNA, a subset of genes shows higher expression levels in immature stem cells consistent with active maintenance of the stem-cell state. Finally, we report the development of an intuitive Web interface as a new community resource to permit visualization of gene expression in HSPCs at single-cell resolution for any gene of choice.
Collapse
|
396
|
Grün D, Muraro MJ, Boisset JC, Wiebrands K, Lyubimova A, Dharmadhikari G, van den Born M, van Es J, Jansen E, Clevers H, de Koning EJP, van Oudenaarden A. De Novo Prediction of Stem Cell Identity using Single-Cell Transcriptome Data. Cell Stem Cell 2016; 19:266-277. [PMID: 27345837 PMCID: PMC4985539 DOI: 10.1016/j.stem.2016.05.010] [Citation(s) in RCA: 394] [Impact Index Per Article: 43.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2016] [Revised: 04/04/2016] [Accepted: 05/12/2016] [Indexed: 02/07/2023]
Abstract
Adult mitotic tissues like the intestine, skin, and blood undergo constant turnover throughout the life of an organism. Knowing the identity of the stem cell is crucial to understanding tissue homeostasis and its aberrations upon disease. Here we present a computational method for the derivation of a lineage tree from single-cell transcriptome data. By exploiting the tree topology and the transcriptome composition, we establish StemID, an algorithm for identifying stem cells among all detectable cell types within a population. We demonstrate that StemID recovers two known adult stem cell populations, Lgr5+ cells in the small intestine and hematopoietic stem cells in the bone marrow. We apply StemID to predict candidate multipotent cell populations in the human pancreas, a tissue with largely uncharacterized turnover dynamics. We hope that StemID will accelerate the search for novel stem cells by providing concrete markers for biological follow-up and validation. StemID infers the lineage tree and identifies stem cells from single-cell mRNA-seq data Direct links of stem cells to distinct sub-types reflect transcriptome plasticity The permissive stem cell transcriptome is characterized by high entropy StemID infers candidate multipotent cell populations in the human pancreas
Collapse
Affiliation(s)
- Dominic Grün
- Hubrecht Institute, Royal Netherlands Academy of Arts and Sciences, 3584 CT Utrecht, the Netherlands; Cancer Genomics Netherlands, University Medical Center Utrecht, 3584 CX Utrecht, the Netherlands; Max Planck Institute of Immunobiology and Epigenetics, 79108 Freiburg, Germany.
| | - Mauro J Muraro
- Hubrecht Institute, Royal Netherlands Academy of Arts and Sciences, 3584 CT Utrecht, the Netherlands; Cancer Genomics Netherlands, University Medical Center Utrecht, 3584 CX Utrecht, the Netherlands
| | - Jean-Charles Boisset
- Hubrecht Institute, Royal Netherlands Academy of Arts and Sciences, 3584 CT Utrecht, the Netherlands; Cancer Genomics Netherlands, University Medical Center Utrecht, 3584 CX Utrecht, the Netherlands
| | - Kay Wiebrands
- Hubrecht Institute, Royal Netherlands Academy of Arts and Sciences, 3584 CT Utrecht, the Netherlands; Cancer Genomics Netherlands, University Medical Center Utrecht, 3584 CX Utrecht, the Netherlands
| | - Anna Lyubimova
- Hubrecht Institute, Royal Netherlands Academy of Arts and Sciences, 3584 CT Utrecht, the Netherlands; Cancer Genomics Netherlands, University Medical Center Utrecht, 3584 CX Utrecht, the Netherlands
| | - Gitanjali Dharmadhikari
- Hubrecht Institute, Royal Netherlands Academy of Arts and Sciences, 3584 CT Utrecht, the Netherlands; Cancer Genomics Netherlands, University Medical Center Utrecht, 3584 CX Utrecht, the Netherlands; Department of Medicine, Section of Nephrology and Section of Endocrinology, Leiden University Medical Center, 2333 ZA Leiden, the Netherlands
| | - Maaike van den Born
- Hubrecht Institute, Royal Netherlands Academy of Arts and Sciences, 3584 CT Utrecht, the Netherlands; Cancer Genomics Netherlands, University Medical Center Utrecht, 3584 CX Utrecht, the Netherlands
| | - Johan van Es
