1
|
Gaspard-Boulinc LC, Gortana L, Walter T, Barillot E, Cavalli FMG. Cell-type deconvolution methods for spatial transcriptomics. Nat Rev Genet 2025:10.1038/s41576-025-00845-y. [PMID: 40369312 DOI: 10.1038/s41576-025-00845-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/14/2025] [Indexed: 05/16/2025]
Abstract
Spatial transcriptomics is a powerful method for studying the spatial organization of cells, which is a critical feature in the development, function and evolution of multicellular life. However, sequencing-based spatial transcriptomics has not yet achieved cellular-level resolution, so advanced deconvolution methods are needed to infer cell-type contributions at each location in the data. Recent progress has led to diverse tools for cell-type deconvolution that are helping to describe tissue architectures in health and disease. In this Review, we describe the varied types of cell-type deconvolution methods for spatial transcriptomics, contrast their capabilities and summarize them in a web-based, interactive table to enable more efficient method selection.
Collapse
Affiliation(s)
- Lucie C Gaspard-Boulinc
- Institut Curie, PSL University, Paris, France
- Institut National de la Santé et de la Recherche Médicale (INSERM), U1331, Paris, France
- Mines Paris, PSL University, CBIO - Centre for Computational Biology, Paris, France
| | - Luca Gortana
- Institut Curie, PSL University, Paris, France
- Institut National de la Santé et de la Recherche Médicale (INSERM), U1331, Paris, France
- Mines Paris, PSL University, CBIO - Centre for Computational Biology, Paris, France
| | - Thomas Walter
- Institut Curie, PSL University, Paris, France
- Institut National de la Santé et de la Recherche Médicale (INSERM), U1331, Paris, France
- Mines Paris, PSL University, CBIO - Centre for Computational Biology, Paris, France
| | - Emmanuel Barillot
- Institut Curie, PSL University, Paris, France
- Institut National de la Santé et de la Recherche Médicale (INSERM), U1331, Paris, France
- Mines Paris, PSL University, CBIO - Centre for Computational Biology, Paris, France
| | - Florence M G Cavalli
- Institut Curie, PSL University, Paris, France.
- Institut National de la Santé et de la Recherche Médicale (INSERM), U1331, Paris, France.
- Mines Paris, PSL University, CBIO - Centre for Computational Biology, Paris, France.
| |
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
|
Yao S, Nguyen TD, Lan Y, Yang W, Chen D, Shao Y, Yang Z. MetaPhenotype: A Transferable Meta-Learning Model for Single-Cell Mass Spectrometry-Based Cell Phenotype Prediction Using Limited Number of Cells. Anal Chem 2024; 96:19238-19247. [PMID: 39570119 DOI: 10.1021/acs.analchem.4c02038] [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] [Indexed: 11/22/2024]
Abstract
Single-cell mass spectrometry (SCMS) is an emerging tool for studying cell heterogeneity according to variation of molecular species in single cells. Although it has become increasingly common to employ machine learning models in SCMS data analysis, such as the classification of cell phenotypes, the existing machine learning models often suffer from low adaptability and transferability. In addition, SCMS studies of rare cells can be restricted by limited number of cell samples. To overcome these limitations, we performed SCMS analyses of melanoma cancer cell lines with two phenotypes (i.e., primary and metastatic cells). We then developed a meta-learning-based model, MetaPhenotype, that can be trained using a small amount of SCMS data to accurately classify cells into primary or metastatic phenotypes. Our results show that compared with standard transfer learning models, MetaPhenotype can rapidly predict and achieve a high accuracy of over 90% with fewer new training samples. Overall, our work opens the possibility of accurate cell phenotype classification based on fewer SCMS samples, thus lowering the demand for sample acquisition.
