51
|
|
52
|
Hegenbarth JC, Lezzoche G, De Windt LJ, Stoll M. Perspectives on Bulk-Tissue RNA Sequencing and Single-Cell RNA Sequencing for Cardiac Transcriptomics. FRONTIERS IN MOLECULAR MEDICINE 2022; 2:839338. [PMID: 39086967 PMCID: PMC11285642 DOI: 10.3389/fmmed.2022.839338] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/19/2021] [Accepted: 01/31/2022] [Indexed: 08/02/2024]
Abstract
The heart has been the center of numerous transcriptomic studies in the past decade. Even though our knowledge of the key organ in our cardiovascular system has significantly increased over the last years, it is still not fully understood yet. In recent years, extensive efforts were made to understand the genetic and transcriptomic contribution to cardiac function and failure in more detail. The advent of Next Generation Sequencing (NGS) technologies has brought many discoveries but it is unable to comprehend the finely orchestrated interactions between and within the various cell types of the heart. With the emergence of single-cell sequencing more than 10 years ago, researchers gained a valuable new tool to enable the exploration of new subpopulations of cells, cell-cell interactions, and integration of multi-omic approaches at a single-cell resolution. Despite this innovation, it is essential to make an informed choice regarding the appropriate technique for transcriptomic studies, especially when working with myocardial tissue. Here, we provide a primer for researchers interested in transcriptomics using NGS technologies.
Collapse
Affiliation(s)
- Jana-Charlotte Hegenbarth
- Department of Molecular Genetics, Faculty of Science and Engineering, Faculty of Health, Medicine and Life Sciences, Maastricht University, Maastricht, Netherlands
| | - Giuliana Lezzoche
- Department of Molecular Genetics, Faculty of Science and Engineering, Faculty of Health, Medicine and Life Sciences, Maastricht University, Maastricht, Netherlands
| | - Leon J. De Windt
- Department of Molecular Genetics, Faculty of Science and Engineering, Faculty of Health, Medicine and Life Sciences, Maastricht University, Maastricht, Netherlands
| | - Monika Stoll
- Department of Biochemistry, CARIM School for Cardiovascular Diseases, Maastricht University, Maastricht, Netherlands
- Department of Genetic Epidemiology, Institute of Human Genetics, University Hospital Münster, Münster, Germany
| |
Collapse
|
53
|
Jackson CA, Vogel C. New horizons in the stormy sea of multimodal single-cell data integration. Mol Cell 2022; 82:248-259. [PMID: 35063095 PMCID: PMC8830781 DOI: 10.1016/j.molcel.2021.12.012] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2021] [Revised: 12/08/2021] [Accepted: 12/13/2021] [Indexed: 01/22/2023]
Abstract
While measurements of RNA expression have dominated the world of single-cell analyses, new single-cell techniques increasingly allow collection of different data modalities, measuring different molecules, structural connections, and intermolecular interactions. Integrating the resulting multimodal single-cell datasets is a new bioinformatics challenge. Equally important, it is a new experimental design challenge for the bench scientist, who is not only choosing from a myriad of techniques for each data modality but also faces new challenges in experimental design. The ultimate goal is to design, execute, and analyze multimodal single-cell experiments that are more than just descriptive but enable the learning of new causal and mechanistic biology. This objective requires strict consideration of the goals behind the analysis, which might range from mapping the heterogeneity of a cellular population to assembling system-wide causal networks that can further our understanding of cellular functions and eventually lead to models of tissues and organs. We review steps and challenges toward this goal. Single-cell transcriptomics is now a mature technology, and methods to measure proteins, lipids, small-molecule metabolites, and other molecular phenotypes at the single-cell level are rapidly developing. Integrating these single-cell readouts so that each cell has measurements of multiple types of data, e.g., transcriptomes, proteomes, and metabolomes, is expected to allow identification of highly specific cellular subpopulations and to provide the basis for inferring causal biological mechanisms.
Collapse
Affiliation(s)
- Christopher A Jackson
- New York University, Department of Biology, Center for Genomics and Systems Biology, New York, NY, USA.
| | - Christine Vogel
- New York University, Department of Biology, Center for Genomics and Systems Biology, New York, NY, USA
| |
Collapse
|
54
|
Vermeersch L, Jariani A, Helsen J, Heineike BM, Verstrepen KJ. Single-Cell RNA Sequencing in Yeast Using the 10× Genomics Chromium Device. Methods Mol Biol 2022; 2477:3-20. [PMID: 35524108 DOI: 10.1007/978-1-0716-2257-5_1] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
Single-cell RNA sequencing (scRNA-seq) is emerging as an essential technique for studying the physiology of individual cells in populations. Although well-established and optimized for mammalian cells, research of microorganisms has been faced with major technical challenges for using scRNA-seq, because of their rigid cell wall, smaller cell size and overall lower total RNA content per cell. Here, we describe an easy-to-implement adaptation of the protocol for the yeast Saccharomyces cerevisiae using the 10× Genomics platform, originally optimized for mammalian cells. Introducing Zymolyase, a cell wall-digesting enzyme, to one of the initial steps of single-cell droplet formation allows efficient in-droplet lysis of yeast cells, without affecting the droplet emulsion and further sample processing. In addition, we also describe the downstream data analysis, which combines established scRNA-seq analysis protocols with specific adaptations for yeast, and R-scripts for further secondary analysis of the data.
Collapse
Affiliation(s)
- Lieselotte Vermeersch
- VIB Laboratory for Systems Biology, VIB-KU Leuven Center for Microbiology, Leuven, Belgium
- CMPG Laboratory of Genetics and Genomics, Department M2S, KU Leuven, Leuven, Belgium
| | - Abbas Jariani
- VIB Laboratory for Systems Biology, VIB-KU Leuven Center for Microbiology, Leuven, Belgium
- CMPG Laboratory of Genetics and Genomics, Department M2S, KU Leuven, Leuven, Belgium
| | - Jana Helsen
- VIB Laboratory for Systems Biology, VIB-KU Leuven Center for Microbiology, Leuven, Belgium
- CMPG Laboratory of Genetics and Genomics, Department M2S, KU Leuven, Leuven, Belgium
- CMPG Laboratory of Predictive Genetics and Multicellular Systems, Department M2S, KU Leuven, Leuven, Belgium
| | - Benjamin M Heineike
- Molecular Biology of Metabolism Laboratory, Francis Crick Institute, London, UK
- Quantitative Gene Expression Research Group, MRC London Institute of Medical Sciences (LMS), London, UK
- Quantitative Gene Expression Research Group, Faculty of Medicine, Imperial College London, Institute of Clinical Sciences (ICS), London, UK
| | - Kevin J Verstrepen
- VIB Laboratory for Systems Biology, VIB-KU Leuven Center for Microbiology, Leuven, Belgium.
- CMPG Laboratory of Genetics and Genomics, Department M2S, KU Leuven, Leuven, Belgium.
| |
Collapse
|
55
|
Dohn R, Xie B, Back R, Selewa A, Eckart H, Rao RP, Basu A. mDrop-Seq: Massively Parallel Single-Cell RNA-Seq of Saccharomyces cerevisiae and Candida albicans. Vaccines (Basel) 2021; 10:vaccines10010030. [PMID: 35062691 PMCID: PMC8779198 DOI: 10.3390/vaccines10010030] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2021] [Revised: 12/18/2021] [Accepted: 12/22/2021] [Indexed: 11/16/2022] Open
Abstract
Advances in high-throughput single-cell RNA sequencing (scRNA-seq) have been limited by technical challenges such as tough cell walls and low RNA quantity that prevent transcriptomic profiling of microbial species at throughput. We present microbial Drop-seq or mDrop-seq, a high-throughput scRNA-seq technique that is demonstrated on two yeast species, Saccharomyces cerevisiae, a popular model organism, and Candida albicans, a common opportunistic pathogen. We benchmarked mDrop-seq for sensitivity and specificity and used it to profile 35,109 S. cerevisiae cells to detect variation in mRNA levels between them. As a proof of concept, we quantified expression differences in heat shock S. cerevisiae using mDrop-seq. We detected differential activation of stress response genes within a seemingly homogenous population of S. cerevisiae under heat shock. We also applied mDrop-seq to C. albicans cells, a polymorphic and clinically relevant species of yeast with a thicker cell wall compared to S. cerevisiae. Single-cell transcriptomes in 39,705 C. albicans cells were characterized using mDrop-seq under different conditions, including exposure to fluconazole, a common anti-fungal drug. We noted differential regulation in stress response and drug target pathways between C. albicans cells, changes in cell cycle patterns and marked increases in histone activity when treated with fluconazole. We demonstrate mDrop-seq to be an affordable and scalable technique that can quantify the variability in gene expression in different yeast species. We hope that mDrop-seq will lead to a better understanding of genetic variation in pathogens in response to stimuli and find immediate applications in investigating drug resistance, infection outcome and developing new drugs and treatment strategies.
Collapse
Affiliation(s)
- Ryan Dohn
- Section of Genetic Medicine, Department of Medicine, University of Chicago, Chicago, IL 60637, USA; (B.X.); (R.B.); (A.S.); (H.E.); (A.B.)
- Committee on Genetics, Genomics and Systems Biology, University of Chicago, Chicago, IL 60637, USA
- Correspondence:
| | - Bingqing Xie
- Section of Genetic Medicine, Department of Medicine, University of Chicago, Chicago, IL 60637, USA; (B.X.); (R.B.); (A.S.); (H.E.); (A.B.)
| | - Rebecca Back
- Section of Genetic Medicine, Department of Medicine, University of Chicago, Chicago, IL 60637, USA; (B.X.); (R.B.); (A.S.); (H.E.); (A.B.)
| | - Alan Selewa
- Section of Genetic Medicine, Department of Medicine, University of Chicago, Chicago, IL 60637, USA; (B.X.); (R.B.); (A.S.); (H.E.); (A.B.)
- Biophysical Sciences Graduate Program, University of Chicago, Chicago, IL 60637, USA
| | - Heather Eckart
- Section of Genetic Medicine, Department of Medicine, University of Chicago, Chicago, IL 60637, USA; (B.X.); (R.B.); (A.S.); (H.E.); (A.B.)
| | - Reeta Prusty Rao
- Department of Biology and Biotechnology, Worcester Polytechnic Institute, Worcester, MA 01609, USA;
| | - Anindita Basu
- Section of Genetic Medicine, Department of Medicine, University of Chicago, Chicago, IL 60637, USA; (B.X.); (R.B.); (A.S.); (H.E.); (A.B.)
- Committee on Genetics, Genomics and Systems Biology, University of Chicago, Chicago, IL 60637, USA
- Biophysical Sciences Graduate Program, University of Chicago, Chicago, IL 60637, USA
| |
Collapse
|
56
|
Olivares-Yañez C, Sánchez E, Pérez-Lara G, Seguel A, Camejo PY, Larrondo LF, Vidal EA, Canessa P. A comprehensive transcription factor and DNA-binding motif resource for the construction of gene regulatory networks in Botrytis cinerea and Trichoderma atroviride. Comput Struct Biotechnol J 2021; 19:6212-6228. [PMID: 34900134 PMCID: PMC8637145 DOI: 10.1016/j.csbj.2021.11.012] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2021] [Revised: 11/11/2021] [Accepted: 11/11/2021] [Indexed: 11/25/2022] Open
Abstract
Botrytis cinerea and Trichoderma atroviride are two relevant fungi in agricultural systems. To gain insights into these organisms' transcriptional gene regulatory networks (GRNs), we generated a manually curated transcription factor (TF) dataset for each of them, followed by a GRN inference utilizing available sequence motifs describing DNA-binding specificity and global gene expression data. As a proof of concept of the usefulness of this resource to pinpoint key transcriptional regulators, we employed publicly available transcriptomics data and a newly generated dual RNA-seq dataset to build context-specific Botrytis and Trichoderma GRNs under two different biological paradigms: exposure to continuous light and Botrytis-Trichoderma confrontation assays. Network analysis of fungal responses to constant light revealed striking differences in the transcriptional landscape of both fungi. On the other hand, we found that the confrontation of both microorganisms elicited a distinct set of differentially expressed genes with changes in T. atroviride exceeding those in B. cinerea. Using our regulatory network data, we were able to determine, in both fungi, central TFs involved in this interaction response, including TFs controlling a large set of extracellular peptidases in the biocontrol agent T. atroviride. In summary, our work provides a comprehensive catalog of transcription factors and regulatory interactions for both organisms. This catalog can now serve as a basis for generating novel hypotheses on transcriptional regulatory circuits in different experimental contexts.
Collapse
Affiliation(s)
- Consuelo Olivares-Yañez
- ANID - Millennium Science Initiative Program - Millennium Institute for Integrative Biology (iBio), Avda. Libertador Bernardo O'Higgins 340, Santiago, Chile.,Centro de Biotecnologia Vegetal, Universidad Andres Bello, Republica 330, Santiago, Chile
| | - Evelyn Sánchez
- ANID - Millennium Science Initiative Program - Millennium Institute for Integrative Biology (iBio), Avda. Libertador Bernardo O'Higgins 340, Santiago, Chile.,Centro de Genomica y Bioinformatica, Facultad de Ciencias, Universidad Mayor, Camino la Pirámide 5750, Huechuraba, Santiago, Chile
| | - Gabriel Pérez-Lara
- ANID - Millennium Science Initiative Program - Millennium Institute for Integrative Biology (iBio), Avda. Libertador Bernardo O'Higgins 340, Santiago, Chile.,Centro de Biotecnologia Vegetal, Universidad Andres Bello, Republica 330, Santiago, Chile
| | - Aldo Seguel
- ANID - Millennium Science Initiative Program - Millennium Institute for Integrative Biology (iBio), Avda. Libertador Bernardo O'Higgins 340, Santiago, Chile.,Departamento de Genetica Molecular y Microbiologia, Facultad de Ciencias Biologicas, Pontificia Universidad Catolica de Chile, Avda. Libertador Bernardo O'Higgins 340, Santiago, Chile
| | - Pamela Y Camejo
- ANID - Millennium Science Initiative Program - Millennium Institute for Integrative Biology (iBio), Avda. Libertador Bernardo O'Higgins 340, Santiago, Chile
| | - Luis F Larrondo
- ANID - Millennium Science Initiative Program - Millennium Institute for Integrative Biology (iBio), Avda. Libertador Bernardo O'Higgins 340, Santiago, Chile.,Departamento de Genetica Molecular y Microbiologia, Facultad de Ciencias Biologicas, Pontificia Universidad Catolica de Chile, Avda. Libertador Bernardo O'Higgins 340, Santiago, Chile
| | - Elena A Vidal
- ANID - Millennium Science Initiative Program - Millennium Institute for Integrative Biology (iBio), Avda. Libertador Bernardo O'Higgins 340, Santiago, Chile.,Centro de Genomica y Bioinformatica, Facultad de Ciencias, Universidad Mayor, Camino la Pirámide 5750, Huechuraba, Santiago, Chile.,Escuela de Biotecnologia, Facultad de Ciencias, Universidad Mayor, Camino la Pirámide 5750, Huechuraba, Santiago, Chile
| | - Paulo Canessa
- ANID - Millennium Science Initiative Program - Millennium Institute for Integrative Biology (iBio), Avda. Libertador Bernardo O'Higgins 340, Santiago, Chile.,Centro de Biotecnologia Vegetal, Universidad Andres Bello, Republica 330, Santiago, Chile
| |
Collapse
|
57
|
Capturing hidden regulation based on noise change of gene expression level from single cell RNA-seq in yeast. Sci Rep 2021; 11:22547. [PMID: 34799619 PMCID: PMC8604932 DOI: 10.1038/s41598-021-01558-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Accepted: 10/29/2021] [Indexed: 11/08/2022] Open
Abstract
Recent progress in high throughput single cell RNA-seq (scRNA-seq) has activated the development of data-driven inferring methods of gene regulatory networks. Most network estimations assume that perturbations produce downstream effects. However, the effects of gene perturbations are sometimes compensated by a gene with redundant functionality (functional compensation). In order to avoid functional compensation, previous studies constructed double gene deletions, but its vast nature of gene combinations was not suitable for comprehensive network estimation. We hypothesized that functional compensation may emerge as a noise change without mean change (noise-only change) due to varying physical properties and strong compensation effects. Here, we show compensated interactions, which are not detected by mean change, are captured by noise-only change quantified from scRNA-seq. We investigated whether noise-only change genes caused by a single deletion of STP1 and STP2, which have strong functional compensation, are enriched in redundantly regulated genes. As a result, noise-only change genes are enriched in their redundantly regulated genes. Furthermore, novel downstream genes detected from noise change are enriched in "transport", which is related to known downstream genes. Herein, we suggest the noise difference comparison has the potential to be applied as a new strategy for network estimation that capture even compensated interaction.
Collapse
|
58
|
Pang AP, Zhang F, Hu X, Luo Y, Wang H, Durrani S, Wu FG, Li BZ, Zhou Z, Lu Z, Lin F. Glutamine involvement in nitrogen regulation of cellulase production in fungi. BIOTECHNOLOGY FOR BIOFUELS 2021; 14:199. [PMID: 34645509 PMCID: PMC8513308 DOI: 10.1186/s13068-021-02046-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/08/2021] [Accepted: 09/23/2021] [Indexed: 05/27/2023]
Abstract
BACKGROUND Cellulase synthesized by fungi can environment-friendly and sustainably degrades cellulose to fermentable sugars for producing cellulosic biofuels, biobased medicine and fine chemicals. Great efforts have been made to study the regulation mechanism of cellulase biosynthesis in fungi with the focus on the carbon sources, while little attention has been paid to the impact and regulation mechanism of nitrogen sources on cellulase production. RESULTS Glutamine displayed the strongest inhibition effect on cellulase biosynthesis in Trichoderma reesei, followed by yeast extract, urea, tryptone, ammonium sulfate and L-glutamate. Cellulase production, cell growth and sporulation in T. reesei RUT-C30 grown on cellulose were all inhibited with the addition of glutamine (a preferred nitrogen source) with no change for mycelium morphology. This inhibition effect was attributed to both L-glutamine itself and the nitrogen excess induced by its presence. In agreement with the reduced cellulase production, the mRNA levels of 44 genes related to the cellulase production were decreased severely in the presence of glutamine. The transcriptional levels of genes involved in other nitrogen transport, ribosomal biogenesis and glutamine biosynthesis were decreased notably by glutamine, while the expression of genes relevant to glutamate biosynthesis, amino acid catabolism, and glutamine catabolism were increased noticeably. Moreover, the transcriptional level of cellulose signaling related proteins ooc1 and ooc2, and the cellular receptor of rapamycin trFKBP12 was increased remarkably, whose deletion exacerbated the cellulase depression influence of glutamine. CONCLUSION Glutamine may well be the metabolite effector in nitrogen repression of cellulase synthesis, like the role of glucose plays in carbon catabolite repression. Glutamine under excess nitrogen condition repressed cellulase biosynthesis significantly as well as cell growth and sporulation in T. reesei RUT-C30. More importantly, the presence of glutamine notably impacted the transport and metabolism of nitrogen. Genes ooc1, ooc2, and trFKBP12 are associated with the cellulase repression impact of glutamine. These findings advance our understanding of nitrogen regulation of cellulase production in filamentous fungi, which would aid in the rational design of strains and fermentation strategies for cellulase production in industry.
Collapse
Affiliation(s)
- Ai-Ping Pang
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, Nanjing, China
| | - Funing Zhang
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, Nanjing, China
| | - Xin Hu
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, Nanjing, China
| | - Yongsheng Luo
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, Nanjing, China
| | - Haiyan Wang
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, Nanjing, China
| | - Samran Durrani
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, Nanjing, China
| | - Fu-Gen Wu
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, Nanjing, China
| | - Bing-Zhi Li
- Key Laboratory of Systems Bioengineering (Ministry of Education), School of Chemical Engineering and Technology, Tianjin University, Tianjin, China
| | - Zhihua Zhou
- Key Laboratory of Synthetic Biology, Institute of Plant Physiology and Ecology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Zuhong Lu
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, Nanjing, China
| | - Fengming Lin
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, Nanjing, China
| |
Collapse
|
59
|
Sieriebriennikov B, Reinberg D, Desplan C. A molecular toolkit for superorganisms. Trends Genet 2021; 37:846-859. [PMID: 34116864 PMCID: PMC8355152 DOI: 10.1016/j.tig.2021.05.005] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2021] [Revised: 05/14/2021] [Accepted: 05/17/2021] [Indexed: 12/16/2022]
Abstract
Social insects, such as ants, bees, wasps, and termites, draw biologists' attention due to their distinctive lifestyles. As experimental systems, they provide unique opportunities to study organismal differentiation, division of labor, longevity, and the evolution of development. Ants are particularly attractive because several ant species can be propagated in the laboratory. However, the same lifestyle that makes social insects interesting also hampers the use of molecular genetic techniques. Here, we summarize the efforts of the ant research community to surmount these hurdles and obtain novel mechanistic insight into the biology of social insects. We review current approaches and propose novel ones involving genomics, transcriptomics, chromatin and DNA methylation profiling, RNA interference (RNAi), and genome editing in ants and discuss future experimental strategies.
Collapse
Affiliation(s)
- Bogdan Sieriebriennikov
- Department of Biochemistry and Molecular Pharmacology, NYU Grossman School of Medicine, New York, NY, USA; Department of Biology, New York University, New York, NY, USA
| | - Danny Reinberg
- Department of Biochemistry and Molecular Pharmacology, NYU Grossman School of Medicine, New York, NY, USA; Howard Hughes Medical Institute, NYU Grossman School of Medicine, New York, NY, USA.
| | - Claude Desplan
- Department of Biology, New York University, New York, NY, USA.
| |
Collapse
|
60
|
Duveau F, Vande Zande P, Metzger BP, Diaz CJ, Walker EA, Tryban S, Siddiq MA, Yang B, Wittkopp PJ. Mutational sources of trans-regulatory variation affecting gene expression in Saccharomyces cerevisiae. eLife 2021; 10:67806. [PMID: 34463616 PMCID: PMC8456550 DOI: 10.7554/elife.67806] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2021] [Accepted: 08/03/2021] [Indexed: 12/15/2022] Open
Abstract
Heritable variation in a gene’s expression arises from mutations impacting cis- and trans-acting components of its regulatory network. Here, we investigate how trans-regulatory mutations are distributed within the genome and within a gene regulatory network by identifying and characterizing 69 mutations with trans-regulatory effects on expression of the same focal gene in Saccharomyces cerevisiae. Relative to 1766 mutations without effects on expression of this focal gene, we found that these trans-regulatory mutations were enriched in coding sequences of transcription factors previously predicted to regulate expression of the focal gene. However, over 90% of the trans-regulatory mutations identified mapped to other types of genes involved in diverse biological processes including chromatin state, metabolism, and signal transduction. These data show how genetic changes in diverse types of genes can impact a gene’s expression in trans, revealing properties of trans-regulatory mutations that provide the raw material for trans-regulatory variation segregating within natural populations.