- Hubrecht Institute, Royal Netherlands Academy of Arts and Sciences, 3584 CT Utrecht, the Netherlands; Cancer Genomics Netherlands, University Medical Center Utrecht, 3584 CX Utrecht, the Netherlands
| | - Erik Jansen
- Hubrecht Institute, Royal Netherlands Academy of Arts and Sciences, 3584 CT Utrecht, the Netherlands; Cancer Genomics Netherlands, University Medical Center Utrecht, 3584 CX Utrecht, the Netherlands
| | - Hans Clevers
- Hubrecht Institute, Royal Netherlands Academy of Arts and Sciences, 3584 CT Utrecht, the Netherlands; Cancer Genomics Netherlands, University Medical Center Utrecht, 3584 CX Utrecht, the Netherlands; Princess Maxima Center for Pediatric Oncology, 3508 AB Utrecht, the Netherlands
| | - Eelco J P de Koning
- Hubrecht Institute, Royal Netherlands Academy of Arts and Sciences, 3584 CT Utrecht, the Netherlands; Cancer Genomics Netherlands, University Medical Center Utrecht, 3584 CX Utrecht, the Netherlands; Department of Medicine, Section of Nephrology and Section of Endocrinology, Leiden University Medical Center, 2333 ZA Leiden, the Netherlands
| | - Alexander van Oudenaarden
- Hubrecht Institute, Royal Netherlands Academy of Arts and Sciences, 3584 CT Utrecht, the Netherlands; Cancer Genomics Netherlands, University Medical Center Utrecht, 3584 CX Utrecht, the Netherlands.
| |
Collapse
|
397
|
Hamey FK, Nestorowa S, Wilson NK, Göttgens B. Advancing haematopoietic stem and progenitor cell biology through single-cell profiling. FEBS Lett 2016; 590:4052-4067. [PMID: 27259698 DOI: 10.1002/1873-3468.12231] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2016] [Revised: 05/27/2016] [Accepted: 05/31/2016] [Indexed: 12/25/2022]
Abstract
Haematopoietic stem and progenitor cells (HSPCs) sit at the top of the haematopoietic hierarchy, and their fate choices need to be carefully controlled to ensure balanced production of all mature blood cell types. As cell fate decisions are made at the level of the individual cells, recent technological advances in measuring gene and protein expression in increasingly large numbers of single cells have been rapidly adopted to study both normal and pathological HSPC function. In this review we emphasise the importance of combining the correct computational models with single-cell experimental techniques, and illustrate how such integrated approaches have been used to resolve heterogeneities in populations, reconstruct lineage differentiation, identify regulatory relationships and link molecular profiling to cellular function.
Collapse
Affiliation(s)
- Fiona K Hamey
- Department of Haematology and Wellcome Trust - MRC Cambridge Stem Cell Institute, University of Cambridge, UK
| | - Sonia Nestorowa
- Department of Haematology and Wellcome Trust - MRC Cambridge Stem Cell Institute, University of Cambridge, UK
| | - Nicola K Wilson
- Department of Haematology and Wellcome Trust - MRC Cambridge Stem Cell Institute, University of Cambridge, UK
| | - Berthold Göttgens
- Department of Haematology and Wellcome Trust - MRC Cambridge Stem Cell Institute, University of Cambridge, UK
| |
Collapse
|
398
|
Ntranos V, Kamath GM, Zhang JM, Pachter L, Tse DN. Fast and accurate single-cell RNA-seq analysis by clustering of transcript-compatibility counts. Genome Biol 2016; 17:112. [PMID: 27230763 PMCID: PMC4881296 DOI: 10.1186/s13059-016-0970-8] [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: 02/24/2016] [Accepted: 04/29/2016] [Indexed: 12/17/2022] Open
Abstract
Current approaches to single-cell transcriptomic analysis are computationally intensive and require assay-specific modeling, which limits their scope and generality. We propose a novel method that compares and clusters cells based on their transcript-compatibility read counts rather than on the transcript or gene quantifications used in standard analysis pipelines. In the reanalysis of two landmark yet disparate single-cell RNA-seq datasets, we show that our method is up to two orders of magnitude faster than previous approaches, provides accurate and in some cases improved results, and is directly applicable to data from a wide variety of assays.
Collapse
Affiliation(s)
- Vasilis Ntranos
- Department of Electrical Engineering and Computer Sciences, University of California, Berkeley, CA, USA
| | - Govinda M Kamath
- Department of Electrical Engineering, Stanford University, Stanford, CA, USA
| | - Jesse M Zhang
- Department of Electrical Engineering, Stanford University, Stanford, CA, USA
| | - Lior Pachter
- Departments of Mathematics and Molecular and Cell Biology, University of California, Berkeley, CA, USA.
| | - David N Tse
- Department of Electrical Engineering and Computer Sciences, University of California, Berkeley, CA, USA. .,Department of Electrical Engineering, Stanford University, Stanford, CA, USA.
| |
Collapse
|
399
|
Single-cell gene expression profiling and cell state dynamics: collecting data, correlating data points and connecting the dots. Curr Opin Biotechnol 2016; 39:207-214. [PMID: 27152696 DOI: 10.1016/j.copbio.2016.04.015] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2016] [Revised: 04/13/2016] [Accepted: 04/14/2016] [Indexed: 01/28/2023]
Abstract
Single-cell analyses of transcript and protein expression profiles-more precisely, single-cell resolution analysis of molecular profiles of cell populations-have now entered the center stage with widespread applications of single-cell qPCR, single-cell RNA-Seq and CyTOF. These high-dimensional population snapshot techniques are complemented by low-dimensional time-resolved, microscopy-based monitoring methods. Both fronts of advance have exposed a rich heterogeneity of cell states within uniform cell populations in many biological contexts, producing a new kind of data that has triggered computational analysis methods for data visualization, dimensionality reduction, and cluster (subpopulation) identification. The next step is now to go beyond collecting data and correlating data points: to connect the dots, that is, to understand what actually underlies the identified data patterns. This entails interpreting the 'clouds of points' in state space as a manifestation of the underlying molecular regulatory network. In that way control of cell state dynamics can be formalized as a quasi-potential landscape, as first proposed by Waddington. We summarize key methods of data acquisition and computational analysis and explain the principles that link the single-cell resolution measurements to dynamical systems theory.
Collapse
|
400
|
Ji Z, Ji H. TSCAN: Pseudo-time reconstruction and evaluation in single-cell RNA-seq analysis. Nucleic Acids Res 2016; 44:e117. [PMID: 27179027 PMCID: PMC4994863 DOI: 10.1093/nar/gkw430] [Citation(s) in RCA: 415] [Impact Index Per Article: 46.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2015] [Accepted: 05/05/2016] [Indexed: 12/21/2022] Open
Abstract
When analyzing single-cell RNA-seq data, constructing a pseudo-temporal path to order
cells based on the gradual transition of their transcriptomes is a useful way to study
gene expression dynamics in a heterogeneous cell population. Currently, a limited number
of computational tools are available for this task, and quantitative methods for comparing
different tools are lacking. Tools for Single Cell Analysis (TSCAN) is a software tool
developed to better support in silico
pseudo-Time reconstruction in
Single-Cell RNA-seq
ANalysis. TSCAN uses a cluster-based minimum spanning tree (MST)
approach to order cells. Cells are first grouped into clusters and an MST is then
constructed to connect cluster centers. Pseudo-time is obtained by projecting each cell
onto the tree, and the ordered sequence of cells can be used to study dynamic changes of
gene expression along the pseudo-time. Clustering cells before MST construction reduces
the complexity of the tree space. This often leads to improved cell ordering. It also
allows users to conveniently adjust the ordering based on prior knowledge. TSCAN has a
graphical user interface (GUI) to support data visualization and user interaction.
Furthermore, quantitative measures are developed to objectively evaluate and compare
different pseudo-time reconstruction methods. TSCAN is available at https://github.com/zji90/TSCAN and as a Bioconductor package.
Collapse
Affiliation(s)
- Zhicheng Ji
- Department of Biostatistics, Johns Hopkins University Bloomberg School of Public Health, 615 North Wolfe Street, Baltimore, MD 21205, USA
| | - Hongkai Ji
- Department of Biostatistics, Johns Hopkins University Bloomberg School of Public Health, 615 North Wolfe Street, Baltimore, MD 21205, USA
| |
Collapse
|