Collapse
Affiliation(s)
- Songyuan Yao
- Department of Chemistry and Biochemistry, University of Oklahoma, Norman, Oklahoma 73019, United States
| | - Tra D Nguyen
- Department of Chemistry and Biochemistry, University of Oklahoma, Norman, Oklahoma 73019, United States
| | - Yunpeng Lan
- Department of Chemistry and Biochemistry, University of Oklahoma, Norman, Oklahoma 73019, United States
| | - Wen Yang
- Stephenson School of Biomedical Engineering, University of Oklahoma, Norman, Oklahoma 73019, United States
| | - Dan Chen
- Department of Chemistry and Biochemistry, University of Oklahoma, Norman, Oklahoma 73019, United States
| | - Yihan Shao
- Department of Chemistry and Biochemistry, University of Oklahoma, Norman, Oklahoma 73019, United States
| | - Zhibo Yang
- Department of Chemistry and Biochemistry, University of Oklahoma, Norman, Oklahoma 73019, United States
| |
Collapse
|
4
|
Liao L, Martin PCN, Kim H, Panahandeh S, Won KJ. Data enhancement in the age of spatial biology. Adv Cancer Res 2024; 163:39-70. [PMID: 39271267 DOI: 10.1016/bs.acr.2024.06.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/15/2024]
Abstract
Unveiling the intricate interplay of cells in their native environment lies at the heart of understanding fundamental biological processes and unraveling disease mechanisms, particularly in complex diseases like cancer. Spatial transcriptomics (ST) offers a revolutionary lens into the spatial organization of gene expression within tissues, empowering researchers to study both cell heterogeneity and microenvironments in health and disease. However, current ST technologies often face limitations in either resolution or the number of genes profiled simultaneously. Integrating ST data with complementary sources, such as single-cell transcriptomics and detailed tissue staining images, presents a powerful solution to overcome these limitations. This review delves into the computational approaches driving the integration of spatial transcriptomics with other data types. By illuminating the key challenges and outlining the current algorithmic solutions, we aim to highlight the immense potential of these methods to revolutionize our understanding of cancer biology.
Collapse
Affiliation(s)
- Linbu Liao
- Biotech Research and Innovation Centre (BRIC), University of Copenhagen, Denmark; Samuel Oschin Cancer Center, Cedars-Sinai Medical Center, Los Angeles, CA, United States
| | - Patrick C N Martin
- Department of Computational Biomedicine, Cedars-Sinai Medical Center, Los Angeles, CA, United States
| | - Hyobin Kim
- Department of Computational Biomedicine, Cedars-Sinai Medical Center, Los Angeles, CA, United States
| | - Sanaz Panahandeh
- Department of Computational Biomedicine, Cedars-Sinai Medical Center, Los Angeles, CA, United States
| | - Kyoung Jae Won
- Department of Computational Biomedicine, Cedars-Sinai Medical Center, Los Angeles, CA, United States.
| |
Collapse
|
5
|
Ma Y, Shi W, Dong Y, Sun Y, Jin Q. Spatial Multi-Omics in Alzheimer's Disease: A Multi-Dimensional Approach to Understanding Pathology and Progression. Curr Issues Mol Biol 2024; 46:4968-4990. [PMID: 38785566 PMCID: PMC11119029 DOI: 10.3390/cimb46050298] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2024] [Revised: 05/14/2024] [Accepted: 05/15/2024] [Indexed: 05/25/2024] Open
Abstract
Alzheimer's Disease (AD) presents a complex neuropathological landscape characterized by hallmark amyloid plaques and neurofibrillary tangles, leading to progressive cognitive decline. Despite extensive research, the molecular intricacies contributing to AD pathogenesis are inadequately understood. While single-cell omics technology holds great promise for application in AD, particularly in deciphering the understanding of different cell types and analyzing rare cell types and transcriptomic expression changes, it is unable to provide spatial distribution information, which is crucial for understanding the pathological processes of AD. In contrast, spatial multi-omics research emerges as a promising and comprehensive approach to analyzing tissue cells, potentially better suited for addressing these issues in AD. This article focuses on the latest advancements in spatial multi-omics technology and compares various techniques. Additionally, we provide an overview of current spatial omics-based research results in AD. These technologies play a crucial role in facilitating new discoveries and advancing translational AD research in the future. Despite challenges such as balancing resolution, increasing throughput, and data analysis, the application of spatial multi-omics holds immense potential in revolutionizing our understanding of human disease processes and identifying new biomarkers and therapeutic targets, thereby potentially contributing to the advancement of AD research.
Collapse
Affiliation(s)
| | | | | | | | - Qiguan Jin
- College of Physical Education, Yangzhou University, Yangzhou 225127, China; (Y.M.); (W.S.); (Y.D.); (Y.S.)
| |
Collapse
|
6
|
Hozumi Y, Tanemura KA, Wei GW. Preprocessing of Single Cell RNA Sequencing Data Using Correlated Clustering and Projection. J Chem Inf Model 2024; 64:2829-2838. [PMID: 37402705 PMCID: PMC11009150 DOI: 10.1021/acs.jcim.3c00674] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/06/2023]
Abstract
Single-cell RNA sequencing (scRNA-seq) is widely used to reveal heterogeneity in cells, which has given us insights into cell-cell communication, cell differentiation, and differential gene expression. However, analyzing scRNA-seq data is a challenge due to sparsity and the large number of genes involved. Therefore, dimensionality reduction and feature selection are important for removing spurious signals and enhancing the downstream analysis. We present Correlated Clustering and Projection (CCP), a new data-domain dimensionality reduction method, for the first time. CCP projects each cluster of similar genes into a supergene defined as the accumulated pairwise nonlinear gene-gene correlations among all cells. Using 14 benchmark data sets, we demonstrate that CCP has significant advantages over classical principal component analysis (PCA) for clustering and/or classification problems with intrinsically high dimensionality. In addition, we introduce the Residue-Similarity index (RSI) as a novel metric for clustering and classification and the R-S plot as a new visualization tool. We show that the RSI correlates with accuracy without requiring the knowledge of the true labels. The R-S plot provides a unique alternative to the uniform manifold approximation and projection (UMAP) and t-distributed stochastic neighbor embedding (t-SNE) for data with a large number of cell types.
Collapse
Affiliation(s)
- Yuta Hozumi
- Department of Mathematics, Michigan State University, East Lansing, Michigan 48824, United States
| | - Kiyoto Aramis Tanemura
- Department of Mathematics, Michigan State University, East Lansing, Michigan 48824, United States
| | - Guo-Wei Wei
- Department of Mathematics, Michigan State University, East Lansing, Michigan 48824, United States
- Department of Electrical and Computer Engineering, Michigan State University, East Lansing, Michigan 48824, United States
- Department of Biochemistry and Molecular Biology, Michigan State University, East Lansing, Michigan 48824, United States
| |
Collapse
|
7
|
Danishuddin, Khan S, Kim JJ. Spatial transcriptomics data and analytical methods: An updated perspective. Drug Discov Today 2024; 29:103889. [PMID: 38244672 DOI: 10.1016/j.drudis.2024.103889] [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: 10/31/2023] [Revised: 01/01/2024] [Accepted: 01/15/2024] [Indexed: 01/22/2024]
Abstract
Spatial transcriptomics (ST) is a newly emerging field that integrates high-resolution imaging and transcriptomic data to enable the high-throughput analysis of the spatial localization of transcripts in diverse biological systems. The rapid progress in this field necessitates the development of innovative computational methods to effectively tackle the distinct challenges posed by the analysis of ST data. These platforms, integrating AI techniques, offer a promising avenue for understanding disease mechanisms and expediting drug discovery. Despite significant advances in the development of ST data analysis techniques, there is an ongoing need to enhance these models for increased biological relevance. In this review, we briefly discuss the ST-related databases and current deep-learning-based models for spatial transcriptome data analyses and highlight their roles and future perspectives in biomedical applications.
Collapse
Affiliation(s)
- Danishuddin
- Department of Biotechnology, Yeungnam University, Gyeongsan, Gyeongbuk 38541, Korea.
| | - Shawez Khan
- National Center for Cancer Immune Therapy (CCIT-DK), Department of Oncology, