1
|
Ye W, Clark E, Talatala E, Davis R, Ramirez‐Solano M, Sheng Q, Yang J, Collins S, Hillel A, Gelbard A. Characterizing the Cellular Constituents of Proximal Airway Disease in Granulomatosis With Polyangiitis. Otolaryngol Head Neck Surg 2025; 172:2009-2017. [PMID: 40062629 PMCID: PMC12120036 DOI: 10.1002/ohn.1197] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2024] [Revised: 01/07/2025] [Accepted: 02/11/2025] [Indexed: 05/31/2025]
Abstract
OBJECTIVE Granulomatosis with polyangiitis (GPA) is a rare multisystem autoimmune vasculitis. 10-20% of patients suffer life-threatening obstruction of their proximal airways. Although progress has been made in the treatment of systemic disease, ameliorating airway disease in GPA remains an unmet need arising from limited understanding of disease pathogenesis. We sought to characterize the cellular constituents of the affected proximal airway mucosa in GPA airway scar. STUDY DESIGN Basic/translational study. SETTING Single tertiary care center. METHODS Using single-cell RNA sequencing, we profiled the cellular constituents of proximal airway samples from GPA and disease comparators (GPA; n = 9, idiopathic subglottic stenosis: iSGS; n = 7, post-intubation proximal stenosis: PIPS; n = 5, and control; n = 10). We report transcriptomes for subglottic epithelial, immune, endothelial, and stromal cell types and map expression of GPA risk genes to tissue types present in the proximal airway. We compared differential gene expression across immune cell populations and performed pseudotime analysis using Monocle 3. RESULTS Similar to iSGS and PIPS, the subglottic mucosa of GPA patients demonstrated an abundant immune infiltrate. 71% of the established GPA risk genes (10 of 14) localized to T cells and macrophages. Differential gene expression and pseudotime analysis revealed a sub-population of CD4-/CD8- inflammatory T cells that only originated from GPA. CONCLUSION We characterized the cellular composition of GPA airway disease and demonstrated that the expression of GPA risk alleles is predominantly localized to immune cell populations. We also identified a subset of inflammatory T cells that is unique to GPA.
Collapse
Affiliation(s)
- Wenda Ye
- Department of Otolaryngology–Head and Neck SurgeryVanderbilt University Medical CenterNashvilleTennesseeUSA
| | - Evan Clark
- Department of Otolaryngology–Head and Neck SurgeryVanderbilt University Medical CenterNashvilleTennesseeUSA
| | - Edward Talatala
- Department of Otolaryngology–Head and Neck SurgeryVanderbilt University Medical CenterNashvilleTennesseeUSA
| | - Ruth Davis
- Department of Otolaryngology–Head and Neck SurgeryUniversity of Wisconsin School of Medicine and Public HealthMadisonWisconsinUSA
| | | | - Quanhu Sheng
- Department of BiostatisticsVanderbilt University Medical CenterNashvilleTennesseeUSA
| | - Jing Yang
- Department of BiostatisticsVanderbilt University Medical CenterNashvilleTennesseeUSA
| | - Sam Collins
- Department of Otolaryngology–Head and Neck SurgeryJohns Hopkins UniversityBaltimoreMarylandUSA
| | - Alexander Hillel
- Department of Otolaryngology–Head and Neck SurgeryJohns Hopkins UniversityBaltimoreMarylandUSA
| | - Alexander Gelbard
- Department of Otolaryngology–Head and Neck SurgeryVanderbilt University Medical CenterNashvilleTennesseeUSA
| |
Collapse
|
2
|
Nie R, Zheng C, Ren L, Teng Y, Sun Y, Wang L, Li J, Cai J. Mitigating Cell Cycle Effects in Multi-Omics Data: Solutions and Analytical Frameworks. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025:e05823. [PMID: 40434003 DOI: 10.1002/advs.202505823] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/02/2025] [Revised: 04/30/2025] [Indexed: 05/29/2025]
Abstract
Cell cycle structures vary significantly across cell types, which exhibit distinct phase compositions. Asynchronous DNA replication and dynamic cellular characteristics during the cell cycle result in considerable heterogeneity in DNA dosage, chromatin accessibility, methylation, and expression. Nonetheless, the consequences of cell cycle disruption in the interpretation of multi-omics data remain unclear. Here, we systematically assessed the influence of distinct cell phase structures on the interpretation of omics features in proliferating cells, and proposed solutions for each omics dataset. For copy number variation (CNV) calling, asynchronous replication timing (RT) interference induces false CNVs in cells with high S-phase ratio (SPR), which are significantly decreased following replication timing domain (RTD) correction. Similar noise is observed in the chromatin accessibility data. Moreover, for DNA methylation and transcriptomic analyses, cell cycle-sorted data outperformed direct comparison in elucidating the biological features of compared cells. Additionally, we established an integrated pipeline to identify differentially expressed genes (DEGs) after cell cycle phasing. Consequently, our study demonstrated extensive cell-cycle heterogeneity, warranting consideration in future studies involving cells with diverse cell-cycle structures. RTD correction or phase-specific comparison could reduce the influence of cell cycle composition on the analysis of the differences observed between stem and differentiated cells.
Collapse
Affiliation(s)
- Rui Nie
- Chinese Academy of Sciences and China National Center for Bioinformation, Beijing, 100101, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Caihong Zheng
- Laboratory of Molecular Biology and Bovine Breeding, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing, 100193, China
| | - Likun Ren
- Chinese Academy of Sciences and China National Center for Bioinformation, Beijing, 100101, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Yue Teng
- Chinese Academy of Sciences and China National Center for Bioinformation, Beijing, 100101, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Yaoyu Sun
- School of Life Sciences, Peking University, Beijing, 100871, China
| | - Lifei Wang
- Department of Chemistry, the University of Hong Kong, Hong Kong, 999077, China
| | - Junya Li
- Laboratory of Molecular Biology and Bovine Breeding, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing, 100193, China
| | - Jun Cai
- Chinese Academy of Sciences and China National Center for Bioinformation, Beijing, 100101, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| |
Collapse
|
3
|
Wang W, Pang Z, Zhang S, Yang P, Pan Y, Qiao L, Yang K, Liu J, Wang R, Liu W. Multi-omics integrated analysis reveals the molecular mechanism of tail fat deposition differences in sheep with different tail types. BMC Genomics 2025; 26:465. [PMID: 40346476 PMCID: PMC12065285 DOI: 10.1186/s12864-025-11658-y] [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: 03/25/2025] [Accepted: 04/30/2025] [Indexed: 05/11/2025] Open
Abstract
BACKGROUND The accumulation of tail fat in sheep is a manifestation of adaptive evolution to the environment. Sheep with different tail types show significant differences in physiological functions and tail fat deposition. Although these differences reflect the developmental mechanism of tail fat under different gene regulation, the situation of sheep tail fat tissue at the single cell level has not been explored, and its molecular mechanism still needs to be further elucidated. RESULTS Here, we characterized the genomic features of sheep with different tail types, detected the transcriptomic differences in tail adipose tissue between fat-tailed and thin-tailed sheep, established a single-cell atlas of sheep tail adipose tissue, and screened potential molecular markers (SESN1, RPRD1A and RASGEF1B) that regulate differences in sheep tail fat deposition through multi-omics integrated analysis. We found that the differential mechanism of sheep tail fat deposition not only involves adipocyte differentiation and proliferation, but is also closely related to cell-specific communication networks (When adipocytes act as signal outputters, LAMININ and other signal pathways are strongly expressed in guangling large tailed sheep and hu sheep), including interactions with immune cells and tissue remodeling to drive the typing of tail fat. In addition, we revealed the differentiation trajectory of sheep tail adipocytes through pseudo-time analysis and constructed the cell communication network of sheep tail adipose tissue. CONCLUSIONS Our results provide insights into the molecular mechanisms of tail fat deposition in sheep with different tail types, and provide a deeper explanation for the development and functional regulation of adipocytes.
Collapse
Affiliation(s)
- Wannian Wang
- Department of Animal Genetics, Breeding and Reproduction, College of Animal Science, Shanxi Agricultural University, Taigu, 030801, China
| | - Zhixv Pang
- Department of Animal Genetics, Breeding and Reproduction, College of Animal Science, Shanxi Agricultural University, Taigu, 030801, China
| | - Siying Zhang
- Department of Animal Genetics, Breeding and Reproduction, College of Animal Science, Shanxi Agricultural University, Taigu, 030801, China
| | - Pengkun Yang
- Department of Animal Genetics, Breeding and Reproduction, College of Animal Science, Shanxi Agricultural University, Taigu, 030801, China
| | - Yangyang Pan
- Department of Animal Genetics, Breeding and Reproduction, College of Animal Science, Shanxi Agricultural University, Taigu, 030801, China
| | - Liying Qiao
- Department of Animal Genetics, Breeding and Reproduction, College of Animal Science, Shanxi Agricultural University, Taigu, 030801, China
| | - Kaijie Yang
- Department of Animal Genetics, Breeding and Reproduction, College of Animal Science, Shanxi Agricultural University, Taigu, 030801, China
| | - Jianhua Liu
- Department of Animal Genetics, Breeding and Reproduction, College of Animal Science, Shanxi Agricultural University, Taigu, 030801, China
- Key Laboratory of Farm Animal Genetic Resources Exploration and Precision Breeding of Shanxi Province, Taigu, 030801, China
| | - Ruizhen Wang
- Academy of Chinese Medical Sciences, Henan University of Chinese Medicine, Zhengzhou, 450046, China
| | - Wenzhong Liu
- Department of Animal Genetics, Breeding and Reproduction, College of Animal Science, Shanxi Agricultural University, Taigu, 030801, China.
- Key Laboratory of Farm Animal Genetic Resources Exploration and Precision Breeding of Shanxi Province, Taigu, 030801, China.
| |
Collapse
|
4
|
Sheinin R, Sharan R, Madi A. scNET: learning context-specific gene and cell embeddings by integrating single-cell gene expression data with protein-protein interactions. Nat Methods 2025; 22:708-716. [PMID: 40097811 PMCID: PMC11978505 DOI: 10.1038/s41592-025-02627-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2024] [Accepted: 02/07/2025] [Indexed: 03/19/2025]
Abstract
Recent advances in single-cell RNA sequencing (scRNA-seq) techniques have provided unprecedented insights into the heterogeneity of various tissues. However, gene expression data alone often fails to capture and identify changes in cellular pathways and complexes, as they are more discernible at the protein level. Moreover, analyzing scRNA-seq data presents further challenges due to inherent characteristics such as high noise levels and zero inflation. In this study, we propose an approach to address these limitations by integrating scRNA-seq datasets with a protein-protein interaction network. Our method utilizes a unique dual-view architecture based on graph neural networks, enabling joint representation of gene expression and protein-protein interaction network data. This approach models gene-to-gene relationships under specific biological contexts and refines cell-cell relations using an attention mechanism. Next, through comprehensive evaluations, we demonstrate that scNET better captures gene annotation, pathway characterization and gene-gene relationship identification, while improving cell clustering and pathway analysis across diverse cell types and biological conditions.
Collapse
Affiliation(s)
- Ron Sheinin
- Blavatnik School of Computer Science and AI, Tel Aviv University, Tel Aviv, Israel
| | - Roded Sharan
- Blavatnik School of Computer Science and AI, Tel Aviv University, Tel Aviv, Israel.
| | - Asaf Madi
- Department of Pathology, Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel.
| |
Collapse
|
5
|
Cortés-Ríos J, Rodriguez-Fernandez M, Sorger PK, Fröhlich F. Dynamic Modeling of Cell Cycle Arrest Through Integrated Single-Cell and Mathematical Modelling Approaches. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.02.20.639240. [PMID: 40060624 PMCID: PMC11888159 DOI: 10.1101/2025.02.20.639240] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 03/18/2025]
Abstract
Highly multiplexed imaging assays allow simultaneous quantification of multiple protein and phosphorylation markers, providing a static snapshots of cell types and states. Pseudo-time techniques can transform these static snapshots of unsynchronized cells into dynamic trajectories, enabling the study of dynamic processes such as development trajectories and the cell cycle. Such ordering also enables training of mathematical models on these data, but technical challenges have hitherto made it difficult to integrate multiple experimental conditions, limiting the predictive power and insights these models can generate. In this work, we propose data processing and model training approaches for integrating multiplexed, multi-condition immunofluorescence data with mathematical modelling. We devise training strategies that are applicable to datasets where cells exhibit oscillatory as well as arrested dynamics and use them to train a cell cycle model on a dataset of MCF-10A mammary epithelial exposed to cell-cycle arresting small molecules. We validate the model by investigating predicted growth factor sensitivities and responses to inhibitors of cells at different initial conditions. We anticipate that our framework will generalise to other highly multiplexed measurement techniques such as mass-cytometry, rendering larger bodies of data accessible to dynamic modelling and paving the way to deeper biological insights.
Collapse
Affiliation(s)
- Javiera Cortés-Ríos
- Institute for Biological and Medical Engineering, Schools of Engineering, Medicine and Biological Sciences, Pontificia Universidad Católica de Chile, Chile
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, University at Buffalo, State University of New York, Buffalo, New York, United States of America
| | - Maria Rodriguez-Fernandez
- Institute for Biological and Medical Engineering, Schools of Engineering, Medicine and Biological Sciences, Pontificia Universidad Católica de Chile, Chile
| | - Peter K Sorger
- Laboratory of Systems Pharmacology and Department of Systems Biology, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Fabian Fröhlich
- Dynamics of Living Systems Laboratory, The Francis Crick Institute, London, United Kingdom
| |
Collapse
|
6
|
Qu R, Cheng X, Sefik E, Stanley Iii JS, Landa B, Strino F, Platt S, Garritano J, Odell ID, Coifman R, Flavell RA, Myung P, Kluger Y. Gene trajectory inference for single-cell data by optimal transport metrics. Nat Biotechnol 2025; 43:258-268. [PMID: 38580861 PMCID: PMC11452571 DOI: 10.1038/s41587-024-02186-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2022] [Accepted: 02/26/2024] [Indexed: 04/07/2024]
Abstract
Single-cell RNA sequencing has been widely used to investigate cell state transitions and gene dynamics of biological processes. Current strategies to infer the sequential dynamics of genes in a process typically rely on constructing cell pseudotime through cell trajectory inference. However, the presence of concurrent gene processes in the same group of cells and technical noise can obscure the true progression of the processes studied. To address this challenge, we present GeneTrajectory, an approach that identifies trajectories of genes rather than trajectories of cells. Specifically, optimal transport distances are calculated between gene distributions across the cell-cell graph to extract gene programs and define their gene pseudotemporal order. Here we demonstrate that GeneTrajectory accurately extracts progressive gene dynamics in myeloid lineage maturation. Moreover, we show that GeneTrajectory deconvolves key gene programs underlying mouse skin hair follicle dermal condensate differentiation that could not be resolved by cell trajectory approaches. GeneTrajectory facilitates the discovery of gene programs that control the changes and activities of biological processes.