Collapse
Affiliation(s)
- Fabien Duveau
- Department of Ecology and Evolutionary Biology, University of Michigan, Ann Arbor, United States.,Laboratory of Biology and Modeling of the Cell, Ecole Normale Supérieure de Lyon, CNRS, Université Claude Bernard Lyon, Université de Lyon, Lyon, France
| | - Petra Vande Zande
- Department of Molecular, Cellular, and Developmental Biology, University of Michigan, Ann Arbor, United States
| | - Brian Ph Metzger
- Department of Ecology and Evolutionary Biology, University of Michigan, Ann Arbor, United States
| | - Crisandra J Diaz
- Department of Molecular, Cellular, and Developmental Biology, University of Michigan, Ann Arbor, United States
| | - Elizabeth A Walker
- Department of Ecology and Evolutionary Biology, University of Michigan, Ann Arbor, United States
| | - Stephen Tryban
- Department of Ecology and Evolutionary Biology, University of Michigan, Ann Arbor, United States
| | - Mohammad A Siddiq
- Department of Ecology and Evolutionary Biology, University of Michigan, Ann Arbor, United States
| | - Bing Yang
- Department of Molecular, Cellular, and Developmental Biology, University of Michigan, Ann Arbor, United States
| | - Patricia J Wittkopp
- Department of Ecology and Evolutionary Biology, University of Michigan, Ann Arbor, United States.,Department of Molecular, Cellular, and Developmental Biology, University of Michigan, Ann Arbor, United States
| |
Collapse
|
61
|
Qin J, Hu Y, Yao JC, Leung RWT, Zhou Y, Qin Y, Wang J. Cell fate conversion prediction by group sparse optimization method utilizing single-cell and bulk OMICs data. Brief Bioinform 2021; 22:6347206. [PMID: 34374760 DOI: 10.1093/bib/bbab311] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2021] [Revised: 07/06/2021] [Accepted: 07/19/2021] [Indexed: 01/09/2023] Open
Abstract
Cell fate conversion by overexpressing defined factors is a powerful tool in regenerative medicine. However, identifying key factors for cell fate conversion requires laborious experimental efforts; thus, many of such conversions have not been achieved yet. Nevertheless, cell fate conversions found in many published studies were incomplete as the expression of important gene sets could not be manipulated thoroughly. Therefore, the identification of master transcription factors for complete and efficient conversion is crucial to render this technology more applicable clinically. In the past decade, systematic analyses on various single-cell and bulk OMICs data have uncovered numerous gene regulatory mechanisms, and made it possible to predict master gene regulators during cell fate conversion. By virtue of the sparse structure of master transcription factors and the group structure of their simultaneous regulatory effects on the cell fate conversion process, this study introduces a novel computational method predicting master transcription factors based on group sparse optimization technique integrating data from multi-OMICs levels, which can be applicable to both single-cell and bulk OMICs data with a high tolerance of data sparsity. When it is compared with current prediction methods by cross-referencing published and validated master transcription factors, it possesses superior performance. In short, this method facilitates fast identification of key regulators, give raise to the possibility of higher successful conversion rate and in the hope of reducing experimental cost.
Collapse
Affiliation(s)
- Jing Qin
- School of Pharmaceutical Sciences (Shenzhen), Sun Yat-sen University, Shenzhen, 518107, China
| | - Yaohua Hu
- Shenzhen Key Laboratory of Advanced Machine Learning and Applications, College of Mathematics and Statistics, Shenzhen University, Shenzhen 518060, China
| | - Jen-Chih Yao
- Research Center for Interneural Computing, China Medical University, Taichung 40402, Taiwan
| | - Ricky Wai Tak Leung
- School of Pharmaceutical Sciences (Shenzhen), Sun Yat-sen University, Shenzhen, 518107, China
| | - Yongqiang Zhou
- School of Pharmaceutical Sciences (Shenzhen), Sun Yat-sen University, Shenzhen, 518107, China
| | - Yiming Qin
- Center for Genomic Sciences & School of Biomedical Sciences, The University of Hong Kong, Pokfulam, Hong Kong
| | - Junwen Wang
- Department of Quantitative Health Sciences and Center for Individualized Medicine, Mayo Clinic, Scottsdale, AZ 85259, USA.,Department of Biomedical Informatics, Arizona State University, Scottsdale, AZ 85259, USA
| |
Collapse
|
62
|
Thibivilliers S, Libault M. Enhancing Our Understanding of Plant Cell-to-Cell Interactions Using Single-Cell Omics. FRONTIERS IN PLANT SCIENCE 2021; 12:696811. [PMID: 34421948 PMCID: PMC8375048 DOI: 10.3389/fpls.2021.696811] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/20/2021] [Accepted: 07/07/2021] [Indexed: 05/05/2023]
Abstract
Plants are composed of cells that physically interact and constantly adapt to their environment. To reveal the contribution of each plant cells to the biology of the entire organism, their molecular, morphological, and physiological attributes must be quantified and analyzed in the context of the morphology of the plant organs. The emergence of single-cell/nucleus omics technologies now allows plant biologists to access different modalities of individual cells including their epigenome and transcriptome to reveal the unique molecular properties of each cell composing the plant and their dynamic regulation during cell differentiation and in response to their environment. In this manuscript, we provide a perspective regarding the challenges and strategies to collect plant single-cell biological datasets and their analysis in the context of cellular interactions. As an example, we provide an analysis of the transcriptional regulation of the Arabidopsis genes controlling the differentiation of the root hair cells at the single-cell level. We also discuss the perspective of the use of spatial profiling to complement existing plant single-cell omics.
Collapse
Affiliation(s)
| | - Marc Libault
- Department of Agronomy and Horticulture, Center for Plant Science Innovation, University of Nebraska-Lincoln, Lincoln, NE, United States
| |
Collapse
|
63
|
Yu CY, Mitrofanova A. Mechanism-Centric Approaches for Biomarker Detection and Precision Therapeutics in Cancer. Front Genet 2021; 12:687813. [PMID: 34408770 PMCID: PMC8365516 DOI: 10.3389/fgene.2021.687813] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Accepted: 06/28/2021] [Indexed: 12/18/2022] Open
Abstract
Biomarker discovery is at the heart of personalized treatment planning and cancer precision therapeutics, encompassing disease classification and prognosis, prediction of treatment response, and therapeutic targeting. However, many biomarkers represent passenger rather than driver alterations, limiting their utilization as functional units for therapeutic targeting. We suggest that identification of driver biomarkers through mechanism-centric approaches, which take into account upstream and downstream regulatory mechanisms, is fundamental to the discovery of functionally meaningful markers. Here, we examine computational approaches that identify mechanism-centric biomarkers elucidated from gene co-expression networks, regulatory networks (e.g., transcriptional regulation), protein-protein interaction (PPI) networks, and molecular pathways. We discuss their objectives, advantages over gene-centric approaches, and known limitations. Future directions highlight the importance of input and model interpretability, method and data integration, and the role of recently introduced technological advantages, such as single-cell sequencing, which are central for effective biomarker discovery and time-cautious precision therapeutics.
Collapse
Affiliation(s)
- Christina Y. Yu
- Department of Biomedical and Health Informatics, School of Health Professions, Rutgers, The State University of New Jersey, Newark, NJ, United States
| | - Antonina Mitrofanova
- Department of Biomedical and Health Informatics, School of Health Professions, Rutgers, The State University of New Jersey, Newark, NJ, United States
- Rutgers Cancer Institute of New Jersey, Rutgers, The State University of New Jersey, New Brunswick, NJ, United States
| |
Collapse
|
64
|
Multiscale models quantifying yeast physiology: towards a whole-cell model. Trends Biotechnol 2021; 40:291-305. [PMID: 34303549 DOI: 10.1016/j.tibtech.2021.06.010] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2021] [Revised: 06/26/2021] [Accepted: 06/28/2021] [Indexed: 12/21/2022]
Abstract
The yeast Saccharomyces cerevisiae is widely used as a cell factory and as an important eukaryal model organism for studying cellular physiology related to human health and disease. Yeast was also the first eukaryal organism for which a genome-scale metabolic model (GEM) was developed. In recent years there has been interest in expanding the modeling framework for yeast by incorporating enzymatic parameters and other heterogeneous cellular networks to obtain a more comprehensive description of cellular physiology. We review the latest developments in multiscale models of yeast, and illustrate how a new generation of multiscale models could significantly enhance the predictive performance and expand the applications of classical GEMs in cell factory design and basic studies of yeast physiology.
Collapse
|
65
|
Differential Expression Levels of Sox9 in Early Neocortical Radial Glial Cells Regulate the Decision between Stem Cell Maintenance and Differentiation. J Neurosci 2021; 41:6969-6986. [PMID: 34266896 PMCID: PMC8372026 DOI: 10.1523/jneurosci.2905-20.2021] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2020] [Revised: 06/25/2021] [Accepted: 06/30/2021] [Indexed: 12/18/2022] Open
Abstract
Radial glial progenitor cells (RGCs) in the dorsal telencephalon directly or indirectly produce excitatory projection neurons and macroglia of the neocortex. Recent evidence shows that the pool of RGCs is more heterogeneous than originally thought and that progenitor subpopulations can generate particular neuronal cell types. Using single-cell RNA sequencing, we have studied gene expression patterns of RGCs with different neurogenic behavior at early stages of cortical development. At this early age, some RGCs rapidly produce postmitotic neurons, whereas others self-renew and undergo neurogenic divisions at a later age. We have identified candidate genes that are differentially expressed among these early RGC subpopulations, including the transcription factor Sox9. Using in utero electroporation in embryonic mice of either sex, we demonstrate that elevated Sox9 expression in progenitors affects RGC cell cycle duration and leads to the generation of upper layer cortical neurons. Our data thus reveal molecular differences between progenitor cells with different neurogenic behavior at early stages of corticogenesis and indicates that Sox9 is critical for the maintenance of RGCs to regulate the generation of upper layer neurons. SIGNIFICANCE STATEMENT The existence of heterogeneity in the pool of RGCs and its relationship with the generation of cellular diversity in the cerebral cortex has been an interesting topic of debate for many years. Here we describe the existence of RGCs with reduced neurogenic behavior at early embryonic ages presenting a particular molecular signature. This molecular signature consists of differential expression of some genes including the transcription factor Sox9, which has been found to be a specific regulator of this subpopulation of progenitor cells. Functional experiments perturbing expression levels of Sox9 reveal its instructive role in the regulation of the neurogenic behavior of RGCs and its relationship with the generation of upper layer projection neurons at later ages.
Collapse
|
66
|
Gao S, Dai Y, Rehman J. A Bayesian inference transcription factor activity model for the analysis of single-cell transcriptomes. Genome Res 2021; 31:1296-1311. [PMID: 34193535 PMCID: PMC8256867 DOI: 10.1101/gr.265595.120] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2020] [Accepted: 05/26/2021] [Indexed: 01/06/2023]
Abstract
Single-cell RNA sequencing (scRNA-seq) has emerged as a powerful experimental approach to study cellular heterogeneity. One of the challenges in scRNA-seq data analysis is integrating different types of biological data to consistently recognize discrete biological functions and regulatory mechanisms of cells, such as transcription factor activities and gene regulatory networks in distinct cell populations. We have developed an approach to infer transcription factor activities from scRNA-seq data that leverages existing biological data on transcription factor binding sites. The Bayesian inference transcription factor activity model (BITFAM) integrates ChIP-seq transcription factor binding information into scRNA-seq data analysis. We show that the inferred transcription factor activities for key cell types identify regulatory transcription factors that are known to mechanistically control cell function and cell fate. The BITFAM approach not only identifies biologically meaningful transcription factor activities, but also provides valuable insights into underlying transcription factor regulatory mechanisms.
Collapse
Affiliation(s)
- Shang Gao
- Department of Bioengineering, University of Illinois at Chicago, Chicago, Illinois 60612, USA
- Department of Medicine, Division of Cardiology, University of Illinois at Chicago, Chicago, Illinois 60612, USA
- Department of Pharmacology and Regenerative Medicine, University of Illinois at Chicago, Chicago, Illinois 60612, USA
| | - Yang Dai
- Department of Bioengineering, University of Illinois at Chicago, Chicago, Illinois 60612, USA
| | - Jalees Rehman
- Department of Bioengineering, University of Illinois at Chicago, Chicago, Illinois 60612, USA
- Department of Medicine, Division of Cardiology, University of Illinois at Chicago, Chicago, Illinois 60612, USA
- Department of Pharmacology and Regenerative Medicine, University of Illinois at Chicago, Chicago, Illinois 60612, USA
- University of Illinois Cancer Center, Chicago, Illinois 60612, USA
| |
Collapse
|
67
|
Tripathi RK, Wilkins O. Single cell gene regulatory networks in plants: Opportunities for enhancing climate change stress resilience. PLANT, CELL & ENVIRONMENT 2021; 44:2006-2017. [PMID: 33522607 PMCID: PMC8359182 DOI: 10.1111/pce.14012] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/01/2020] [Revised: 01/21/2021] [Accepted: 01/22/2021] [Indexed: 05/05/2023]
Abstract
Global warming poses major challenges for plant survival and agricultural productivity. Thus, efforts to enhance stress resilience in plants are key strategies for protecting food security. Gene regulatory networks (GRNs) are a critical mechanism conferring stress resilience. Until recently, predicting GRNs of the individual cells that make up plants and other multicellular organisms was impeded by aggregate population scale measurements of transcriptome and other genome-scale features. With the advancement of high-throughput single cell RNA-seq and other single cell assays, learning GRNs for individual cells is now possible, in principle. In this article, we report on recent advances in experimental and analytical methodologies for single cell sequencing assays especially as they have been applied to the study of plants. We highlight recent advances and ongoing challenges for scGRN prediction, and finally, we highlight the opportunity to use scGRN discovery for studying and ultimately enhancing abiotic stress resilience in plants.
Collapse
Affiliation(s)
- Rajiv K. Tripathi
- Department of Biological SciencesUniversity of ManitobaWinnipegManitobaCanada
| | - Olivia Wilkins
- Department of Biological SciencesUniversity of ManitobaWinnipegManitobaCanada
| |
Collapse
|
68
|
Alsaigh T, Di Bartolo BA, Mulangala J, Figtree GA, Leeper NJ. Bench-to-Bedside in Vascular Medicine: Optimizing the Translational Pipeline for Patients With Peripheral Artery Disease. Circ Res 2021; 128:1927-1943. [PMID: 34110900 PMCID: PMC8208504 DOI: 10.1161/circresaha.121.318265] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Peripheral arterial disease is a growing worldwide problem with a wide spectrum of clinical severity and is projected to consume >$21 billion per year in the United States alone. While vascular researchers have brought several therapies to the clinic in recent years, few of these approaches have leveraged advances in high-throughput discovery screens, novel translational models, or innovative trial designs. In the following review, we discuss recent advances in unbiased genomics and broader omics technology platforms, along with preclinical vascular models designed to enhance our understanding of disease pathobiology and prioritize targets for additional investigation. Furthermore, we summarize novel approaches to clinical studies in subjects with claudication and ischemic ulceration, with an emphasis on streamlining and accelerating bench-to-bedside translation. By providing a framework designed to enhance each aspect of future clinical development programs, we hope to enrich the pipeline of therapies that may prevent loss of life and limb for those with peripheral arterial disease.
Collapse
Affiliation(s)
- Tom Alsaigh
- Department of Surgery, Division of Vascular Surgery, Stanford University School of Medicine, Stanford, California, United States of America
| | - Belinda A. Di Bartolo
- Cardiothoracic and Vascular Health, Kolling Institute and Department of Cardiology, Royal North Shore Hospital, Northern Sydney Local Health District, Australia
| | | | - Gemma A. Figtree
- Cardiothoracic and Vascular Health, Kolling Institute and Department of Cardiology, Royal North Shore Hospital, Northern Sydney Local Health District, Australia
| | - Nicholas J. Leeper
- Department of Surgery, Division of Vascular Surgery, Stanford University School of Medicine, Stanford, California, United States of America
| |
Collapse
|
69
|
Bartlett T. Fusion of single-cell transcriptome and DNA-binding data, for genomic network inference in cortical development. BMC Bioinformatics 2021; 22:301. [PMID: 34088262 PMCID: PMC8176738 DOI: 10.1186/s12859-021-04201-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2020] [Accepted: 05/12/2021] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Network models are well-established as very useful computational-statistical tools in cell biology. However, a genomic network model based only on gene expression data can, by definition, only infer gene co-expression networks. Hence, in order to infer gene regulatory patterns, it is necessary to also include data related to binding of regulatory factors to DNA. RESULTS We propose a new dynamic genomic network model, for inferring patterns of genomic regulatory influence in dynamic processes such as development. Our model fuses experiment-specific gene expression data with publicly available DNA-binding data. The method we propose is computationally efficient, and can be applied to genome-wide data with tens of thousands of transcripts. Thus, our method is well suited for use as an exploratory tool for genome-wide data. We apply our method to data from human fetal cortical development, and our findings confirm genomic regulatory patterns which are recognised as being fundamental to neuronal development. CONCLUSIONS Our method provides a mathematical/computational toolbox which, when coupled with targeted experiments, will reveal and confirm important new functional genomic regulatory processes in mammalian development.
Collapse
Affiliation(s)
- Thomas Bartlett
- University College London, Gower Street, London, WC1E 6BT, UK.
| |
Collapse
|
70
|
Arita Y, Kim G, Li Z, Friesen H, Turco G, Wang RY, Climie D, Usaj M, Hotz M, Stoops EH, Baryshnikova A, Boone C, Botstein D, Andrews BJ, McIsaac RS. A genome-scale yeast library with inducible expression of individual genes. Mol Syst Biol 2021; 17:e10207. [PMID: 34096681 PMCID: PMC8182650 DOI: 10.15252/msb.202110207] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2021] [Revised: 04/27/2021] [Accepted: 04/30/2021] [Indexed: 11/09/2022] Open
Abstract
The ability to switch a gene from off to on and monitor dynamic changes provides a powerful approach for probing gene function and elucidating causal regulatory relationships. Here, we developed and characterized YETI (Yeast Estradiol strains with Titratable Induction), a collection in which > 5,600 yeast genes are engineered for transcriptional inducibility with single-gene precision at their native loci and without plasmids. Each strain contains SGA screening markers and a unique barcode, enabling high-throughput genetics. We characterized YETI using growth phenotyping and BAR-seq screens, and we used a YETI allele to identify the regulon of Rof1, showing that it acts to repress transcription. We observed that strains with inducible essential genes that have low native expression can often grow without inducer. Analysis of data from eukaryotic and prokaryotic systems shows that native expression is a variable that can bias promoter-perturbing screens, including CRISPRi. We engineered a second expression system, Z3 EB42, that gives lower expression than Z3 EV, a feature enabling conditional activation and repression of lowly expressed essential genes that grow without inducer in the YETI library.
Collapse
Affiliation(s)
- Yuko Arita
- Terrence Donnelly Centre for Cellular and Biomolecular ResearchUniversity of TorontoTorontoONCanada
- RIKEN Centre for Sustainable Resource ScienceWakoSaitamaJapan
| | - Griffin Kim
- Calico Life Sciences LLCSouth San FranciscoCAUSA
| | - Zhijian Li
- Terrence Donnelly Centre for Cellular and Biomolecular ResearchUniversity of TorontoTorontoONCanada
| | - Helena Friesen
- Terrence Donnelly Centre for Cellular and Biomolecular ResearchUniversity of TorontoTorontoONCanada
| | - Gina Turco
- Calico Life Sciences LLCSouth San FranciscoCAUSA
| | | | - Dale Climie
- Terrence Donnelly Centre for Cellular and Biomolecular ResearchUniversity of TorontoTorontoONCanada
| | - Matej Usaj
- Terrence Donnelly Centre for Cellular and Biomolecular ResearchUniversity of TorontoTorontoONCanada
| | - Manuel Hotz
- Calico Life Sciences LLCSouth San FranciscoCAUSA
| | | | | | - Charles Boone
- Terrence Donnelly Centre for Cellular and Biomolecular ResearchUniversity of TorontoTorontoONCanada
- RIKEN Centre for Sustainable Resource ScienceWakoSaitamaJapan
- Department of Molecular GeneticsUniversity of TorontoTorontoONCanada
| | | | - Brenda J Andrews
- Terrence Donnelly Centre for Cellular and Biomolecular ResearchUniversity of TorontoTorontoONCanada
- Department of Molecular GeneticsUniversity of TorontoTorontoONCanada
| | | |
Collapse
|
71
|
Ma F, Salomé PA, Merchant SS, Pellegrini M. Single-cell RNA sequencing of batch Chlamydomonas cultures reveals heterogeneity in their diurnal cycle phase. THE PLANT CELL 2021; 33:1042-1057. [PMID: 33585940 PMCID: PMC8226295 DOI: 10.1093/plcell/koab025] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/16/2020] [Accepted: 01/13/2021] [Indexed: 05/02/2023]
Abstract
The photosynthetic unicellular alga Chlamydomonas (Chlamydomonas reinhardtii) is a versatile reference for algal biology because of its ease of culture in the laboratory. Genomic and systems biology approaches have previously described transcriptome responses to environmental changes using bulk data, thus representing the average behavior from pools of cells. Here, we apply single-cell RNA sequencing (scRNA-seq) to probe the heterogeneity of Chlamydomonas cell populations under three environments and in two genotypes differing by the presence of a cell wall. First, we determined that RNA can be extracted from single algal cells with or without a cell wall, offering the possibility to sample natural algal communities. Second, scRNA-seq successfully separated single cells into nonoverlapping cell clusters according to their growth conditions. Cells exposed to iron or nitrogen deficiency were easily distinguished despite a shared tendency to arrest photosynthesis and cell division to economize resources. Notably, these groups of cells not only recapitulated known patterns observed with bulk RNA-seq but also revealed their inherent heterogeneity. A substantial source of variation between cells originated from their endogenous diurnal phase, although cultures were grown in constant light. We exploited this result to show that circadian iron responses may be conserved from algae to land plants. We document experimentally that bulk RNA-seq data represent an average of typically hidden heterogeneity in the population.
Collapse
Affiliation(s)
- Feiyang Ma
- Department of Molecular, Cell and Developmental Biology, University of California, Los Angeles, California 90095, USA
| | - Patrice A Salomé
- Department of Chemistry and Biochemistry, University of California, Los Angeles, California 90095, USA
- Institute for Genomics and Proteomics, University of California, Los Angeles, California 90095, USA
| | - Sabeeha S Merchant
- Department of Chemistry and Biochemistry, University of California, Los Angeles, California 90095, USA
- Institute for Genomics and Proteomics, University of California, Los Angeles, California 90095, USA
- Departments of Molecular and Cell Biology and Plant and Microbial Biology, University of California, Berkeley, California 94720, USA
- Lawrence Berkeley National Laboratory, Berkeley, California 94720, USA
| | - Matteo Pellegrini
- Department of Molecular, Cell and Developmental Biology, University of California, Los Angeles, California 90095, USA
- Institute for Genomics and Proteomics, University of California, Los Angeles, California 90095, USA
| |
Collapse
|
72
|
Abstract
Lysine is the first limiting essential amino acid in rice because it is present in the lowest quantity compared to all the other amino acids. Amino acids are the building block of proteins and play an essential role in maintaining the human body’s healthy functioning. Rice is a staple food for more than half of the global population; thus, increasing the lysine content in rice will help improve global health. In this paper, we studied the lysine biosynthesis pathway in rice (Oryza sativa) to identify the regulators of the lysine reporter gene LYSA (LOC_Os02g24354). Genetically intervening at the regulators has the potential to increase the overall lysine content in rice. We modeled the lysine biosynthesis pathway in rice seedlings under normal and saline (NaCl) stress conditions using Bayesian networks. We estimated the model parameters using experimental data and identified the gene DAPF(LOC_Os12g37960) as a positive regulator of the lysine reporter gene LYSA under both normal and saline stress conditions. Based on this analysis, we conclude that the gene DAPF is a potent candidate for genetic intervention. Upregulating DAPF using methods such as CRISPR-Cas9 gene editing strategy has the potential to upregulate the lysine reporter gene LYSA and increase the overall lysine content in rice.
Collapse
|
73
|
|
74
|
Auslander N, Gussow AB, Koonin EV. Incorporating Machine Learning into Established Bioinformatics Frameworks. Int J Mol Sci 2021; 22:2903. [PMID: 33809353 PMCID: PMC8000113 DOI: 10.3390/ijms22062903] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2021] [Revised: 03/08/2021] [Accepted: 03/10/2021] [Indexed: 12/23/2022] Open
Abstract
The exponential growth of biomedical data in recent years has urged the application of numerous machine learning techniques to address emerging problems in biology and clinical research. By enabling the automatic feature extraction, selection, and generation of predictive models, these methods can be used to efficiently study complex biological systems. Machine learning techniques are frequently integrated with bioinformatic methods, as well as curated databases and biological networks, to enhance training and validation, identify the best interpretable features, and enable feature and model investigation. Here, we review recently developed methods that incorporate machine learning within the same framework with techniques from molecular evolution, protein structure analysis, systems biology, and disease genomics. We outline the challenges posed for machine learning, and, in particular, deep learning in biomedicine, and suggest unique opportunities for machine learning techniques integrated with established bioinformatics approaches to overcome some of these challenges.
Collapse
Affiliation(s)
| | | | - Eugene V. Koonin
- National Center for Biotechnology Information, National Library of Medicine, National Institutes of Health, Bethesda, MD 20894, USA;
| |
Collapse
|
75
|
Revealing lineage-related signals in single-cell gene expression using random matrix theory. Proc Natl Acad Sci U S A 2021; 118:1913931118. [PMID: 33836557 PMCID: PMC7980374 DOI: 10.1073/pnas.1913931118] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Gene expression profiles of a cellular population, generated by single-cell RNA sequencing, contains rich information about biological state, including cell type, cell cycle phase, gene regulatory patterns, and location within the tissue of origin. A major challenge is to disentangle information about these different biological states from each other, including distinguishing from cell lineage, since the correlation of cellular expression patterns is necessarily contaminated by ancestry. Here, we use a recent advance in random matrix theory, discovered in the context of protein phylogeny, to identify differentiation or ancestry-related processes in single-cell data. Qin and Colwell [C. Qin, L. J. Colwell, Proc. Natl. Acad. Sci. U.S.A. 115, 690-695 (2018)] showed that ancestral relationships in protein sequences create a power-law signature in the covariance eigenvalue distribution. We demonstrate the existence of such signatures in scRNA-seq data and that the genes driving them are indeed related to differentiation and developmental pathways. We predict the existence of similar power-law signatures for cells along linear trajectories and demonstrate this for linearly differentiating systems. Furthermore, we generalize to show that the same signatures can arise for cells along tissue-specific spatial trajectories. We illustrate these principles in diverse tissues and organisms, including the mammalian epidermis and lung, Drosophila whole-embryo, adult Hydra, dendritic cells, the intestinal epithelium, and cells undergoing induced pluripotent stem cells (iPSC) reprogramming. We show how these results can be used to interpret the gradual dynamics of lineage structure along iPSC reprogramming. Together, we provide a framework that can be used to identify signatures of specific biological processes in single-cell data without prior knowledge and identify candidate genes associated with these processes.
Collapse
|
76
|
Single-Cell RNA Sequencing of the Adult Mammalian Heart-State-of-the-Art and Future Perspectives. Curr Heart Fail Rep 2021; 18:64-70. [PMID: 33629280 PMCID: PMC7954708 DOI: 10.1007/s11897-021-00504-3] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 01/22/2021] [Indexed: 11/28/2022]
Abstract
Purpose of the Review Cardiovascular disease remains the leading cause of death worldwide, resulting in cardiac dysfunction and, subsequently, heart failure (HF). Single-cell RNA sequencing (scRNA-seq) is a rapidly developing tool for studying the transcriptional heterogeneity in both healthy and diseased hearts. Wide applications of techniques like scRNA-seq could significantly contribute to uncovering the molecular mechanisms involved in the onset and progression to HF and contribute to the development of new, improved therapies. This review discusses several studies that successfully applied scRNA-seq to the mouse and human heart using various methods of tissue processing and downstream analysis. Recent Findings The application of scRNA-seq in the cardiovascular field is continuously expanding, providing new detailed insights into cardiac pathophysiology. Summary Increased understanding of cardiac pathophysiology on the single-cell level will contribute to the development of novel, more effective therapeutic strategies. Here, we summarise the possible application of scRNA-seq to the adult mammalian heart.
Collapse
|
77
|
Turki T, Taguchi YH. Discriminating the single-cell gene regulatory networks of human pancreatic islets: A novel deep learning application. Comput Biol Med 2021; 132:104257. [PMID: 33740535 DOI: 10.1016/j.compbiomed.2021.104257] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2020] [Revised: 02/01/2021] [Accepted: 02/03/2021] [Indexed: 12/24/2022]
Abstract
Analysis of single-cell pancreatic data can play an important role in understanding various metabolic diseases and health conditions. Due to the sparsity and noise present in such single-cell gene expression data, inference of single-cell gene regulatory networks remains a challenge. Since recent studies have reported the reliable inference of single-cell gene regulatory networks (SCGRNs), the current study focused on discriminating the SCGRNs of T2D patients from those of healthy controls. By accurately distinguishing SCGRNs of healthy pancreas from those of T2D pancreas, it would be possible to annotate, organize, visualize, and identify common patterns of SCGRNs in metabolic diseases. Such annotated SCGRNs could play an important role in accelerating the process of building large data repositories. This study aimed to contribute to the development of a novel deep learning (DL) application. First, we generated a dataset consisting of 224 SCGRNs belonging to both T2D and healthy pancreas and made it freely available. Next, we chose seven DL architectures, including VGG16, VGG19, Xception, ResNet50, ResNet101, DenseNet121, and DenseNet169, trained each of them on the dataset, and checked their prediction based on a test set. Of note, we evaluated the DL architectures on a single NVIDIA GeForce RTX 2080Ti GPU. Experimental results on the whole dataset, using several performance measures, demonstrated the superiority of VGG19 DL model in the automatic classification of SCGRNs, derived from the single-cell pancreatic data.
Collapse
Affiliation(s)
- Turki Turki
- Department of Computer Science, King Abdulaziz University, Jeddah, 21589, Saudi Arabia.
| | - Y-H Taguchi
- Department of Physics, Chuo University, Tokyo, 112-8551, Japan.
| |
Collapse
|
78
|
Sun S, Gresham D. Cellular quiescence in budding yeast. Yeast 2021; 38:12-29. [PMID: 33350503 DOI: 10.1002/yea.3545] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2020] [Revised: 12/11/2020] [Accepted: 12/14/2020] [Indexed: 12/20/2022] Open
Abstract
Cellular quiescence, the temporary and reversible exit from proliferative growth, is the predominant state of all cells. However, our understanding of the biological processes and molecular mechanisms that underlie cell quiescence remains incomplete. As with the mitotic cell cycle, budding and fission yeast are preeminent model systems for studying cellular quiescence owing to their rich experimental toolboxes and the evolutionary conservation across eukaryotes of pathways and processes that control quiescence. Here, we review current knowledge of cell quiescence in budding yeast and how it pertains to cellular quiescence in other organisms, including multicellular animals. Quiescence entails large-scale remodeling of virtually every cellular process, organelle, gene expression, and metabolic state that is executed dynamically as cells undergo the initiation, maintenance, and exit from quiescence. We review these major transitions, our current understanding of their molecular bases, and highlight unresolved questions. We summarize the primary methods employed for quiescence studies in yeast and discuss their relative merits. Understanding cell quiescence has important consequences for human disease as quiescent single-celled microbes are notoriously difficult to kill and quiescent human cells play important roles in diseases such as cancer. We argue that research on cellular quiescence will be accelerated through the adoption of common criteria, and methods, for defining cell quiescence. An integrated approach to studying cell quiescence, and a focus on the behavior of individual cells, will yield new insights into the pathways and processes that underlie cell quiescence leading to a more complete understanding of the life cycle of cells. TAKE AWAY: Quiescent cells are viable cells that have reversibly exited the cell cycle Quiescence is induced in response to a variety of nutrient starvation signals Quiescence is executed dynamically through three phases: initiation, maintenance, and exit Quiescence entails large-scale remodeling of gene expression, organelles, and metabolism Single-cell approaches are required to address heterogeneity among quiescent cells.
Collapse
Affiliation(s)
- Siyu Sun
- Center for Genomics and Systems Biology, New York University, New York, New York, 10003, USA.,Department of Biology, New York University, New York, New York, 10003, USA
| | - David Gresham
- Center for Genomics and Systems Biology, New York University, New York, New York, 10003, USA.,Department of Biology, New York University, New York, New York, 10003, USA
| |
Collapse
|
79
|
Fang L, Li Y, Ma L, Xu Q, Tan F, Chen G. GRNdb: decoding the gene regulatory networks in diverse human and mouse conditions. Nucleic Acids Res 2021; 49:D97-D103. [PMID: 33151298 PMCID: PMC7779055 DOI: 10.1093/nar/gkaa995] [Citation(s) in RCA: 44] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2020] [Revised: 10/10/2020] [Accepted: 10/13/2020] [Indexed: 12/11/2022] Open
Abstract
Gene regulatory networks (GRNs) formed by transcription factors (TFs) and their downstream target genes play essential roles in gene expression regulation. Moreover, GRNs can be dynamic changing across different conditions, which are crucial for understanding the underlying mechanisms of disease pathogenesis. However, no existing database provides comprehensive GRN information for various human and mouse normal tissues and diseases at the single-cell level. Based on the known TF-target relationships and the large-scale single-cell RNA-seq data collected from public databases as well as the bulk data of The Cancer Genome Atlas and the Genotype-Tissue Expression project, we systematically predicted the GRNs of 184 different physiological and pathological conditions of human and mouse involving >633 000 cells and >27 700 bulk samples. We further developed GRNdb, a freely accessible and user-friendly database (http://www.grndb.com/) for searching, comparing, browsing, visualizing, and downloading the predicted information of 77 746 GRNs, 19 687 841 TF-target pairs, and related binding motifs at single-cell/bulk resolution. GRNdb also allows users to explore the gene expression profile, correlations, and the associations between expression levels and the patient survival of diverse cancers. Overall, GRNdb provides a valuable and timely resource to the scientific community to elucidate the functions and mechanisms of gene expression regulation in various conditions.
