1
|
Jin S, Plikus MV, Nie Q. CellChat for systematic analysis of cell-cell communication from single-cell transcriptomics. Nat Protoc 2025; 20:180-219. [PMID: 39289562 DOI: 10.1038/s41596-024-01045-4] [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: 07/31/2023] [Accepted: 06/27/2024] [Indexed: 09/19/2024]
Abstract
Recent advances in single-cell sequencing technologies offer an opportunity to explore cell-cell communication in tissues systematically and with reduced bias. A key challenge is integrating known molecular interactions and measurements into a framework to identify and analyze complex cell-cell communication networks. Previously, we developed a computational tool, named CellChat, that infers and analyzes cell-cell communication networks from single-cell transcriptomic data within an easily interpretable framework. CellChat quantifies the signaling communication probability between two cell groups using a simplified mass-action-based model, which incorporates the core interaction between ligands and receptors with multisubunit structure along with modulation by cofactors. Importantly, CellChat performs a systematic and comparative analysis of cell-cell communication using a variety of quantitative metrics and machine-learning approaches. CellChat v2 is an updated version that includes additional comparison functionalities, an expanded database of ligand-receptor pairs along with rich functional annotations, and an Interactive CellChat Explorer. Here we provide a step-by-step protocol for using CellChat v2 on single-cell transcriptomic data, including inference and analysis of cell-cell communication from one dataset and identification of altered intercellular communication, signals and cell populations from different datasets across biological conditions. The R implementation of CellChat v2 toolkit and its tutorials together with the graphic outputs are available at https://github.com/jinworks/CellChat . This protocol typically takes ~5 min depending on dataset size and requires a basic understanding of R and single-cell data analysis but no specialized bioinformatics training for its implementation.
Collapse
Affiliation(s)
- Suoqin Jin
- School of Mathematics and Statistics, Wuhan University, Wuhan, China.
- Hubei Key Laboratory of Computational Science, Wuhan University, Wuhan, China.
| | - Maksim V Plikus
- NSF-Simons Center for Multiscale Cell Fate Research, University of California, Irvine, Irvine, CA, USA
- Department of Developmental and Cell Biology, University of California, Irvine, Irvine, CA, USA
| | - Qing Nie
- NSF-Simons Center for Multiscale Cell Fate Research, University of California, Irvine, Irvine, CA, USA.
- Department of Developmental and Cell Biology, University of California, Irvine, Irvine, CA, USA.
- Department of Mathematics, University of California, Irvine, Irvine, CA, USA.
| |
Collapse
|
2
|
Sun F, Li H, Sun D, Fu S, Gu L, Shao X, Wang Q, Dong X, Duan B, Xing F, Wu J, Xiao M, Zhao F, Han JDJ, Liu Q, Fan X, Li C, Wang C, Shi T. Single-cell omics: experimental workflow, data analyses and applications. SCIENCE CHINA. LIFE SCIENCES 2025; 68:5-102. [PMID: 39060615 DOI: 10.1007/s11427-023-2561-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/07/2023] [Accepted: 04/18/2024] [Indexed: 07/28/2024]
Abstract
Cells are the fundamental units of biological systems and exhibit unique development trajectories and molecular features. Our exploration of how the genomes orchestrate the formation and maintenance of each cell, and control the cellular phenotypes of various organismsis, is both captivating and intricate. Since the inception of the first single-cell RNA technology, technologies related to single-cell sequencing have experienced rapid advancements in recent years. These technologies have expanded horizontally to include single-cell genome, epigenome, proteome, and metabolome, while vertically, they have progressed to integrate multiple omics data and incorporate additional information such as spatial scRNA-seq and CRISPR screening. Single-cell omics represent a groundbreaking advancement in the biomedical field, offering profound insights into the understanding of complex diseases, including cancers. Here, we comprehensively summarize recent advances in single-cell omics technologies, with a specific focus on the methodology section. This overview aims to guide researchers in selecting appropriate methods for single-cell sequencing and related data analysis.
Collapse
Affiliation(s)
- Fengying Sun
- Department of Clinical Laboratory, the Affiliated Wuhu Hospital of East China Normal University (The Second People's Hospital of Wuhu City), Wuhu, 241000, China
| | - Haoyan Li
- Pharmaceutical Informatics Institute, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China
| | - Dongqing Sun
- Key Laboratory of Spine and Spinal Cord Injury Repair and Regeneration (Tongji University), Ministry of Education, Orthopaedic Department, Tongji Hospital, Bioinformatics Department, School of Life Sciences and Technology, Tongji University, Shanghai, 200082, China
- Frontier Science Center for Stem Cells, School of Life Sciences and Technology, Tongji University, Shanghai, 200092, China
| | - Shaliu Fu
- Key Laboratory of Spine and Spinal Cord Injury Repair and Regeneration (Tongji University), Ministry of Education, Orthopaedic Department, Tongji Hospital, Bioinformatics Department, School of Life Sciences and Technology, Tongji University, Shanghai, 200082, China
- Translational Medical Center for Stem Cell Therapy and Institute for Regenerative Medicine, Shanghai East Hospital, Bioinformatics Department, School of Life Sciences and Technology, Tongji University, Shanghai, 200082, China
- Research Institute of Intelligent Computing, Zhejiang Lab, Hangzhou, 311121, China
- Shanghai Research Institute for Intelligent Autonomous Systems, Shanghai, 201210, China
| | - Lei Gu
- Center for Single-cell Omics, School of Public Health, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Xin Shao
- Pharmaceutical Informatics Institute, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China
- National Key Laboratory of Chinese Medicine Modernization, Innovation Center of Yangtze River Delta, Zhejiang University, Jiaxing, 314103, China
| | - Qinqin Wang
- Center for Single-cell Omics, School of Public Health, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Xin Dong
- Key Laboratory of Spine and Spinal Cord Injury Repair and Regeneration (Tongji University), Ministry of Education, Orthopaedic Department, Tongji Hospital, Bioinformatics Department, School of Life Sciences and Technology, Tongji University, Shanghai, 200082, China
- Frontier Science Center for Stem Cells, School of Life Sciences and Technology, Tongji University, Shanghai, 200092, China
| | - Bin Duan
- Key Laboratory of Spine and Spinal Cord Injury Repair and Regeneration (Tongji University), Ministry of Education, Orthopaedic Department, Tongji Hospital, Bioinformatics Department, School of Life Sciences and Technology, Tongji University, Shanghai, 200082, China
- Translational Medical Center for Stem Cell Therapy and Institute for Regenerative Medicine, Shanghai East Hospital, Bioinformatics Department, School of Life Sciences and Technology, Tongji University, Shanghai, 200082, China
- Research Institute of Intelligent Computing, Zhejiang Lab, Hangzhou, 311121, China
- Shanghai Research Institute for Intelligent Autonomous Systems, Shanghai, 201210, China
| | - Feiyang Xing
- Key Laboratory of Spine and Spinal Cord Injury Repair and Regeneration (Tongji University), Ministry of Education, Orthopaedic Department, Tongji Hospital, Bioinformatics Department, School of Life Sciences and Technology, Tongji University, Shanghai, 200082, China
- Frontier Science Center for Stem Cells, School of Life Sciences and Technology, Tongji University, Shanghai, 200092, China
| | - Jun Wu
- Center for Bioinformatics and Computational Biology, Shanghai Key Laboratory of Regulatory Biology, the Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai, 200241, China
| | - Minmin Xiao
- Department of Clinical Laboratory, the Affiliated Wuhu Hospital of East China Normal University (The Second People's Hospital of Wuhu City), Wuhu, 241000, China.
| | - Fangqing Zhao
- Beijing Institutes of Life Science, Chinese Academy of Sciences, Beijing, 100101, China.
| | - Jing-Dong J Han
- Peking-Tsinghua Center for Life Sciences, Academy for Advanced Interdisciplinary Studies, Center for Quantitative Biology (CQB), Peking University, Beijing, 100871, China.
| | - Qi Liu
- Key Laboratory of Spine and Spinal Cord Injury Repair and Regeneration (Tongji University), Ministry of Education, Orthopaedic Department, Tongji Hospital, Bioinformatics Department, School of Life Sciences and Technology, Tongji University, Shanghai, 200082, China.
- Translational Medical Center for Stem Cell Therapy and Institute for Regenerative Medicine, Shanghai East Hospital, Bioinformatics Department, School of Life Sciences and Technology, Tongji University, Shanghai, 200082, China.
- Research Institute of Intelligent Computing, Zhejiang Lab, Hangzhou, 311121, China.
- Shanghai Research Institute for Intelligent Autonomous Systems, Shanghai, 201210, China.
| | - Xiaohui Fan
- Pharmaceutical Informatics Institute, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China.
- National Key Laboratory of Chinese Medicine Modernization, Innovation Center of Yangtze River Delta, Zhejiang University, Jiaxing, 314103, China.
- Zhejiang Key Laboratory of Precision Diagnosis and Therapy for Major Gynecological Diseases, Women's Hospital, Zhejiang University School of Medicine, Hangzhou, 310006, China.
| | - Chen Li
- Center for Single-cell Omics, School of Public Health, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China.
| | - Chenfei Wang
- Key Laboratory of Spine and Spinal Cord Injury Repair and Regeneration (Tongji University), Ministry of Education, Orthopaedic Department, Tongji Hospital, Bioinformatics Department, School of Life Sciences and Technology, Tongji University, Shanghai, 200082, China.
- Frontier Science Center for Stem Cells, School of Life Sciences and Technology, Tongji University, Shanghai, 200092, China.
| | - Tieliu Shi
- Department of Clinical Laboratory, the Affiliated Wuhu Hospital of East China Normal University (The Second People's Hospital of Wuhu City), Wuhu, 241000, China.
- Center for Bioinformatics and Computational Biology, Shanghai Key Laboratory of Regulatory Biology, the Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai, 200241, China.
- Key Laboratory of Advanced Theory and Application in Statistics and Data Science-MOE, School of Statistics, East China Normal University, Shanghai, 200062, China.
| |
Collapse
|
3
|
Wang X, Almet AA, Nie Q. Detecting global and local hierarchical structures in cell-cell communication using CrossChat. Nat Commun 2024; 15:10542. [PMID: 39627184 PMCID: PMC11615294 DOI: 10.1038/s41467-024-54821-x] [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: 05/01/2024] [Accepted: 11/20/2024] [Indexed: 12/06/2024] Open
Abstract
Cell-cell communication (CCC) occurs across different biological scales, ranging from interactions between large groups of cells to interactions between individual cells, forming a hierarchical structure. Globally, CCC may exist between clusters or only subgroups of a cluster with varying size, while locally, a group of cells as sender or receiver may exhibit distinct signaling properties. Current existing methods infer CCC from single-cell RNA-seq or Spatial Transcriptomics only between predefined cell groups, neglecting the existing hierarchical structure within CCC that are determined by signaling molecules, in particular, ligands and receptors. Here, we develop CrossChat, a novel computational framework designed to infer and analyze the hierarchical cell-cell communication structures using two complementary approaches: a global hierarchical structure using a multi-resolution clustering method, and multiple local hierarchical structures using a tree detection method. This framework provides a comprehensive approach to understand the hierarchical relationships within CCC that govern complex tissue functions. By applying our method to two nonspatial scRNA-seq datasets sampled from COVID-19 patients and mouse embryonic skin, and two spatial transcriptomics datasets generated from Stereo-seq of mouse embryo and 10x Visium of mouse wounded skin, we showcase CrossChat's functionalities for analyzing both global and local hierarchical structures within cell-cell communication.
Collapse
Affiliation(s)
- Xinyi Wang
- Department of Mathematics, University of California, Irvine, CA, USA
| | - Axel A Almet
- Department of Mathematics, University of California, Irvine, CA, USA.
- The NSF-Simons Center for Multiscale Cell Fate Research, University of California, Irvine, CA, USA.
| | - Qing Nie
- Department of Mathematics, University of California, Irvine, CA, USA.
- The NSF-Simons Center for Multiscale Cell Fate Research, University of California, Irvine, CA, USA.
- Department of Developmental and Cell Biology, University of California, Irvine, CA, USA.
| |
Collapse
|
4
|
Chap BS, Rayroux N, Grimm AJ, Ghisoni E, Dangaj Laniti D. Crosstalk of T cells within the ovarian cancer microenvironment. Trends Cancer 2024; 10:1116-1130. [PMID: 39341696 DOI: 10.1016/j.trecan.2024.09.001] [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: 06/28/2024] [Revised: 09/02/2024] [Accepted: 09/03/2024] [Indexed: 10/01/2024]
Abstract
Ovarian cancer (OC) represents ecosystems of highly diverse tumor microenvironments (TMEs). The presence of tumor-infiltrating lymphocytes (TILs) is linked to enhanced immune responses and long-term survival. In this review we present emerging evidence suggesting that cellular crosstalk tightly regulates the distribution of TILs within the TME, underscoring the need to better understand key cellular networks that promote or impede T cell infiltration in OC. We also capture the emergent methodologies and computational techniques that enable the dissection of cell-cell crosstalk. Finally, we present innovative ex vivo TME models that can be leveraged to map and perturb cellular communications to enhance T cell infiltration and immune reactivity.
Collapse
Affiliation(s)
- Bovannak S Chap
- Department of Oncology, Lausanne University Hospital, University of Lausanne, Lausanne, Switzerland; Ludwig Institute for Cancer Research, Lausanne Branch, University of Lausanne (UNIL), Lausanne, Switzerland; Agora Cancer Research Center, Lausanne, Switzerland
| | - Nicolas Rayroux
- Department of Oncology, Lausanne University Hospital, University of Lausanne, Lausanne, Switzerland; Ludwig Institute for Cancer Research, Lausanne Branch, University of Lausanne (UNIL), Lausanne, Switzerland; Agora Cancer Research Center, Lausanne, Switzerland
| | - Alizée J Grimm
- Department of Oncology, Lausanne University Hospital, University of Lausanne, Lausanne, Switzerland; Ludwig Institute for Cancer Research, Lausanne Branch, University of Lausanne (UNIL), Lausanne, Switzerland; Agora Cancer Research Center, Lausanne, Switzerland
| | - Eleonora Ghisoni
- Department of Oncology, Lausanne University Hospital, University of Lausanne, Lausanne, Switzerland; Ludwig Institute for Cancer Research, Lausanne Branch, University of Lausanne (UNIL), Lausanne, Switzerland; Agora Cancer Research Center, Lausanne, Switzerland
| | - Denarda Dangaj Laniti
- Department of Oncology, Lausanne University Hospital, University of Lausanne, Lausanne, Switzerland; Ludwig Institute for Cancer Research, Lausanne Branch, University of Lausanne (UNIL), Lausanne, Switzerland; Agora Cancer Research Center, Lausanne, Switzerland.
| |
Collapse
|
5
|
Guo Y, Li T, Gong B, Hu Y, Wang S, Yang L, Zheng C. From Images to Genes: Radiogenomics Based on Artificial Intelligence to Achieve Non-Invasive Precision Medicine in Cancer Patients. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024:e2408069. [PMID: 39535476 DOI: 10.1002/advs.202408069] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/15/2024] [Revised: 10/19/2024] [Indexed: 11/16/2024]
Abstract
With the increasing demand for precision medicine in cancer patients, radiogenomics emerges as a promising frontier. Radiogenomics is originally defined as a methodology for associating gene expression information from high-throughput technologies with imaging phenotypes. However, with advancements in medical imaging, high-throughput omics technologies, and artificial intelligence, both the concept and application of radiogenomics have significantly broadened. In this review, the history of radiogenomics is enumerated, related omics technologies, the five basic workflows and their applications across tumors, the role of AI in radiogenomics, the opportunities and challenges from tumor heterogeneity, and the applications of radiogenomics in tumor immune microenvironment. The application of radiogenomics in positron emission tomography and the role of radiogenomics in multi-omics studies is also discussed. Finally, the challenges faced by clinical transformation, along with future trends in this field is discussed.
Collapse
Affiliation(s)
- Yusheng Guo
- Department of Radiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Hubei Key Laboratory of Molecular Imaging, Wuhan, 430022, China
| | - Tianxiang Li
- Department of Ultrasound, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical. Sciences, Peking Union Medical College, Beijing, 100730, China
| | - Bingxin Gong
- Department of Radiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Hubei Key Laboratory of Molecular Imaging, Wuhan, 430022, China
| | - Yan Hu
- Research Institute of Trustworthy Autonomous Systems and Department of Computer Science and Engineering, Southern University of Science and Technology, Shenzhen, 518055, China
| | - Sichen Wang
- School of Life Science and Technology, Computational Biology Research Center, Harbin Institute of Technology, Harbin, 150001, China
| | - Lian Yang
- Department of Radiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Hubei Key Laboratory of Molecular Imaging, Wuhan, 430022, China
| | - Chuansheng Zheng
- Department of Radiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Hubei Key Laboratory of Molecular Imaging, Wuhan, 430022, China
| |
Collapse
|
6
|
Wang J, Wang S, Li Q, Liu F, Wan Y, Liang H. Bibliometric and visual analysis of single-cell multiomics in neurodegenerative disease arrest studies. Front Neurol 2024; 15:1450663. [PMID: 39440247 PMCID: PMC11493674 DOI: 10.3389/fneur.2024.1450663] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2024] [Accepted: 09/26/2024] [Indexed: 10/25/2024] Open
Abstract
Background Neurodegenerative diseases are progressive disorders that severely diminish the quality of life of patients. However, research on neurodegenerative diseases needs to be refined and deepened. Single-cell polyomics is a technique for obtaining transcriptomic, proteomic, and other information from a single cell. In recent years, the heat of single-cell multiomics as an emerging research tool for brain science has gradually increased. Therefore, the aim of this study was to analyze the current status and trends of studies related to the application of single-cell multiomics in neurodegenerative diseases through bibliometrics. Result A total of 596 publications were included in the bibliometric analysis. Between 2015 and 2022, the number of publications increased annually, with the total number of citations increasing significantly, exhibiting the fastest rate of growth between 2019 and 2022. The country/region collaboration map shows that the United States has the most publications and cumulative citations, and that China and the United States have the most collaborations. The institutions that produced the greatest number of articles were Harvard Medical School, Skupin, Alexander, and Wiendl. Among the authors, Heinz had the highest output. Mathys, H accumulated the most citations and was the authoritative author in the field. The journal Nature Communications has published the most literature in this field. A keyword analysis reveals that neurodegenerative diseases and lesions (e.g., Alzheimer's disease, amyloid beta) are the core and foundation of the field. Conversely, single-cell multiomics related research (e.g., single-cell RNA sequencing, bioinformatics) and brain nerve cells (e.g., microglia, astrocytes, neural stem cells) are the hot frontiers of this specialty. Among the references, the article "Single-cell transcriptomic analysis of Alzheimer's disease" is the most frequently cited (1,146 citations), and the article "Cell types in the mouse cortex and hippocampus revealed by single-cell RNA-seq" was the most cited article in the field. Conclusion The objective of this study is to employ bibliometric methods to visualize studies related to single-cell multiomics in neurodegenerative diseases. This will enable us to summarize the current state of research and to reveal key trends and emerging hotspots in the field.
Collapse
Affiliation(s)
- Jieyan Wang
- Department of Urology, People’s Hospital of Longhua, Shenzhen, China
| | - Shuqing Wang
- First Clinical Medical School, Southern Medical University, Guangzhou, China
| | - Qingyu Li
- First Clinical Medical School, Southern Medical University, Guangzhou, China
| | - Fei Liu
- First Clinical Medical School, Southern Medical University, Guangzhou, China
| | - Yantong Wan
- Guangdong Provincial Key Laboratory of Proteomics, Department of Pathophysiology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Hui Liang
- Department of Urology, People’s Hospital of Longhua, Shenzhen, China
| |
Collapse
|
7
|
Zhang W, Xu Y, Zheng X, Shen J, Li Y. Identifying cell types by lasso-constraint regularized Gaussian graphical model based on weighted distance penalty. Brief Bioinform 2024; 25:bbae572. [PMID: 39541187 PMCID: PMC11562834 DOI: 10.1093/bib/bbae572] [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: 08/11/2024] [Revised: 10/10/2024] [Accepted: 10/29/2024] [Indexed: 11/16/2024] Open
Abstract
Single-cell RNA sequencing (scRNA-seq) technology is one of the most cost-effective and efficacious methods for revealing cellular heterogeneity and diversity. Precise identification of cell types is essential for establishing a robust foundation for downstream analyses and is a prerequisite for understanding heterogeneous mechanisms. However, the accuracy of existing methods warrants improvement, and highly accurate methods often impose stringent equipment requirements. Moreover, most unsupervised learning-based approaches are constrained by the need to input the number of cell types a prior, which limits their widespread application. In this paper, we propose a novel algorithm framework named WLGG. Initially, to capture the underlying nonlinear information, we introduce a weighted distance penalty term utilizing the Gaussian kernel function, which maps data from a low-dimensional nonlinear space to a high-dimensional linear space. We subsequently impose a Lasso constraint on the regularized Gaussian graphical model to enhance its ability to capture linear data characteristics. Additionally, we utilize the Eigengap strategy to predict the number of cell types and obtain predicted labels via spectral clustering. The experimental results on 14 test datasets demonstrate the superior clustering accuracy of the WLGG algorithm over 16 alternative methods. Furthermore, downstream analysis, including marker gene identification, pseudotime inference, and functional enrichment analysis based on the similarity matrix and predicted labels from the WLGG algorithm, substantiates the reliability of WLGG and offers valuable insights into biological dynamic biological processes and regulatory mechanisms.
Collapse
Affiliation(s)
- Wei Zhang
- School of Mathematics and Physics, Wuhan Institute of Technology, Wuhan 430205, China
| | - Yaxin Xu
- Peng Cheng Laboratory, and School of Microelectronics, Southern University of Science and Technology, Shenzhen 518055, China
| | - Xiaoying Zheng
- School of Mathematics and Physics, Wuhan Institute of Technology, Wuhan 430205, China
| | - Juan Shen
- School of Mathematics and Physics, Wuhan Institute of Technology, Wuhan 430205, China
| | - Yuanyuan Li
- School of Mathematics and Physics, Wuhan Institute of Technology, Wuhan 430205, China
| |
Collapse
|
8
|
Ding Q, Yang W, Xue G, Liu H, Cai Y, Que J, Jin X, Luo M, Pang F, Yang Y, Lin Y, Liu Y, Sun H, Tan R, Wang P, Xu Z, Jiang Q. Dimension reduction, cell clustering, and cell-cell communication inference for single-cell transcriptomics with DcjComm. Genome Biol 2024; 25:241. [PMID: 39252099 PMCID: PMC11382422 DOI: 10.1186/s13059-024-03385-6] [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: 01/09/2024] [Accepted: 08/30/2024] [Indexed: 09/11/2024] Open
Abstract
Advances in single-cell transcriptomics provide an unprecedented opportunity to explore complex biological processes. However, computational methods for analyzing single-cell transcriptomics still have room for improvement especially in dimension reduction, cell clustering, and cell-cell communication inference. Herein, we propose a versatile method, named DcjComm, for comprehensive analysis of single-cell transcriptomics. DcjComm detects functional modules to explore expression patterns and performs dimension reduction and clustering to discover cellular identities by the non-negative matrix factorization-based joint learning model. DcjComm then infers cell-cell communication by integrating ligand-receptor pairs, transcription factors, and target genes. DcjComm demonstrates superior performance compared to state-of-the-art methods.
Collapse
Affiliation(s)
- Qian Ding
- Center for Bioinformatics, School of Life Science and Technology, Harbin Institute of Technology, Harbin, 150000, China
| | - Wenyi Yang
- Center for Bioinformatics, School of Life Science and Technology, Harbin Institute of Technology, Harbin, 150000, China
| | - Guangfu Xue
- Center for Bioinformatics, School of Life Science and Technology, Harbin Institute of Technology, Harbin, 150000, China
| | - Hongxin Liu
- Center for Bioinformatics, School of Life Science and Technology, Harbin Institute of Technology, Harbin, 150000, China
| | - Yideng Cai
- Center for Bioinformatics, School of Life Science and Technology, Harbin Institute of Technology, Harbin, 150000, China
| | - Jinhao Que
- Center for Bioinformatics, School of Life Science and Technology, Harbin Institute of Technology, Harbin, 150000, China
| | - Xiyun Jin
- School of Interdisciplinary Medicine and Engineering, Harbin Medical University, Harbin, 150076, China
| | - Meng Luo
- Center for Bioinformatics, School of Life Science and Technology, Harbin Institute of Technology, Harbin, 150000, China
| | - Fenglan Pang
- Center for Bioinformatics, School of Life Science and Technology, Harbin Institute of Technology, Harbin, 150000, China
| | - Yuexin Yang
- Center for Bioinformatics, School of Life Science and Technology, Harbin Institute of Technology, Harbin, 150000, China
| | - Yi Lin
- School of Interdisciplinary Medicine and Engineering, Harbin Medical University, Harbin, 150076, China
| | - Yusong Liu
- School of Interdisciplinary Medicine and Engineering, Harbin Medical University, Harbin, 150076, China
| | - Haoxiu Sun
- School of Interdisciplinary Medicine and Engineering, Harbin Medical University, Harbin, 150076, China
| | - Renjie Tan
- School of Interdisciplinary Medicine and Engineering, Harbin Medical University, Harbin, 150076, China
| | - Pingping Wang
- School of Interdisciplinary Medicine and Engineering, Harbin Medical University, Harbin, 150076, China.
| | - Zhaochun Xu
- School of Interdisciplinary Medicine and Engineering, Harbin Medical University, Harbin, 150076, China.
| | - Qinghua Jiang
- Center for Bioinformatics, School of Life Science and Technology, Harbin Institute of Technology, Harbin, 150000, China.
- School of Interdisciplinary Medicine and Engineering, Harbin Medical University, Harbin, 150076, China.
- State Key Laboratory of Frigid Zone Cardiovascular Diseases (SKLFZCD), Harbin Medical University, Harbin, 150076, China.
| |
Collapse
|
9
|
Su J, Song Y, Zhu Z, Huang X, Fan J, Qiao J, Mao F. Cell-cell communication: new insights and clinical implications. Signal Transduct Target Ther 2024; 9:196. [PMID: 39107318 PMCID: PMC11382761 DOI: 10.1038/s41392-024-01888-z] [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: 12/29/2023] [Revised: 05/09/2024] [Accepted: 06/02/2024] [Indexed: 09/11/2024] Open
Abstract
Multicellular organisms are composed of diverse cell types that must coordinate their behaviors through communication. Cell-cell communication (CCC) is essential for growth, development, differentiation, tissue and organ formation, maintenance, and physiological regulation. Cells communicate through direct contact or at a distance using ligand-receptor interactions. So cellular communication encompasses two essential processes: cell signal conduction for generation and intercellular transmission of signals, and cell signal transduction for reception and procession of signals. Deciphering intercellular communication networks is critical for understanding cell differentiation, development, and metabolism. First, we comprehensively review the historical milestones in CCC studies, followed by a detailed description of the mechanisms of signal molecule transmission and the importance of the main signaling pathways they mediate in maintaining biological functions. Then we systematically introduce a series of human diseases caused by abnormalities in cell communication and their progress in clinical applications. Finally, we summarize various methods for monitoring cell interactions, including cell imaging, proximity-based chemical labeling, mechanical force analysis, downstream analysis strategies, and single-cell technologies. These methods aim to illustrate how biological functions depend on these interactions and the complexity of their regulatory signaling pathways to regulate crucial physiological processes, including tissue homeostasis, cell development, and immune responses in diseases. In addition, this review enhances our understanding of the biological processes that occur after cell-cell binding, highlighting its application in discovering new therapeutic targets and biomarkers related to precision medicine. This collective understanding provides a foundation for developing new targeted drugs and personalized treatments.
Collapse
Affiliation(s)
- Jimeng Su
- Institute of Medical Innovation and Research, Peking University Third Hospital, Beijing, China
- Cancer Center, Peking University Third Hospital, Beijing, China
- College of Animal Science and Technology, Yangzhou University, Yangzhou, Jiangsu, China
| | - Ying Song
- Institute of Medical Innovation and Research, Peking University Third Hospital, Beijing, China
- Cancer Center, Peking University Third Hospital, Beijing, China
| | - Zhipeng Zhu
- Institute of Medical Innovation and Research, Peking University Third Hospital, Beijing, China
- Cancer Center, Peking University Third Hospital, Beijing, China
| | - Xinyue Huang
- Biomedical Research Institute, Shenzhen Peking University-the Hong Kong University of Science and Technology Medical Center, Shenzhen, China
| | - Jibiao Fan
- College of Animal Science and Technology, Yangzhou University, Yangzhou, Jiangsu, China
| | - Jie Qiao
- State Key Laboratory of Female Fertility Promotion, Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, China.
- National Clinical Research Center for Obstetrics and Gynecology (Peking University Third Hospital), Beijing, China.
- Key Laboratory of Assisted Reproduction (Peking University), Ministry of Education, Beijing, China.
- Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive Technology, Beijing, China.
| | - Fengbiao Mao
- Institute of Medical Innovation and Research, Peking University Third Hospital, Beijing, China.
- Cancer Center, Peking University Third Hospital, Beijing, China.
| |
Collapse
|
10
|
Masters H, Wang S, Tu C, Nguyen Q, Sha Y, Karikomi MK, Fung PSR, Tran B, Martel C, Kwang N, Neel M, Jaime OG, Espericueta V, Johnson BA, Kessenbrock K, Nie Q, Monuki ES. Sequential emergence and contraction of epithelial subtypes in the prenatal human choroid plexus revealed by a stem cell model. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.06.12.598747. [PMID: 38948782 PMCID: PMC11212933 DOI: 10.1101/2024.06.12.598747] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/02/2024]
Abstract
Despite the major roles of choroid plexus epithelial cells (CPECs) in brain homeostasis and repair, their developmental lineage and diversity remain undefined. In simplified differentiations from human pluripotent stem cells, derived CPECs (dCPECs) displayed canonical properties and dynamic multiciliated phenotypes that interacted with Aβ uptake. Single dCPEC transcriptomes over time correlated well with human organoid and fetal CPECs, while pseudotemporal and cell cycle analyses highlighted the direct CPEC origin from neuroepithelial cells. In addition, time series analyses defined metabolic (type 1) and ciliogenic dCPECs (type 2) at early timepoints, followed by type 1 diversification into anabolic-secretory (type 1a) and catabolic-absorptive subtypes (type 1b) as type 2 cells contracted. These temporal patterns were then confirmed in independent derivations and mapped to prenatal stages using human tissues. In addition to defining the prenatal lineage of human CPECs, these findings suggest new dynamic models of ChP support for the developing human brain.
Collapse
|
11
|
Armingol E, Baghdassarian HM, Lewis NE. The diversification of methods for studying cell-cell interactions and communication. Nat Rev Genet 2024; 25:381-400. [PMID: 38238518 PMCID: PMC11139546 DOI: 10.1038/s41576-023-00685-8] [Citation(s) in RCA: 15] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/01/2023] [Indexed: 05/20/2024]
Abstract
No cell lives in a vacuum, and the molecular interactions between cells define most phenotypes. Transcriptomics provides rich information to infer cell-cell interactions and communication, thus accelerating the discovery of the roles of cells within their communities. Such research relies heavily on algorithms that infer which cells are interacting and the ligands and receptors involved. Specific pressures on different research niches are driving the evolution of next-generation computational tools, enabling new conceptual opportunities and technological advances. More sophisticated algorithms now account for the heterogeneity and spatial organization of cells, multiple ligand types and intracellular signalling events, and enable the use of larger and more complex datasets, including single-cell and spatial transcriptomics. Similarly, new high-throughput experimental methods are increasing the number and resolution of interactions that can be analysed simultaneously. Here, we explore recent progress in cell-cell interaction research and highlight the diversification of the next generation of tools, which have yielded a rich ecosystem of tools for different applications and are enabling invaluable discoveries.
Collapse
Affiliation(s)
- Erick Armingol
- Bioinformatics and Systems Biology Graduate Program, University of California, San Diego, La Jolla, CA, USA.
- Department of Paediatrics, University of California, San Diego, La Jolla, CA, USA.
| | - Hratch M Baghdassarian
- Bioinformatics and Systems Biology Graduate Program, University of California, San Diego, La Jolla, CA, USA
- Department of Paediatrics, University of California, San Diego, La Jolla, CA, USA
| | - Nathan E Lewis
- Department of Paediatrics, University of California, San Diego, La Jolla, CA, USA.
- Department of Bioengineering, University of California, San Diego, La Jolla, CA, USA.
| |
Collapse
|
12
|
Sun M, Rong J, Zhou M, Liu Y, Sun S, Liu L, Cai D, Liang F, Zhao L. Astrocyte-Microglia Crosstalk: A Novel Target for the Treatment of Migraine. Aging Dis 2024; 15:1277-1288. [PMID: 37450927 PMCID: PMC11081170 DOI: 10.14336/ad.2023.0623] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2022] [Accepted: 06/23/2023] [Indexed: 07/18/2023] Open
Abstract
Migraine is a pervasive neurologic disease closely related to neurogenic inflammation. The astrocytes and microglia in the central nervous system are vital in inducing neurogenic inflammation in migraine. Recently, it has been found that there may be a crosstalk phenomenon between microglia and astrocytes, which plays a crucial part in the pathology and treatment of Alzheimer's disease and other central nervous system diseases closely related to inflammation, thus becoming a novel hotspot in neuroimmune research. However, the role of the crosstalk between microglia and astrocytes in the pathogenesis and treatment of migraine is yet to be discussed. Based on the preliminary literature reports, we have reviewed relevant evidence of the crosstalk between microglia and astrocytes in the pathogenesis of migraine and summarized the crosstalk pathways, thereby hoping to provide novel ideas for future research and treatment.
Collapse
Affiliation(s)
- Mingsheng Sun
- College of Acupuncture and Tuina, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Jing Rong
- College of Acupuncture and Tuina, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Mengdi Zhou
- College of Acupuncture and Tuina, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Yi Liu
- College of Acupuncture and Tuina, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Shiqi Sun
- College of Acupuncture and Tuina, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Lu Liu
- College of Acupuncture and Tuina, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Dingjun Cai
- College of Acupuncture and Tuina, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Fanrong Liang
- College of Acupuncture and Tuina, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Ling Zhao
- College of Acupuncture and Tuina, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| |
Collapse
|
13
|
Vasighizaker A, Hora S, Zeng R, Rueda L. SEGCECO: Subgraph Embedding of Gene expression matrix for prediction of CEll-cell COmmunication. Brief Bioinform 2024; 25:bbae160. [PMID: 38605638 PMCID: PMC11009470 DOI: 10.1093/bib/bbae160] [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: 11/07/2023] [Revised: 01/30/2024] [Accepted: 03/13/2024] [Indexed: 04/13/2024] Open
Abstract
Recent advances in single-cell RNA sequencing technology have eased analyses of signaling networks of cells. Recently, cell-cell interaction has been studied based on various link prediction approaches on graph-structured data. These approaches have assumptions about the likelihood of node interaction, thus showing high performance for only some specific networks. Subgraph-based methods have solved this problem and outperformed other approaches by extracting local subgraphs from a given network. In this work, we present a novel method, called Subgraph Embedding of Gene expression matrix for prediction of CEll-cell COmmunication (SEGCECO), which uses an attributed graph convolutional neural network to predict cell-cell communication from single-cell RNA-seq data. SEGCECO captures the latent and explicit attributes of undirected, attributed graphs constructed from the gene expression profile of individual cells. High-dimensional and sparse single-cell RNA-seq data make converting the data into a graphical format a daunting task. We successfully overcome this limitation by applying SoptSC, a similarity-based optimization method in which the cell-cell communication network is built using a cell-cell similarity matrix which is learned from gene expression data. We performed experiments on six datasets extracted from the human and mouse pancreas tissue. Our comparative analysis shows that SEGCECO outperforms latent feature-based approaches, and the state-of-the-art method for link prediction, WLNM, with 0.99 ROC and 99% prediction accuracy. The datasets can be found at https://www.ncbi.nlm.nih.gov/geo/query/acc.cgi?acc=GSE84133 and the code is publicly available at Github https://github.com/sheenahora/SEGCECO and Code Ocean https://codeocean.com/capsule/8244724/tree.
Collapse
Affiliation(s)
| | - Sheena Hora
- Software Development Department, Amazon, USA
| | - Raymond Zeng
- School of Computer Science, University of Windsor, Windsor, ON, Canada
| | - Luis Rueda
- School of Computer Science, University of Windsor, Windsor, ON, Canada
| |
Collapse
|
14
|
Peng L, Gao P, Xiong W, Li Z, Chen X. Identifying potential ligand-receptor interactions based on gradient boosted neural network and interpretable boosting machine for intercellular communication analysis. Comput Biol Med 2024; 171:108110. [PMID: 38367445 DOI: 10.1016/j.compbiomed.2024.108110] [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: 10/30/2023] [Revised: 01/24/2024] [Accepted: 02/04/2024] [Indexed: 02/19/2024]
Abstract
Cell-cell communication is essential to many key biological processes. Intercellular communication is generally mediated by ligand-receptor interactions (LRIs). Thus, building a comprehensive and high-quality LRI resource can significantly improve intercellular communication analysis. Meantime, due to lack of a "gold standard" dataset, it remains a challenge to evaluate LRI-mediated intercellular communication results. Here, we introduce CellGiQ, a high-confident LRI prediction framework for intercellular communication analysis. Highly confident LRIs are first inferred by LRI feature extraction with BioTriangle, LRI selection using LightGBM, and LRI classification based on ensemble of gradient boosted neural network and interpretable boosting machine. Subsequently, known and identified high-confident LRIs are filtered by combining single-cell RNA sequencing (scRNA-seq) data and further applied to intercellular communication inference through a quartile scoring strategy. To validation the predictions, CellGiQ exploited several evaluation strategies: using AUC and AUPR, it surpassed six competing LRI prediction models on four LRI datasets; through Venn diagrams and molecular docking, its predicted LRIs were validated by five other popular intercellular communication inference methods; based on the overlapping LRIs, it computed high Jaccard index with six other state-of-the-art intercellular communication prediction tools within human HNSCC tissues; by comparing with classical models and literature retrieve, its inferred HNSCC-related intercellular communication results was further validated. The novelty of this study is to identify high-confident LRIs based on machine learning as well as design several LRI validation ways, providing reference for computational LRI prediction. CellGiQ provides an open-source and useful tool to decompose LRI-mediated intercellular communication at single cell resolution. CellGiQ is freely available at https://github.com/plhhnu/CellGiQ.
Collapse
Affiliation(s)
- Lihong Peng
- College of Life Science and Chemistry, Hunan University of Technology, Zhuzhou, 412007, Hunan, China
| | - Pengfei Gao
- College of Life Science and Chemistry, Hunan University of Technology, Zhuzhou, 412007, Hunan, China
| | - Wei Xiong
- College of Life Science and Chemistry, Hunan University of Technology, Zhuzhou, 412007, Hunan, China
| | - Zejun Li
- School of Computer Science and Engineering, Hunan Institute of Technology, Hengyang, 421002, Hunan, China.
| | - Xing Chen
- School of Science, Jiangnan University, Wuxi, 214122, Jiangsu, China.
| |
Collapse
|
15
|
Xu Y, Zhang W, Zheng X, Cai X. Combining Global-Constrained Concept Factorization and a Regularized Gaussian Graphical Model for Clustering Single-Cell RNA-seq Data. Interdiscip Sci 2024; 16:1-15. [PMID: 37815679 DOI: 10.1007/s12539-023-00587-7] [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: 02/06/2023] [Revised: 09/14/2023] [Accepted: 09/17/2023] [Indexed: 10/11/2023]
Abstract
Single-cell RNA sequencing technology is one of the most cost-effective ways to uncover transcriptomic heterogeneity. With the rapid rise of this technology, enormous amounts of scRNA-seq data have been produced. Due to the high dimensionality, noise, sparsity and missing features of the available scRNA-seq data, accurately clustering the scRNA-seq data for downstream analysis is a significant challenge. Many computational methods have been designed to address this issue; nevertheless, the efficacy of the available methods is still inadequate. In addition, most similarity-based methods require a number of clusters as input, which is difficult to achieve in real applications. In this study, we developed a novel computational method for clustering scRNA-seq data by considering both global and local information, named GCFG. This method characterizes the global properties of data by applying concept factorization, and the regularized Gaussian graphical model is utilized to evaluate the local embedding relationship of data. To learn the cell-cell similarity matrix, we integrated the two components, and an iterative optimization algorithm was developed. The categorization of single cells is obtained by applying Louvain, a modularity-based community discovery algorithm, to the similarity matrix. The behavior of the GCFG approach is assessed on 14 real scRNA-seq datasets in terms of ACC and ARI, and comparison results with 17 other competitive methods suggest that GCFG is effective and robust.
Collapse
Affiliation(s)
- Yaxin Xu
- School of Sciences, East China Jiaotong University, Nanchang, 330013, China
| | - Wei Zhang
- School of Sciences, East China Jiaotong University, Nanchang, 330013, China.
| | - Xiaoying Zheng
- Operations Research and Planning Department, Naval University of Engineering, Wuhan, 430033, China
| | - Xianxian Cai
- School of Sciences, East China Jiaotong University, Nanchang, 330013, China
| |
Collapse
|
16
|
Liu Y, Zhang Y, Chang X, Liu X. MDIC3: Matrix decomposition to infer cell-cell communication. PATTERNS (NEW YORK, N.Y.) 2024; 5:100911. [PMID: 38370122 PMCID: PMC10873161 DOI: 10.1016/j.patter.2023.100911] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/12/2022] [Revised: 05/31/2023] [Accepted: 12/08/2023] [Indexed: 02/20/2024]
Abstract
Crosstalk among cells is vital for maintaining the biological function and intactness of systems. Most existing methods for investigating cell-cell communications are based on ligand-receptor (L-R) expression, and they focus on the study between two cells. Thus, the final communication inference results are particularly sensitive to the completeness and accuracy of the prior biological knowledge. Because existing L-R research focuses mainly on humans, most existing methods can only examine cell-cell communication for humans. As far as we know, there is currently no effective method to overcome this species limitation. Here, we propose MDIC3 (matrix decomposition to infer cell-cell communication), an unsupervised tool to investigate cell-cell communication in any species, and the results are not limited by specific L-R pairs or signaling pathways. By comparing it with existing methods for the inference of cell-cell communication, MDIC3 obtained better performance in both humans and mice.
Collapse
Affiliation(s)
- Yi Liu
- Key Laboratory of Systems Health Science of Zhejiang Province, School of Life Science, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou 310024, China
- School of Mathematics and Statistics, Shandong University, Weihai 364209, China
| | - Yuelei Zhang
- School of Mathematics and Statistics, Shandong University, Weihai 364209, China
| | - Xiao Chang
- Institute of Statistics and Applied Mathematics, Anhui University of Finance and Economics, Bengbu 233030, China
| | - Xiaoping Liu
- Key Laboratory of Systems Health Science of Zhejiang Province, School of Life Science, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou 310024, China
| |
Collapse
|
17
|
Xu H, Wang T, Miao Y, Qian M, Yang Y, Wang S. MK-BMC: a Multi-Kernel framework with Boosted distance metrics for Microbiome data for Classification. Bioinformatics 2024; 40:btad757. [PMID: 38200571 PMCID: PMC10789312 DOI: 10.1093/bioinformatics/btad757] [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: 06/09/2023] [Revised: 10/30/2023] [Accepted: 01/09/2024] [Indexed: 01/12/2024] Open
Abstract
MOTIVATION Research on human microbiome has suggested associations with human health, opening opportunities to predict health outcomes using microbiome. Studies have also suggested that diverse forms of taxa such as rare taxa that are evolutionally related and abundant taxa that are evolutionally unrelated could be associated with or predictive of a health outcome. Although prediction models were developed for microbiome data, no prediction models currently exist that use multiple forms of microbiome-outcome associations. RESULTS We developed MK-BMC, a Multi-Kernel framework with Boosted distance Metrics for Classification using microbiome data. We propose to first boost widely used distance metrics for microbiome data using taxon-level association signal strengths to up-weight taxa that are potentially associated with an outcome of interest. We then propose a multi-kernel prediction model with one kernel capturing one form of association between taxa and the outcome, where a kernel measures similarities of microbiome compositions between pairs of samples being transformed from a proposed boosted distance metric. We demonstrated superior prediction performance of (i) boosted distance metrics for microbiome data over original ones and (ii) MK-BMC over competing methods through extensive simulations. We applied MK-BMC to predict thyroid, obesity, and inflammatory bowel disease status using gut microbiome data from the American Gut Project and observed much-improved prediction performance over that of competing methods. The learned kernel weights help us understand contributions of individual microbiome signal forms nicely. AVAILABILITY AND IMPLEMENTATION Source code together with a sample input dataset is available at https://github.com/HXu06/MK-BMC.
Collapse
Affiliation(s)
- Huang Xu
- Department of Statistics and Finance, University of Science and Technology of China, Hefei 230026, China
| | - Tian Wang
- Department of Biostatistics, Mailman School of Public Health, Columbia University, New York, NY 10032, United States
| | - Yuqi Miao
- Department of Biostatistics, Mailman School of Public Health, Columbia University, New York, NY 10032, United States
| | - Min Qian
- Department of Biostatistics, Mailman School of Public Health, Columbia University, New York, NY 10032, United States
| | - Yaning Yang
- Department of Statistics and Finance, University of Science and Technology of China, Hefei 230026, China
| | - Shuang Wang
- Department of Biostatistics, Mailman School of Public Health, Columbia University, New York, NY 10032, United States
| |
Collapse
|
18
|
Wang X, Almet AA, Nie Q. The promising application of cell-cell interaction analysis in cancer from single-cell and spatial transcriptomics. Semin Cancer Biol 2023; 95:42-51. [PMID: 37454878 PMCID: PMC10627116 DOI: 10.1016/j.semcancer.2023.07.001] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Revised: 03/02/2023] [Accepted: 07/13/2023] [Indexed: 07/18/2023]
Abstract
Cell-cell interactions instruct cell fate and function. These interactions are hijacked to promote cancer development. Single-cell transcriptomics and spatial transcriptomics have become powerful new tools for researchers to profile the transcriptional landscape of cancer at unparalleled genetic depth. In this review, we discuss the rapidly growing array of computational tools to infer cell-cell interactions from non-spatial single-cell RNA-sequencing and the limited but growing number of methods for spatial transcriptomics data. Downstream analyses of these computational tools and applications to cancer studies are highlighted. We finish by suggesting several directions for further extensions that anticipate the increasing availability of multi-omics cancer data.
Collapse
Affiliation(s)
- Xinyi Wang
- Department of Mathematics, University of California, Irvine, Irvine, CA, United States
| | - Axel A Almet
- Department of Mathematics, University of California, Irvine, Irvine, CA, United States; The NSF-Simons Center for Multiscale Cell Fate Research, University of California, Irvine, Irvine, CA, United States.
| | - Qing Nie
- Department of Mathematics, University of California, Irvine, Irvine, CA, United States; The NSF-Simons Center for Multiscale Cell Fate Research, University of California, Irvine, Irvine, CA, United States; Department of Developmental and Cell Biology, University of California, Irvine, Irvine, CA, United States.
| |
Collapse
|
19
|
Luo J, Deng M, Zhang X, Sun X. ESICCC as a systematic computational framework for evaluation, selection, and integration of cell-cell communication inference methods. Genome Res 2023; 33:1788-1805. [PMID: 37827697 PMCID: PMC10691505 DOI: 10.1101/gr.278001.123] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2023] [Accepted: 09/21/2023] [Indexed: 10/14/2023]
Abstract
Cell-cell communication (CCC) is critical for determining cell fates and functions in multicellular organisms. With the advent of single-cell RNA-sequencing (scRNA-seq) and spatial transcriptomics (ST), an increasing number of CCC inference methods have been developed. Nevertheless, a thorough comparison of their performances is yet to be conducted. To fill this gap, we developed a systematic benchmark framework called ESICCC to evaluate 18 ligand-receptor (LR) inference methods and five ligand/receptor-target inference methods using a total of 116 data sets, including 15 ST data sets, 15 sets of cell line perturbation data, two sets of cell type-specific expression/proteomics data, and 84 sets of sampled or unsampled scRNA-seq data. We evaluated and compared the agreement, accuracy, robustness, and usability of these methods. Regarding accuracy evaluation, RNAMagnet, CellChat, and scSeqComm emerge as the three best-performing methods for intercellular ligand-receptor inference based on scRNA-seq data, whereas stMLnet and HoloNet are the best methods for predicting ligand/receptor-target regulation using ST data. To facilitate the practical applications, we provide a decision-tree-style guideline for users to easily choose best tools for their specific research concerns in CCC inference, and develop an ensemble pipeline CCCbank that enables versatile combinations of methods and databases. Moreover, our comparative results also uncover several critical influential factors for CCC inference, such as prior interaction information, ligand-receptor scoring algorithm, intracellular signaling complexity, and spatial relationship, which may be considered in the future studies to advance the development of new methodologies.
Collapse
Affiliation(s)
- Jiaxin Luo
- Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou 510080, China
- School of Mathematics, Sun Yat-sen University, Guangzhou 510275, China
| | - Minghua Deng
- School of Mathematical Sciences, Peking University, Beijing, 100871, China
| | - Xuegong Zhang
- Bioinformatics Division of BNRIST and Department of Automation, MOE Key Lab of Bioinformatics, Tsinghua University, Beijing, 100084, China
| | - Xiaoqiang Sun
- School of Mathematics, Sun Yat-sen University, Guangzhou 510275, China;
| |
Collapse
|
20
|
Luo X, Liu Z, Xu R. Adult tissue-specific stem cell interaction: novel technologies and research advances. Front Cell Dev Biol 2023; 11:1220694. [PMID: 37808078 PMCID: PMC10551553 DOI: 10.3389/fcell.2023.1220694] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2023] [Accepted: 09/11/2023] [Indexed: 10/10/2023] Open
Abstract
Adult tissue-specific stem cells play a dominant role in tissue homeostasis and regeneration. Various in vivo markers of adult tissue-specific stem cells have been increasingly reported by lineage tracing in genetic mouse models, indicating that marked cells differentiation is crucial during homeostasis and regeneration. How adult tissue-specific stem cells with indicated markers contact the adjacent lineage with indicated markers is of significance to be studied. Novel methods bring future findings. Recent advances in lineage tracing, synthetic receptor systems, proximity labeling, and transcriptomics have enabled easier and more accurate cell behavior visualization and qualitative and quantitative analysis of cell-cell interactions than ever before. These technological innovations have prompted researchers to re-evaluate previous experimental results, providing increasingly compelling experimental results for understanding the mechanisms of cell-cell interactions. This review aimed to describe the recent methodological advances of dual enzyme lineage tracing system, the synthetic receptor system, proximity labeling, single-cell RNA sequencing and spatial transcriptomics in the study of adult tissue-specific stem cells interactions. An enhanced understanding of the mechanisms of adult tissue-specific stem cells interaction is important for tissue regeneration and maintenance of homeostasis in organisms.
Collapse
Affiliation(s)
| | | | - Ruoshi Xu
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Department of Cariology and Endodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| |
Collapse
|
21
|
Peng L, Tan J, Xiong W, Zhang L, Wang Z, Yuan R, Li Z, Chen X. Deciphering ligand-receptor-mediated intercellular communication based on ensemble deep learning and the joint scoring strategy from single-cell transcriptomic data. Comput Biol Med 2023; 163:107137. [PMID: 37364528 DOI: 10.1016/j.compbiomed.2023.107137] [Citation(s) in RCA: 26] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2023] [Revised: 05/18/2023] [Accepted: 06/04/2023] [Indexed: 06/28/2023]
Abstract
BACKGROUND Cell-cell communication in a tumor microenvironment is vital to tumorigenesis, tumor progression and therapy. Intercellular communication inference helps understand molecular mechanisms of tumor growth, progression and metastasis. METHODS Focusing on ligand-receptor co-expressions, in this study, we developed an ensemble deep learning framework, CellComNet, to decipher ligand-receptor-mediated cell-cell communication from single-cell transcriptomic data. First, credible LRIs are captured by integrating data arrangement, feature extraction, dimension reduction, and LRI classification based on an ensemble of heterogeneous Newton boosting machine and deep neural network. Next, known and identified LRIs are screened based on single-cell RNA sequencing (scRNA-seq) data in certain tissues. Finally, cell-cell communication is inferred by incorporating scRNA-seq data, the screened LRIs, a joint scoring strategy that combines expression thresholding and expression product of ligands and receptors. RESULTS The proposed CellComNet framework was compared with four competing protein-protein interaction prediction models (PIPR, XGBoost, DNNXGB, and OR-RCNN) and obtained the best AUCs and AUPRs on four LRI datasets, elucidating the optimal LRI classification ability. CellComNet was further applied to analyze intercellular communication in human melanoma and head and neck squamous cell carcinoma (HNSCC) tissues. The results demonstrate that cancer-associated fibroblasts highly communicate with melanoma cells and endothelial cells strong communicate with HNSCC cells. CONCLUSIONS The proposed CellComNet framework efficiently identified credible LRIs and significantly improved cell-cell communication inference performance. We anticipate that CellComNet can contribute to anticancer drug design and tumor-targeted therapy.
Collapse
Affiliation(s)
- Lihong Peng
- School of Computer Science, Hunan University of Technology, Zhuzhou, 412007, Hunan, China; College of Life Sciences and Chemistry, Hunan University of Technology, Zhuzhou, 412007, Hunan, China
| | - Jingwei Tan
- School of Computer Science, Hunan University of Technology, Zhuzhou, 412007, Hunan, China
| | - Wei Xiong
- School of Computer Science, Hunan University of Technology, Zhuzhou, 412007, Hunan, China
| | - Li Zhang
- School of Information and Control Engineering, China University of Mining and Technology, Xuzhou, 221116, Jiangsu, China
| | - Zhao Wang
- School of Computer Science, Hunan University of Technology, Zhuzhou, 412007, Hunan, China
| | - Ruya Yuan
- School of Computer Science, Hunan University of Technology, Zhuzhou, 412007, Hunan, China
| | - Zejun Li
- School of Computer Science, Hunan Institute of Technology, Hengyang, 421002, Hunan, China.
| | - Xing Chen
- School of Science, Jiangnan University, Wuxi, 214122, Jiangsu, China.
| |
Collapse
|
22
|
Maffuid K, Cao Y. Decoding the Complexity of Immune-Cancer Cell Interactions: Empowering the Future of Cancer Immunotherapy. Cancers (Basel) 2023; 15:4188. [PMID: 37627216 PMCID: PMC10453128 DOI: 10.3390/cancers15164188] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2023] [Revised: 08/16/2023] [Accepted: 08/16/2023] [Indexed: 08/27/2023] Open
Abstract
The tumor and tumor microenvironment (TME) consist of a complex network of cells, including malignant, immune, fibroblast, and vascular cells, which communicate with each other. Disruptions in cell-cell communication within the TME, caused by a multitude of extrinsic and intrinsic factors, can contribute to tumorigenesis, hinder the host immune system, and enable tumor evasion. Understanding and addressing intercellular miscommunications in the TME are vital for combating these processes. The effectiveness of immunotherapy and the heterogeneous response observed among patients can be attributed to the intricate cellular communication between immune cells and cancer cells. To unravel these interactions, various experimental, statistical, and computational techniques have been developed. These include ligand-receptor analysis, intercellular proximity labeling approaches, and imaging-based methods, which provide insights into the distorted cell-cell interactions within the TME. By characterizing these interactions, we can enhance the design of cancer immunotherapy strategies. In this review, we present recent advancements in the field of mapping intercellular communication, with a particular focus on immune-tumor cellular interactions. By modeling these interactions, we can identify critical factors and develop strategies to improve immunotherapy response and overcome treatment resistance.
Collapse
Affiliation(s)
- Kaitlyn Maffuid
- Division of Pharmacotherapy and Experimental Therapeutics, School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA;
| | - Yanguang Cao
- Division of Pharmacotherapy and Experimental Therapeutics, School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA;
- Lineberger Comprehensive Cancer Center, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| |
Collapse
|
23
|
Xie Z, Li X, Mora A. A Comparison of Cell-Cell Interaction Prediction Tools Based on scRNA-seq Data. Biomolecules 2023; 13:1211. [PMID: 37627276 PMCID: PMC10452151 DOI: 10.3390/biom13081211] [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: 06/18/2023] [Revised: 07/29/2023] [Accepted: 07/31/2023] [Indexed: 08/27/2023] Open
Abstract
Computational prediction of cell-cell interactions (CCIs) is becoming increasingly important for understanding disease development and progression. We present a benchmark study of available CCI prediction tools based on single-cell RNA sequencing (scRNA-seq) data. By comparing prediction outputs with a manually curated gold standard for idiopathic pulmonary fibrosis (IPF), we evaluated prediction performance and processing time of several CCI prediction tools, including CCInx, CellChat, CellPhoneDB, iTALK, NATMI, scMLnet, SingleCellSignalR, and an ensemble of tools. According to our results, CellPhoneDB and NATMI are the best performer CCI prediction tools, among the ones analyzed, when we define a CCI as a source-target-ligand-receptor tetrad. In addition, we recommend specific tools according to different types of research projects and discuss the possible future paths in the field.
Collapse
Affiliation(s)
- Zihong Xie
- Joint School of Life Sciences, Guangzhou Medical University and Guangzhou Institutes of Biomedicine and Health (Chinese Academy of Sciences), Guangzhou 511436, China;
| | - Xuri Li
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-Sen University, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou 510060, China
| | - Antonio Mora
- Joint School of Life Sciences, Guangzhou Medical University and Guangzhou Institutes of Biomedicine and Health (Chinese Academy of Sciences), Guangzhou 511436, China;
| |
Collapse
|
24
|
Cheng C, Chen W, Jin H, Chen X. A Review of Single-Cell RNA-Seq Annotation, Integration, and Cell-Cell Communication. Cells 2023; 12:1970. [PMID: 37566049 PMCID: PMC10417635 DOI: 10.3390/cells12151970] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2023] [Revised: 07/10/2023] [Accepted: 07/21/2023] [Indexed: 08/12/2023] Open
Abstract
Single-cell RNA sequencing (scRNA-seq) has emerged as a powerful tool for investigating cellular biology at an unprecedented resolution, enabling the characterization of cellular heterogeneity, identification of rare but significant cell types, and exploration of cell-cell communications and interactions. Its broad applications span both basic and clinical research domains. In this comprehensive review, we survey the current landscape of scRNA-seq analysis methods and tools, focusing on count modeling, cell-type annotation, data integration, including spatial transcriptomics, and the inference of cell-cell communication. We review the challenges encountered in scRNA-seq analysis, including issues of sparsity or low expression, reliability of cell annotation, and assumptions in data integration, and discuss the potential impact of suboptimal clustering and differential expression analysis tools on downstream analyses, particularly in identifying cell subpopulations. Finally, we discuss recent advancements and future directions for enhancing scRNA-seq analysis. Specifically, we highlight the development of novel tools for annotating single-cell data, integrating and interpreting multimodal datasets covering transcriptomics, epigenomics, and proteomics, and inferring cellular communication networks. By elucidating the latest progress and innovation, we provide a comprehensive overview of the rapidly advancing field of scRNA-seq analysis.
Collapse
Affiliation(s)
- Changde Cheng
- Department of Computational Biology, St. Jude Children’s Research Hospital, Memphis, TN 38105, USA;
| | - Wenan Chen
- Center for Applied Bioinformatics, St. Jude Children’s Research Hospital, Memphis, TN 38105, USA; (W.C.); (H.J.)
| | - Hongjian Jin
- Center for Applied Bioinformatics, St. Jude Children’s Research Hospital, Memphis, TN 38105, USA; (W.C.); (H.J.)
| | - Xiang Chen
- Department of Computational Biology, St. Jude Children’s Research Hospital, Memphis, TN 38105, USA;
| |
Collapse
|
25
|
Jin J, Yu S, Lu P, Cao P. Deciphering plant cell-cell communications using single-cell omics data. Comput Struct Biotechnol J 2023; 21:3690-3695. [PMID: 37576747 PMCID: PMC10412842 DOI: 10.1016/j.csbj.2023.06.016] [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: 10/27/2022] [Revised: 06/16/2023] [Accepted: 06/16/2023] [Indexed: 08/15/2023] Open
Abstract
Plants have various cell types that respond to different environmental factors, and cell-cell communication is the fundamental process that controls these plant responses. The emergence of single-cell techniques provides opportunities to explore features unique to each cell type and construct a comprehensive cell-cell communication (CCC) network. Although the most current successes of CCC inference were achieved in animal research, computational methods can also be directly applied to plants. This review describes the current major models for cell-cell communication inference and summarizes the computational tools based on single-cell omics datasets. In addition, we discuss the limitations of plant cell-cell communication research and propose new directions to expand the field in meaningful ways.
Collapse
Affiliation(s)
- Jingjing Jin
- China Tobacco Gene Research Center, Zhengzhou Tobacco Research Institute of CNTC, Zhengzhou 450001, China
| | - Shizhou Yu
- Molecular Genetics Key Laboratory of China Tobacco, Guizhou Academy of Tobacco Science, Guiyang 550081, China
| | - Peng Lu
- China Tobacco Gene Research Center, Zhengzhou Tobacco Research Institute of CNTC, Zhengzhou 450001, China
| | - Peijian Cao
- China Tobacco Gene Research Center, Zhengzhou Tobacco Research Institute of CNTC, Zhengzhou 450001, China
- School of Agricultural Sciences, Zhengzhou University, Zhengzhou 450001, China
| |
Collapse
|
26
|
He C, Zhou P, Nie Q. exFINDER: identify external communication signals using single-cell transcriptomics data. Nucleic Acids Res 2023; 51:e58. [PMID: 37026478 PMCID: PMC10250247 DOI: 10.1093/nar/gkad262] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2022] [Revised: 03/22/2023] [Accepted: 03/28/2023] [Indexed: 04/08/2023] Open
Abstract
Cells make decisions through their communication with other cells and receiving signals from their environment. Using single-cell transcriptomics, computational tools have been developed to infer cell-cell communication through ligands and receptors. However, the existing methods only deal with signals sent by the measured cells in the data, the received signals from the external system are missing in the inference. Here, we present exFINDER, a method that identifies such external signals received by the cells in the single-cell transcriptomics datasets by utilizing the prior knowledge of signaling pathways. In particular, exFINDER can uncover external signals that activate the given target genes, infer the external signal-target signaling network (exSigNet), and perform quantitative analysis on exSigNets. The applications of exFINDER to scRNA-seq datasets from different species demonstrate the accuracy and robustness of identifying external signals, revealing critical transition-related signaling activities, inferring critical external signals and targets, clustering signal-target paths, and evaluating relevant biological events. Overall, exFINDER can be applied to scRNA-seq data to reveal the external signal-associated activities and maybe novel cells that send such signals.
Collapse
Affiliation(s)
- Changhan He
- Department of Mathematics, University of California, Irvine, Irvine, CA 92697, USA
| | - Peijie Zhou
- Department of Mathematics, University of California, Irvine, Irvine, CA 92697, USA
| | - Qing Nie
- Department of Mathematics, University of California, Irvine, Irvine, CA 92697, USA
- Department of Cell and Developmental Biology, University of California, Irvine, Irvine, CA 92697, USA
| |
Collapse
|
27
|
Van de Sande B, Lee JS, Mutasa-Gottgens E, Naughton B, Bacon W, Manning J, Wang Y, Pollard J, Mendez M, Hill J, Kumar N, Cao X, Chen X, Khaladkar M, Wen J, Leach A, Ferran E. Applications of single-cell RNA sequencing in drug discovery and development. Nat Rev Drug Discov 2023; 22:496-520. [PMID: 37117846 PMCID: PMC10141847 DOI: 10.1038/s41573-023-00688-4] [Citation(s) in RCA: 81] [Impact Index Per Article: 40.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/10/2023] [Indexed: 04/30/2023]
Abstract
Single-cell technologies, particularly single-cell RNA sequencing (scRNA-seq) methods, together with associated computational tools and the growing availability of public data resources, are transforming drug discovery and development. New opportunities are emerging in target identification owing to improved disease understanding through cell subtyping, and highly multiplexed functional genomics screens incorporating scRNA-seq are enhancing target credentialling and prioritization. ScRNA-seq is also aiding the selection of relevant preclinical disease models and providing new insights into drug mechanisms of action. In clinical development, scRNA-seq can inform decision-making via improved biomarker identification for patient stratification and more precise monitoring of drug response and disease progression. Here, we illustrate how scRNA-seq methods are being applied in key steps in drug discovery and development, and discuss ongoing challenges for their implementation in the pharmaceutical industry.
Collapse
Affiliation(s)
| | | | | | - Bart Naughton
- Computational Neurobiology, Eisai, Cambridge, MA, USA
| | - Wendi Bacon
- EMBL-EBI, Wellcome Genome Campus, Hinxton, UK
- The Open University, Milton Keynes, UK
| | | | - Yong Wang
- Precision Bioinformatics, Prometheus Biosciences, San Diego, CA, USA
| | | | - Melissa Mendez
- Genomic Sciences, GlaxoSmithKline, Collegeville, PA, USA
| | - Jon Hill
- Global Computational Biology and Digital Sciences, Boehringer Ingelheim Pharmaceuticals Inc., Ridgefield, CT, USA
| | - Namit Kumar
- Informatics & Predictive Sciences, Bristol Myers Squibb, San Diego, CA, USA
| | - Xiaohong Cao
- Genomic Research Center, AbbVie Inc., Cambridge, MA, USA
| | - Xiao Chen
- Magnet Biomedicine, Cambridge, MA, USA
| | - Mugdha Khaladkar
- Human Genetics and Computational Biology, GlaxoSmithKline, Collegeville, PA, USA
| | - Ji Wen
- Oncology Research and Development Unit, Pfizer, La Jolla, CA, USA
| | | | | |
Collapse
|
28
|
Stabell AR, Lee GE, Jia Y, Wong KN, Wang S, Ling J, Nguyen SD, Sen GL, Nie Q, Atwood SX. Single-cell transcriptomics of human-skin-equivalent organoids. Cell Rep 2023; 42:112511. [PMID: 37195865 PMCID: PMC10348600 DOI: 10.1016/j.celrep.2023.112511] [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: 07/11/2022] [Revised: 03/07/2023] [Accepted: 04/28/2023] [Indexed: 05/19/2023] Open
Abstract
Several methods for generating human-skin-equivalent (HSE) organoid cultures are in use to study skin biology; however, few studies thoroughly characterize these systems. To fill this gap, we use single-cell transcriptomics to compare in vitro HSEs, xenograft HSEs, and in vivo epidermis. By combining differential gene expression, pseudotime analyses, and spatial localization, we reconstruct HSE keratinocyte differentiation trajectories that recapitulate known in vivo epidermal differentiation pathways and show that HSEs contain major in vivo cellular states. However, HSEs also develop unique keratinocyte states, an expanded basal stem cell program, and disrupted terminal differentiation. Cell-cell communication modeling shows aberrant epithelial-to-mesenchymal transition (EMT)-associated signaling pathways that alter upon epidermal growth factor (EGF) supplementation. Last, xenograft HSEs at early time points post transplantation significantly rescue many in vitro deficits while undergoing a hypoxic response that drives an alternative differentiation lineage. This study highlights the strengths and limitations of organoid cultures and identifies areas for potential innovation.
Collapse
Affiliation(s)
- Adam R Stabell
- Department of Developmental and Cell Biology, University of California, Irvine, Irvine, CA 92697, USA; NSF-Simons Center for Multiscale Cell Fate Research, University of California, Irvine, Irvine, CA 92697, USA
| | - Grace E Lee
- Department of Developmental and Cell Biology, University of California, Irvine, Irvine, CA 92697, USA
| | - Yunlong Jia
- Department of Developmental and Cell Biology, University of California, Irvine, Irvine, CA 92697, USA
| | - Kirsten N Wong
- Department of Developmental and Cell Biology, University of California, Irvine, Irvine, CA 92697, USA; NSF-Simons Center for Multiscale Cell Fate Research, University of California, Irvine, Irvine, CA 92697, USA
| | - Shuxiong Wang
- Department of Mathematics, University of California, Irvine, Irvine, CA 92697, USA
| | - Ji Ling
- Department of Dermatology, University of California, San Diego, La Jolla, CA 92093, USA
| | - Sandrine D Nguyen
- Department of Developmental and Cell Biology, University of California, Irvine, Irvine, CA 92697, USA
| | - George L Sen
- Department of Dermatology, University of California, San Diego, La Jolla, CA 92093, USA; Department of Cellular and Molecular Medicine, UCSD Stem Cell Program, University of California, San Diego, La Jolla, CA 92093, USA
| | - Qing Nie
- Department of Developmental and Cell Biology, University of California, Irvine, Irvine, CA 92697, USA; NSF-Simons Center for Multiscale Cell Fate Research, University of California, Irvine, Irvine, CA 92697, USA; Department of Mathematics, University of California, Irvine, Irvine, CA 92697, USA; Center for Complex Biological Systems, Chao Family Comprehensive Cancer Center, University of California, Irvine, Irvine, CA 92697, USA
| | - Scott X Atwood
- Department of Developmental and Cell Biology, University of California, Irvine, Irvine, CA 92697, USA; NSF-Simons Center for Multiscale Cell Fate Research, University of California, Irvine, Irvine, CA 92697, USA; Center for Complex Biological Systems, Chao Family Comprehensive Cancer Center, University of California, Irvine, Irvine, CA 92697, USA; Department of Dermatology, University of California, Irvine, Irvine, CA 92697, USA.
| |
Collapse
|
29
|
Rommelfanger MK, Behrends M, Chen Y, Martinez J, Bens M, Xiong L, Rudolph KL, MacLean AL. Gene regulatory network inference with popInfer reveals dynamic regulation of hematopoietic stem cell quiescence upon diet restriction and aging. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.04.18.537360. [PMID: 37131596 PMCID: PMC10153203 DOI: 10.1101/2023.04.18.537360] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/04/2023]
Abstract
Inference of gene regulatory networks (GRNs) can reveal cell state transitions from single-cell genomics data. However, obstacles to temporal inference from snapshot data are difficult to overcome. Single-nuclei multiomics data offer means to bridge this gap and derive temporal information from snapshot data using joint measurements of gene expression and chromatin accessibility in the same single cells. We developed popInfer to infer networks that characterize lineage-specific dynamic cell state transitions from joint gene expression and chromatin accessibility data. Benchmarking against alternative methods for GRN inference, we showed that popInfer achieves higher accuracy in the GRNs inferred. popInfer was applied to study single-cell multiomics data characterizing hematopoietic stem cells (HSCs) and the transition from HSC to a multipotent progenitor cell state during murine hematopoiesis across age and dietary conditions. From networks predicted by popInfer, we discovered gene interactions controlling entry to/exit from HSC quiescence that are perturbed in response to diet or aging.
Collapse
Affiliation(s)
- Megan K. Rommelfanger
- Department of Quantitative and Computational Biology, University of Southern California, Los Angeles, CA 90089, USA
| | - Marthe Behrends
- Research Group on Stem Cell and Metabolism Aging, Leibniz Institute on Aging, Fritz Lipmann Institute (FLI), Jena, Germany
| | - Yulin Chen
- Research Group on Stem Cell and Metabolism Aging, Leibniz Institute on Aging, Fritz Lipmann Institute (FLI), Jena, Germany
| | - Jonathan Martinez
- Department of Quantitative and Computational Biology, University of Southern California, Los Angeles, CA 90089, USA
| | - Martin Bens
- Core Facility Next Generation Sequencing, Leibniz Institute on Aging, Fritz Lipmann Institute (FLI), Jena, Germany
| | - Lingyun Xiong
- Department of Quantitative and Computational Biology, University of Southern California, Los Angeles, CA 90089, USA
- Department of Stem Cell Biology and Regenerative Medicine, Broad-CIRM Center, Keck School of Medicine, University of Southern California, Los Angeles, CA 90089, USA
| | - K. Lenhard Rudolph
- Research Group on Stem Cell and Metabolism Aging, Leibniz Institute on Aging, Fritz Lipmann Institute (FLI), Jena, Germany
- Medical Faculty, Jena University Hospital, Friedrich Schiller University, Jena, Germany
| | - Adam L. MacLean
- Department of Quantitative and Computational Biology, University of Southern California, Los Angeles, CA 90089, USA
| |
Collapse
|
30
|
Yang Y, Li G, Zhong Y, Xu Q, Lin YT, Roman-Vicharra C, Chapkin RS, Cai JJ. scTenifoldXct: A semi-supervised method for predicting cell-cell interactions and mapping cellular communication graphs. Cell Syst 2023; 14:302-311.e4. [PMID: 36787742 PMCID: PMC10121998 DOI: 10.1016/j.cels.2023.01.004] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2022] [Revised: 08/22/2022] [Accepted: 01/20/2023] [Indexed: 02/16/2023]
Abstract
We present scTenifoldXct, a semi-supervised computational tool for detecting ligand-receptor (LR)-mediated cell-cell interactions and mapping cellular communication graphs. Our method is based on manifold alignment, using LR pairs as inter-data correspondences to embed ligand and receptor genes expressed in interacting cells into a unified latent space. Neural networks are employed to minimize the distance between corresponding genes while preserving the structure of gene regression networks. We apply scTenifoldXct to real datasets for testing and demonstrate that our method detects interactions with high consistency compared with other methods. More importantly, scTenifoldXct uncovers weak but biologically relevant interactions overlooked by other methods. We also demonstrate how scTenifoldXct can be used to compare different samples, such as healthy vs. diseased and wild type vs. knockout, to identify differential interactions, thereby revealing functional implications associated with changes in cellular communication status.
Collapse
Affiliation(s)
- Yongjian Yang
- Department of Electrical and Computer Engineering, Texas A&M University, College Station, TX 77843, USA
| | - Guanxun Li
- Department of Statistics, Texas A&M University, College Station, TX 77843, USA
| | - Yan Zhong
- Key Laboratory of Advanced Theory and Application in Statistics and Data Science-MOE, School of Statistics, East China Normal University, 3663 North Zhongshan Road, Shanghai 200062, China
| | - Qian Xu
- Department of Veterinary Integrative Biosciences, Texas A&M University, College Station, TX 77843, USA
| | - Yu-Te Lin
- Graduate Institute of Biomedical Electronics and Bioinformatics, National Taiwan University, Taipei, Taiwan
| | - Cristhian Roman-Vicharra
- Department of Veterinary Integrative Biosciences, Texas A&M University, College Station, TX 77843, USA
| | - Robert S Chapkin
- Department of Nutrition and the Program in Integrative Nutrition & Complex Diseases, Texas A&M University, College Station, TX 77843, USA.
| | - James J Cai
- Department of Electrical and Computer Engineering, Texas A&M University, College Station, TX 77843, USA; Department of Veterinary Integrative Biosciences, Texas A&M University, College Station, TX 77843, USA; Interdisciplinary Program of Genetics, Texas A&M University, College Station, TX 77843, USA.
| |
Collapse
|
31
|
He C, Zhou P, Nie Q. exFINDER: identify external communication signals using single-cell transcriptomics data. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.03.24.533888. [PMID: 37034624 PMCID: PMC10081188 DOI: 10.1101/2023.03.24.533888] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/15/2023]
Abstract
Cells make decisions through their communication with other cells and receiving signals from their environment. Using single-cell transcriptomics, computational tools have been developed to infer cell-cell communication through ligands and receptors. However, the existing methods only deal with signals sent by the measured cells in the data, the received signals from the external system are missing in the inference. Here, we present exFINDER, a method that identifies such external signals received by the cells in the single-cell transcriptomics datasets by utilizing the prior knowledge of signaling pathways. In particular, exFINDER can uncover external signals that activate the given target genes, infer the external signal-target signaling network (exSigNet), and perform quantitative analysis on exSigNets. The applications of exFINDER to scRNA-seq datasets from different species demonstrate the accuracy and robustness of identifying external signals, revealing critical transition-related signaling activities, inferring critical external signals and targets, clustering signal-target paths, and evaluating relevant biological events. Overall, exFINDER can be applied to scRNA-seq data to reveal the external signal-associated activities and maybe novel cells that send such signals.
Collapse
Affiliation(s)
- Changhan He
- Department of Mathematics, University of California, Irvine, Irvine, CA 92697, USA
| | - Peijie Zhou
- Department of Mathematics, University of California, Irvine, Irvine, CA 92697, USA
| | - Qing Nie
- Department of Mathematics, University of California, Irvine, Irvine, CA 92697, USA
- Department of Cell and Developmental Biology, University of California, Irvine, Irvine, CA 92697, USA
| |
Collapse
|
32
|
Vahid MR, Kurlovs AH, Andreani T, Augé F, Olfati-Saber R, de Rinaldis E, Rapaport F, Savova V. DiSiR: fast and robust method to identify ligand-receptor interactions at subunit level from single-cell RNA-sequencing data. NAR Genom Bioinform 2023; 5:lqad030. [PMID: 36968431 PMCID: PMC10034587 DOI: 10.1093/nargab/lqad030] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2022] [Revised: 01/29/2023] [Accepted: 03/09/2023] [Indexed: 03/25/2023] Open
Abstract
Most cell-cell interactions and crosstalks are mediated by ligand-receptor interactions. The advent of single-cell RNA-sequencing (scRNA-seq) techniques has enabled characterizing tissue heterogeneity at single-cell level. In the past few years, several methods have been developed to study ligand-receptor interactions at cell type level using scRNA-seq data. However, there is still no easy way to query the activity of a specific user-defined signaling pathway in a targeted way or to map the interactions of the same subunit with different ligands as part of different receptor complexes. Here, we present DiSiR, a fast and easy-to-use permutation-based software framework to investigate how individual cells are interacting with each other by analyzing signaling pathways of multi-subunit ligand-activated receptors from scRNA-seq data, not only for available curated databases of ligand-receptor interactions, but also for interactions that are not listed in these databases. We show that, when utilized to infer ligand-receptor interactions from both simulated and real datasets, DiSiR outperforms other well-known permutation-based methods, e.g. CellPhoneDB and ICELLNET. Finally, to demonstrate DiSiR's utility in exploring data and generating biologically relevant hypotheses, we apply it to COVID lung and rheumatoid arthritis (RA) synovium scRNA-seq datasets and highlight potential differences between inflammatory pathways at cell type level for control versus disease samples.
Collapse
Affiliation(s)
- Milad R Vahid
- Sanofi R&D Data and Data Science, Artificial Intelligence & Deep Analytics, Omics Data Science, 450 Water Street, Cambridge, MA 02142, USA
| | - Andre H Kurlovs
- Sanofi R&D, Precision Medicine and Computational Biology, 350 Water Street, Cambridge, MA 02142, USA
| | - Tommaso Andreani
- Sanofi R&D Data and Data Science, Artificial Intelligence & Deep Analytics, Omics Data Science, Frankfurt am Main 65926, Germany
| | - Franck Augé
- Sanofi R&D Data and Data Science, Artificial Intelligence & Deep Analytics, Omics Data Science, 1, Av Pierre Brossolette 91385, Chilly-Mazarin, France
| | - Reza Olfati-Saber
- Sanofi R&D Data and Data Science, Artificial Intelligence & Deep Analytics, Omics Data Science, 450 Water Street, Cambridge, MA 02142, USA
| | - Emanuele de Rinaldis
- Sanofi R&D, Precision Medicine and Computational Biology, 350 Water Street, Cambridge, MA 02142, USA
| | - Franck Rapaport
- Sanofi R&D Data and Data Science, Artificial Intelligence & Deep Analytics, Omics Data Science, 450 Water Street, Cambridge, MA 02142, USA
- Sanofi R&D, Precision Medicine and Computational Biology, 350 Water Street, Cambridge, MA 02142, USA
| | - Virginia Savova
- Sanofi R&D, Precision Medicine and Computational Biology, 350 Water Street, Cambridge, MA 02142, USA
| |
Collapse
|
33
|
Zhao W, Johnston KG, Ren H, Xu X, Nie Q. Inferring neuron-neuron communications from single-cell transcriptomics through NeuronChat. Nat Commun 2023; 14:1128. [PMID: 36854676 PMCID: PMC9974942 DOI: 10.1038/s41467-023-36800-w] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2022] [Accepted: 02/15/2023] [Indexed: 03/02/2023] Open
Abstract
Neural communication networks form the fundamental basis for brain function. These communication networks are enabled by emitted ligands such as neurotransmitters, which activate receptor complexes to facilitate communication. Thus, neural communication is fundamentally dependent on the transcriptome. Here we develop NeuronChat, a method and package for the inference, visualization and analysis of neural-specific communication networks among pre-defined cell groups using single-cell expression data. We incorporate a manually curated molecular interaction database of neural signaling for both human and mouse, and benchmark NeuronChat on several published datasets to validate its ability in predicting neural connectivity. Then, we apply NeuronChat to three different neural tissue datasets to illustrate its functionalities in identifying interneural communication networks, revealing conserved or context-specific interactions across different biological contexts, and predicting communication pattern changes in diseased brains with autism spectrum disorder. Finally, we demonstrate NeuronChat can utilize spatial transcriptomics data to infer and visualize neural-specific cell-cell communication.
Collapse
Affiliation(s)
- Wei Zhao
- Department of Mathematics and the NSF-Simons Center for Multiscale Cell Fate Research, University of California, Irvine, CA, 92697, USA
| | - Kevin G Johnston
- Department of Mathematics and the NSF-Simons Center for Multiscale Cell Fate Research, University of California, Irvine, CA, 92697, USA
| | - Honglei Ren
- Department of Mathematics and the NSF-Simons Center for Multiscale Cell Fate Research, University of California, Irvine, CA, 92697, USA
| | - Xiangmin Xu
- Department of Anatomy and Neurobiology, School of Medicine, University of California, Irvine, CA, 92697, USA
- Department of Biomedical Engineering, University of California, Irvine, CA, 92697, USA
- Department of Computer Science, University of California, Irvine, CA, 92697, USA
- The Center for Neural Circuit Mapping, University of California, Irvine, CA, 92697, USA
| | - Qing Nie
- Department of Mathematics and the NSF-Simons Center for Multiscale Cell Fate Research, University of California, Irvine, CA, 92697, USA.
- Department of Biomedical Engineering, University of California, Irvine, CA, 92697, USA.
- The Center for Neural Circuit Mapping, University of California, Irvine, CA, 92697, USA.
- Department of Developmental and Cell Biology, University of California, Irvine, CA, 92697, USA.
| |
Collapse
|
34
|
Subedi S, Park YP. Single-cell pair-wise relationships untangled by composite embedding model. iScience 2023; 26:106025. [PMID: 36824286 PMCID: PMC9941206 DOI: 10.1016/j.isci.2023.106025] [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: 09/26/2022] [Revised: 11/24/2022] [Accepted: 01/17/2023] [Indexed: 01/25/2023] Open
Abstract
In multicellular organisms, cell identity and functions are primed and refined through interactions with other surrounding cells. Here, we propose a scalable machine learning method, termed SPRUCE, which is designed to systematically ascertain common cell-cell communication patterns embedded in single-cell RNA-seq data. We applied our approach to investigate tumor microenvironments consolidating multiple breast cancer datasets and found seven frequently observed interaction signatures and underlying gene-gene interaction networks. Our results implicate that a part of tumor heterogeneity, especially within the same subtype, is better understood by differential interaction patterns rather than the static expression of known marker genes.
Collapse
Affiliation(s)
- Sishir Subedi
- Bioinformatics Graduate Program, University of British Columbia, Vancouver, BC, Canada
- BC Cancer Research, Part of Provincial Health Care Authority, Vancouver, BC, Canada
| | - Yongjin P. Park
- BC Cancer Research, Part of Provincial Health Care Authority, Vancouver, BC, Canada
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC, Canada
- Department of Statistics, University of British Columbia, Vancouver, BC, Canada
| |
Collapse
|
35
|
Cang Z, Zhao Y, Almet AA, Stabell A, Ramos R, Plikus MV, Atwood SX, Nie Q. Screening cell-cell communication in spatial transcriptomics via collective optimal transport. Nat Methods 2023; 20:218-228. [PMID: 36690742 PMCID: PMC9911355 DOI: 10.1038/s41592-022-01728-4] [Citation(s) in RCA: 77] [Impact Index Per Article: 38.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2021] [Accepted: 11/21/2022] [Indexed: 01/24/2023]
Abstract
Spatial transcriptomic technologies and spatially annotated single-cell RNA sequencing datasets provide unprecedented opportunities to dissect cell-cell communication (CCC). However, incorporation of the spatial information and complex biochemical processes required in the reconstruction of CCC remains a major challenge. Here, we present COMMOT (COMMunication analysis by Optimal Transport) to infer CCC in spatial transcriptomics, which accounts for the competition between different ligand and receptor species as well as spatial distances between cells. A collective optimal transport method is developed to handle complex molecular interactions and spatial constraints. Furthermore, we introduce downstream analysis tools to infer spatial signaling directionality and genes regulated by signaling using machine learning models. We apply COMMOT to simulation data and eight spatial datasets acquired with five different technologies to show its effectiveness and robustness in identifying spatial CCC in data with varying spatial resolutions and gene coverages. Finally, COMMOT identifies new CCCs during skin morphogenesis in a case study of human epidermal development.
Collapse
Affiliation(s)
- Zixuan Cang
- Department of Mathematics and Center for Research in Scientific Computation, North Carolina State University, Raleigh, NC, USA
| | - Yanxiang Zhao
- Department of Mathematics, The George Washington University, Washington, DC, USA
| | - Axel A Almet
- Department of Mathematics, University of California, Irvine, Irvine, CA, USA
- The NSF-Simons Center for Multiscale Cell Fate Research, University of California, Irvine, Irvine, CA, USA
| | - Adam Stabell
- The NSF-Simons Center for Multiscale Cell Fate Research, University of California, Irvine, Irvine, CA, USA
- Department of Developmental and Cell Biology, University of California, Irvine, Irvine, CA, USA
| | - Raul Ramos
- The NSF-Simons Center for Multiscale Cell Fate Research, University of California, Irvine, Irvine, CA, USA
- Department of Developmental and Cell Biology, University of California, Irvine, Irvine, CA, USA
| | - Maksim V Plikus
- The NSF-Simons Center for Multiscale Cell Fate Research, University of California, Irvine, Irvine, CA, USA
- Department of Developmental and Cell Biology, University of California, Irvine, Irvine, CA, USA
| | - Scott X Atwood
- The NSF-Simons Center for Multiscale Cell Fate Research, University of California, Irvine, Irvine, CA, USA
- Department of Developmental and Cell Biology, University of California, Irvine, Irvine, CA, USA
| | - Qing Nie
- Department of Mathematics, University of California, Irvine, Irvine, CA, USA.
- The NSF-Simons Center for Multiscale Cell Fate Research, University of California, Irvine, Irvine, CA, USA.
- Department of Developmental and Cell Biology, University of California, Irvine, Irvine, CA, USA.
| |
Collapse
|
36
|
Zhao W, Johnston KG, Ren H, Xu X, Nie Q. Inferring neuron-neuron communications from single-cell transcriptomics through NeuronChat. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.01.12.523826. [PMID: 36712056 PMCID: PMC9882151 DOI: 10.1101/2023.01.12.523826] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
Neural communication networks form the fundamental basis for brain function. These communication networks are enabled by emitted ligands such as neurotransmitters, which activate receptor complexes to facilitate communication. Thus, neural communication is fundamentally dependent on the transcriptome. Here we develop NeuronChat, a method and package for the inference, visualization and analysis of neural-specific communication networks among pre-defined cell groups using single-cell expression data. We incorporate a manually curated molecular interaction database of neural signaling for both human and mouse, and benchmark NeuronChat on several published datasets to validate its ability in predicting neural connectivity. Then, we apply NeuronChat to three different neural tissue datasets to illustrate its functionalities in identifying interneural communication networks, revealing conserved or context-specific interactions across different biological contexts, and predicting communication pattern changes in diseased brains with autism spectrum disorder. Finally, we demonstrate NeuronChat can utilize spatial transcriptomics data to infer and visualize neural-specific cell-cell communication.
Collapse
Affiliation(s)
- Wei Zhao
- Department of Mathematics and the NSF-Simons Center for Multiscale Cell Fate Research, University of California, Irvine, CA 92697
| | - Kevin G. Johnston
- Department of Mathematics and the NSF-Simons Center for Multiscale Cell Fate Research, University of California, Irvine, CA 92697
| | - Honglei Ren
- Department of Mathematics and the NSF-Simons Center for Multiscale Cell Fate Research, University of California, Irvine, CA 92697
| | - Xiangmin Xu
- Department of Anatomy and Neurobiology, School of Medicine, University of California, Irvine, CA 92697
- Department of Biomedical Engineering, University of California, Irvine, CA 92697
- Department of Computer Science, University of California, Irvine, CA 92697
- The Center for the Neurobiology of Learning and Memory, University of California, Irvine, CA 92697
- The Center for Neural Circuit Mapping, University of California, Irvine, CA 92697
| | - Qing Nie
- Department of Mathematics and the NSF-Simons Center for Multiscale Cell Fate Research, University of California, Irvine, CA 92697
- Department of Developmental and Cell Biology, University of California, Irvine, CA 92697
- Department of Biomedical Engineering, University of California, Irvine, CA 92697
- The Center for Neural Circuit Mapping, University of California, Irvine, CA 92697
| |
Collapse
|
37
|
He GJ, Galvis J, Cheung TH, Le Grand F. Single-Cell Transcriptomic Analysis of Mononuclear Cell Populations in Skeletal Muscle. Methods Mol Biol 2023; 2640:369-395. [PMID: 36995608 DOI: 10.1007/978-1-0716-3036-5_26] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/31/2023]
Abstract
Skeletal muscle possesses a remarkable regenerative capacity, mainly relying on a population of undifferentiated and unipotent muscle progenitors, called muscle stem cells (MuSCs) or satellite cells, and their interplay with various cell types within the niche. Investigating the cellular composition of skeletal muscle tissues and the heterogeneity among various cell populations is crucial to the unbiased understanding of how cellular networks work in harmony at the population level in the context of skeletal muscle homeostasis, regeneration, aging, and diseases. As opposed to probing the average profile in a cell population, single-cell RNA-seq has unlocked access to the transcriptomic landscape characterization of individual cells in a highly parallel manner. This chapter describes the workflow for single-cell transcriptomic analysis of mononuclear cells in skeletal muscle by taking advantage of the droplet-based single-cell RNA-seq platform, Chromium Single Cell 3' solution from 10x Genomics®. Using this protocol, we can reveal insights into muscle-resident cell-type identities, which can be exploited to study the muscle stem cell niche further.
Collapse
Affiliation(s)
- Gary J He
- Division of Life Science, Center for Stem Cell Research, HKUST-Nan Fung Life Sciences Joint Laboratory, State Key Laboratory of Molecular Neuroscience, Molecular Neuroscience Center, Hong Kong University of Science and Technology, Kowloon, Hong Kong, China
| | - Johanna Galvis
- Division of Life Science, Center for Stem Cell Research, HKUST-Nan Fung Life Sciences Joint Laboratory, State Key Laboratory of Molecular Neuroscience, Molecular Neuroscience Center, Hong Kong University of Science and Technology, Kowloon, Hong Kong, China
| | - Tom H Cheung
- Division of Life Science, Center for Stem Cell Research, HKUST-Nan Fung Life Sciences Joint Laboratory, State Key Laboratory of Molecular Neuroscience, Molecular Neuroscience Center, Hong Kong University of Science and Technology, Kowloon, Hong Kong, China.
| | - Fabien Le Grand
- Institut NeuroMyoGène, Université Claude Bernard Lyon 1, Lyon, France.
| |
Collapse
|
38
|
Su M, Pan T, Chen QZ, Zhou WW, Gong Y, Xu G, Yan HY, Li S, Shi QZ, Zhang Y, He X, Jiang CJ, Fan SC, Li X, Cairns MJ, Wang X, Li YS. Data analysis guidelines for single-cell RNA-seq in biomedical studies and clinical applications. Mil Med Res 2022; 9:68. [PMID: 36461064 PMCID: PMC9716519 DOI: 10.1186/s40779-022-00434-8] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/27/2022] [Accepted: 11/18/2022] [Indexed: 12/03/2022] Open
Abstract
The application of single-cell RNA sequencing (scRNA-seq) in biomedical research has advanced our understanding of the pathogenesis of disease and provided valuable insights into new diagnostic and therapeutic strategies. With the expansion of capacity for high-throughput scRNA-seq, including clinical samples, the analysis of these huge volumes of data has become a daunting prospect for researchers entering this field. Here, we review the workflow for typical scRNA-seq data analysis, covering raw data processing and quality control, basic data analysis applicable for almost all scRNA-seq data sets, and advanced data analysis that should be tailored to specific scientific questions. While summarizing the current methods for each analysis step, we also provide an online repository of software and wrapped-up scripts to support the implementation. Recommendations and caveats are pointed out for some specific analysis tasks and approaches. We hope this resource will be helpful to researchers engaging with scRNA-seq, in particular for emerging clinical applications.
Collapse
Affiliation(s)
- Min Su
- State Key Laboratory of Reproductive Medicine, Nanjing Medical University, Nanjing, 211166 China
| | - Tao Pan
- College of Biomedical Information and Engineering, the First Affiliated Hospital of Hainan Medical University, Hainan Medical University, Haikou, 571199 Hainan China
| | - Qiu-Zhen Chen
- State Key Laboratory of Reproductive Medicine, Nanjing Medical University, Nanjing, 211166 China
| | - Wei-Wei Zhou
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, 150081 Heilongjiang China
| | - Yi Gong
- State Key Laboratory of Reproductive Medicine, Nanjing Medical University, Nanjing, 211166 China
- Department of Immunology, Nanjing Medical University, Nanjing, 211166 China
| | - Gang Xu
- College of Biomedical Information and Engineering, the First Affiliated Hospital of Hainan Medical University, Hainan Medical University, Haikou, 571199 Hainan China
| | - Huan-Yu Yan
- State Key Laboratory of Reproductive Medicine, Nanjing Medical University, Nanjing, 211166 China
| | - Si Li
- College of Biomedical Information and Engineering, the First Affiliated Hospital of Hainan Medical University, Hainan Medical University, Haikou, 571199 Hainan China
| | - Qiao-Zhen Shi
- State Key Laboratory of Reproductive Medicine, Nanjing Medical University, Nanjing, 211166 China
| | - Ya Zhang
- College of Biomedical Information and Engineering, the First Affiliated Hospital of Hainan Medical University, Hainan Medical University, Haikou, 571199 Hainan China
| | - Xiao He
- Department of Laboratory Medicine, Women and Children’s Hospital of Chongqing Medical University, Chongqing, 401174 China
| | | | - Shi-Cai Fan
- Shenzhen Institute for Advanced Study, University of Electronic Science and Technology of China, Shenzhen, 518110 Guangdong China
| | - Xia Li
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, 150081 Heilongjiang China
| | - Murray J. Cairns
- School of Biomedical Sciences and Pharmacy, Faculty of Health and Medicine, the University of Newcastle, University Drive, Callaghan, NSW 2308 Australia
- Precision Medicine Research Program, Hunter Medical Research Institute, New Lambton Heights, NSW 2305 Australia
| | - Xi Wang
- State Key Laboratory of Reproductive Medicine, Nanjing Medical University, Nanjing, 211166 China
| | - Yong-Sheng Li
- College of Biomedical Information and Engineering, the First Affiliated Hospital of Hainan Medical University, Hainan Medical University, Haikou, 571199 Hainan China
| |
Collapse
|
39
|
Lin X, Gao L, Whitener N, Ahmed A, Wei Z. A model-based constrained deep learning clustering approach for spatially resolved single-cell data. Genome Res 2022; 32:1906-1917. [PMID: 36198490 PMCID: PMC9712636 DOI: 10.1101/gr.276477.121] [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: 12/08/2021] [Accepted: 09/28/2022] [Indexed: 11/25/2022]
Abstract
Spatially resolved scRNA-seq (sp-scRNA-seq) technologies provide the potential to comprehensively profile gene expression patterns in tissue context. However, the development of computational methods lags behind the advances in these technologies, which limits the fulfillment of their potential. In this study, we develop a deep learning approach for clustering sp-scRNA-seq data, named Deep Spatially constrained Single-cell Clustering (DSSC). In this model, we integrate the spatial information of cells into the clustering process in two steps: (1) the spatial information is encoded by using a graphical neural network model, and (2) cell-to-cell constraints are built based on the spatial expression pattern of the marker genes and added in the model to guide the clustering process. Then, a deep embedding clustering is performed on the bottleneck layer of autoencoder by Kullback-Leibler (KL) divergence along with the learning of feature representation. DSSC is the first model that can use information from both spatial coordinates and marker genes to guide cell/spot clustering. Extensive experiments on both simulated and real data sets show that DSSC boosts clustering performance significantly compared with the state-of-the-art methods. It has robust performance across different data sets with various cell type/tissue organization and/or cell type/tissue spatial dependency. We conclude that DSSC is a promising tool for clustering sp-scRNA-seq data.