Collapse
Affiliation(s)
- Rihao Qu
- Computational Biology & Bioinformatics Program, Yale University, New Haven, CT, USA
- Department of Pathology, Yale University School of Medicine, New Haven, CT, USA
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT, USA
| | - Xiuyuan Cheng
- Department of Mathematics, Duke University, Durham, NC, USA
| | - Esen Sefik
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT, USA
| | | | - Boris Landa
- Program in Applied Mathematics, Yale University, New Haven, CT, USA
| | | | - Sarah Platt
- Department of Pathology, Yale University School of Medicine, New Haven, CT, USA
- Department of Dermatology, Yale University School of Medicine, New Haven, CT, USA
| | - James Garritano
- Program in Applied Mathematics, Yale University, New Haven, CT, USA
| | - Ian D Odell
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT, USA
- Department of Dermatology, Yale University School of Medicine, New Haven, CT, USA
| | - Ronald Coifman
- Program in Applied Mathematics, Yale University, New Haven, CT, USA
- Department of Mathematics, Yale University, New Haven, CT, USA
- Department of Electrical Engineering, Yale University, New Haven, CT, USA
| | - Richard A Flavell
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT, USA
- Howard Hughes Medical Institute, Yale University School of Medicine, New Haven, CT, USA
| | - Peggy Myung
- Department of Pathology, Yale University School of Medicine, New Haven, CT, USA
- Department of Dermatology, Yale University School of Medicine, New Haven, CT, USA
| | - Yuval Kluger
- Computational Biology & Bioinformatics Program, Yale University, New Haven, CT, USA.
- Department of Pathology, Yale University School of Medicine, New Haven, CT, USA.
- Program in Applied Mathematics, Yale University, New Haven, CT, USA.
| |
Collapse
|
7
|
O’Connor SA, Garcia L, Hoover R, Patel AP, Bartelle BB, Hugnot JP, Paddison PJ, Plaisier CL. Classifying cell cycle states and a quiescent-like G0 state using single-cell transcriptomics. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2024.04.16.589816. [PMID: 38659838 PMCID: PMC11042294 DOI: 10.1101/2024.04.16.589816] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/26/2024]
Abstract
Single-cell transcriptomics has unveiled a vast landscape of cellular heterogeneity in which the cell cycle is a significant component. We trained a high-resolution cell cycle classifier (ccAFv2) using single cell RNA-seq (scRNA-seq) characterized human neural stem cells. The ccAFv2 classifies six cell cycle states (G1, Late G1, S, S/G2, G2/M, and M/Early G1) and a quiescent-like G0 state (Neural G0), and it incorporates a tunable parameter to filter out less certain classifications. The ccAFv2 classifier performed better than or equivalent to other state-of-the-art methods even while classifying more cell cycle states, including G0. We demonstrate that the ccAFv2 classifier effectively generalizes the S, S/G2, G2/M, and M/Early G1 states across cell types derived from all three germ layers. While the G0, G1, and Late G1 states perform well in neuroepithelial cell types, their accuracy is lower in other cell types. However, misclassifications are confined to the G0, G1, and Late G1 states. We showcased the versatility of ccAFv2 by successfully applying it to classify cells, nuclei, and spatial transcriptomics data in humans and mice, using various normalization methods and gene identifiers. We provide methods to regress the cell cycle expression patterns out of single cell or nuclei data to uncover underlying biological signals. The classifier can be used either as an R package integrated with Seurat or a PyPI package integrated with SCANPY. We proved that ccAFv2 has enhanced accuracy, flexibility, and adaptability across various experimental conditions, establishing ccAFv2 as a powerful tool for dissecting complex biological systems, unraveling cellular heterogeneity, and deciphering the molecular mechanisms by which proliferation and quiescence affect cellular processes.
Collapse
Affiliation(s)
- Samantha A. O’Connor
- School of Biological and Health Systems Engineering, Arizona State University, Tempe AZ, USA
| | - Leonor Garcia
- Institut de Génomique Fonctionnelle, Université de Montpellier, CNRS, INSERM, 141 rue de la Cardonille, 34091, Montpellier, France
| | - Rori Hoover
- School of Biological and Health Systems Engineering, Arizona State University, Tempe AZ, USA
| | - Anoop P. Patel
- Brotman-Baty Institute for Precision Medicine, University of Washington, Seattle, WA, USA
- Department of Neurosurgery, Preston Robert Tisch Brain Tumor Center, Duke University, Durham, NC, USA
| | - Benjamin B. Bartelle
- School of Biological and Health Systems Engineering, Arizona State University, Tempe AZ, USA
| | - Jean-Philippe Hugnot
- Institut de Génomique Fonctionnelle, Université de Montpellier, CNRS, INSERM, 141 rue de la Cardonille, 34091, Montpellier, France
| | - Patrick J. Paddison
- Human Biology Division, Fred Hutchinson Cancer Research Center, Seattle WA, USA
| | - Christopher L. Plaisier
- School of Biological and Health Systems Engineering, Arizona State University, Tempe AZ, USA
| |
Collapse
|
8
|
Shen N, Korthauer K. vmrseq: probabilistic modeling of single-cell methylation heterogeneity. Genome Biol 2024; 25:321. [PMID: 39736632 DOI: 10.1186/s13059-024-03457-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2023] [Accepted: 12/09/2024] [Indexed: 01/01/2025] Open
Abstract
Single-cell DNA methylation measurements reveal genome-scale inter-cellular epigenetic heterogeneity, but extreme sparsity and noise challenges rigorous analysis. Previous methods to detect variably methylated regions (VMRs) have relied on predefined regions or sliding windows and report regions insensitive to heterogeneity level present in input. We present vmrseq, a statistical method that overcomes these challenges to detect VMRs with increased accuracy in synthetic benchmarks and improved feature selection in case studies. vmrseq also highlights context-dependent correlations between methylation and gene expression, supporting previous findings and facilitating novel hypotheses on epigenetic regulation. vmrseq is available at https://github.com/nshen7/vmrseq .
Collapse
Affiliation(s)
- Ning Shen
- Department of Statistics, University of British Columbia, Vancouver, Canada
- Centre for Molecular Medicine and Therapeutics, BC Children's Hospital Research Institute, Vancouver, Canada
| | - Keegan Korthauer
- Department of Statistics, University of British Columbia, Vancouver, Canada.
- Centre for Molecular Medicine and Therapeutics, BC Children's Hospital Research Institute, Vancouver, Canada.
| |
Collapse
|
9
|
Gao H, Hua K, Wu X, Wei L, Chen S, Yin Q, Jiang R, Zhang X. Building a learnable universal coordinate system for single-cell atlas with a joint-VAE model. Commun Biol 2024; 7:977. [PMID: 39134617 PMCID: PMC11319358 DOI: 10.1038/s42003-024-06564-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2023] [Accepted: 07/05/2024] [Indexed: 08/15/2024] Open
Abstract
A universal coordinate system that can ensemble the huge number of cells and capture their heterogeneities is of vital importance for constructing large-scale cell atlases as references for molecular and cellular studies. Studies have shown that cells exhibit multifaceted heterogeneities in their transcriptomic features at multiple resolutions. This nature of complexity makes it hard to design a fixed coordinate system through a combination of known features. It is desirable to build a learnable universal coordinate model that can capture major heterogeneities and serve as a controlled generative model for data augmentation. We developed UniCoord, a specially-tuned joint-VAE model to represent single-cell transcriptomic data in a lower-dimensional latent space with high interpretability. Each latent dimension can represent either discrete or continuous feature, and either supervised by prior knowledge or unsupervised. The latent dimensions can be easily reconfigured to generate pseudo transcriptomic profiles with desired properties. UniCoord can also be used as a pre-trained model to analyze new data with unseen cell types and thus can serve as a feasible framework for cell annotation and comparison. UniCoord provides a prototype for a learnable universal coordinate framework to enable better analysis and generation of cells with highly orchestrated functions and heterogeneities.
Collapse
Affiliation(s)
- Haoxiang Gao
- MOE Key Laboratory of Bioinformatics and Bioinformatics Division, BNRIST, Department of Automation, Tsinghua University, Beijing, China
| | - Kui Hua
- MOE Key Laboratory of Bioinformatics and Bioinformatics Division, BNRIST, Department of Automation, Tsinghua University, Beijing, China
| | - Xinze Wu
- MOE Key Laboratory of Bioinformatics and Bioinformatics Division, BNRIST, Department of Automation, Tsinghua University, Beijing, China
| | - Lei Wei
- MOE Key Laboratory of Bioinformatics and Bioinformatics Division, BNRIST, Department of Automation, Tsinghua University, Beijing, China.
| | - Sijie Chen
- MOE Key Laboratory of Bioinformatics and Bioinformatics Division, BNRIST, Department of Automation, Tsinghua University, Beijing, China
| | - Qijin Yin
- MOE Key Laboratory of Bioinformatics and Bioinformatics Division, BNRIST, Department of Automation, Tsinghua University, Beijing, China
| | - Rui Jiang
- MOE Key Laboratory of Bioinformatics and Bioinformatics Division, BNRIST, Department of Automation, Tsinghua University, Beijing, China
| | - Xuegong Zhang
- MOE Key Laboratory of Bioinformatics and Bioinformatics Division, BNRIST, Department of Automation, Tsinghua University, Beijing, China.
- School of Life Sciences and School of Medicine, Center for Synthetic and Systems Biology, Tsinghua University, Beijing, China.
| |
Collapse
|
10
|
Liu J, Ma J, Wen J, Zhou X. A Cell Cycle-Aware Network for Data Integration and Label Transferring of Single-Cell RNA-Seq and ATAC-Seq. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2401815. [PMID: 38887194 PMCID: PMC11336957 DOI: 10.1002/advs.202401815] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/20/2024] [Revised: 04/22/2024] [Indexed: 06/20/2024]
Abstract
In recent years, the integration of single-cell multi-omics data has provided a more comprehensive understanding of cell functions and internal regulatory mechanisms from a non-single omics perspective, but it still suffers many challenges, such as omics-variance, sparsity, cell heterogeneity, and confounding factors. As it is known, the cell cycle is regarded as a confounder when analyzing other factors in single-cell RNA-seq data, but it is not clear how it will work on the integrated single-cell multi-omics data. Here, a cell cycle-aware network (CCAN) is developed to remove cell cycle effects from the integrated single-cell multi-omics data while keeping the cell type-specific variations. This is the first computational model to study the cell-cycle effects in the integration of single-cell multi-omics data. Validations on several benchmark datasets show the outstanding performance of CCAN in a variety of downstream analyses and applications, including removing cell cycle effects and batch effects of scRNA-seq datasets from different protocols, integrating paired and unpaired scRNA-seq and scATAC-seq data, accurately transferring cell type labels from scRNA-seq to scATAC-seq data, and characterizing the differentiation process from hematopoietic stem cells to different lineages in the integration of differentiation data.
Collapse
Affiliation(s)
- Jiajia Liu
- Center for Computational Systems MedicineMcWilliams School of Biomedical InformaticsThe University of Texas Health Science Center at HoustonHoustonTX77030USA
| | - Jian Ma
- Department of Electronic Information and Computer EngineeringThe Engineering & Technical College of Chengdu University of TechnologyLeshanSichuan614000China
| | - Jianguo Wen
- Center for Computational Systems MedicineMcWilliams School of Biomedical InformaticsThe University of Texas Health Science Center at HoustonHoustonTX77030USA
| | - Xiaobo Zhou
- Center for Computational Systems MedicineMcWilliams School of Biomedical InformaticsThe University of Texas Health Science Center at HoustonHoustonTX77030USA
- McGovern Medical SchoolThe University of Texas Health Science Center at HoustonHoustonTX77030USA
- School of DentistryThe University of Texas Health Science Center at HoustonHoustonTX77030USA
| |
Collapse
|
11
|
Wu Y, Shi Z, Zhou X, Zhang P, Yang X, Ding J, Wu H. scHiCyclePred: a deep learning framework for predicting cell cycle phases from single-cell Hi-C data using multi-scale interaction information. Commun Biol 2024; 7:923. [PMID: 39085477 PMCID: PMC11291681 DOI: 10.1038/s42003-024-06626-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2023] [Accepted: 07/24/2024] [Indexed: 08/02/2024] Open
Abstract
The emergence of single-cell Hi-C (scHi-C) technology has provided unprecedented opportunities for investigating the intricate relationship between cell cycle phases and the three-dimensional (3D) structure of chromatin. However, accurately predicting cell cycle phases based on scHi-C data remains a formidable challenge. Here, we present scHiCyclePred, a prediction model that integrates multiple feature sets to leverage scHi-C data for predicting cell cycle phases. scHiCyclePred extracts 3D chromatin structure features by incorporating multi-scale interaction information. The comparative analysis illustrates that scHiCyclePred surpasses existing methods such as Nagano_method and CIRCLET across various metrics including accuracy (ACC), F1 score, Precision, Recall, and balanced accuracy (BACC). In addition, we evaluate scHiCyclePred against the previously published CIRCLET using the dataset of complex tissues (Liu_dataset). Experimental results reveal significant improvements with scHiCyclePred exhibiting improvements of 0.39, 0.52, 0.52, and 0.39 over the CIRCLET in terms of ACC, F1 score, Precision, and Recall metrics, respectively. Furthermore, we conduct analyses on three-dimensional chromatin dynamics and gene features during the cell cycle, providing a more comprehensive understanding of cell cycle dynamics through chromatin structure. scHiCyclePred not only offers insights into cell biology but also holds promise for catalyzing breakthroughs in disease research. Access scHiCyclePred on GitHub at https:// github.com/HaoWuLab-Bioinformatics/ scHiCyclePred .
Collapse
Affiliation(s)
- Yingfu Wu
- School of Software, Shandong University, Jinan, Shandong, China
- Shenzhen Research Institute of Shandong University, Shenzhen, Guangdong, China
- College of Information Engineering, Northwest A&F University, Yangling, Shaanxi, China
| | - Zhenqi Shi
- School of Software, Shandong University, Jinan, Shandong, China
| | - Xiangfei Zhou
- School of Software, Shandong University, Jinan, Shandong, China
| | - Pengyu Zhang
- College of Information Engineering, Northwest A&F University, Yangling, Shaanxi, China
| | - Xiuhui Yang
- School of Software, Shandong University, Jinan, Shandong, China
| | - Jun Ding
- Department of Medicine, Meakins-Christie Laboratories, McGill University, Montreal, QC, Canada.
| | - Hao Wu
- School of Software, Shandong University, Jinan, Shandong, China.
- Shenzhen Research Institute of Shandong University, Shenzhen, Guangdong, China.
| |
Collapse
|
12
|
Niu Y, Luo J, Zong C. Single-cell total-RNA profiling unveils regulatory hubs of transcription factors. Nat Commun 2024; 15:5941. [PMID: 39009595 PMCID: PMC11251146 DOI: 10.1038/s41467-024-50291-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2023] [Accepted: 07/03/2024] [Indexed: 07/17/2024] Open
Abstract
Recent development of RNA velocity uses master equations to establish the kinetics of the life cycle of RNAs from unspliced RNA to spliced RNA (i.e., mature RNA) to degradation. To feed this kinetic analysis, simultaneous measurement of unspliced RNA and spliced RNA in single cells is greatly desired. However, the majority of single-cell RNA-seq chemistry primarily captures mature RNA species to measure gene expressions. Here, we develop a one-step total-RNA chemistry-based single-cell RNA-seq method: snapTotal-seq. We benchmark this method with multiple single-cell RNA-seq assays in their performance in kinetic analysis of cell cycle by RNA velocity. Next, with LASSO regression between transcription factors, we identify the critical regulatory hubs mediating the cell cycle dynamics. We also apply snapTotal-seq to profile the oncogene-induced senescence and identify the key regulatory hubs governing the entry of senescence. Furthermore, from the comparative analysis of unspliced RNA and spliced RNA, we identify a significant portion of genes whose expression changes occur in spliced RNA but not to the same degree in unspliced RNA, indicating these gene expression changes are mainly controlled by post-transcriptional regulation. Overall, we demonstrate that snapTotal-seq can provide enriched information about gene regulation, especially during the transition between cell states.
Collapse
Affiliation(s)
- Yichi Niu
- Department of Molecular and Human Genetics, Houston, TX, USA
- Genetics & Genomics Program, Houston, TX, USA
| | - Jiayi Luo
- Department of Molecular and Human Genetics, Houston, TX, USA
- Cancer and Cell Biology Program, Houston, TX, USA
| | - Chenghang Zong
- Department of Molecular and Human Genetics, Houston, TX, USA.
- Genetics & Genomics Program, Houston, TX, USA.
- Cancer and Cell Biology Program, Houston, TX, USA.
- Integrative Molecular and Biomedical Sciences Program, Houston, TX, USA.
- Dan L Duncan Comprehensive Cancer Center, Houston, TX, USA.
- McNair Medical Institute, Baylor College of Medicine, Houston, TX, USA.
| |
Collapse
|
13
|
Watson S, Porter H, Sudbery I, Thompson R. Modification of Seurat v4 for the Development of a Phase Assignment Tool Able to Distinguish between G2 and Mitotic Cells. Int J Mol Sci 2024; 25:4589. [PMID: 38731808 PMCID: PMC11083997 DOI: 10.3390/ijms25094589] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2024] [Revised: 04/02/2024] [Accepted: 04/12/2024] [Indexed: 05/13/2024] Open
Abstract
Single-cell RNA sequencing (scRNAseq) is a rapidly advancing field enabling the characterisation of heterogeneous gene expression profiles within a population. The cell cycle phase is a major contributor to gene expression variance between cells and computational analysis tools have been developed to assign cell cycle phases to cells within scRNAseq datasets. Whilst these tools can be extremely useful, all have the drawback that they classify cells as only G1, S or G2/M. Existing discrete cell phase assignment tools are unable to differentiate between G2 and M and continuous-phase-assignment tools are unable to identify a region corresponding specifically to mitosis in a pseudo-timeline for continuous assignment along the cell cycle. In this study, bulk RNA sequencing was used to identify differentially expressed genes between mitotic and interphase cells isolated based on phospho-histone H3 expression using fluorescence-activated cell sorting. These gene lists were used to develop a methodology which can distinguish G2 and M phase cells in scRNAseq datasets. The phase assignment tools present in Seurat were modified to allow for cell cycle phase assignment of all stages of the cell cycle to identify a mitotic-specific cell population.
Collapse
Affiliation(s)
- Steven Watson
- School of Medicine and Population Health, University of Sheffield, Sheffield S10 2TN, UK
| | - Harry Porter
- School of Medicine, University of Nottingham, Nottingham NG5 1PB, UK
| | - Ian Sudbery
- School of Biosciences, University of Sheffield, Sheffield S10 2TN, UK
- Sheffield Institute for Nucleic Acid Research (SInFoNiA), Sheffield S10 2TN, UK
| | - Ruth Thompson
- School of Medicine and Population Health, University of Sheffield, Sheffield S10 2TN, UK
- Sheffield Institute for Nucleic Acid Research (SInFoNiA), Sheffield S10 2TN, UK
| |
Collapse
|
14
|
Karlebach G, Robinson PN. Computing Minimal Boolean Models of Gene Regulatory Networks. J Comput Biol 2024; 31:117-127. [PMID: 37889991 DOI: 10.1089/cmb.2023.0122] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/29/2023] Open
Abstract
Models of gene regulatory networks (GRNs) capture the dynamics of the regulatory processes that occur within the cell as a means to understanding the variability observed in gene expression between different conditions. Arguably the simplest mathematical construct used for modeling is the Boolean network, which dictates a set of logical rules for transition between states described as Boolean vectors. Due to the complexity of gene regulation and the limitations of experimental technologies, in most cases knowledge about regulatory interactions and Boolean states is partial. In addition, the logical rules themselves are not known a priori. Our goal in this work is to create an algorithm that finds the network that fits the data optimally, and identify the network states that correspond to the noise-free data. We present a novel methodology for integrating experimental data and performing a search for the optimal consistent structure via optimization of a linear objective function under a set of linear constraints. In addition, we extend our methodology into a heuristic that alleviates the computational complexity of the problem for datasets that are generated by single-cell RNA-Sequencing (scRNA-Seq). We demonstrate the effectiveness of these tools using simulated data, and in addition a publicly available scRNA-Seq dataset and the GRN that is associated with it. Our methodology will enable researchers to obtain a better understanding of the dynamics of GRNs and their biological role.
Collapse
Affiliation(s)
- Guy Karlebach
- The Jackson Laboratory for Genomic Medicine, Farmington, Connecticut, USA
| | - Peter N Robinson
- The Jackson Laboratory for Genomic Medicine, Farmington, Connecticut, USA
| |
Collapse
|
15
|
Pan X, Zhang X. Studying temporal dynamics of single cells: expression, lineage and regulatory networks. Biophys Rev 2024; 16:57-67. [PMID: 38495440 PMCID: PMC10937865 DOI: 10.1007/s12551-023-01090-5] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2023] [Accepted: 06/27/2023] [Indexed: 03/19/2024] Open
Abstract
Learning how multicellular organs are developed from single cells to different cell types is a fundamental problem in biology. With the high-throughput scRNA-seq technology, computational methods have been developed to reveal the temporal dynamics of single cells from transcriptomic data, from phenomena on cell trajectories to the underlying mechanism that formed the trajectory. There are several distinct families of computational methods including Trajectory Inference (TI), Lineage Tracing (LT), and Gene Regulatory Network (GRN) Inference which are involved in such studies. This review summarizes these computational approaches which use scRNA-seq data to study cell differentiation and cell fate specification as well as the advantages and limitations of different methods. We further discuss how GRNs can potentially affect cell fate decisions and trajectory structures. Supplementary Information The online version contains supplementary material available at 10.1007/s12551-023-01090-5.
Collapse
Affiliation(s)
- Xinhai Pan
- School of Computational Science and Engineering, Georgia Institute of Technology, Atlanta, GA 30332 USA
| | - Xiuwei Zhang
- School of Computational Science and Engineering, Georgia Institute of Technology, Atlanta, GA 30332 USA
| |
Collapse
|
16
|
Guo X, Chen L. From G1 to M: a comparative study of methods for identifying cell cycle phases. Brief Bioinform 2024; 25:bbad517. [PMID: 38261342 PMCID: PMC10805071 DOI: 10.1093/bib/bbad517] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2023] [Revised: 11/08/2023] [Accepted: 12/13/2023] [Indexed: 01/24/2024] Open
Abstract
Accurate identification of cell cycle phases in single-cell RNA-sequencing (scRNA-seq) data is crucial for biomedical research. Many methods have been developed to tackle this challenge, employing diverse approaches to predict cell cycle phases. In this review article, we delve into the standard processes in identifying cell cycle phases within scRNA-seq data and present several representative methods for comparison. To rigorously assess the accuracy of these methods, we propose an error function and employ multiple benchmarking datasets encompassing human and mouse data. Our evaluation results reveal a key finding: the fit between the reference data and the dataset being analyzed profoundly impacts the effectiveness of cell cycle phase identification methods. Therefore, researchers must carefully consider the compatibility between the reference data and their dataset to achieve optimal results. Furthermore, we explore the potential benefits of incorporating benchmarking data with multiple known cell cycle phases into the analysis. Merging such data with the target dataset shows promise in enhancing prediction accuracy. By shedding light on the accuracy and performance of cell cycle phase prediction methods across diverse datasets, this review aims to motivate and guide future methodological advancements. Our findings offer valuable insights for researchers seeking to improve their understanding of cellular dynamics through scRNA-seq analysis, ultimately fostering the development of more robust and widely applicable cell cycle identification methods.
Collapse
Affiliation(s)
- Xinyu Guo
- Department of Quantitative and Computational Biology, University of Southern California, 1050 Childs Way, Los Angeles, CA 90089, United States
| | - Liang Chen
- Department of Quantitative and Computational Biology, University of Southern California, 1050 Childs Way, Los Angeles, CA 90089, United States
| |
Collapse
|
17
|
Lang PF, Penas DR, Banga JR, Weindl D, Novak B. Reusable rule-based cell cycle model explains compartment-resolved dynamics of 16 observables in RPE-1 cells. PLoS Comput Biol 2024; 20:e1011151. [PMID: 38190398 PMCID: PMC10773963 DOI: 10.1371/journal.pcbi.1011151] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2023] [Accepted: 11/24/2023] [Indexed: 01/10/2024] Open
Abstract
The mammalian cell cycle is regulated by a well-studied but complex biochemical reaction system. Computational models provide a particularly systematic and systemic description of the mechanisms governing mammalian cell cycle control. By combining both state-of-the-art multiplexed experimental methods and powerful computational tools, this work aims at improving on these models along four dimensions: model structure, validation data, validation methodology and model reusability. We developed a comprehensive model structure of the full cell cycle that qualitatively explains the behaviour of human retinal pigment epithelial-1 cells. To estimate the model parameters, time courses of eight cell cycle regulators in two compartments were reconstructed from single cell snapshot measurements. After optimisation with a parallel global optimisation metaheuristic we obtained excellent agreements between simulations and measurements. The PEtab specification of the optimisation problem facilitates reuse of model, data and/or optimisation results. Future perturbation experiments will improve parameter identifiability and allow for testing model predictive power. Such a predictive model may aid in drug discovery for cell cycle-related disorders.
Collapse
Affiliation(s)
- Paul F. Lang
- Department of Biochemistry, University of Oxford, Oxford, United Kingdom
| | - David R. Penas
- Computational Biology Lab, MBG-CSIC (Spanish National Research Council), Pontevedra, Spain
| | - Julio R. Banga
- Computational Biology Lab, MBG-CSIC (Spanish National Research Council), Pontevedra, Spain
| | - Daniel Weindl
- Computational Health Center, Helmholtz Zentrum München Deutsches Forschungszentrum für Gesundheit und Umwelt (GmbH), Neuherberg, Germany
| | - Bela Novak
- Department of Biochemistry, University of Oxford, Oxford, United Kingdom
| |
Collapse
|
18
|
Zheng W, Min W, Wang S. TsImpute: an accurate two-step imputation method for single-cell RNA-seq data. Bioinformatics 2023; 39:btad731. [PMID: 38039139 PMCID: PMC10724850 DOI: 10.1093/bioinformatics/btad731] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2023] [Revised: 11/22/2023] [Accepted: 11/30/2023] [Indexed: 12/03/2023] Open
Abstract
MOTIVATION Single-cell RNA sequencing (scRNA-seq) technology has enabled discovering gene expression patterns at single cell resolution. However, due to technical limitations, there are usually excessive zeros, called "dropouts," in scRNA-seq data, which may mislead the downstream analysis. Therefore, it is crucial to impute these dropouts to recover the biological information. RESULTS We propose a two-step imputation method called tsImpute to impute scRNA-seq data. At the first step, tsImpute adopts zero-inflated negative binomial distribution to discriminate dropouts from true zeros and performs initial imputation by calculating the expected expression level. At the second step, it conducts clustering with this modified expression matrix, based on which the final distance weighted imputation is performed. Numerical results based on both simulated and real data show that tsImpute achieves favorable performance in terms of gene expression recovery, cell clustering, and differential expression analysis. AVAILABILITY AND IMPLEMENTATION The R package of tsImpute is available at https://github.com/ZhengWeihuaYNU/tsImpute.
Collapse
Affiliation(s)
- Weihua Zheng
- Department of Computer Science and Engineering, School of Information Science and Engineering, Yunnan University, Kunming 650504, China
| | - Wenwen Min
- Department of Computer Science and Engineering, School of Information Science and Engineering, Yunnan University, Kunming 650504, China
- Yunnan Key Laboratory of Intelligent Systems and Computing, Yunnan University, Kunming 650504, China
| | - Shunfang Wang
- Department of Computer Science and Engineering, School of Information Science and Engineering, Yunnan University, Kunming 650504, China
- Yunnan Key Laboratory of Intelligent Systems and Computing, Yunnan University, Kunming 650504, China
| |
Collapse
|
19
|
Karin J, Bornfeld Y, Nitzan M. scPrisma infers, filters and enhances topological signals in single-cell data using spectral template matching. Nat Biotechnol 2023; 41:1645-1654. [PMID: 36849830 PMCID: PMC10635821 DOI: 10.1038/s41587-023-01663-5] [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] [Received: 04/07/2022] [Accepted: 01/06/2023] [Indexed: 03/01/2023]
Abstract
Single-cell RNA sequencing has been instrumental in uncovering cellular spatiotemporal context. This task is challenging as cells simultaneously encode multiple, potentially cross-interfering, biological signals. Here we propose scPrisma, a spectral computational method that uses topological priors to decouple, enhance and filter different classes of biological processes in single-cell data, such as periodic and linear signals. We apply scPrisma to the analysis of the cell cycle in HeLa cells, circadian rhythm and spatial zonation in liver lobules, diurnal cycle in Chlamydomonas and circadian rhythm in the suprachiasmatic nucleus in the brain. scPrisma can be used to distinguish mixed cellular populations by specific characteristics such as cell type and uncover regulatory networks and cell-cell interactions specific to predefined biological signals, such as the circadian rhythm. We show scPrisma's flexibility in incorporating prior knowledge, inference of topologically informative genes and generalization to additional diverse templates and systems. scPrisma can be used as a stand-alone workflow for signal analysis and as a prior step for downstream single-cell analysis.
Collapse
Affiliation(s)
- Jonathan Karin
- School of Computer Science and Engineering, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Yonathan Bornfeld
- School of Computer Science and Engineering, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Mor Nitzan
- School of Computer Science and Engineering, The Hebrew University of Jerusalem, Jerusalem, Israel.
- Racah Institute of Physics, The Hebrew University of Jerusalem, Jerusalem, Israel.
- Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel.
| |
Collapse
|
20
|
Islam MT, Xing L. Leveraging cell-cell similarity for high-performance spatial and temporal cellular mappings from gene expression data. PATTERNS (NEW YORK, N.Y.) 2023; 4:100840. [PMID: 37876896 PMCID: PMC10591141 DOI: 10.1016/j.patter.2023.100840] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/14/2022] [Revised: 06/29/2023] [Accepted: 08/10/2023] [Indexed: 10/26/2023]
Abstract
Single-cell trajectory mapping and spatial reconstruction are two important developments in life science and provide a unique means to decode heterogeneous tissue formation, cellular dynamics, and tissue developmental processes. The success of these techniques depends critically on the performance of analytical tools used for high-dimensional (HD) gene expression data processing. Existing methods discern the patterns of the data without explicitly considering the underlying biological characteristics of the system, often leading to suboptimal solutions. Here, we present a cell-cell similarity-driven framework of genomic data analysis for high-fidelity spatial and temporal cellular mappings. The approach exploits the similarity features of the cells to discover discriminative patterns of the data. We show that for a wide variety of datasets, the proposed approach drastically improves the accuracies of spatial and temporal mapping analyses compared with state-of-the-art techniques.
Collapse
Affiliation(s)
- Md Tauhidul Islam
- Department of Radiation Oncology, Stanford University, Stanford, CA 94305, USA
| | - Lei Xing
- Department of Radiation Oncology, Stanford University, Stanford, CA 94305, USA
| |
Collapse
|
21
|
Wang Y, Yang C, Wang Z, Wang Y, Yan Q, Feng Y, Liu Y, Huang J, Zhou J. Epithelial Galectin-3 Induced the Mitochondrial Complex Inhibition and Cell Cycle Arrest of CD8 + T Cells in Severe/Critical COVID-19. Int J Mol Sci 2023; 24:12780. [PMID: 37628961 PMCID: PMC10454470 DOI: 10.3390/ijms241612780] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2023] [Revised: 07/20/2023] [Accepted: 08/02/2023] [Indexed: 08/27/2023] Open
Abstract
Previous research suggested that the dramatical decrease in CD8+ T cells is a contributing factor in the poor prognosis and disease progression of COVID-19 patients. However, the underlying mechanisms are not fully understood. In this study, we conducted Single-cell RNA sequencing (scRNA-seq) and single-cell T cell receptor sequencing (scTCR-seq) analysis, which revealed a proliferative-exhausted MCM+FASLGlow CD8+ T cell phenotype in severe/critical COVID-19 patients. These CD8+ T cells were characterized by G2/M cell cycle arrest, downregulation of respiratory chain complex genes, and inhibition of mitochondrial biogenesis. CellChat analysis of infected lung epithelial cells and CD8+ T cells found that the galectin signaling pathway played a crucial role in CD8+ T cell reduction and dysfunction. To further elucidate the mechanisms, we established SARS-CoV-2 ORF3a-transfected A549 cells, and co-cultured them with CD8+ T cells for ex vivo experiments. Our results showed that epithelial galectin-3 inhibited the transcription of the mitochondrial respiratory chain complex III/IV genes of CD8+ T cells by suppressing the nuclear translocation of nuclear respiratory factor 1 (NRF1). Further findings showed that the suppression of NRF1 translocation was associated with ERK-related and Akt-related signaling pathways. Importantly, the galectin-3 inhibitor, TD-139, promoted nuclear translocation of NRF1, thus enhancing the expression of the mitochondrial respiratory chain complex III/IV genes and the mitochondrial biogenesis of CD8+ T cells. Our study provided new insights into the immunopathogenesis of COVID-19 and identified potential therapeutic targets for the prevention and treatment of severe/critical COVID-19 patients.
Collapse
Affiliation(s)
- Yudie Wang
- Department of Biology and Genetics, College of Life Sciences and Health, Wuhan University of Science and Technology, Wuhan 430065, China
| | - Cheng Yang
- Department of Biology and Genetics, College of Life Sciences and Health, Wuhan University of Science and Technology, Wuhan 430065, China
| | - Zhongyi Wang
- Department of Biology and Genetics, College of Life Sciences and Health, Wuhan University of Science and Technology, Wuhan 430065, China
| | - Yi Wang
- Department of Biology and Genetics, College of Life Sciences and Health, Wuhan University of Science and Technology, Wuhan 430065, China
| | - Qing Yan
- Department of Biology and Genetics, College of Life Sciences and Health, Wuhan University of Science and Technology, Wuhan 430065, China
| | - Ying Feng
- Department of Biology and Genetics, College of Life Sciences and Health, Wuhan University of Science and Technology, Wuhan 430065, China
| | - Yanping Liu
- Department of Biology and Genetics, College of Life Sciences and Health, Wuhan University of Science and Technology, Wuhan 430065, China
| | - Juan Huang
- Department of Hematology, Maternal and Child Health Hospital of Hubei Province, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430070, China
| | - Jingjiao Zhou
- Department of Biology and Genetics, College of Life Sciences and Health, Wuhan University of Science and Technology, Wuhan 430065, China
| |
Collapse
|
22
|
Cai C, Yue Y, Yue B. Single-cell RNA sequencing in skeletal muscle developmental biology. Biomed Pharmacother 2023; 162:114631. [PMID: 37003036 DOI: 10.1016/j.biopha.2023.114631] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2023] [Revised: 03/29/2023] [Accepted: 03/29/2023] [Indexed: 04/01/2023] Open
Abstract
Skeletal muscle is the most extensive tissue in mammals, and they perform several functions; it is derived from paraxial mesodermal somites and undergoes hyperplasia and hypertrophy to form multinucleated, contractile, and functional muscle fibers. Skeletal muscle is a complex heterogeneous tissue composed of various cell types that establish communication strategies to exchange biological information; therefore, characterizing the cellular heterogeneity and transcriptional signatures of skeletal muscle is central to understanding its ontogeny's details. Studies of skeletal myogenesis have focused primarily on myogenic cells' proliferation, differentiation, migration, and fusion and ignored the intricate network of cells with specific biological functions. The rapid development of single-cell sequencing technology has recently enabled the exploration of skeletal muscle cell types and molecular events during development. This review summarizes the progress in single-cell RNA sequencing and its applications in skeletal myogenesis, which will provide insights into skeletal muscle pathophysiology.
Collapse
Affiliation(s)
- Cuicui Cai
- Key Laboratory of Qinghai-Tibetan Plateau Animal Genetic Resource Reservation and Utilization, Sichuan Province and Ministry of Education, Southwest Minzu University, Chengdu 610225, China; Guyuan Branch, Ningxia Academy of Agriculture and Forestry Sciences, Guyuan 7560000, China
| | - Yuan Yue
- Department of Pathobiology and Immunology, Hebei University of Chinese Medicine, Shijiazhuang 050200, China
| | - Binglin Yue
- Key Laboratory of Qinghai-Tibetan Plateau Animal Genetic Resource Reservation and Utilization, Sichuan Province and Ministry of Education, Southwest Minzu University, Chengdu 610225, China.
| |
Collapse
|
23
|
Pu J, Wang B, Liu X, Chen L, Li SC. SMURF: embedding single-cell RNA-seq data with matrix factorization preserving self-consistency. Brief Bioinform 2023; 24:7008800. [PMID: 36715274 DOI: 10.1093/bib/bbad026] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2022] [Revised: 12/17/2022] [Accepted: 01/09/2023] [Indexed: 01/31/2023] Open
Abstract
The advance in single-cell RNA-sequencing (scRNA-seq) sheds light on cell-specific transcriptomic studies of cell developments, complex diseases and cancers. Nevertheless, scRNA-seq techniques suffer from 'dropout' events, and imputation tools are proposed to address the sparsity. Here, rather than imputation, we propose a tool, SMURF, to extract the low-dimensional embeddings from cells and genes utilizing matrix factorization with a mixture of Poisson-Gamma divergent as objective while preserving self-consistency. SMURF exhibits feasible cell subpopulation discovery efficacy with obtained cell embeddings on replicated in silico and eight web lab scRNA datasets with ground truth cell types. Furthermore, SMURF can reduce the cell embedding to a 1D-oval space to recover the time course of cell cycle. SMURF can also serve as an imputation tool; the in silico data assessment shows that SMURF parades the most robust gene expression recovery power with low root mean square error and high Pearson correlation. Moreover, SMURF recovers the gene distribution for the WM989 Drop-seq data. SMURF is available at https://github.com/deepomicslab/SMURF.
Collapse
Affiliation(s)
- Juhua Pu
- State Key Laboratory of Software Development Environment, Beihang University, Beijing, China
- Beihang Hangzhou Innovation Institute Yuhang, Xixi Octagon City, Yuhang District, Hangzhou 310023, China
| | - Bingchen Wang
- State Key Laboratory of Software Development Environment, Beihang University, Beijing, China
- Beihang Hangzhou Innovation Institute Yuhang, Xixi Octagon City, Yuhang District, Hangzhou 310023, China
| | - Xingwu Liu
- School of Mathematical Sciences, Dalian University of Technology, Dalian, Liaoning, China
| | - Lingxi Chen
- Department of Computer Science, City University of Hong Kong, 83 Tat Chee Ave, Kowloon Tong, Hong Kong, China
| | - Shuai Cheng Li
- Department of Computer Science, City University of Hong Kong, 83 Tat Chee Ave, Kowloon Tong, Hong Kong, China
| |
Collapse
|
24
|
Li L, Zhao Y, Li H, Zhang S. BLTSA: pseudotime prediction for single cells by branched local tangent space alignment. Bioinformatics 2023; 39:7000337. [PMID: 36692140 PMCID: PMC9923702 DOI: 10.1093/bioinformatics/btad054] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2022] [Revised: 12/11/2022] [Accepted: 01/23/2023] [Indexed: 01/25/2023] Open
Abstract
MOTIVATION The development of single-cell RNA sequencing (scRNA-seq) technology makes it possible to study the cellular dynamic processes such as cell cycle and cell differentiation. Due to the difficulties in generating genuine time-series scRNA-seq data, it is of great importance to computationally infer the pseudotime of the cells along differentiation trajectory based on their gene expression patterns. The existing pseudotime prediction methods often suffer from the high level noise of single-cell data, thus it is still necessary to study the single-cell trajectory inference methods. RESULTS In this study, we propose a branched local tangent space alignment (BLTSA) method to infer single-cell pseudotime for multi-furcation trajectories. By assuming that single cells are sampled from a low-dimensional self-intersecting manifold, BLTSA first identifies the tip and branching cells in the trajectory based on cells' local Euclidean neighborhoods. Local coordinates within the tangent spaces are then determined by each cell's local neighborhood after clustering all the cells to different branches iteratively. The global coordinates for all the single cells are finally obtained by aligning the local coordinates based on the tangent spaces. We evaluate the performance of BLTSA on four simulation datasets and five real datasets. The experimental results show that BLTSA has obvious advantages over other comparison methods. AVAILABILITY AND IMPLEMENTATION R codes are available at https://github.com/LiminLi-xjtu/BLTSA. SUPPLEMENTARY INFORMATION Supplementary data are available at Bioinformatics online.
Collapse
Affiliation(s)
- Limin Li
- School of Mathematics and Statistics, Xi'an Jiaotong University, Xi'an 710049, China
| | - Yameng Zhao
- School of Mathematics and Statistics, Xi'an Jiaotong University, Xi'an 710049, China
| | - Huiran Li
- School of Mathematics and Statistics, Xi'an Jiaotong University, Xi'an 710049, China
| | - Shuqin Zhang
- School of Mathematical Sciences, Fudan University, Shanghai 200433, China
| |
Collapse
|
25
|
Khozyainova AA, Valyaeva AA, Arbatsky MS, Isaev SV, Iamshchikov PS, Volchkov EV, Sabirov MS, Zainullina VR, Chechekhin VI, Vorobev RS, Menyailo ME, Tyurin-Kuzmin PA, Denisov EV. Complex Analysis of Single-Cell RNA Sequencing Data. BIOCHEMISTRY. BIOKHIMIIA 2023; 88:231-252. [PMID: 37072324 PMCID: PMC10000364 DOI: 10.1134/s0006297923020074] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/23/2022] [Revised: 12/13/2022] [Accepted: 12/13/2022] [Indexed: 03/12/2023]
Abstract
Single-cell RNA sequencing (scRNA-seq) is a revolutionary tool for studying the physiology of normal and pathologically altered tissues. This approach provides information about molecular features (gene expression, mutations, chromatin accessibility, etc.) of cells, opens up the possibility to analyze the trajectories/phylogeny of cell differentiation and cell-cell interactions, and helps in discovery of new cell types and previously unexplored processes. From a clinical point of view, scRNA-seq facilitates deeper and more detailed analysis of molecular mechanisms of diseases and serves as a basis for the development of new preventive, diagnostic, and therapeutic strategies. The review describes different approaches to the analysis of scRNA-seq data, discusses the advantages and disadvantages of bioinformatics tools, provides recommendations and examples of their successful use, and suggests potential directions for improvement. We also emphasize the need for creating new protocols, including multiomics ones, for the preparation of DNA/RNA libraries of single cells with the purpose of more complete understanding of individual cells.
Collapse
Affiliation(s)
- Anna A Khozyainova
- Laboratory of Cancer Progression Biology, Cancer Research Institute, Tomsk National Research Medical Center, Russian Academy of Sciences, Tomsk, 634050, Russia.
| | - Anna A Valyaeva
- Faculty of Bioengineering and Bioinformatics, Lomonosov Moscow State University, Moscow, 119991, Russia
- Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Moscow, 119991, Russia
| | - Mikhail S Arbatsky
- Laboratory of Artificial Intelligence and Bioinformatics, The Russian Clinical Research Center for Gerontology, Pirogov Russian National Medical University, Moscow, 129226, Russia
- School of Public Administration, Lomonosov Moscow State University, Moscow, 119991, Russia
- Faculty of Fundamental Medicine, Lomonosov Moscow State University, Moscow, 119991, Russia
| | - Sergey V Isaev
- Research Institute of Personalized Medicine, National Center for Personalized Medicine of Endocrine Diseases, National Medical Research Center for Endocrinology, Moscow, 117036, Russia
| | - Pavel S Iamshchikov
- Laboratory of Cancer Progression Biology, Cancer Research Institute, Tomsk National Research Medical Center, Russian Academy of Sciences, Tomsk, 634050, Russia
- Laboratory of Complex Analysis of Big Bioimage Data, National Research Tomsk State University, Tomsk, 634050, Russia
| | - Egor V Volchkov
- Department of Oncohematology, Dmitry Rogachev National Research Center of Pediatric Hematology, Oncology and Immunology, Moscow, 117198, Russia
| | - Marat S Sabirov
- Laboratory of Bioinformatics and Molecular Genetics, Koltzov Institute of Developmental Biology of the Russian Academy of Sciences, Moscow, 119334, Russia
| | - Viktoria R Zainullina
- Laboratory of Cancer Progression Biology, Cancer Research Institute, Tomsk National Research Medical Center, Russian Academy of Sciences, Tomsk, 634050, Russia
| | - Vadim I Chechekhin
- Faculty of Fundamental Medicine, Lomonosov Moscow State University, Moscow, 119991, Russia
| | - Rostislav S Vorobev
- Laboratory of Cancer Progression Biology, Cancer Research Institute, Tomsk National Research Medical Center, Russian Academy of Sciences, Tomsk, 634050, Russia
| | - Maxim E Menyailo
- Laboratory of Cancer Progression Biology, Cancer Research Institute, Tomsk National Research Medical Center, Russian Academy of Sciences, Tomsk, 634050, Russia
| | - Pyotr A Tyurin-Kuzmin
- Faculty of Fundamental Medicine, Lomonosov Moscow State University, Moscow, 119991, Russia
| | - Evgeny V Denisov
- Laboratory of Cancer Progression Biology, Cancer Research Institute, Tomsk National Research Medical Center, Russian Academy of Sciences, Tomsk, 634050, Russia
| |
Collapse
|
26
|
Chen L, Li S. Incorporating cell hierarchy to decipher the functional diversity of single cells. Nucleic Acids Res 2023; 51:e9. [PMID: 36373664 PMCID: PMC9881154 DOI: 10.1093/nar/gkac1044] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2022] [Revised: 10/13/2022] [Accepted: 10/21/2022] [Indexed: 11/16/2022] Open
Abstract
Cells possess functional diversity hierarchically. However, most single-cell analyses neglect the nested structures while detecting and visualizing the functional diversity. Here, we incorporate cell hierarchy to study functional diversity at subpopulation, club (i.e., sub-subpopulation), and cell layers. Accordingly, we implement a package, SEAT, to construct cell hierarchies utilizing structure entropy by minimizing the global uncertainty in cell-cell graphs. With cell hierarchies, SEAT deciphers functional diversity in 36 datasets covering scRNA, scDNA, scATAC, and scRNA-scATAC multiome. First, SEAT finds optimal cell subpopulations with high clustering accuracy. It identifies cell types or fates from omics profiles and boosts accuracy from 0.34 to 1. Second, SEAT detects insightful functional diversity among cell clubs. The hierarchy of breast cancer cells reveals that the specific tumor cell club drives AREG-EGFT signaling. We identify a dense co-accessibility network of cis-regulatory elements specified by one cell club in GM12878. Third, the cell order from the hierarchy infers periodic pseudo-time of cells, improving accuracy from 0.79 to 0.89. Moreover, we incorporate cell hierarchy layers as prior knowledge to refine nonlinear dimension reduction, enabling us to visualize hierarchical cell layouts in low-dimensional space.
Collapse
Affiliation(s)
- Lingxi Chen
- Department of Computer Science, City University of Hong Kong, 83 Tat Chee Ave, Kowloon Tong, Hong Kong, China
- City University of Hong Kong Shenzhen Research Institute, Shenzhen, 518057, Guangdong, China
| | - Shuai Cheng Li
- Department of Computer Science, City University of Hong Kong, 83 Tat Chee Ave, Kowloon Tong, Hong Kong, China
- City University of Hong Kong Shenzhen Research Institute, Shenzhen, 518057, Guangdong, China
| |
Collapse
|
27
|
Gavish A, Chain B, Salame TM, Antebi YE, Nevo S, Reich-Zeliger S, Friedman N. From pseudo to real-time dynamics of T cell thymic differentiation. iScience 2022; 26:105826. [PMID: 36624839 PMCID: PMC9823121 DOI: 10.1016/j.isci.2022.105826] [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: 06/09/2022] [Revised: 11/14/2022] [Accepted: 12/09/2022] [Indexed: 12/23/2022] Open
Abstract
Numerous methods have recently emerged for ordering single cells along developmental trajectories. However, accurate depiction of developmental dynamics can only be achieved after rescaling the trajectory according to the relative time spent at each developmental point. We formulate a model which estimates local cell densities and fluxes, and incorporates cell division and apoptosis rates, to infer the real-time dimension of the developmental trajectory. We validate the model using mathematical simulations and apply it to experimental high dimensional cytometry data obtained from the mouse thymus to construct the true time profile of the thymocyte developmental process. Our method can easily be implemented in any of the existing tools for trajectory inference.
Collapse
Affiliation(s)
- Avishai Gavish
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, Israel,Corresponding author
| | - Benny Chain
- Division of Infection and Immunity, University College London, London, UK
| | - Tomer M. Salame
- Life Sciences Core Facilities, Weizmann Institute of Science, Rehovot, Israel
| | - Yaron E. Antebi
- Department of Molecular Genetics, Weizmann Institute of Science, Rehovot, Israel
| | - Shir Nevo
- Department of Immunology, Weizmann Institute of Science, Rehovot, Israel
| | - Shlomit Reich-Zeliger
- Department of Immunology, Weizmann Institute of Science, Rehovot, Israel,Corresponding author
| | - Nir Friedman
- Department of Immunology, Weizmann Institute of Science, Rehovot, Israel
| |
Collapse
|
28
|
Auerbach BJ, FitzGerald GA, Li M. Tempo: an unsupervised Bayesian algorithm for circadian phase inference in single-cell transcriptomics. Nat Commun 2022; 13:6580. [PMID: 36323668 PMCID: PMC9630322 DOI: 10.1038/s41467-022-34185-w] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2022] [Accepted: 10/17/2022] [Indexed: 11/05/2022] Open
Abstract
The circadian clock is a 24 h cellular timekeeping mechanism that regulates human physiology. Answering several fundamental questions in circadian biology will require joint measures of single-cell circadian phases and transcriptomes. However, no widespread experimental approaches exist for this purpose. While computational approaches exist to infer cell phase directly from single-cell RNA-sequencing data, existing methods yield poor circadian phase estimates, and do not quantify estimation uncertainty, which is essential for interpretation of results from very sparse single-cell RNA-sequencing data. To address these unmet needs, we introduce Tempo, a Bayesian variational inference approach that incorporates domain knowledge of the clock and quantifies phase estimation uncertainty. Through simulations and analyses of real data, we demonstrate that Tempo yields more accurate estimates of circadian phase than existing methods and provides well-calibrated uncertainty quantifications. Tempo will facilitate large-scale studies of single-cell circadian transcription.
Collapse
Affiliation(s)
- Benjamin J Auerbach
- Graduate Group in Genomics and Computational Biology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, 19104, USA.
| | - Garret A FitzGerald
- Institute for Translational Medicine and Therapeutics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, 19104, USA
| | - Mingyao Li
- Department of Biostatistics, Epidemiology and Informatics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, 19104, USA.
| |
Collapse
|
29
|
Liu Z, Li H, Dang Q, Weng S, Duo M, Lv J, Han X. Integrative insights and clinical applications of single-cell sequencing in cancer immunotherapy. Cell Mol Life Sci 2022; 79:577. [PMID: 36316529 PMCID: PMC11803023 DOI: 10.1007/s00018-022-04608-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2022] [Revised: 10/12/2022] [Accepted: 10/20/2022] [Indexed: 11/03/2022]
Abstract
Recently, immunotherapy has gained increasing popularity in oncology. Several immunotherapies obtained remarkable clinical effects, but the efficacy varied, and only subsets of cancer patients benefited. Breaking the constraints and improving immunotherapy efficacy is extremely important in precision medicine. Whereas traditional sequencing approaches mask the characteristics of individual cells, single-cell sequencing provides multiple dimensions of cellular characterization at the single-cell level, including genomic, transcriptomic, epigenomic, proteomic, and multi-omics. Hence, the complexity of the tumor microenvironment, the universality of tumor heterogeneity, cell composition and cell-cell interactions, cell lineage tracking, and tumor drug resistance mechanisms are revealed in-depth. However, the clinical transformation of single-cell technology is not to the point of in-depth study, especially in the application of immunotherapy. The newly discovered vital cells and tremendous biomarkers facilitate the development of more efficient individualized therapeutic regimens to guide clinical treatment and predict prognosis. This review provided an overview of the progress in distinct single-cell sequencing methods and emerging strategies. For perspective, the expanding utility of combining single-cell sequencing and other technologies was discussed.
Collapse
Affiliation(s)
- Zaoqu Liu
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan, China
- Interventional Institute of Zhengzhou University, Zhengzhou, 450052, Henan, China
- Interventional Treatment and Clinical Research Center of Henan Province, Zhengzhou, 450052, Henan, China
| | - Huanyun Li
- Genetic and Prenatal Diagnosis Center, Department of Obstetrics and Gynecology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan, China
| | - Qin Dang
- Department of Colorectal Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan, China
| | - Siyuan Weng
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan, China
| | - Mengjie Duo
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan, China
| | - Jinxiang Lv
- Department of Gastroenterology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan, China
| | - Xinwei Han
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan, China.
- Interventional Institute of Zhengzhou University, Zhengzhou, 450052, Henan, China.
- Interventional Treatment and Clinical Research Center of Henan Province, Zhengzhou, 450052, Henan, China.
| |
Collapse
|
30
|
Gorin G, Fang M, Chari T, Pachter L. RNA velocity unraveled. PLoS Comput Biol 2022; 18:e1010492. [PMID: 36094956 PMCID: PMC9499228 DOI: 10.1371/journal.pcbi.1010492] [Citation(s) in RCA: 75] [Impact Index Per Article: 25.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2022] [Revised: 09/22/2022] [Accepted: 08/14/2022] [Indexed: 11/24/2022] Open
Abstract
We perform a thorough analysis of RNA velocity methods, with a view towards understanding the suitability of the various assumptions underlying popular implementations. In addition to providing a self-contained exposition of the underlying mathematics, we undertake simulations and perform controlled experiments on biological datasets to assess workflow sensitivity to parameter choices and underlying biology. Finally, we argue for a more rigorous approach to RNA velocity, and present a framework for Markovian analysis that points to directions for improvement and mitigation of current problems.
Collapse
Affiliation(s)
- Gennady Gorin
- Division of Chemistry and Chemical Engineering, California Institute of Technology, Pasadena, California, United States of America
| | - Meichen Fang
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, California, United States of America
| | - Tara Chari
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, California, United States of America
| | - Lior Pachter
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, California, United States of America
- Department of Computing and Mathematical Sciences, California Institute of Technology, Pasadena, California, United States of America
| |
Collapse
|
31
|
Tegowski M, Flamand MN, Meyer KD. scDART-seq reveals distinct m 6A signatures and mRNA methylation heterogeneity in single cells. Mol Cell 2022; 82:868-878.e10. [PMID: 35081365 PMCID: PMC8857065 DOI: 10.1016/j.molcel.2021.12.038] [Citation(s) in RCA: 102] [Impact Index Per Article: 34.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2021] [Revised: 12/22/2021] [Accepted: 12/23/2021] [Indexed: 12/14/2022]
Abstract
N6-methyladenosine (m6A) is an abundant RNA modification that plays critical roles in RNA regulation and cellular function. Global m6A profiling has revealed important aspects of m6A distribution and function, but to date such studies have been restricted to large populations of cells. Here, we develop a method to identify m6A sites transcriptome-wide in single cells. We uncover surprising heterogeneity in the presence and abundance of m6A sites across individual cells and identify differentially methylated mRNAs across the cell cycle. Additionally, we show that cellular subpopulations can be distinguished based on their RNA methylation signatures, independent from gene expression. These studies reveal fundamental features of m6A that have been missed by m6A profiling of bulk cells and suggest the presence of cell-intrinsic mechanisms for m6A deposition.
Collapse
Affiliation(s)
- Matthew Tegowski
- Department of Biochemistry, Duke University School of Medicine, Durham, NC 27710, USA
| | - Mathieu N Flamand
- Department of Biochemistry, Duke University School of Medicine, Durham, NC 27710, USA
| | - Kate D Meyer
- Department of Biochemistry, Duke University School of Medicine, Durham, NC 27710, USA; Department of Neurobiology, Duke University School of Medicine, Durham, NC 27710, USA.
| |
Collapse
|
32
|
Deshpande A, Chu LF, Stewart R, Gitter A. Network inference with Granger causality ensembles on single-cell transcriptomics. Cell Rep 2022; 38:110333. [PMID: 35139376 PMCID: PMC9093087 DOI: 10.1016/j.celrep.2022.110333] [Citation(s) in RCA: 50] [Impact Index Per Article: 16.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2019] [Revised: 02/19/2021] [Accepted: 01/12/2022] [Indexed: 12/20/2022] Open
Abstract
Cellular gene expression changes throughout a dynamic biological process, such as differentiation. Pseudotimes estimate cells' progress along a dynamic process based on their individual gene expression states. Ordering the expression data by pseudotime provides information about the underlying regulator-gene interactions. Because the pseudotime distribution is not uniform, many standard mathematical methods are inapplicable for analyzing the ordered gene expression states. Here we present single-cell inference of networks using Granger ensembles (SINGE), an algorithm for gene regulatory network inference from ordered single-cell gene expression data. SINGE uses kernel-based Granger causality regression to smooth irregular pseudotimes and missing expression values. It aggregates predictions from an ensemble of regression analyses to compile a ranked list of candidate interactions between transcriptional regulators and target genes. In two mouse embryonic stem cell differentiation datasets, SINGE outperforms other contemporary algorithms. However, a more detailed examination reveals caveats about poor performance for individual regulators and uninformative pseudotimes.
Collapse
Affiliation(s)
- Atul Deshpande
- Department of Electrical and Computer Engineering, University of Wisconsin - Madison, Madison, WI 53706, USA; Morgridge Institute for Research, Madison, WI 53715, USA
| | - Li-Fang Chu
- Morgridge Institute for Research, Madison, WI 53715, USA
| | - Ron Stewart
- Morgridge Institute for Research, Madison, WI 53715, USA
| | - Anthony Gitter
- Morgridge Institute for Research, Madison, WI 53715, USA; Department of Biostatistics and Medical Informatics, University of Wisconsin - Madison, Madison, WI 53792, USA.
| |
Collapse
|
33
|
Zinovyev A, Sadovsky M, Calzone L, Fouché A, Groeneveld CS, Chervov A, Barillot E, Gorban AN. Modeling Progression of Single Cell Populations Through the Cell Cycle as a Sequence of Switches. Front Mol Biosci 2022; 8:793912. [PMID: 35178429 PMCID: PMC8846220 DOI: 10.3389/fmolb.2021.793912] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2021] [Accepted: 12/15/2021] [Indexed: 11/13/2022] Open
Abstract
Cell cycle is a biological process underlying the existence and propagation of life in time and space. It has been an object for mathematical modeling for long, with several alternative mechanistic modeling principles suggested, describing in more or less details the known molecular mechanisms. Recently, cell cycle has been investigated at single cell level in snapshots of unsynchronized cell populations, exploiting the new methods for transcriptomic and proteomic molecular profiling. This raises a need for simplified semi-phenomenological cell cycle models, in order to formalize the processes underlying the cell cycle, at a higher abstracted level. Here we suggest a modeling framework, recapitulating the most important properties of the cell cycle as a limit trajectory of a dynamical process characterized by several internal states with switches between them. In the simplest form, this leads to a limit cycle trajectory, composed by linear segments in logarithmic coordinates describing some extensive (depending on system size) cell properties. We prove a theorem connecting the effective embedding dimensionality of the cell cycle trajectory with the number of its linear segments. We also develop a simplified kinetic model with piecewise-constant kinetic rates describing the dynamics of lumps of genes involved in S-phase and G2/M phases. We show how the developed cell cycle models can be applied to analyze the available single cell datasets and simulate certain properties of the observed cell cycle trajectories. Based on our model, we can predict with good accuracy the cell line doubling time from the length of cell cycle trajectory.
Collapse
Affiliation(s)
- Andrei Zinovyev
- Institut Curie, PSL Research University, Paris, France
- INSERM, Paris, France
- MINES ParisTech, PSL Research University, CBIO-Centre for Computational Biology, Paris, France
- *Correspondence: Andrei Zinovyev,
| | - Michail Sadovsky
- Institute of Computational Modeling (RAS), Krasnoyarsk, Russia
- Laboratory of Medical Cybernetics, V.F.Voino-Yasenetsky Krasnoyarsk State Medical University, Krasnoyarsk, Russia
- Federal Research and Clinic Center of FMBA of Russia, Krasnoyarsk, Russia
- Laboratory of Advanced Methods for High-Dimensional Data Analysis, Lobachevsky University, Nizhniy Novgorod, Russia
| | - Laurence Calzone
- Institut Curie, PSL Research University, Paris, France
- INSERM, Paris, France
- MINES ParisTech, PSL Research University, CBIO-Centre for Computational Biology, Paris, France
| | - Aziz Fouché
- Institut Curie, PSL Research University, Paris, France
- INSERM, Paris, France
- MINES ParisTech, PSL Research University, CBIO-Centre for Computational Biology, Paris, France
| | - Clarice S. Groeneveld
- Cartes d’Identité des Tumeurs (CIT) Program, Ligue Nationale Contre le Cancer, Paris, France
- Oncologie Moleculaire, UMR144, Institut Curie, Paris, France
| | - Alexander Chervov
- Institut Curie, PSL Research University, Paris, France
- INSERM, Paris, France
- MINES ParisTech, PSL Research University, CBIO-Centre for Computational Biology, Paris, France
| | - Emmanuel Barillot
- Institut Curie, PSL Research University, Paris, France
- INSERM, Paris, France
- MINES ParisTech, PSL Research University, CBIO-Centre for Computational Biology, Paris, France
| | - Alexander N. Gorban
- Laboratory of Advanced Methods for High-Dimensional Data Analysis, Lobachevsky University, Nizhniy Novgorod, Russia
- Department of Mathematics, University of Leicester, Leicester, United Kingdom
| |
Collapse
|
34
|
Zheng SC, Stein-O'Brien G, Augustin JJ, Slosberg J, Carosso GA, Winer B, Shin G, Bjornsson HT, Goff LA, Hansen KD. Universal prediction of cell-cycle position using transfer learning. Genome Biol 2022; 23:41. [PMID: 35101061 PMCID: PMC8802487 DOI: 10.1186/s13059-021-02581-y] [Citation(s) in RCA: 52] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2021] [Accepted: 12/17/2021] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND The cell cycle is a highly conserved, continuous process which controls faithful replication and division of cells. Single-cell technologies have enabled increasingly precise measurements of the cell cycle both as a biological process of interest and as a possible confounding factor. Despite its importance and conservation, there is no universally applicable approach to infer position in the cell cycle with high-resolution from single-cell RNA-seq data. RESULTS Here, we present tricycle, an R/Bioconductor package, to address this challenge by leveraging key features of the biology of the cell cycle, the mathematical properties of principal component analysis of periodic functions, and the use of transfer learning. We estimate a cell-cycle embedding using a fixed reference dataset and project new data into this reference embedding, an approach that overcomes key limitations of learning a dataset-dependent embedding. Tricycle then predicts a cell-specific position in the cell cycle based on the data projection. The accuracy of tricycle compares favorably to gold-standard experimental assays, which generally require specialized measurements in specifically constructed in vitro systems. Using internal controls which are available for any dataset, we show that tricycle predictions generalize to datasets with multiple cell types, across tissues, species, and even sequencing assays. CONCLUSIONS Tricycle generalizes across datasets and is highly scalable and applicable to atlas-level single-cell RNA-seq data.
Collapse
Affiliation(s)
- Shijie C Zheng
- Department of Biostatistics, Johns Hopkins Bloomberg School of Public Health, Baltimore, USA
| | - Genevieve Stein-O'Brien
- Department of Genetic Medicine, Johns Hopkins School of Medicine, Baltimore, USA
- Department of Neuroscience, Johns Hopkins School of Medicine, Baltimore, USA
- Kavli Neurodiscovery Institute, Johns Hopkins University, Baltimore, USA
- Division of Biostatistics and Bioinformatics, Department of Oncology, Johns Hopkins School of Medicine, Baltimore, USA
| | - Jonathan J Augustin
- Department of Genetic Medicine, Johns Hopkins School of Medicine, Baltimore, USA
| | - Jared Slosberg
- Department of Genetic Medicine, Johns Hopkins School of Medicine, Baltimore, USA
| | - Giovanni A Carosso
- Department of Genetic Medicine, Johns Hopkins School of Medicine, Baltimore, USA
| | - Briana Winer
- Department of Genetic Medicine, Johns Hopkins School of Medicine, Baltimore, USA
| | - Gloria Shin
- Department of Genetic Medicine, Johns Hopkins School of Medicine, Baltimore, USA
| | - Hans T Bjornsson
- Department of Genetic Medicine, Johns Hopkins School of Medicine, Baltimore, USA
- Department of Pediatrics, Johns Hopkins School of Medicine, Baltimore, USA
- Faculty of Medicine, Univeristy of Iceland, Reykjavik, Iceland
- Landspitali University Hospital, Reykjavik, Iceland
| | - Loyal A Goff
- Department of Genetic Medicine, Johns Hopkins School of Medicine, Baltimore, USA.
- Department of Neuroscience, Johns Hopkins School of Medicine, Baltimore, USA.
- Kavli Neurodiscovery Institute, Johns Hopkins University, Baltimore, USA.
| | - Kasper D Hansen
- Department of Biostatistics, Johns Hopkins Bloomberg School of Public Health, Baltimore, USA.
- Department of Genetic Medicine, Johns Hopkins School of Medicine, Baltimore, USA.
| |
Collapse
|
35
|
Liu J, Yang M, Zhao W, Zhou X. CCPE: cell cycle pseudotime estimation for single cell RNA-seq data. Nucleic Acids Res 2022; 50:704-716. [PMID: 34931240 PMCID: PMC8789092 DOI: 10.1093/nar/gkab1236] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2021] [Revised: 11/22/2021] [Accepted: 12/15/2021] [Indexed: 11/25/2022] Open
Abstract
Pseudotime analysis from scRNA-seq data enables to characterize the continuous progression of various biological processes, such as the cell cycle. Cell cycle plays an important role in cell fate decisions and differentiation and is often regarded as a confounder in scRNA-seq data analysis when analyzing the role of other factors. Therefore, accurate prediction of cell cycle pseudotime and identification of cell cycle stages are important steps for characterizing the development-related biological processes. Here, we develop CCPE, a novel cell cycle pseudotime estimation method to characterize cell cycle timing and identify cell cycle phases from scRNA-seq data. CCPE uses a discriminative helix to characterize the circular process of the cell cycle and estimates each cell's pseudotime along the cell cycle. We evaluated the performance of CCPE based on a variety of simulated and real scRNA-seq datasets. Our results indicate that CCPE is an effective method for cell cycle estimation and competitive in various applications compared with other existing methods. CCPE successfully identified cell cycle marker genes and is robust to dropout events in scRNA-seq data. Accurate prediction of the cell cycle using CCPE can also effectively facilitate the removal of cell cycle effects across cell types or conditions.
Collapse
Affiliation(s)
- Jiajia Liu
- College of Electronic and Information Engineering, Tongji University, Shanghai, Shanghai 201804, China
- Center for Computational Systems Medicine, School of Biomedical Informatics, The University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| | - Mengyuan Yang
- Center for Computational Systems Medicine, School of Biomedical Informatics, The University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| | - Weiling Zhao
- Center for Computational Systems Medicine, School of Biomedical Informatics, The University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| | - Xiaobo Zhou
- Center for Computational Systems Medicine, School of Biomedical Informatics, The University of Texas Health Science Center at Houston, Houston, TX 77030, USA
- McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX 77030, USA
- School of Dentistry, The University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| |
Collapse
|
36
|
Monti M, Fiorentino J, Milanetti E, Gosti G, Tartaglia GG. Prediction of Time Series Gene Expression and Structural Analysis of Gene Regulatory Networks Using Recurrent Neural Networks. ENTROPY (BASEL, SWITZERLAND) 2022; 24:141. [PMID: 35205437 PMCID: PMC8871363 DOI: 10.3390/e24020141] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/13/2021] [Revised: 01/14/2022] [Accepted: 01/15/2022] [Indexed: 11/17/2022]
Abstract
Methods for time series prediction and classification of gene regulatory networks (GRNs) from gene expression data have been treated separately so far. The recent emergence of attention-based recurrent neural network (RNN) models boosted the interpretability of RNN parameters, making them appealing for the understanding of gene interactions. In this work, we generated synthetic time series gene expression data from a range of archetypal GRNs and we relied on a dual attention RNN to predict the gene temporal dynamics. We show that the prediction is extremely accurate for GRNs with different architectures. Next, we focused on the attention mechanism of the RNN and, using tools from graph theory, we found that its graph properties allow one to hierarchically distinguish different architectures of the GRN. We show that the GRN responded differently to the addition of noise in the prediction by the RNN and we related the noise response to the analysis of the attention mechanism. In conclusion, this work provides a way to understand and exploit the attention mechanism of RNNs and it paves the way to RNN-based methods for time series prediction and inference of GRNs from gene expression data.
Collapse
Affiliation(s)
- Michele Monti
- RNA System Biology Lab, Department of Neuroscience and Brain Technologies, Istituto Italiano di Tecnologia, Via Morego 30, 16163 Genoa, Italy
- Centre for Genomic Regulation (CRG), The Barcelona Institute for Science and Technology, Dr. Aiguader 88, 08003 Barcelona, Spain
| | - Jonathan Fiorentino
- Center for Life Nano- & Neuro-Science, Istituto Italiano di Tecnologia, Viale Regina Elena 291, 00161 Rome, Italy; (J.F.); (E.M.); (G.G.)
| | - Edoardo Milanetti
- Center for Life Nano- & Neuro-Science, Istituto Italiano di Tecnologia, Viale Regina Elena 291, 00161 Rome, Italy; (J.F.); (E.M.); (G.G.)
- Department of Physics, Sapienza University of Rome, 00185 Rome, Italy
| | - Giorgio Gosti
- Center for Life Nano- & Neuro-Science, Istituto Italiano di Tecnologia, Viale Regina Elena 291, 00161 Rome, Italy; (J.F.); (E.M.); (G.G.)
- Department of Physics, Sapienza University of Rome, 00185 Rome, Italy
| | - Gian Gaetano Tartaglia
- RNA System Biology Lab, Department of Neuroscience and Brain Technologies, Istituto Italiano di Tecnologia, Via Morego 30, 16163 Genoa, Italy
- Centre for Genomic Regulation (CRG), The Barcelona Institute for Science and Technology, Dr. Aiguader 88, 08003 Barcelona, Spain
- Center for Life Nano- & Neuro-Science, Istituto Italiano di Tecnologia, Viale Regina Elena 291, 00161 Rome, Italy; (J.F.); (E.M.); (G.G.)
- Department of Biology and Biotechnology Charles Darwin, Sapienza University of Rome, 00185 Rome, Italy
| |
Collapse
|
37
|
Kelly V, Al-Rawi A, Lewis D, Kustatscher G, Ly T. Low Cell Number Proteomic Analysis Using In-Cell Protease Digests Reveals a Robust Signature for Cell Cycle State Classification. Mol Cell Proteomics 2022; 21:100169. [PMID: 34742921 PMCID: PMC8760417 DOI: 10.1016/j.mcpro.2021.100169] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2021] [Revised: 09/02/2021] [Accepted: 10/25/2021] [Indexed: 12/04/2022] Open
Abstract
Comprehensive proteome analysis of rare cell phenotypes remains a significant challenge. We report a method for low cell number MS-based proteomics using protease digestion of mildly formaldehyde-fixed cells in cellulo, which we call the "in-cell digest." We combined this with averaged MS1 precursor library matching to quantitatively characterize proteomes from low cell numbers of human lymphoblasts. About 4500 proteins were detected from 2000 cells, and 2500 proteins were quantitated from 200 lymphoblasts. The ease of sample processing and high sensitivity makes this method exceptionally suited for the proteomic analysis of rare cell states, including immune cell subsets and cell cycle subphases. To demonstrate the method, we characterized the proteome changes across 16 cell cycle states (CCSs) isolated from an asynchronous TK6 cells, avoiding synchronization. States included late mitotic cells present at extremely low frequency. We identified 119 pseudoperiodic proteins that vary across the cell cycle. Clustering of the pseudoperiodic proteins showed abundance patterns consistent with "waves" of protein degradation in late S, at the G2&M border, midmitosis, and at mitotic exit. These clusters were distinguished by significant differences in predicted nuclear localization and interaction with the anaphase-promoting complex/cyclosome. The dataset also identifies putative anaphase-promoting complex/cyclosome substrates in mitosis and the temporal order in which they are targeted for degradation. We demonstrate that a protein signature made of these 119 high-confidence cell cycle-regulated proteins can be used to perform unbiased classification of proteomes into CCSs. We applied this signature to 296 proteomes that encompass a range of quantitation methods, cell types, and experimental conditions. The analysis confidently assigns a CCS for 49 proteomes, including correct classification for proteomes from synchronized cells. We anticipate that this robust cell cycle protein signature will be crucial for classifying cell states in single-cell proteomes.
Collapse
Affiliation(s)
- Van Kelly
- Wellcome Centre for Cell Biology, School of Biological Sciences, University of Edinburgh, Edinburgh, UK; Institute of Quantitative Biology, Biochemistry and Biotechnology, University of Edinburgh, Edinburgh, UK
| | - Aymen Al-Rawi
- Wellcome Centre for Cell Biology, School of Biological Sciences, University of Edinburgh, Edinburgh, UK
| | - David Lewis
- Wellcome Centre for Cell Biology, School of Biological Sciences, University of Edinburgh, Edinburgh, UK
| | - Georg Kustatscher
- Institute of Quantitative Biology, Biochemistry and Biotechnology, University of Edinburgh, Edinburgh, UK
| | - Tony Ly
- Wellcome Centre for Cell Biology, School of Biological Sciences, University of Edinburgh, Edinburgh, UK; Centre for Gene Regulation and Expression, School of Life Sciences, University of Dundee, Dundee, UK.
| |
Collapse
|
38
|
Servidei T, Lucchetti D, Navarra P, Sgambato A, Riccardi R, Ruggiero A. Cell-of-Origin and Genetic, Epigenetic, and Microenvironmental Factors Contribute to the Intra-Tumoral Heterogeneity of Pediatric Intracranial Ependymoma. Cancers (Basel) 2021; 13:6100. [PMID: 34885210 PMCID: PMC8657076 DOI: 10.3390/cancers13236100] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2021] [Revised: 11/24/2021] [Accepted: 12/01/2021] [Indexed: 02/07/2023] Open
Abstract
Intra-tumoral heterogeneity (ITH) is a complex multifaceted phenomenon that posits major challenges for the clinical management of cancer patients. Genetic, epigenetic, and microenvironmental factors are concurrent drivers of diversity among the distinct populations of cancer cells. ITH may also be installed by cancer stem cells (CSCs), that foster unidirectional hierarchy of cellular phenotypes or, alternatively, shift dynamically between distinct cellular states. Ependymoma (EPN), a molecularly heterogeneous group of tumors, shows a specific spatiotemporal distribution that suggests a link between ependymomagenesis and alterations of the biological processes involved in embryonic brain development. In children, EPN most often arises intra-cranially and is associated with an adverse outcome. Emerging evidence shows that EPN displays large intra-patient heterogeneity. In this review, after touching on EPN inter-tumoral heterogeneity, we focus on the sources of ITH in pediatric intra-cranial EPN in the framework of the CSC paradigm. We also examine how single-cell technology has shed new light on the complexity and developmental origins of EPN and the potential impact that this understanding may have on the therapeutic strategies against this deadly pediatric malignancy.
Collapse
Affiliation(s)
- Tiziana Servidei
- Pediatric Oncology Unit, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Catholic University of the Sacred Hearth, 00168 Rome, Italy; (R.R.); (A.R.)
| | - Donatella Lucchetti
- Dipartimento Universitario di Medicina e Chirurgia Traslazionale, Università Cattolica del Sacro Cuore, 00168 Rome, Italy; (D.L.); (A.S.)
| | - Pierluigi Navarra
- Department of Healthcare Surveillance and Bioethics, Section of Pharmacology, Università Cattolica del Sacro Cuore-Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Rome, Italy;
| | - Alessandro Sgambato
- Dipartimento Universitario di Medicina e Chirurgia Traslazionale, Università Cattolica del Sacro Cuore, 00168 Rome, Italy; (D.L.); (A.S.)
- Centro di Riferimento Oncologico della Basilicata (IRCCS-CROB), 85028 Rionero in Vulture, PZ, Italy
| | - Riccardo Riccardi
- Pediatric Oncology Unit, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Catholic University of the Sacred Hearth, 00168 Rome, Italy; (R.R.); (A.R.)
| | - Antonio Ruggiero
- Pediatric Oncology Unit, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Catholic University of the Sacred Hearth, 00168 Rome, Italy; (R.R.); (A.R.)
| |
Collapse
|
39
|
Auerbach BJ, Hu J, Reilly MP, Li M. Applications of single-cell genomics and computational strategies to study common disease and population-level variation. Genome Res 2021; 31:1728-1741. [PMID: 34599006 PMCID: PMC8494214 DOI: 10.1101/gr.275430.121] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The advent and rapid development of single-cell technologies have made it possible to study cellular heterogeneity at an unprecedented resolution and scale. Cellular heterogeneity underlies phenotypic differences among individuals, and studying cellular heterogeneity is an important step toward our understanding of the disease molecular mechanism. Single-cell technologies offer opportunities to characterize cellular heterogeneity from different angles, but how to link cellular heterogeneity with disease phenotypes requires careful computational analysis. In this article, we will review the current applications of single-cell methods in human disease studies and describe what we have learned so far from existing studies about human genetic variation. As single-cell technologies are becoming widely applicable in human disease studies, population-level studies have become a reality. We will describe how we should go about pursuing and designing these studies, particularly how to select study subjects, how to determine the number of cells to sequence per subject, and the needed sequencing depth per cell. We also discuss computational strategies for the analysis of single-cell data and describe how single-cell data can be integrated with bulk tissue data and data generated from genome-wide association studies. Finally, we point out open problems and future research directions.
Collapse
Affiliation(s)
- Benjamin J Auerbach
- Graduate Group in Genomics and Computational Biology, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania 19104, USA
| | - Jian Hu
- Department of Biostatistics, Epidemiology and Informatics, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania 19104, USA
| | - Muredach P Reilly
- Division of Cardiology, Department of Medicine, Columbia University Irving Medical Center, New York, New York 10032, USA
| | - Mingyao Li
- Department of Biostatistics, Epidemiology and Informatics, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania 19104, USA
| |
Collapse
|
40
|
Bonora G, Ramani V, Singh R, Fang H, Jackson DL, Srivatsan S, Qiu R, Lee C, Trapnell C, Shendure J, Duan Z, Deng X, Noble WS, Disteche CM. Single-cell landscape of nuclear configuration and gene expression during stem cell differentiation and X inactivation. Genome Biol 2021; 22:279. [PMID: 34579774 PMCID: PMC8474932 DOI: 10.1186/s13059-021-02432-w] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2020] [Accepted: 07/07/2021] [Indexed: 11/25/2022] Open
Abstract
BACKGROUND Mammalian development is associated with extensive changes in gene expression, chromatin accessibility, and nuclear structure. Here, we follow such changes associated with mouse embryonic stem cell differentiation and X inactivation by integrating, for the first time, allele-specific data from these three modalities obtained by high-throughput single-cell RNA-seq, ATAC-seq, and Hi-C. RESULTS Allele-specific contact decay profiles obtained by single-cell Hi-C clearly show that the inactive X chromosome has a unique profile in differentiated cells that have undergone X inactivation. Loss of this inactive X-specific structure at mitosis is followed by its reappearance during the cell cycle, suggesting a "bookmark" mechanism. Differentiation of embryonic stem cells to follow the onset of X inactivation is associated with changes in contact decay profiles that occur in parallel on both the X chromosomes and autosomes. Single-cell RNA-seq and ATAC-seq show evidence of a delay in female versus male cells, due to the presence of two active X chromosomes at early stages of differentiation. The onset of the inactive X-specific structure in single cells occurs later than gene silencing, consistent with the idea that chromatin compaction is a late event of X inactivation. Single-cell Hi-C highlights evidence of discrete changes in nuclear structure characterized by the acquisition of very long-range contacts throughout the nucleus. Novel computational approaches allow for the effective alignment of single-cell gene expression, chromatin accessibility, and 3D chromosome structure. CONCLUSIONS Based on trajectory analyses, three distinct nuclear structure states are detected reflecting discrete and profound simultaneous changes not only to the structure of the X chromosomes, but also to that of autosomes during differentiation. Our study reveals that long-range structural changes to chromosomes appear as discrete events, unlike progressive changes in gene expression and chromatin accessibility.
Collapse
Affiliation(s)
- Giancarlo Bonora
- Department of Genome Sciences, University of Washington, Seattle, WA, USA
| | - Vijay Ramani
- Department of Biochemistry & Biophysics, University of California San Francisco, San Francisco, CA, USA
| | - Ritambhara Singh
- Department of Computer Science, Brown University, Providence, RI, USA
- Center for Computational Molecular Biology, Brown University, Providence, RI, USA
| | - He Fang
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA, USA
| | - Dana L Jackson
- Department of Genome Sciences, University of Washington, Seattle, WA, USA
| | - Sanjay Srivatsan
- Department of Genome Sciences, University of Washington, Seattle, WA, USA
| | - Ruolan Qiu
- Department of Genome Sciences, University of Washington, Seattle, WA, USA
| | - Choli Lee
- Department of Genome Sciences, University of Washington, Seattle, WA, USA
| | - Cole Trapnell
- Department of Genome Sciences, University of Washington, Seattle, WA, USA
- Brotman Baty Institute for Precision Medicine, Seattle, WA, USA
- Allen Discovery Center for Cell Lineage Tracing, Seattle, WA, USA
| | - Jay Shendure
- Department of Genome Sciences, University of Washington, Seattle, WA, USA
- Brotman Baty Institute for Precision Medicine, Seattle, WA, USA
- Allen Discovery Center for Cell Lineage Tracing, Seattle, WA, USA
- Howard Hughes Medical Institute, Seattle, WA, USA
| | - Zhijun Duan
- Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, USA
- Division of Hematology, Department of Medicine, University of Washington, Seattle, USA
| | - Xinxian Deng
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA, USA.
| | - William S Noble
- Department of Genome Sciences, University of Washington, Seattle, WA, USA.
- Paul G. Allen School of Computer Science and Engineering, University of Washington, Seattle, WA, USA.
| | - Christine M Disteche
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA, USA.
- Brotman Baty Institute for Precision Medicine, Seattle, WA, USA.
- Department of Medicine, University of Washington, Seattle, WA, USA.
| |
Collapse
|
41
|
Briggs EM, Warren FSL, Matthews KR, McCulloch R, Otto TD. Application of single-cell transcriptomics to kinetoplastid research. Parasitology 2021; 148:1223-1236. [PMID: 33678213 PMCID: PMC8311972 DOI: 10.1017/s003118202100041x] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2020] [Revised: 02/22/2021] [Accepted: 02/24/2021] [Indexed: 12/13/2022]
Abstract
Kinetoplastid parasites are responsible for both human and animal diseases across the globe where they have a great impact on health and economic well-being. Many species and life cycle stages are difficult to study due to limitations in isolation and culture, as well as to their existence as heterogeneous populations in hosts and vectors. Single-cell transcriptomics (scRNA-seq) has the capacity to overcome many of these difficulties, and can be leveraged to disentangle heterogeneous populations, highlight genes crucial for propagation through the life cycle, and enable detailed analysis of host–parasite interactions. Here, we provide a review of studies that have applied scRNA-seq to protozoan parasites so far. In addition, we provide an overview of sample preparation and technology choice considerations when planning scRNA-seq experiments, as well as challenges faced when analysing the large amounts of data generated. Finally, we highlight areas of kinetoplastid research that could benefit from scRNA-seq technologies.
Collapse
Affiliation(s)
- Emma M. Briggs
- Institute for Immunology and Infection Research, School of Biological Sciences, University of Edinburgh, Edinburgh, UK
- Wellcome Centre for Integrative Parasitology, Institute of Infection, Immunity and Inflammation, University of Glasgow, Glasgow, UK
| | - Felix S. L. Warren
- Wellcome Centre for Integrative Parasitology, Institute of Infection, Immunity and Inflammation, University of Glasgow, Glasgow, UK
| | - Keith R. Matthews
- Institute for Immunology and Infection Research, School of Biological Sciences, University of Edinburgh, Edinburgh, UK
| | - Richard McCulloch
- Wellcome Centre for Integrative Parasitology, Institute of Infection, Immunity and Inflammation, University of Glasgow, Glasgow, UK
| | - Thomas D. Otto
- Wellcome Centre for Integrative Parasitology, Institute of Infection, Immunity and Inflammation, University of Glasgow, Glasgow, UK
| |
Collapse
|
42
|
Chen Y, Zhang Y, Li JYH, Ouyang Z. LISA2: Learning Complex Single-Cell Trajectory and Expression Trends. Front Genet 2021; 12:681206. [PMID: 34512717 PMCID: PMC8428276 DOI: 10.3389/fgene.2021.681206] [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: 03/16/2021] [Accepted: 06/01/2021] [Indexed: 12/20/2022] Open
Abstract
Single-cell transcriptional and epigenomics profiles have been applied in a variety of tissues and diseases for discovering new cell types, differentiation trajectories, and gene regulatory networks. Many methods such as Monocle 2/3, URD, and STREAM have been developed for tree-based trajectory building. Here, we propose a fast and flexible trajectory learning method, LISA2, for single-cell data analysis. This new method has two distinctive features: (1) LISA2 utilizes specified leaves and root to reduce the complexity for building the developmental trajectory, especially for some special cases such as rare cell populations and adjacent terminal cell states; and (2) LISA2 is applicable for both transcriptomics and epigenomics data. LISA2 visualizes complex trajectories using 3D Landmark ISOmetric feature MAPping (L-ISOMAP). We apply LISA2 to simulation and real datasets in cerebellum, diencephalon, and hematopoietic stem cells including both single-cell transcriptomics data and single-cell assay for transposase-accessible chromatin data. LISA2 is efficient in estimating single-cell trajectory and expression trends for different kinds of molecular state of cells.
Collapse
Affiliation(s)
- Yang Chen
- Department of Biostatistics and Epidemiology, School of Public Health and Health Sciences, University of Massachusetts, Amherst, MA, United States
| | - Yuping Zhang
- Department of Statistics, University of Connecticut, Storrs, CT, United States
- Institute for Systems Genomics, University of Connecticut, Storrs, CT, United States
| | - James Y. H. Li
- Institute for Systems Genomics, University of Connecticut, Storrs, CT, United States
- Department of Genetics and Genome Sciences, School of Medicine, University of Connecticut, Farmington, CT, United States
| | - Zhengqing Ouyang
- Department of Biostatistics and Epidemiology, School of Public Health and Health Sciences, University of Massachusetts, Amherst, MA, United States
| |
Collapse
|
43
|
Griffiths JI, Chen J, Cosgrove PA, O’Dea A, Sharma P, Ma C, Trivedi M, Kalinsky K, Wisinski KB, O’Regan R, Makhoul I, Spring LM, Bardia A, Adler FR, Cohen AL, Chang JT, Khan QJ, Bild AH. Serial single-cell genomics reveals convergent subclonal evolution of resistance as early-stage breast cancer patients progress on endocrine plus CDK4/6 therapy. NATURE CANCER 2021; 2:658-671. [PMID: 34712959 PMCID: PMC8547038 DOI: 10.1038/s43018-021-00215-7] [Citation(s) in RCA: 44] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Combining cyclin-dependent kinase (CDK) inhibitors with endocrine therapy improves outcomes for metastatic estrogen receptor positive (ER+) breast cancer patients but its value in earlier stage patients is unclear. We examined evolutionary trajectories of early-stage breast cancer tumors, using single cell RNA sequencing (scRNAseq) of serial biopsies from the FELINE clinical trial (#NCT02712723) of endocrine therapy (letrozole) alone or combined with the CDK inhibitor ribociclib. Despite differences in subclonal diversity evolution across patients and treatments, common resistance phenotypes emerged. Resistant tumors treated with combination therapy showed accelerated loss of estrogen signaling with convergent up-regulation of JNK signaling through growth factor receptors. In contrast, cancer cells maintaining estrogen signaling during mono- or combination therapy showed potentiation of CDK4/6 activation and ERK upregulation through ERBB4 signaling. These results indicate that combination therapy in early-stage ER+ breast cancer leads to emergence of resistance through a shift from estrogen to alternative growth signal-mediated proliferation.
Collapse
Affiliation(s)
- Jason I. Griffiths
- Department of Medical Oncology & Therapeutics Research, City of Hope National Medical Center, 1500 East Duarte Road, Duarte, CA, 91010, USA.,Department of Mathematics, University of Utah 155 South 1400 East, Salt Lake City, UT, 84112, USA
| | - Jinfeng Chen
- Department of Medical Oncology & Therapeutics Research, City of Hope National Medical Center, 1500 East Duarte Road, Duarte, CA, 91010, USA
| | - Patrick A. Cosgrove
- Department of Medical Oncology & Therapeutics Research, City of Hope National Medical Center, 1500 East Duarte Road, Duarte, CA, 91010, USA
| | - Anne O’Dea
- Division of Medical Oncology, University of Kansas Medical Center, Westwood, KS, 66160, USA
| | - Priyanka Sharma
- Division of Medical Oncology, University of Kansas Medical Center, Westwood, KS, 66160, USA
| | - Cynthia Ma
- Division of Oncology, Washington University School of Medicine, St. Louis, MO, 63130, USA
| | - Meghna Trivedi
- Department of Medicine, Columbia University Irving Medical Center, NY, 10032, USA
| | - Kevin Kalinsky
- Department of Medicine, Columbia University Irving Medical Center, NY, 10032, USA
| | - Kari B. Wisinski
- Department of Medicine, University of Wisconsin School of Medicine and Public Health, Carbone Cancer Center, WI, 53726, USA
| | - Ruth O’Regan
- Department of Medicine, University of Wisconsin School of Medicine and Public Health, Carbone Cancer Center, WI, 53726, USA
| | - Issam Makhoul
- Division of Internal Medical Oncology, University of Arkansas for Medical Sciences, AR, 72205, USA
| | - Laura M. Spring
- Department of Medicine, Massachusetts General Hospital Cancer Center and Harvard Medical School, MA, 02114, USA
| | - Aditya Bardia
- Department of Medicine, Massachusetts General Hospital Cancer Center and Harvard Medical School, MA, 02114, USA
| | - Frederick R. Adler
- Department of Mathematics, University of Utah 155 South 1400 East, Salt Lake City, UT, 84112, USA.,School of Biological Sciences, University of Utah 257 South 1400 East, Salt Lake City, UT, 84112, USA
| | - Adam L. Cohen
- Department of Internal Medicine, Huntsman Cancer Institute, University of Utah, Salt Lake City, UT 84112, USA
| | - Jeffrey T. Chang
- Department of Integrative Biology and Pharmacology, School of Medicine, School of Biomedical Informatics, UT Health Sciences Center at Houston, Houston, TX, 77030, USA
| | - Qamar J. Khan
- Division of Medical Oncology, University of Kansas Medical Center, Westwood, KS, 66160, USA.,To whom correspondence should be addressed: Andrea Bild () and Qamar Khan ()
| | - Andrea H. Bild
- Department of Medical Oncology & Therapeutics Research, City of Hope National Medical Center, 1500 East Duarte Road, Duarte, CA, 91010, USA.,To whom correspondence should be addressed: Andrea Bild () and Qamar Khan ()
| |
Collapse
|
44
|
Liu J, Fan Z, Zhao W, Zhou X. Machine Intelligence in Single-Cell Data Analysis: Advances and New Challenges. Front Genet 2021; 12:655536. [PMID: 34135939 PMCID: PMC8203333 DOI: 10.3389/fgene.2021.655536] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2021] [Accepted: 04/26/2021] [Indexed: 12/18/2022] Open
Abstract
The rapid development of single-cell technologies allows for dissecting cellular heterogeneity at different omics layers with an unprecedented resolution. In-dep analysis of cellular heterogeneity will boost our understanding of complex biological systems or processes, including cancer, immune system and chronic diseases, thereby providing valuable insights for clinical and translational research. In this review, we will focus on the application of machine learning methods in single-cell multi-omics data analysis. We will start with the pre-processing of single-cell RNA sequencing (scRNA-seq) data, including data imputation, cross-platform batch effect removal, and cell cycle and cell-type identification. Next, we will introduce advanced data analysis tools and methods used for copy number variance estimate, single-cell pseudo-time trajectory analysis, phylogenetic tree inference, cell-cell interaction, regulatory network inference, and integrated analysis of scRNA-seq and spatial transcriptome data. Finally, we will present the latest analyzing challenges, such as multi-omics integration and integrated analysis of scRNA-seq data.
Collapse
Affiliation(s)
- Jiajia Liu
- College of Electronic and Information Engineering, Tongji University, Shanghai, China
- School of Biomedical Informatics, The University of Texas Health Science Centre at Houston, Houston, TX, United States
| | - Zhiwei Fan
- School of Biomedical Informatics, The University of Texas Health Science Centre at Houston, Houston, TX, United States
- West China School of Public Health, West China Fourth Hospital, Sichuan University, Chengdu, China
| | - Weiling Zhao
- School of Biomedical Informatics, The University of Texas Health Science Centre at Houston, Houston, TX, United States
| | - Xiaobo Zhou
- School of Biomedical Informatics, The University of Texas Health Science Centre at Houston, Houston, TX, United States
| |
Collapse
|
45
|
Cheng X, Lai H, Luo W, Zhang M, Miao J, Song W, Xing S, Wang J, Gao WQ. Single-cell analysis reveals urothelial cell heterogeneity and regenerative cues following cyclophosphamide-induced bladder injury. Cell Death Dis 2021; 12:446. [PMID: 33953164 PMCID: PMC8099875 DOI: 10.1038/s41419-021-03740-6] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2021] [Revised: 04/16/2021] [Accepted: 04/16/2021] [Indexed: 02/06/2023]
Abstract
Cyclophosphamide is a commonly used chemotherapeutic drug to treat cancer with side effects that trigger bladder injury and hemorrhagic cystitis. Although previous studies have demonstrated that certain cell subsets and communications are activated to drive the repair and regeneration of bladder, it is not well understood how distinct bladder cell subsets function synergistically in this process. Here, we used droplet-based single-cell RNA sequencing (scRNA-seq) to profile the cell types within the murine bladder mucous layer under normal and injured conditions. Our analysis showed that superficial cells are directly repaired by cycling intermediate cells. We further identified two resident mesenchymal lineages (Acta2+ myofibroblasts and Cd34+ fibroblasts). The delineation of cell-cell communications revealed that Acta2+ myofibroblasts upregulated Fgf7 expression during acute injury, which activated Fgfr signaling in progenitor cells within the basal/intermediate layers to promote urothelial cell growth and repair. Overall, our study contributes to a more comprehensive understanding of the cellular dynamics during cyclophosphamide-induced bladder injury and may help identify important niche factors contributing to the regeneration of injured bladders.
Collapse
Affiliation(s)
- Xiaomu Cheng
- State Key Laboratory of Oncogenes and Related Genes, Renji-Med X Clinical Stem Cell Research Center, Ren Ji Hospital, School of Medicine and School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, China.,Med-X Research Institute, Shanghai Jiao Tong University, Shanghai, China
| | - Huadong Lai
- Med-X Research Institute, Shanghai Jiao Tong University, Shanghai, China
| | - Wenqin Luo
- State Key Laboratory of Oncogenes and Related Genes, Renji-Med X Clinical Stem Cell Research Center, Ren Ji Hospital, School of Medicine and School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, China
| | - Man Zhang
- Med-X Research Institute, Shanghai Jiao Tong University, Shanghai, China
| | - Juju Miao
- Med-X Research Institute, Shanghai Jiao Tong University, Shanghai, China
| | - Weichen Song
- Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Shunpeng Xing
- Department of Critical Care Medicine, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China.
| | - Jia Wang
- State Key Laboratory of Oncogenes and Related Genes, Renji-Med X Clinical Stem Cell Research Center, Ren Ji Hospital, School of Medicine and School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, China.
| | - Wei-Qiang Gao
- State Key Laboratory of Oncogenes and Related Genes, Renji-Med X Clinical Stem Cell Research Center, Ren Ji Hospital, School of Medicine and School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, China. .,Med-X Research Institute, Shanghai Jiao Tong University, Shanghai, China.
| |
Collapse
|
46
|
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: 1.0] [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
|
47
|
Phillips NE, Hugues A, Yeung J, Durandau E, Nicolas D, Naef F. The circadian oscillator analysed at the single-transcript level. Mol Syst Biol 2021; 17:e10135. [PMID: 33719202 PMCID: PMC7957410 DOI: 10.15252/msb.202010135] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2020] [Revised: 01/05/2021] [Accepted: 01/19/2021] [Indexed: 12/31/2022] Open
Abstract
The circadian clock is an endogenous and self-sustained oscillator that anticipates daily environmental cycles. While rhythmic gene expression of circadian genes is well-described in populations of cells, the single-cell mRNA dynamics of multiple core clock genes remain largely unknown. Here we use single-molecule fluorescence in situ hybridisation (smFISH) at multiple time points to measure pairs of core clock transcripts, Rev-erbα (Nr1d1), Cry1 and Bmal1, in mouse fibroblasts. The mean mRNA level oscillates over 24 h for all three genes, but mRNA numbers show considerable spread between cells. We develop a probabilistic model for multivariate mRNA counts using mixtures of negative binomials, which accounts for transcriptional bursting, circadian time and cell-to-cell heterogeneity, notably in cell size. Decomposing the mRNA variability into distinct noise sources shows that clock time contributes a small fraction of the total variability in mRNA number between cells. Thus, our results highlight the intrinsic biological challenges in estimating circadian phase from single-cell mRNA counts and suggest that circadian phase in single cells is encoded post-transcriptionally.
Collapse
Affiliation(s)
- Nicholas E Phillips
- Institute of BioengineeringSchool of Life SciencesEcole Polytechnique Fédérale de LausanneLausanneSwitzerland
| | - Alice Hugues
- Institute of BioengineeringSchool of Life SciencesEcole Polytechnique Fédérale de LausanneLausanneSwitzerland
- Master de BiologieÉcole Normale Supérieure de LyonUniversité Claude Bernard Lyon IUniversité de LyonLyonFrance
| | - Jake Yeung
- Institute of BioengineeringSchool of Life SciencesEcole Polytechnique Fédérale de LausanneLausanneSwitzerland
| | - Eric Durandau
- Institute of BioengineeringSchool of Life SciencesEcole Polytechnique Fédérale de LausanneLausanneSwitzerland
| | - Damien Nicolas
- Institute of BioengineeringSchool of Life SciencesEcole Polytechnique Fédérale de LausanneLausanneSwitzerland
| | - Felix Naef
- Institute of BioengineeringSchool of Life SciencesEcole Polytechnique Fédérale de LausanneLausanneSwitzerland
| |
Collapse
|
48
|
Xie K, Liu Z, Chen N, Chen T. redPATH: Reconstructing the Pseudo Development Time of Cell Lineages in Single-cell RNA-seq Data and Applications in Cancer. GENOMICS PROTEOMICS & BIOINFORMATICS 2021; 19:292-305. [PMID: 33607293 PMCID: PMC8602773 DOI: 10.1016/j.gpb.2020.06.014] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/30/2019] [Revised: 03/27/2020] [Accepted: 09/08/2020] [Indexed: 11/28/2022]
Abstract
The recent advancement of single-cell RNA sequencing (scRNA-seq) technologies facilitates the study of cell lineages in developmental processes and cancer. In this study, we developed a computational method, called redPATH, to reconstruct the pseudo developmental time of cell lineages using a consensus asymmetric Hamiltonian path algorithm. Besides, we developed a novel approach to visualize the trajectory development and implemented visualization methods to provide biological insights. We validated the performance of redPATH by segmenting different stages of cell development on multiple neural stem cell and cancer datasets, as well as other single-cell transcriptome data. In particular, we identified a stem cell-like subpopulation in malignant glioma cells. These cells express known proliferative markers, such as GFAP, ATP1A2, IGFBPL1, and ALDOC, and remain silenced for quiescent markers such as ID3. Furthermore, we identified MCL1 as a significant gene that regulates cell apoptosis and CSF1R for reprogramming macrophages to control tumor growth. In conclusion, redPATH is a comprehensive tool for analyzing scRNA-seq datasets along the pseudo developmental time. redPATH is available at https://github.com/tinglabs/redPATH.
Collapse
Affiliation(s)
- Kaikun Xie
- Institute for Artificial Intelligence, State Key Lab of Intelligent Technology and Systems, Department of Computer Science and Technology, Tsinghua University, Beijing 100084, China; Tsinghua-Fuzhou Institute of Digital Technology, Beijing National Research Center for Information Science and Technology, Tsinghua University, Beijing 100084, China
| | - Zehua Liu
- Institute for Artificial Intelligence, State Key Lab of Intelligent Technology and Systems, Department of Computer Science and Technology, Tsinghua University, Beijing 100084, China; Tsinghua-Fuzhou Institute of Digital Technology, Beijing National Research Center for Information Science and Technology, Tsinghua University, Beijing 100084, China; Center for Computational and Integrative Biology, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA; Department of Molecular Biology, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA; Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, MA 02142, USA
| | - Ning Chen
- Institute for Artificial Intelligence, State Key Lab of Intelligent Technology and Systems, Department of Computer Science and Technology, Tsinghua University, Beijing 100084, China; Tsinghua-Fuzhou Institute of Digital Technology, Beijing National Research Center for Information Science and Technology, Tsinghua University, Beijing 100084, China.
| | - Ting Chen
- Institute for Artificial Intelligence, State Key Lab of Intelligent Technology and Systems, Department of Computer Science and Technology, Tsinghua University, Beijing 100084, China; Tsinghua-Fuzhou Institute of Digital Technology, Beijing National Research Center for Information Science and Technology, Tsinghua University, Beijing 100084, China.
| |
Collapse
|
49
|
Afeworki Y, Wollenzien H, Kareta MS. Transcriptional Profiling During Neural Conversion. Methods Mol Biol 2021; 2352:171-181. [PMID: 34324187 PMCID: PMC9131516 DOI: 10.1007/978-1-0716-1601-7_12] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
The processes that underlie neuronal conversion ultimately involve a reorganization of transcriptional networks to establish a neuronal cell fate. As such, transcriptional profiling is a key component toward understanding this process. In this chapter, we will discuss methods of elucidating transcriptional networks during neuronal reprogramming and considerations that should be incorporated in experimental design.
Collapse
Affiliation(s)
- Yohannes Afeworki
- Functional Genomics and Bioinformatics Core, Sanford Research, Sioux Falls, SD, USA
| | - Hannah Wollenzien
- Division of Basic Biomedical Sciences, University of South Dakota, Vermillion, SD, USA
- Genetics and Genomics Group, Sanford Research, Sioux Falls, SD, USA
| | - Michael S Kareta
- Functional Genomics and Bioinformatics Core, Sanford Research, Sioux Falls, SD, USA.
- Division of Basic Biomedical Sciences, University of South Dakota, Vermillion, SD, USA.
- Genetics and Genomics Group, Sanford Research, Sioux Falls, SD, USA.
- Cellular Therapies and Stem Cell Biology Group, Sanford Research, Sioux Falls, SD, USA.
- Department of Pediatrics, Sanford School of Medicine, Sioux Falls, SD, USA.
- Department of Chemistry and Biochemistry, South Dakota State University, Brookings, SD, USA.
| |
Collapse
|
50
|
Yu X, Abbas-Aghababazadeh F, Chen YA, Fridley BL. Statistical and Bioinformatics Analysis of Data from Bulk and Single-Cell RNA Sequencing Experiments. Methods Mol Biol 2021; 2194:143-175. [PMID: 32926366 PMCID: PMC7771369 DOI: 10.1007/978-1-0716-0849-4_9] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
High-throughput sequencing (HTS) has revolutionized researchers' ability to study the human transcriptome, particularly as it relates to cancer. Recently, HTS technology has advanced to the point where now one is able to sequence individual cells (i.e., "single-cell sequencing"). Prior to single-cell sequencing technology, HTS would be completed on RNA extracted from a tissue sample consisting of multiple cell types (i.e., "bulk sequencing"). In this chapter, we review the various bioinformatics and statistical methods used in the processing, quality control, and analysis of bulk and single-cell RNA sequencing methods. Additionally, we discuss how these methods are also being used to study tumor heterogeneity.
Collapse
Affiliation(s)
- Xiaoqing Yu
- Department of Biostatistics and Bioinformatics, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL, USA
| | - Farnoosh Abbas-Aghababazadeh
- Department of Biostatistics and Bioinformatics, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL, USA
| | - Y Ann Chen
- Department of Biostatistics and Bioinformatics, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL, USA
| | - Brooke L Fridley
- Department of Biostatistics and Bioinformatics, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL, USA.
| |
Collapse
|