Collapse
Affiliation(s)
- Li Fang
- Center for Bioinformatics and Computational Biology, and Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences, School of Life Sciences, East China Normal University, Shanghai 200241, China
| | - Yunjin Li
- Center for Bioinformatics and Computational Biology, and Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences, School of Life Sciences, East China Normal University, Shanghai 200241, China
| | - Lu Ma
- Center for Bioinformatics and Computational Biology, and Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences, School of Life Sciences, East China Normal University, Shanghai 200241, China
| | - Qiyue Xu
- Center for Bioinformatics and Computational Biology, and Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences, School of Life Sciences, East China Normal University, Shanghai 200241, China
| | - Fei Tan
- Shanghai Skin Disease Hospital, School of Medicine, Tongji University, Shanghai 200443, China
| | - Geng Chen
- Center for Bioinformatics and Computational Biology, and Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences, School of Life Sciences, East China Normal University, Shanghai 200241, China
- Shanghai Applied Protein Technology Co., Ltd. (APTBIO), Shanghai 200233, China
| |
Collapse
|
80
|
Ingham VA, Elg S, Nagi SC, Dondelinger F. Capturing the transcription factor interactome in response to sub-lethal insecticide exposure. CURRENT RESEARCH IN INSECT SCIENCE 2021; 1:None. [PMID: 34977825 PMCID: PMC8702396 DOI: 10.1016/j.cris.2021.100018] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/07/2020] [Revised: 06/15/2021] [Accepted: 07/21/2021] [Indexed: 12/02/2022]
Abstract
The increasing levels of pesticide resistance in agricultural pests and disease vectors represents a threat to both food security and global health. As insecticide resistance intensity strengthens and spreads, the likelihood of a pest encountering a sub-lethal dose of pesticide dramatically increases. Here, we apply dynamic Bayesian networks to a transcriptome time-course generated using sub-lethal pyrethroid exposure on a highly resistant Anopheles coluzzii population. The model accounts for circadian rhythm and ageing effects allowing high confidence identification of transcription factors with key roles in pesticide response. The associations generated by this model show high concordance with lab-based validation and identifies 44 transcription factors putatively regulating insecticide-responsive transcripts. We identify six key regulators, with each displaying differing enrichment terms, demonstrating the complexity of pesticide response. The considerable overlap of resistance mechanisms in agricultural pests and disease vectors strongly suggests that these findings are relevant in a wide variety of pest species.
Collapse
|
81
|
Tjärnberg A, Mahmood O, Jackson CA, Saldi GA, Cho K, Christiaen LA, Bonneau RA. Optimal tuning of weighted kNN- and diffusion-based methods for denoising single cell genomics data. PLoS Comput Biol 2021; 17:e1008569. [PMID: 33411784 PMCID: PMC7817019 DOI: 10.1371/journal.pcbi.1008569] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2020] [Revised: 01/20/2021] [Accepted: 11/28/2020] [Indexed: 12/26/2022] Open
Abstract
The analysis of single-cell genomics data presents several statistical challenges, and extensive efforts have been made to produce methods for the analysis of this data that impute missing values, address sampling issues and quantify and correct for noise. In spite of such efforts, no consensus on best practices has been established and all current approaches vary substantially based on the available data and empirical tests. The k-Nearest Neighbor Graph (kNN-G) is often used to infer the identities of, and relationships between, cells and is the basis of many widely used dimensionality-reduction and projection methods. The kNN-G has also been the basis for imputation methods using, e.g., neighbor averaging and graph diffusion. However, due to the lack of an agreed-upon optimal objective function for choosing hyperparameters, these methods tend to oversmooth data, thereby resulting in a loss of information with regard to cell identity and the specific gene-to-gene patterns underlying regulatory mechanisms. In this paper, we investigate the tuning of kNN- and diffusion-based denoising methods with a novel non-stochastic method for optimally preserving biologically relevant informative variance in single-cell data. The framework, Denoising Expression data with a Weighted Affinity Kernel and Self-Supervision (DEWÄKSS), uses a self-supervised technique to tune its parameters. We demonstrate that denoising with optimal parameters selected by our objective function (i) is robust to preprocessing methods using data from established benchmarks, (ii) disentangles cellular identity and maintains robust clusters over dimension-reduction methods, (iii) maintains variance along several expression dimensions, unlike previous heuristic-based methods that tend to oversmooth data variance, and (iv) rarely involves diffusion but rather uses a fixed weighted kNN graph for denoising. Together, these findings provide a new understanding of kNN- and diffusion-based denoising methods. Code and example data for DEWÄKSS is available at https://gitlab.com/Xparx/dewakss/-/tree/Tjarnberg2020branch.
Collapse
Affiliation(s)
- Andreas Tjärnberg
- Center for Developmental Genetics, New York University, New York, New York, USA
- Center For Genomics and Systems Biology, NYU, New York, New York, USA
- Department of Biology, NYU, New York, New York, USA
| | - Omar Mahmood
- Center For Data Science, NYU, New York, New York, USA
| | - Christopher A. Jackson
- Center For Genomics and Systems Biology, NYU, New York, New York, USA
- Department of Biology, NYU, New York, New York, USA
| | | | - Kyunghyun Cho
- Flatiron Institute, Center for Computational Biology, Simons Foundation, New York, New York, USA
- Courant Institute of Mathematical Sciences, Computer Science Department, New York University, New York, New York, USA
| | - Lionel A. Christiaen
- Center for Developmental Genetics, New York University, New York, New York, USA
- Department of Biology, NYU, New York, New York, USA
| | - Richard A. Bonneau
- Center For Genomics and Systems Biology, NYU, New York, New York, USA
- Department of Biology, NYU, New York, New York, USA
- Center For Data Science, NYU, New York, New York, USA
- Flatiron Institute, Center for Computational Biology, Simons Foundation, New York, New York, USA
- Courant Institute of Mathematical Sciences, Computer Science Department, New York University, New York, New York, USA
| |
Collapse
|
82
|
Wright NR, Rønnest NP, Sonnenschein N. Single-Cell Technologies to Understand the Mechanisms of Cellular Adaptation in Chemostats. Front Bioeng Biotechnol 2020; 8:579841. [PMID: 33392163 PMCID: PMC7775484 DOI: 10.3389/fbioe.2020.579841] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2020] [Accepted: 11/30/2020] [Indexed: 11/13/2022] Open
Abstract
There is a growing interest in continuous manufacturing within the bioprocessing community. In this context, the chemostat process is an important unit operation. The current application of chemostat processes in industry is limited although many high yielding processes are reported in literature. In order to reach the full potential of the chemostat in continuous manufacture, the output should be constant. However, adaptation is often observed resulting in changed productivities over time. The observed adaptation can be coupled to the selective pressure of the nutrient-limited environment in the chemostat. We argue that population heterogeneity should be taken into account when studying adaptation in the chemostat. We propose to investigate adaptation at the single-cell level and discuss the potential of different single-cell technologies, which could be used to increase the understanding of the phenomena. Currently, none of the discussed single-cell technologies fulfill all our criteria but in combination they may reveal important information, which can be used to understand and potentially control the adaptation.
Collapse
Affiliation(s)
- Naia Risager Wright
- Novo Nordisk A/S, Bagsvaerd, Denmark
- Department of Biotechnology and Biomedicine, Technical University of Denmark, Kongens Lyngby, Denmark
| | | | - Nikolaus Sonnenschein
- Department of Biotechnology and Biomedicine, Technical University of Denmark, Kongens Lyngby, Denmark
| |
Collapse
|
83
|
Molecular and evolutionary processes generating variation in gene expression. Nat Rev Genet 2020; 22:203-215. [PMID: 33268840 DOI: 10.1038/s41576-020-00304-w] [Citation(s) in RCA: 121] [Impact Index Per Article: 30.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/21/2020] [Indexed: 12/18/2022]
Abstract
Heritable variation in gene expression is common within and between species. This variation arises from mutations that alter the form or function of molecular gene regulatory networks that are then filtered by natural selection. High-throughput methods for introducing mutations and characterizing their cis- and trans-regulatory effects on gene expression (particularly, transcription) are revealing how different molecular mechanisms generate regulatory variation, and studies comparing these mutational effects with variation seen in the wild are teasing apart the role of neutral and non-neutral evolutionary processes. This integration of molecular and evolutionary biology allows us to understand how the variation in gene expression we see today came to be and to predict how it is most likely to evolve in the future.
Collapse
|
84
|
Imdahl F, Saliba AE. Advances and challenges in single-cell RNA-seq of microbial communities. Curr Opin Microbiol 2020; 57:102-110. [PMID: 33160164 DOI: 10.1016/j.mib.2020.10.001] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2020] [Revised: 09/16/2020] [Accepted: 10/02/2020] [Indexed: 12/17/2022]
Abstract
Microbes have developed complex strategies to respond to their environment and escape the immune system by individualizing their behavior. While single-cell RNA sequencing has become instrumental for studying mammalian cells, its use with fungi, protozoa and bacteria is still in its infancy. In this review, we highlight the major progress towards mapping the molecular states of microbes at the single-cell level using genome-wide transcriptomics and describe how these technologies can be extended to probe thousands of species at high throughput. We envision that mammalian and microbial single-cell profiling could soon be integrated for the study of microbial communities in health and disease.
Collapse
Affiliation(s)
- Fabian Imdahl
- Helmholtz Institute for RNA-based Infection Research (HIRI), Helmholtz-Center for Infection Research (HZI), Würzburg, Germany
| | - Antoine-Emmanuel Saliba
- Helmholtz Institute for RNA-based Infection Research (HIRI), Helmholtz-Center for Infection Research (HZI), Würzburg, Germany.
| |
Collapse
|
85
|
Lenz AR, Galán-Vásquez E, Balbinot E, de Abreu FP, Souza de Oliveira N, da Rosa LO, de Avila e Silva S, Camassola M, Dillon AJP, Perez-Rueda E. Gene Regulatory Networks of Penicillium echinulatum 2HH and Penicillium oxalicum 114-2 Inferred by a Computational Biology Approach. Front Microbiol 2020; 11:588263. [PMID: 33193246 PMCID: PMC7652724 DOI: 10.3389/fmicb.2020.588263] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2020] [Accepted: 09/23/2020] [Indexed: 11/29/2022] Open
Abstract
Penicillium echinulatum 2HH and Penicillium oxalicum 114-2 are well-known cellulase fungal producers. However, few studies addressing global mechanisms for gene regulation of these two important organisms are available so far. A recent finding that the 2HH wild-type is closely related to P. oxalicum leads to a combined study of these two species. Firstly, we provide a global gene regulatory network for P. echinulatum 2HH and P. oxalicum 114-2, based on TF-TG orthology relationships, considering three related species with well-known regulatory interactions combined with TFBSs prediction. The network was then analyzed in terms of topology, identifying TFs as hubs, and modules. Based on this approach, we explore numerous identified modules, such as the expression of cellulolytic and xylanolytic systems, where XlnR plays a key role in positive regulation of the xylanolytic system. It also regulates positively the cellulolytic system by acting indirectly through the cellodextrin induction system. This remarkable finding suggests that the XlnR-dependent cellulolytic and xylanolytic regulatory systems are probably conserved in both P. echinulatum and P. oxalicum. Finally, we explore the functional congruency on the genes clustered in terms of communities, where the genes related to cellular nitrogen, compound metabolic process and macromolecule metabolic process were the most abundant. Therefore, our approach allows us to confer a degree of accuracy regarding the existence of each inferred interaction.
Collapse
Affiliation(s)
- Alexandre Rafael Lenz
- Unidad Académica Yucatán, Instituto de Investigaciones en Matemáticas Aplicadas y en Sistemas, Universidad Nacional Autónoma de Mexico, Mérida, Mexico
- Laboratório de Bioinformática e Biologia Computacional, Instituto de Biotecnologia, Universidade de Caxias do Sul, Caxias do Sul, Brazil
- Departamento de Ciências Exatas e da Terra, Universidade do Estado da Bahia, Salvador, Brazil
| | - Edgardo Galán-Vásquez
- Departamento de Ingeniería de Sistemas Computacionales y Automatización, Instituto de Investigaciones en Matemàticas Aplicadas y en Sistemas, Universidad Nacional Autónoma de Mexico, Ciudad Universitaria, Mexico
| | - Eduardo Balbinot
- Laboratório de Bioinformática e Biologia Computacional, Instituto de Biotecnologia, Universidade de Caxias do Sul, Caxias do Sul, Brazil
| | - Fernanda Pessi de Abreu
- Laboratório de Bioinformática e Biologia Computacional, Instituto de Biotecnologia, Universidade de Caxias do Sul, Caxias do Sul, Brazil
| | - Nikael Souza de Oliveira
- Laboratório de Bioinformática e Biologia Computacional, Instituto de Biotecnologia, Universidade de Caxias do Sul, Caxias do Sul, Brazil
- Laboratório de Enzimas e Biomassas, Instituto de Biotecnologia, Universidade de Caxias do Sul, Caxias do Sul, Brazil
| | - Letícia Osório da Rosa
- Laboratório de Enzimas e Biomassas, Instituto de Biotecnologia, Universidade de Caxias do Sul, Caxias do Sul, Brazil
| | - Scheila de Avila e Silva
- Laboratório de Bioinformática e Biologia Computacional, Instituto de Biotecnologia, Universidade de Caxias do Sul, Caxias do Sul, Brazil
| | - Marli Camassola
- Laboratório de Enzimas e Biomassas, Instituto de Biotecnologia, Universidade de Caxias do Sul, Caxias do Sul, Brazil
| | - Aldo José Pinheiro Dillon
- Laboratório de Enzimas e Biomassas, Instituto de Biotecnologia, Universidade de Caxias do Sul, Caxias do Sul, Brazil
| | - Ernesto Perez-Rueda
- Unidad Académica Yucatán, Instituto de Investigaciones en Matemáticas Aplicadas y en Sistemas, Universidad Nacional Autónoma de Mexico, Mérida, Mexico
- Facultad de Ciencias, Centro de Genómica y Bioinformática, Universidad Mayor, Santiago, Chile
| |
Collapse
|
86
|
Wu N, Yin F, Ou-Yang L, Zhu Z, Xie W. Joint learning of multiple gene networks from single-cell gene expression data. Comput Struct Biotechnol J 2020; 18:2583-2595. [PMID: 33033579 PMCID: PMC7527714 DOI: 10.1016/j.csbj.2020.09.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2020] [Revised: 08/31/2020] [Accepted: 09/01/2020] [Indexed: 11/24/2022] Open
Abstract
Inferring gene networks from gene expression data is important for understanding functional organizations within cells. With the accumulation of single-cell RNA sequencing (scRNA-seq) data, it is possible to infer gene networks at single cell level. However, due to the characteristics of scRNA-seq data, such as cellular heterogeneity and high sparsity caused by dropout events, traditional network inference methods may not be suitable for scRNA-seq data. In this study, we introduce a novel joint Gaussian copula graphical model (JGCGM) to jointly estimate multiple gene networks for multiple cell subgroups from scRNA-seq data. Our model can deal with non-Gaussian data with missing values, and identify the common and unique network structures of multiple cell subgroups, which is suitable for scRNA-seq data. Extensive experiments on synthetic data demonstrate that our proposed model outperforms other compared state-of-the-art network inference models. We apply our model to real scRNA-seq data sets to infer gene networks of different cell subgroups. Hub genes in the estimated gene networks are found to be biological significance.
Collapse
Affiliation(s)
- Nuosi Wu
- College of Electronics and Information Engineering, Shenzhen University, Shenzhen, China
| | - Fu Yin
- College of Electronics and Information Engineering, Shenzhen University, Shenzhen, China
| | - Le Ou-Yang
- College of Electronics and Information Engineering, Shenzhen University, Shenzhen, China
- Guangdong Key Laboratory of Intelligent Information Processing, Shenzhen Key Laboratory of Media Security, and Guangdong Laboratory of Artificial Intelligence and Digital Economy(SZ), Shenzhen University, Shenzhen, China
- Shenzhen Institute of Artificial Intelligence and Robotics for Society, Shenzhen, China
| | - Zexuan Zhu
- College of Computer Science and Software Engineering, Shenzhen University, Shenzhen, China
| | - Weixin Xie
- College of Electronics and Information Engineering, Shenzhen University, Shenzhen, China
| |
Collapse
|
87
|
Katebi A, Kohar V, Lu M. Random Parametric Perturbations of Gene Regulatory Circuit Uncover State Transitions in Cell Cycle. iScience 2020; 23:101150. [PMID: 32450514 PMCID: PMC7251928 DOI: 10.1016/j.isci.2020.101150] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2019] [Revised: 03/05/2020] [Accepted: 05/05/2020] [Indexed: 02/03/2023] Open
Abstract
Many biological processes involve precise cellular state transitions controlled by complex gene regulation. Here, we use budding yeast cell cycle as a model system and explore how a gene regulatory circuit encodes essential information of state transitions. We present a generalized random circuit perturbation method for circuits containing heterogeneous regulation types and its usage to analyze both steady and oscillatory states from an ensemble of circuit models with random kinetic parameters. The stable steady states form robust clusters with a circular structure that are associated with cell cycle phases. This circular structure in the clusters is consistent with single-cell RNA sequencing data. The oscillatory states specify the irreversible state transitions along cell cycle progression. Furthermore, we identify possible mechanisms to understand the irreversible state transitions from the steady states. We expect this approach to be robust and generally applicable to unbiasedly predict dynamical transitions of a gene regulatory circuit.
Collapse
Affiliation(s)
- Ataur Katebi
- The Jackson Laboratory, 600 Main Street, Bar Harbor, ME 04609, USA
| | - Vivek Kohar
- The Jackson Laboratory, 600 Main Street, Bar Harbor, ME 04609, USA
| | - Mingyang Lu
- The Jackson Laboratory, 600 Main Street, Bar Harbor, ME 04609, USA.
| |
Collapse
|
88
|
Abstract
The fermentation industry is known to be very conservative, relying on traditional yeast management. Yet, in the modern fast-paced world, change comes about in facets such as climate change altering the quality and quantity of harvests, changes due to government regulations e.g., the use of pesticides or SO2, the need to become more sustainable, and of course by changes in consumer preferences. As a silent companion of the fermentation industry, the wine yeast Saccharomyces cerevisiae has followed mankind through millennia, changing from a Kulturfolger, into a domesticated species for the production of bread, beer, and wine and further on into a platform strain for the production of biofuels, enzymes, flavors, or pharmaceuticals. This success story is based on the ‘awesome power of yeast genetics’. Central to this is the very efficient homologous recombination (HR) machinery of S. cerevisiae that allows highly-specific genome edits. This microsurgery tool is so reliable that yeast has put a generally recognized as safe (GRAS) label onto itself and entrusted to itself the life-changing decision of mating type-switching. Later, yeast became its own genome editor, interpreted as domestication events, to adapt to harsh fermentation conditions. In biotechnology, yeast HR has been used with tremendous success over the last 40 years. Here we discuss several types of yeast genome edits then focus on HR and its inherent potential for evolving novel wine yeast strains and styles relevant for changing markets.
Collapse
|
89
|
Jariani A, Vermeersch L, Cerulus B, Perez-Samper G, Voordeckers K, Van Brussel T, Thienpont B, Lambrechts D, Verstrepen KJ. A new protocol for single-cell RNA-seq reveals stochastic gene expression during lag phase in budding yeast. eLife 2020; 9:e55320. [PMID: 32420869 PMCID: PMC7259953 DOI: 10.7554/elife.55320] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2020] [Accepted: 05/15/2020] [Indexed: 12/17/2022] Open
Abstract
Current methods for single-cell RNA sequencing (scRNA-seq) of yeast cells do not match the throughput and relative simplicity of the state-of-the-art techniques that are available for mammalian cells. In this study, we report how 10x Genomics' droplet-based single-cell RNA sequencing technology can be modified to allow analysis of yeast cells. The protocol, which is based on in-droplet spheroplasting of the cells, yields an order-of-magnitude higher throughput in comparison to existing methods. After extensive validation of the method, we demonstrate its use by studying the dynamics of the response of isogenic yeast populations to a shift in carbon source, revealing the heterogeneity and underlying molecular processes during this shift. The method we describe opens new avenues for studies focusing on yeast cells, as well as other cells with a degradable cell wall.
Collapse
Affiliation(s)
- Abbas Jariani
- Laboratory for Systems Biology, VIB-KU Leuven Center for MicrobiologyLeuvenBelgium
- Laboratory of Genetics and Genomics, CMPG, Department M2S, KU LeuvenLeuvenBelgium
| | - Lieselotte Vermeersch
- Laboratory for Systems Biology, VIB-KU Leuven Center for MicrobiologyLeuvenBelgium
- Laboratory of Genetics and Genomics, CMPG, Department M2S, KU LeuvenLeuvenBelgium
| | - Bram Cerulus
- Laboratory for Systems Biology, VIB-KU Leuven Center for MicrobiologyLeuvenBelgium
- Laboratory of Genetics and Genomics, CMPG, Department M2S, KU LeuvenLeuvenBelgium
| | - Gemma Perez-Samper
- Laboratory for Systems Biology, VIB-KU Leuven Center for MicrobiologyLeuvenBelgium
- Laboratory of Genetics and Genomics, CMPG, Department M2S, KU LeuvenLeuvenBelgium
| | - Karin Voordeckers
- Laboratory for Systems Biology, VIB-KU Leuven Center for MicrobiologyLeuvenBelgium
- Laboratory of Genetics and Genomics, CMPG, Department M2S, KU LeuvenLeuvenBelgium
| | - Thomas Van Brussel
- Laboratory for Translational Genetics, Department of Human Genetics, KU LeuvenLeuvenBelgium
- VIB Center for Cancer Biology, VIBLeuvenBelgium
| | - Bernard Thienpont
- Laboratory for Translational Genetics, Department of Human Genetics, KU LeuvenLeuvenBelgium
- VIB Center for Cancer Biology, VIBLeuvenBelgium
- Laboratory for Functional Epigenetics, Department of Genetics, KU LeuvenLeuvenBelgium
| | - Diether Lambrechts
- Laboratory for Translational Genetics, Department of Human Genetics, KU LeuvenLeuvenBelgium
- VIB Center for Cancer Biology, VIBLeuvenBelgium
| | - Kevin J Verstrepen
- Laboratory for Systems Biology, VIB-KU Leuven Center for MicrobiologyLeuvenBelgium
- Laboratory of Genetics and Genomics, CMPG, Department M2S, KU LeuvenLeuvenBelgium
| |
Collapse
|
90
|
Jackson C, Gresham D. A Bright IDEA. Mol Syst Biol 2020; 16:e9502. [PMID: 32253808 PMCID: PMC7136649 DOI: 10.15252/msb.20209502] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
Transcription factors (TFs) control the rate of mRNA production. Technological advances have made the task of measuring mRNA levels for all genes straightforward, but identifying causal relationships between TFs and their target genes remains an unsolved problem in biology. In their recent study, McIsaac and colleagues (Hackett et al, 2020) apply a method for inducing the overexpression of a TF and studying the dynamics with which all transcripts respond. Using time series analysis, they are able to resolve direct effects of TFs from secondary effects. This new experimental and analytical approach provides an efficient means of defining gene regulatory relationships for all TFs.
Collapse
Affiliation(s)
- Christopher Jackson
- Center for Genomics and Systems BiologyDepartment of BiologyNew York UniversityNew YorkNYUSA
| | - David Gresham
- Center for Genomics and Systems BiologyDepartment of BiologyNew York UniversityNew YorkNYUSA
| |
Collapse
|
91
|
van der Wijst MGP, de Vries DH, Groot HE, Trynka G, Hon CC, Bonder MJ, Stegle O, Nawijn MC, Idaghdour Y, van der Harst P, Ye CJ, Powell J, Theis FJ, Mahfouz A, Heinig M, Franke L. The single-cell eQTLGen consortium. eLife 2020; 9:e52155. [PMID: 32149610 PMCID: PMC7077978 DOI: 10.7554/elife.52155] [Citation(s) in RCA: 121] [Impact Index Per Article: 30.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2019] [Accepted: 03/03/2020] [Indexed: 12/17/2022] Open
Abstract
In recent years, functional genomics approaches combining genetic information with bulk RNA-sequencing data have identified the downstream expression effects of disease-associated genetic risk factors through so-called expression quantitative trait locus (eQTL) analysis. Single-cell RNA-sequencing creates enormous opportunities for mapping eQTLs across different cell types and in dynamic processes, many of which are obscured when using bulk methods. Rapid increase in throughput and reduction in cost per cell now allow this technology to be applied to large-scale population genetics studies. To fully leverage these emerging data resources, we have founded the single-cell eQTLGen consortium (sc-eQTLGen), aimed at pinpointing the cellular contexts in which disease-causing genetic variants affect gene expression. Here, we outline the goals, approach and potential utility of the sc-eQTLGen consortium. We also provide a set of study design considerations for future single-cell eQTL studies.
Collapse
Affiliation(s)
- MGP van der Wijst
- Department of Genetics, Oncode Institute, University of Groningen, University Medical Center GroningenGroningenNetherlands
| | - DH de Vries
- Department of Genetics, Oncode Institute, University of Groningen, University Medical Center GroningenGroningenNetherlands
| | - HE Groot
- Department of Cardiology, University of Groningen, University Medical Center GroningenGroningenNetherlands
| | - G Trynka
- Wellcome Sanger InstituteHinxtonUnited Kingdom
- Open TargetsHinxtonUnited Kingdom
| | - CC Hon
- RIKEN Center for Integrative Medical SciencesYokahamaJapan
| | - MJ Bonder
- Division of Computational Genomics and Systems Genetics, German Cancer Research Center (DKFZ)HeidelbergGermany
- Genome Biology Unit, European Molecular Biology LaboratoryHeidelbergGermany
| | - O Stegle
- Division of Computational Genomics and Systems Genetics, German Cancer Research Center (DKFZ)HeidelbergGermany
- Genome Biology Unit, European Molecular Biology LaboratoryHeidelbergGermany
| | - MC Nawijn
- Department of Pathology and Medical Biology, GRIAC Research Institute, University of Groningen, University Medical Center GroningenGroningenNetherlands
| | - Y Idaghdour
- Program in Biology, Public Health Research Center, New York University Abu DhabiAbu DhabiUnited Arab Emirates
| | - P van der Harst
- Department of Cardiology, University of Groningen, University Medical Center GroningenGroningenNetherlands
| | - CJ Ye
- Institute for Human Genetics, Bakar Computational Health Sciences Institute, Bakar ImmunoX Initiative, Department of Medicine, Department of Bioengineering and Therapeutic Sciences, Department of Epidemiology and Biostatistics, Chan Zuckerberg Biohub, University of California San FranciscoSan FranciscoUnited States
| | - J Powell
- Garvan-Weizmann Centre for Cellular Genomics, Garvan Institute, UNSW Cellular Genomics Futures Institute, University of New South WalesSydneyAustralia
| | - FJ Theis
- Institute of Computational Biology, Helmholtz Zentrum MünchenNeuherbergGermany
- Department of Mathematics, Technical University of MunichGarching bei MünchenGermany
| | - A Mahfouz
- Leiden Computational Biology Center, Leiden University Medical CenterLeidenNetherlands
- Delft Bioinformatics Lab, Delft University of TechnologyDelftNetherlands
| | - M Heinig
- Institute of Computational Biology, Helmholtz Zentrum MünchenNeuherbergGermany
- Department of Informatics, Technical University of MunichGarching bei MünchenGermany
| | - L Franke
- Department of Genetics, Oncode Institute, University of Groningen, University Medical Center GroningenGroningenNetherlands
| |
Collapse
|
92
|
Jackson CA, Castro DM, Saldi GA, Bonneau R, Gresham D. Gene regulatory network reconstruction using single-cell RNA sequencing of barcoded genotypes in diverse environments. eLife 2020; 9:e51254. [PMID: 31985403 PMCID: PMC7004572 DOI: 10.7554/elife.51254] [Citation(s) in RCA: 92] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2019] [Accepted: 01/10/2020] [Indexed: 11/13/2022] Open
Abstract
Understanding how gene expression programs are controlled requires identifying regulatory relationships between transcription factors and target genes. Gene regulatory networks are typically constructed from gene expression data acquired following genetic perturbation or environmental stimulus. Single-cell RNA sequencing (scRNAseq) captures the gene expression state of thousands of individual cells in a single experiment, offering advantages in combinatorial experimental design, large numbers of independent measurements, and accessing the interaction between the cell cycle and environmental responses that is hidden by population-level analysis of gene expression. To leverage these advantages, we developed a method for scRNAseq in budding yeast (Saccharomyces cerevisiae). We pooled diverse transcriptionally barcoded gene deletion mutants in 11 different environmental conditions and determined their expression state by sequencing 38,285 individual cells. We benchmarked a framework for learning gene regulatory networks from scRNAseq data that incorporates multitask learning and constructed a global gene regulatory network comprising 12,228 interactions.
Collapse
Affiliation(s)
- Christopher A Jackson
- Center For Genomics and Systems BiologyNew York UniversityNew YorkUnited States
- Department of BiologyNew York UniversityNew YorkUnited States
| | | | | | - Richard Bonneau
- Center For Genomics and Systems BiologyNew York UniversityNew YorkUnited States
- Department of BiologyNew York UniversityNew YorkUnited States
- Courant Institute of Mathematical Sciences, Computer Science DepartmentNew York UniversityNew YorkUnited States
- Center For Data ScienceNew York UniversityNew YorkUnited States
- Flatiron Institute, Center for Computational BiologySimons FoundationNew YorkUnited States
| | - David Gresham
- Center For Genomics and Systems BiologyNew York UniversityNew YorkUnited States
- Department of BiologyNew York UniversityNew YorkUnited States
| |
Collapse
|
93
|
Nadal-Ribelles M, Islam S, Wei W, Latorre P, Nguyen M, de Nadal E, Posas F, Steinmetz LM. Yeast Single-cell RNA-seq, Cell by Cell and Step by Step. Bio Protoc 2019; 9:e3359. [PMID: 33654857 DOI: 10.21769/bioprotoc.3359] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2019] [Revised: 07/29/2019] [Accepted: 07/31/2019] [Indexed: 11/02/2022] Open
Abstract
Single-cell RNA-seq (scRNA-seq) has become an established method for uncovering the intrinsic complexity within populations. Even within seemingly homogenous populations of isogenic yeast cells, there is a high degree of heterogeneity that originates from a compact and pervasively transcribed genome. Research with microorganisms such as yeast represents a major challenge for single-cell transcriptomics, due to their small size, rigid cell wall, and low RNA content per cell. Because of these technical challenges, yeast-specific scRNA-seq methodologies have recently started to appear, each one of them relying on different cell-isolation and library-preparation methods. Consequently, each approach harbors unique strengths and weaknesses that need to be considered. We have recently developed a yeast single-cell RNA-seq protocol (yscRNA-seq), which is inexpensive, high-throughput and easy-to-implement, tailored to the unique needs of yeast. yscRNA-seq provides a unique platform that combines single-cell phenotyping via index sorting with the incorporation of unique molecule identifiers on transcripts that allows to digitally count the number of molecules in a strand- and isoform-specific manner. Here, we provide a detailed, step-by-step description of the experimental and computational steps of yscRNA-seq protocol. This protocol will ease the implementation of yscRNA-seq in other laboratories and provide guidelines for the development of novel technologies.
Collapse
Affiliation(s)
- Mariona Nadal-Ribelles
- Department of Genetics, Stanford University, School of Medicine, California, USA.,Stanford Genome Technology Center, Stanford University, California, USA.,Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology, Barcelona, Spain.,Departament de Ciències Experimentals i de la Salut, Cell Signaling Research Group, Universitat Pompeu Fabra (UPF), Barcelona, Spain
| | - Saiful Islam
- Department of Genetics, Stanford University, School of Medicine, California, USA.,Stanford Genome Technology Center, Stanford University, California, USA
| | - Wu Wei
- Department of Genetics, Stanford University, School of Medicine, California, USA.,Stanford Genome Technology Center, Stanford University, California, USA.,CAS Key Laboratory of Computational Biology, CAS-MPG Partner Institute for Computational Biology, Shanghai Institute of Nutrition and Health, Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Pablo Latorre
- Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology, Barcelona, Spain.,Departament de Ciències Experimentals i de la Salut, Cell Signaling Research Group, Universitat Pompeu Fabra (UPF), Barcelona, Spain
| | - Michelle Nguyen
- Department of Genetics, Stanford University, School of Medicine, California, USA.,Stanford Genome Technology Center, Stanford University, California, USA
| | - Eulàlia de Nadal
- Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology, Barcelona, Spain.,Departament de Ciències Experimentals i de la Salut, Cell Signaling Research Group, Universitat Pompeu Fabra (UPF), Barcelona, Spain
| | - Francesc Posas
- Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology, Barcelona, Spain.,Departament de Ciències Experimentals i de la Salut, Cell Signaling Research Group, Universitat Pompeu Fabra (UPF), Barcelona, Spain
| | - Lars M Steinmetz
- Department of Genetics, Stanford University, School of Medicine, California, USA.,Stanford Genome Technology Center, Stanford University, California, USA.,European Molecular Biology Laboratory, Genome Biology Unit, Heidelberg, Germany
| |
Collapse
|
94
|
Blencowe M, Arneson D, Ding J, Chen YW, Saleem Z, Yang X. Network modeling of single-cell omics data: challenges, opportunities, and progresses. Emerg Top Life Sci 2019; 3:379-398. [PMID: 32270049 PMCID: PMC7141415 DOI: 10.1042/etls20180176] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2019] [Revised: 06/07/2019] [Accepted: 06/24/2019] [Indexed: 01/07/2023]
Abstract
Single-cell multi-omics technologies are rapidly evolving, prompting both methodological advances and biological discoveries at an unprecedented speed. Gene regulatory network modeling has been used as a powerful approach to elucidate the complex molecular interactions underlying biological processes and systems, yet its application in single-cell omics data modeling has been met with unique challenges and opportunities. In this review, we discuss these challenges and opportunities, and offer an overview of the recent development of network modeling approaches designed to capture dynamic networks, within-cell networks, and cell-cell interaction or communication networks. Finally, we outline the remaining gaps in single-cell gene network modeling and the outlooks of the field moving forward.
Collapse
Affiliation(s)
- Montgomery Blencowe
- Department of Integrative Biology and Physiology, University of California, Los Angeles, 610 Charles E. Young Drive East, Los Angeles, CA 90095, U.S.A
| | - Douglas Arneson
- Department of Integrative Biology and Physiology, University of California, Los Angeles, 610 Charles E. Young Drive East, Los Angeles, CA 90095, U.S.A
- Bioinformatics Interdepartmental Program, University of California, Los Angeles, 610 Charles E. Young Drive East, Los Angeles, CA 90095, U.S.A
| | - Jessica Ding
- Department of Integrative Biology and Physiology, University of California, Los Angeles, 610 Charles E. Young Drive East, Los Angeles, CA 90095, U.S.A
| | - Yen-Wei Chen
- Department of Integrative Biology and Physiology, University of California, Los Angeles, 610 Charles E. Young Drive East, Los Angeles, CA 90095, U.S.A
- Molecular Toxicology Program, University of California, Los Angeles, 610 Charles E. Young Drive East, Los Angeles, CA 90095, U.S.A
| | - Zara Saleem
- Department of Integrative Biology and Physiology, University of California, Los Angeles, 610 Charles E. Young Drive East, Los Angeles, CA 90095, U.S.A
| | - Xia Yang
- Department of Integrative Biology and Physiology, University of California, Los Angeles, 610 Charles E. Young Drive East, Los Angeles, CA 90095, U.S.A
- Bioinformatics Interdepartmental Program, University of California, Los Angeles, 610 Charles E. Young Drive East, Los Angeles, CA 90095, U.S.A
- Molecular Toxicology Program, University of California, Los Angeles, 610 Charles E. Young Drive East, Los Angeles, CA 90095, U.S.A
- Institute for Quantitative and Computational Biosciences, University of California, Los Angeles, 610 Charles E. Young Drive East, Los Angeles, CA 90095, U.S.A
| |
Collapse
|