Collapse
Affiliation(s)
- Xiang Lin
- Department of Computer Science, New Jersey Institute of Technology, Newark, New Jersey 07102, USA
| | - Le Gao
- Department of Computer Science, New Jersey Institute of Technology, Newark, New Jersey 07102, USA
| | - Nathan Whitener
- Department of Computer Science, Wake Forest University, Winston-Salem, North Carolina 27109, USA
| | - Ashley Ahmed
- Department of Chemistry and Chemical Biology and Biological Sciences, College of Arts and Sciences, Cornell University, Ithaca, New York 14853, USA
| | - Zhi Wei
- Department of Computer Science, New Jersey Institute of Technology, Newark, New Jersey 07102, USA
| |
Collapse
|
40
|
Subramanian A, Zakeri P, Mousa M, Alnaqbi H, Alshamsi FY, Bettoni L, Damiani E, Alsafar H, Saeys Y, Carmeliet P. Angiogenesis goes computational - The future way forward to discover new angiogenic targets? Comput Struct Biotechnol J 2022; 20:5235-5255. [PMID: 36187917 PMCID: PMC9508490 DOI: 10.1016/j.csbj.2022.09.019] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2022] [Revised: 09/09/2022] [Accepted: 09/09/2022] [Indexed: 11/26/2022] Open
Abstract
Multi-omics technologies are being increasingly utilized in angiogenesis research. Yet, computational methods have not been widely used for angiogenic target discovery and prioritization in this field, partly because (wet-lab) vascular biologists are insufficiently familiar with computational biology tools and the opportunities they may offer. With this review, written for vascular biologists who lack expertise in computational methods, we aspire to break boundaries between both fields and to illustrate the potential of these tools for future angiogenic target discovery. We provide a comprehensive survey of currently available computational approaches that may be useful in prioritizing candidate genes, predicting associated mechanisms, and identifying their specificity to endothelial cell subtypes. We specifically highlight tools that use flexible, machine learning frameworks for large-scale data integration and gene prioritization. For each purpose-oriented category of tools, we describe underlying conceptual principles, highlight interesting applications and discuss limitations. Finally, we will discuss challenges and recommend some guidelines which can help to optimize the process of accurate target discovery.
Collapse
Affiliation(s)
- Abhishek Subramanian
- Laboratory of Angiogenesis & Vascular Metabolism, Center for Cancer Biology, VIB, Leuven, Belgium
- Laboratory of Angiogenesis & Vascular Metabolism, Department of Oncology, KU Leuven, Leuven, Belgium
| | - Pooya Zakeri
- Laboratory of Angiogenesis & Vascular Heterogeneity, Department of Biomedicine, Aarhus University, Aarhus, Denmark
- Centre for Brain and Disease Research, Flanders Institute for Biotechnology (VIB), Leuven, Belgium
- Department of Neurosciences and Leuven Brain Institute, KU Leuven, Leuven, Belgium
| | - Mira Mousa
- Center for Biotechnology, Khalifa University of Science and Technology, Abu Dhabi, United Arab Emirates
| | - Halima Alnaqbi
- Center for Biotechnology, Khalifa University of Science and Technology, Abu Dhabi, United Arab Emirates
| | - Fatima Yousif Alshamsi
- Center for Biotechnology, Khalifa University of Science and Technology, Abu Dhabi, United Arab Emirates
- Department of Biomedical Engineering, Khalifa University of Science and Technology, Abu Dhabi, United Arab Emirates
| | - Leo Bettoni
- Laboratory of Angiogenesis & Vascular Metabolism, Center for Cancer Biology, VIB, Leuven, Belgium
- Laboratory of Angiogenesis & Vascular Metabolism, Department of Oncology, KU Leuven, Leuven, Belgium
| | - Ernesto Damiani
- Robotics and Intelligent Systems Institute, Khalifa University, Abu Dhabi, United Arab Emirates
| | - Habiba Alsafar
- Center for Biotechnology, Khalifa University of Science and Technology, Abu Dhabi, United Arab Emirates
- Department of Biomedical Engineering, Khalifa University of Science and Technology, Abu Dhabi, United Arab Emirates
| | - Yvan Saeys
- Data Mining and Modelling for Biomedicine Group, VIB Center for Inflammation Research, Ghent, Belgium
- Department of Applied Mathematics, Computer Science and Statistics, Ghent University, Ghent, Belgium
| | - Peter Carmeliet
- Laboratory of Angiogenesis & Vascular Metabolism, Center for Cancer Biology, VIB, Leuven, Belgium
- Laboratory of Angiogenesis & Vascular Metabolism, Department of Oncology, KU Leuven, Leuven, Belgium
- Laboratory of Angiogenesis & Vascular Heterogeneity, Department of Biomedicine, Aarhus University, Aarhus, Denmark
- Center for Biotechnology, Khalifa University of Science and Technology, Abu Dhabi, United Arab Emirates
| |
Collapse
|
41
|
Ding Q, Yang W, Luo M, Xu C, Xu Z, Pang F, Cai Y, Anashkina AA, Su X, Chen N, Jiang Q. CBLRR: a cauchy-based bounded constraint low-rank representation method to cluster single-cell RNA-seq data. Brief Bioinform 2022; 23:6649282. [DOI: 10.1093/bib/bbac300] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Revised: 06/17/2022] [Accepted: 07/02/2022] [Indexed: 11/14/2022] Open
Abstract
Abstract
The rapid development of single-cel+l RNA sequencing (scRNA-seq) technology provides unprecedented opportunities for exploring biological phenomena at the single-cell level. The discovery of cell types is one of the major applications for researchers to explore the heterogeneity of cells. Some computational methods have been proposed to solve the problem of scRNA-seq data clustering. However, the unavoidable technical noise and notorious dropouts also reduce the accuracy of clustering methods. Here, we propose the cauchy-based bounded constraint low-rank representation (CBLRR), which is a low-rank representation-based method by introducing cauchy loss function (CLF) and bounded nuclear norm regulation, aiming to alleviate the above issue. Specifically, as an effective loss function, the CLF is proven to enhance the robustness of the identification of cell types. Then, we adopt the bounded constraint to ensure the entry values of single-cell data within the restricted interval. Finally, the performance of CBLRR is evaluated on 15 scRNA-seq datasets, and compared with other state-of-the-art methods. The experimental results demonstrate that CBLRR performs accurately and robustly on clustering scRNA-seq data. Furthermore, CBLRR is an effective tool to cluster cells, and provides great potential for downstream analysis of single-cell data. The source code of CBLRR is available online at https://github.com/Ginnay/CBLRR.
Collapse
Affiliation(s)
- Qian Ding
- School of Life Science and Technology, Harbin Institute of Technology , Harbin, Heilongjiang, China
| | - Wenyi Yang
- School of Life Science and Technology, Harbin Institute of Technology , Harbin, Heilongjiang, China
| | - Meng Luo
- School of Life Science and Technology, Harbin Institute of Technology , Harbin, Heilongjiang, China
| | - Chang Xu
- School of Life Science and Technology, Harbin Institute of Technology , Harbin, Heilongjiang, China
| | - Zhaochun Xu
- School of Life Science and Technology, Harbin Institute of Technology , Harbin, Heilongjiang, China
| | - Fenglan Pang
- School of Life Science and Technology, Harbin Institute of Technology , Harbin, Heilongjiang, China
| | - Yideng Cai
- School of Life Science and Technology, Harbin Institute of Technology , Harbin, Heilongjiang, China
| | - Anastasia A Anashkina
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences , Moscow, Russia
| | - Xi Su
- Foshan Maternity & Child Healthcare Hospital, Southern Medical University , Foshan, Guangdong, China
| | - Na Chen
- Department of Hematology, Shandong Provincial Hospital Affiliated to Shandong First Medical University , Jinan, Shandong, China
| | - Qinghua Jiang
- School of Life Science and Technology, Harbin Institute of Technology , Harbin, Heilongjiang, China
| |
Collapse
|
42
|
Armingol E, Baghdassarian HM, Martino C, Perez-Lopez A, Aamodt C, Knight R, Lewis NE. Context-aware deconvolution of cell-cell communication with Tensor-cell2cell. Nat Commun 2022; 13:3665. [PMID: 35760817 PMCID: PMC9237099 DOI: 10.1038/s41467-022-31369-2] [Citation(s) in RCA: 34] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2021] [Accepted: 06/14/2022] [Indexed: 12/23/2022] Open
Abstract
Cell interactions determine phenotypes, and intercellular communication is shaped by cellular contexts such as disease state, organismal life stage, and tissue microenvironment. Single-cell technologies measure the molecules mediating cell-cell communication, and emerging computational tools can exploit these data to decipher intercellular communication. However, current methods either disregard cellular context or rely on simple pairwise comparisons between samples, thus limiting the ability to decipher complex cell-cell communication across multiple time points, levels of disease severity, or spatial contexts. Here we present Tensor-cell2cell, an unsupervised method using tensor decomposition, which deciphers context-driven intercellular communication by simultaneously accounting for multiple stages, states, or locations of the cells. To do so, Tensor-cell2cell uncovers context-driven patterns of communication associated with different phenotypic states and determined by unique combinations of cell types and ligand-receptor pairs. As such, Tensor-cell2cell robustly improves upon and extends the analytical capabilities of existing tools. We show Tensor-cell2cell can identify multiple modules associated with distinct communication processes (e.g., participating cell-cell and ligand-receptor pairs) linked to severities of Coronavirus Disease 2019 and to Autism Spectrum Disorder. Thus, we introduce an effective and easy-to-use strategy for understanding complex communication patterns across diverse conditions.
Collapse
Affiliation(s)
- Erick Armingol
- Bioinformatics and Systems Biology Graduate Program, University of California, San Diego, La Jolla, CA, 92093, USA
- Department of Pediatrics, University of California, San Diego, La Jolla, CA, 92093, USA
| | - Hratch M Baghdassarian
- Bioinformatics and Systems Biology Graduate Program, University of California, San Diego, La Jolla, CA, 92093, USA
- Department of Pediatrics, University of California, San Diego, La Jolla, CA, 92093, USA
| | - Cameron Martino
- Bioinformatics and Systems Biology Graduate Program, University of California, San Diego, La Jolla, CA, 92093, USA
- Department of Pediatrics, University of California, San Diego, La Jolla, CA, 92093, USA
- Center for Microbiome Innovation, University of California San Diego, La Jolla, CA, 92093, USA
| | - Araceli Perez-Lopez
- Biomedicine Research Unit, Facultad de Estudios Superiores Iztacala, Universidad Nacional Autónoma de México, Tlalnepantla, México, 54090, México
| | - Caitlin Aamodt
- Department of Pediatrics, University of California, San Diego, La Jolla, CA, 92093, USA
| | - Rob Knight
- Department of Pediatrics, University of California, San Diego, La Jolla, CA, 92093, USA
- Center for Microbiome Innovation, University of California San Diego, La Jolla, CA, 92093, USA
- Department of Computer Science and Engineering, University of California San Diego, La Jolla, CA, 92093, USA
- Department of Bioengineering, University of California, San Diego, La Jolla, CA, 92093, USA
| | - Nathan E Lewis
- Department of Pediatrics, University of California, San Diego, La Jolla, CA, 92093, USA.
- Department of Bioengineering, University of California, San Diego, La Jolla, CA, 92093, USA.
| |
Collapse
|
43
|
Peng L, Wang F, Wang Z, Tan J, Huang L, Tian X, Liu G, Zhou L. Cell-cell communication inference and analysis in the tumour microenvironments from single-cell transcriptomics: data resources and computational strategies. Brief Bioinform 2022; 23:6618236. [PMID: 35753695 DOI: 10.1093/bib/bbac234] [Citation(s) in RCA: 51] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2022] [Revised: 05/15/2022] [Accepted: 05/19/2022] [Indexed: 12/12/2022] Open
Abstract
Carcinomas are complex ecosystems composed of cancer, stromal and immune cells. Communication between these cells and their microenvironments induces cancer progression and causes therapy resistance. In order to improve the treatment of cancers, it is essential to quantify crosstalk between and within various cell types in a tumour microenvironment. Focusing on the coordinated expression patterns of ligands and cognate receptors, cell-cell communication can be inferred through ligand-receptor interactions (LRIs). In this manuscript, we carry out the following work: (i) introduce pipeline for ligand-receptor-mediated intercellular communication estimation from single-cell transcriptomics and list a few available LRI-related databases and visualization tools; (ii) demonstrate seven classical intercellular communication scoring strategies, highlight four types of representative intercellular communication inference methods, including network-based approaches, machine learning-based approaches, spatial information-based approaches and other approaches; (iii) summarize the evaluation and validation avenues for intercellular communication inference and analyze the advantages and limitations for the above four types of cell-cell communication methods; (iv) comment several major challenges while provide further research directions for intercellular communication analysis in the tumour microenvironments. We anticipate that this work helps to better understand intercellular crosstalk and to further develop powerful cell-cell communication estimation tools for tumor-targeted therapy.
Collapse
Affiliation(s)
- Lihong Peng
- School of Computer Science, Hunan University of Technology, 412007, Hunan, China.,College of Life Sciences and Chemistry, Hunan University of Technology, 412007, Hunan, China
| | - Feixiang Wang
- School of Computer Science, Hunan University of Technology, 412007, Hunan, China
| | - Zhao Wang
- School of Computer Science, Hunan University of Technology, 412007, Hunan, China
| | - Jingwei Tan
- School of Computer Science, Hunan University of Technology, 412007, Hunan, China
| | - Li Huang
- Academy of Arts and Design, Tsinghua University, 10084, Beijing, China.,The Future Laboratory, Tsinghua University, 10084, Beijing, China
| | - Xiongfei Tian
- School of Computer Science, Hunan University of Technology, 412007, Hunan, China
| | - Guangyi Liu
- School of Computer Science, Hunan University of Technology, 412007, Hunan, China
| | - Liqian Zhou
- School of Computer Science, Hunan University of Technology, 412007, Hunan, China
| |
Collapse
|
44
|
Karikomi M, Zhou P, Nie Q. DURIAN: an integrative deconvolution and imputation method for robust signaling analysis of single-cell transcriptomics data. Brief Bioinform 2022; 23:6609525. [PMID: 35709795 PMCID: PMC9294432 DOI: 10.1093/bib/bbac223] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2022] [Revised: 04/29/2022] [Accepted: 05/11/2022] [Indexed: 01/31/2023] Open
Abstract
Single-cell RNA sequencing trades read-depth for dimensionality, often leading to loss of critical signaling gene information that is typically present in bulk data sets. We introduce DURIAN (Deconvolution and mUltitask-Regression-based ImputAtioN), an integrative method for recovery of gene expression in single-cell data. Through systematic benchmarking, we demonstrate the accuracy, robustness and empirical convergence of DURIAN using both synthetic and published data sets. We show that use of DURIAN improves single-cell clustering, low-dimensional embedding, and recovery of intercellular signaling networks. Our study resolves several inconsistent results of cell-cell communication analysis using single-cell or bulk data independently. The method has broad application in biomarker discovery and cell signaling analysis using single-cell transcriptomics data sets.
Collapse
Affiliation(s)
| | - Peijie Zhou
- Corresponding authors: Peijie Zhou, 540P Rowland Hall, University of California Irvine, Irvine CA 92697, USA. Tel: 949-824-5530; Fax: 949-8247993; ; Qing Nie, 540F Rowland Hall, University of California Irvine, Irvine CA 92697, USA. Tel: 949-824-5530; Fax: 949-8247993;
| | - Qing Nie
- Corresponding authors: Peijie Zhou, 540P Rowland Hall, University of California Irvine, Irvine CA 92697, USA. Tel: 949-824-5530; Fax: 949-8247993; ; Qing Nie, 540F Rowland Hall, University of California Irvine, Irvine CA 92697, USA. Tel: 949-824-5530; Fax: 949-8247993;
| |
Collapse
|
45
|
Gan Y, Guo C, Guo W, Xu G, Zou G. Entropy-based inference of transition states and cellular trajectory for single-cell transcriptomics. Brief Bioinform 2022; 23:6607748. [PMID: 35696651 DOI: 10.1093/bib/bbac225] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2022] [Revised: 05/11/2022] [Accepted: 05/12/2022] [Indexed: 11/12/2022] Open
Abstract
The development of single-cell RNA-seq (scRNA-seq) technology allows researchers to characterize the cell types, states and transitions during dynamic biological processes at single-cell resolution. One of the critical tasks is to infer pseudo-time trajectory. However, the existence of transition cells in the intermediate state of complex biological processes poses a challenge for the trajectory inference. Here, we propose a new single-cell trajectory inference method based on transition entropy, named scTite, to identify transitional states and reconstruct cell trajectory from scRNA-seq data. Taking into account the continuity of cellular processes, we introduce a new metric called transition entropy to measure the uncertainty of a cell belonging to different cell clusters, and then identify cell states and transition cells. Specifically, we adopt different strategies to infer the trajectory for the identified cell states and transition cells, and combine them to obtain a detailed cell trajectory. For the identified cell clusters, we utilize the Wasserstein distance based on the probability distribution to calculate distance between clusters, and construct the minimum spanning tree. Meanwhile, we adopt the signaling entropy and partial correlation coefficient to determine transition paths, which contain a group of transition cells with the largest similarity. Then the transitional paths and the MST are combined to infer a refined cell trajectory. We apply scTite to four real scRNA-seq datasets and an integrated dataset, and conduct extensive performance comparison with nine existing trajectory inference methods. The experimental results demonstrate that the proposed method can reconstruct the cell trajectory more accurately than the compared algorithms. The scTite software package is available at https://github.com/dblab2022/scTite.
Collapse
Affiliation(s)
- Yanglan Gan
- School of Computer Science and Technology, Donghua University, 201600, Shanghai, China
| | - Cheng Guo
- School of Computer Science and Technology, Donghua University, 201600, Shanghai, China
| | - Wenjing Guo
- School of Computer Science and Technology, Donghua University, 201600, Shanghai, China
| | - Guangwei Xu
- School of Computer Science and Technology, Donghua University, 201600, Shanghai, China
| | - Guobing Zou
- School of Computer Science and Technology, Shanghai University, 200444, Shanghai, China
| |
Collapse
|
46
|
Dimitrov D, Türei D, Garrido-Rodriguez M, Burmedi PL, Nagai JS, Boys C, Ramirez Flores RO, Kim H, Szalai B, Costa IG, Valdeolivas A, Dugourd A, Saez-Rodriguez J. Comparison of methods and resources for cell-cell communication inference from single-cell RNA-Seq data. Nat Commun 2022; 13:3224. [PMID: 35680885 PMCID: PMC9184522 DOI: 10.1038/s41467-022-30755-0] [Citation(s) in RCA: 187] [Impact Index Per Article: 62.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2021] [Accepted: 05/17/2022] [Indexed: 12/18/2022] Open
Abstract
The growing availability of single-cell data, especially transcriptomics, has sparked an increased interest in the inference of cell-cell communication. Many computational tools were developed for this purpose. Each of them consists of a resource of intercellular interactions prior knowledge and a method to predict potential cell-cell communication events. Yet the impact of the choice of resource and method on the resulting predictions is largely unknown. To shed light on this, we systematically compare 16 cell-cell communication inference resources and 7 methods, plus the consensus between the methods' predictions. Among the resources, we find few unique interactions, a varying degree of overlap, and an uneven coverage of specific pathways and tissue-enriched proteins. We then examine all possible combinations of methods and resources and show that both strongly influence the predicted intercellular interactions. Finally, we assess the agreement of cell-cell communication methods with spatial colocalisation, cytokine activities, and receptor protein abundance and find that predictions are generally coherent with those data modalities. To facilitate the use of the methods and resources described in this work, we provide LIANA, a LIgand-receptor ANalysis frAmework as an open-source interface to all the resources and methods.
Collapse
Affiliation(s)
- Daniel Dimitrov
- Heidelberg University, Faculty of Medicine, and Heidelberg University Hospital, Institute for Computational Biomedicine, BioQuant, Heidelberg, Germany
| | - Dénes Türei
- Heidelberg University, Faculty of Medicine, and Heidelberg University Hospital, Institute for Computational Biomedicine, BioQuant, Heidelberg, Germany
| | - Martin Garrido-Rodriguez
- Heidelberg University, Faculty of Medicine, and Heidelberg University Hospital, Institute for Computational Biomedicine, BioQuant, Heidelberg, Germany
| | - Paul L Burmedi
- Heidelberg University, Faculty of Medicine, and Heidelberg University Hospital, Institute for Computational Biomedicine, BioQuant, Heidelberg, Germany
| | - James S Nagai
- Institute for Computational Genomics, Faculty of Medicine, RWTH Aachen University, Aachen, 52074, Germany
- Joint Research Center for Computational Biomedicine, RWTH Aachen University Hospital, Aachen, Germany
| | - Charlotte Boys
- Heidelberg University, Faculty of Medicine, and Heidelberg University Hospital, Institute for Computational Biomedicine, BioQuant, Heidelberg, Germany
| | - Ricardo O Ramirez Flores
- Heidelberg University, Faculty of Medicine, and Heidelberg University Hospital, Institute for Computational Biomedicine, BioQuant, Heidelberg, Germany
| | - Hyojin Kim
- Heidelberg University, Faculty of Medicine, and Heidelberg University Hospital, Institute for Computational Biomedicine, BioQuant, Heidelberg, Germany
| | - Bence Szalai
- Faculty of Medicine, Department of Physiology, Semmelweis University, Budapest, Hungary
| | - Ivan G Costa
- Institute for Computational Genomics, Faculty of Medicine, RWTH Aachen University, Aachen, 52074, Germany
- Joint Research Center for Computational Biomedicine, RWTH Aachen University Hospital, Aachen, Germany
| | - Alberto Valdeolivas
- Roche Pharma Research and Early Development, Pharmaceutical Sciences, Roche Innovation Center Basel, Basel, Switzerland
| | - Aurélien Dugourd
- Heidelberg University, Faculty of Medicine, and Heidelberg University Hospital, Institute for Computational Biomedicine, BioQuant, Heidelberg, Germany
| | - Julio Saez-Rodriguez
- Heidelberg University, Faculty of Medicine, and Heidelberg University Hospital, Institute for Computational Biomedicine, BioQuant, Heidelberg, Germany.
| |
Collapse
|
47
|
Deng M, Wang Y, Yan Y. Mining cell–cell signaling in single-cell transcriptomics atlases. Curr Opin Cell Biol 2022; 76:102101. [DOI: 10.1016/j.ceb.2022.102101] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2021] [Revised: 03/16/2022] [Accepted: 04/24/2022] [Indexed: 12/22/2022]
|
48
|
Zhang M, Zhai W, Miao J, Cheng X, Luo W, Song W, Wang J, Gao W. Single cell analysis reveals intra-tumour heterogeneity, microenvironment and potential diagnosis markers for clear cell renal cell carcinoma. Clin Transl Med 2022; 12:e713. [PMID: 35605062 PMCID: PMC9126499 DOI: 10.1002/ctm2.713] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2021] [Revised: 12/29/2021] [Accepted: 01/05/2022] [Indexed: 11/12/2022] Open
Affiliation(s)
- Man Zhang
- State Key Laboratory of Oncogenes and Related Genes, Renji‐Med‐X Stem Cell Research Center, Renji Hospital, School of Medicine and School of Biomedical EngineeringShanghai Jiao Tong University, Shanghai, China
- School of Biomedical Engineering and Med‐X Research InstituteShanghai Jiao Tong UniversityShanghaiChina
| | - Wei Zhai
- Department of Urology, Renji Hospital, School of MedicineShanghai Jiao Tong UniversityShanghaiChina
| | - Juju Miao
- State Key Laboratory of Oncogenes and Related Genes, Renji‐Med‐X Stem Cell Research Center, Renji Hospital, School of Medicine and School of Biomedical EngineeringShanghai Jiao Tong University, Shanghai, China
- School of Biomedical Engineering and Med‐X Research InstituteShanghai Jiao Tong UniversityShanghaiChina
- Present address:
School of Life SciencesWestlake UniversityHangzhouChina
| | - Xiaomu Cheng
- State Key Laboratory of Oncogenes and Related Genes, Renji‐Med‐X Stem Cell Research Center, Renji Hospital, School of Medicine and School of Biomedical EngineeringShanghai Jiao Tong University, Shanghai, China
- School of Biomedical Engineering and Med‐X Research InstituteShanghai Jiao Tong UniversityShanghaiChina
| | - Wenqin Luo
- State Key Laboratory of Oncogenes and Related Genes, Renji‐Med‐X Stem Cell Research Center, Renji Hospital, School of Medicine and School of Biomedical EngineeringShanghai Jiao Tong University, Shanghai, China
- School of Biomedical Engineering and Med‐X Research InstituteShanghai Jiao Tong UniversityShanghaiChina
- Present address:
Department of Colorectal SurgeryFudan University Shanghai Cancer CenterShanghaiChina
| | - Weichen Song
- Shanghai Mental Health Center, School of MedicineShanghai Jiao Tong UniversityShanghaiChina
| | - Jia Wang
- Department of Interventional RadiologyFudan University Shanghai Cancer CenterShanghaiChina
| | - Wei‐Qiang Gao
- State Key Laboratory of Oncogenes and Related Genes, Renji‐Med‐X Stem Cell Research Center, Renji Hospital, School of Medicine and School of Biomedical EngineeringShanghai Jiao Tong University, Shanghai, China
- School of Biomedical Engineering and Med‐X Research InstituteShanghai Jiao Tong UniversityShanghaiChina
| |
Collapse
|
49
|
Schonfeldova B, Zec K, Udalova IA. Synovial single-cell heterogeneity, zonation and interactions: a patchwork of effectors in arthritis. Rheumatology (Oxford) 2022; 61:913-925. [PMID: 34559213 PMCID: PMC8889290 DOI: 10.1093/rheumatology/keab721] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2021] [Revised: 08/27/2021] [Accepted: 09/13/2021] [Indexed: 02/07/2023] Open
Abstract
Despite extensive research, there is still no treatment that would lead to remission in all patients with rheumatoid arthritis as our understanding of the affected site, the synovium, is still incomplete. Recently, single-cell technologies helped to decipher the cellular heterogeneity of the synovium; however, certain synovial cell populations, such as endothelial cells or peripheral neurons, remain to be profiled on a single-cell level. Furthermore, associations between certain cellular states and inflammation were found; whether these cells cause the inflammation remains to be answered. Similarly, cellular zonation and interactions between individual effectors in the synovium are yet to be fully determined. A deeper understanding of cell signalling and interactions in the synovium is crucial for a better design of therapeutics with the goal of complete remission in all patients.
Collapse
Affiliation(s)
- Barbora Schonfeldova
- The Kennedy Institute of Rheumatology, Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Science, University of Oxford, Oxford, UK
| | - Kristina Zec
- The Kennedy Institute of Rheumatology, Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Science, University of Oxford, Oxford, UK
| | - Irina A Udalova
- The Kennedy Institute of Rheumatology, Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Science, University of Oxford, Oxford, UK
| |
Collapse
|
50
|
Wang M, Song WM, Ming C, Wang Q, Zhou X, Xu P, Krek A, Yoon Y, Ho L, Orr ME, Yuan GC, Zhang B. Guidelines for bioinformatics of single-cell sequencing data analysis in Alzheimer's disease: review, recommendation, implementation and application. Mol Neurodegener 2022; 17:17. [PMID: 35236372 PMCID: PMC8889402 DOI: 10.1186/s13024-022-00517-z] [Citation(s) in RCA: 49] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2021] [Accepted: 01/18/2022] [Indexed: 12/13/2022] Open
Abstract
Alzheimer's disease (AD) is the most common form of dementia, characterized by progressive cognitive impairment and neurodegeneration. Extensive clinical and genomic studies have revealed biomarkers, risk factors, pathways, and targets of AD in the past decade. However, the exact molecular basis of AD development and progression remains elusive. The emerging single-cell sequencing technology can potentially provide cell-level insights into the disease. Here we systematically review the state-of-the-art bioinformatics approaches to analyze single-cell sequencing data and their applications to AD in 14 major directions, including 1) quality control and normalization, 2) dimension reduction and feature extraction, 3) cell clustering analysis, 4) cell type inference and annotation, 5) differential expression, 6) trajectory inference, 7) copy number variation analysis, 8) integration of single-cell multi-omics, 9) epigenomic analysis, 10) gene network inference, 11) prioritization of cell subpopulations, 12) integrative analysis of human and mouse sc-RNA-seq data, 13) spatial transcriptomics, and 14) comparison of single cell AD mouse model studies and single cell human AD studies. We also address challenges in using human postmortem and mouse tissues and outline future developments in single cell sequencing data analysis. Importantly, we have implemented our recommended workflow for each major analytic direction and applied them to a large single nucleus RNA-sequencing (snRNA-seq) dataset in AD. Key analytic results are reported while the scripts and the data are shared with the research community through GitHub. In summary, this comprehensive review provides insights into various approaches to analyze single cell sequencing data and offers specific guidelines for study design and a variety of analytic directions. The review and the accompanied software tools will serve as a valuable resource for studying cellular and molecular mechanisms of AD, other diseases, or biological systems at the single cell level.
Collapse
Affiliation(s)
- Minghui Wang
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, 1470 Madison Avenue, Room S8-111, New York, NY 10029 USA
- Mount Sinai Center for Transformative Disease Modeling, Icahn School of Medicine at Mount Sinai, 1470 Madison Avenue, Room S8-111, New York, NY 10029 USA
| | - Won-min Song
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, 1470 Madison Avenue, Room S8-111, New York, NY 10029 USA
- Mount Sinai Center for Transformative Disease Modeling, Icahn School of Medicine at Mount Sinai, 1470 Madison Avenue, Room S8-111, New York, NY 10029 USA
| | - Chen Ming
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, 1470 Madison Avenue, Room S8-111, New York, NY 10029 USA
- Mount Sinai Center for Transformative Disease Modeling, Icahn School of Medicine at Mount Sinai, 1470 Madison Avenue, Room S8-111, New York, NY 10029 USA
| | - Qian Wang
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, 1470 Madison Avenue, Room S8-111, New York, NY 10029 USA
- Mount Sinai Center for Transformative Disease Modeling, Icahn School of Medicine at Mount Sinai, 1470 Madison Avenue, Room S8-111, New York, NY 10029 USA
| | - Xianxiao Zhou
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, 1470 Madison Avenue, Room S8-111, New York, NY 10029 USA
- Mount Sinai Center for Transformative Disease Modeling, Icahn School of Medicine at Mount Sinai, 1470 Madison Avenue, Room S8-111, New York, NY 10029 USA
| | - Peng Xu
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, 1470 Madison Avenue, Room S8-111, New York, NY 10029 USA
- Mount Sinai Center for Transformative Disease Modeling, Icahn School of Medicine at Mount Sinai, 1470 Madison Avenue, Room S8-111, New York, NY 10029 USA
| | - Azra Krek
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, 1470 Madison Avenue, Room S8-111, New York, NY 10029 USA
- Institute for Personalized Medicine, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY 10029 USA
| | - Yonejung Yoon
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, 1470 Madison Avenue, Room S8-111, New York, NY 10029 USA
- Mount Sinai Center for Transformative Disease Modeling, Icahn School of Medicine at Mount Sinai, 1470 Madison Avenue, Room S8-111, New York, NY 10029 USA
| | - Lap Ho
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, 1470 Madison Avenue, Room S8-111, New York, NY 10029 USA
- Mount Sinai Center for Transformative Disease Modeling, Icahn School of Medicine at Mount Sinai, 1470 Madison Avenue, Room S8-111, New York, NY 10029 USA
| | - Miranda E. Orr
- Department of Internal Medicine, Section of Gerontology and Geriatric Medicine, Wake Forest School of Medicine, Winston-Salem, North Carolina USA
- Sticht Center for Healthy Aging and Alzheimer’s Prevention, Wake Forest School of Medicine, Winston-Salem, North Carolina USA
| | - Guo-Cheng Yuan
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, 1470 Madison Avenue, Room S8-111, New York, NY 10029 USA
- Institute for Personalized Medicine, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY 10029 USA
| | - Bin Zhang
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, 1470 Madison Avenue, Room S8-111, New York, NY 10029 USA
- Mount Sinai Center for Transformative Disease Modeling, Icahn School of Medicine at Mount Sinai, 1470 Madison Avenue, Room S8-111, New York, NY 10029 USA
- Icahn Institute of Genomics and Multiscale Biology, Icahn School of Medicine at Mount Sinai, 1470 Madison Avenue, Room S8-111, New York, NY 10029 USA
- Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, 1470 Madison Avenue, Room S8-111, New York, NY 10029 USA
| |
Collapse
|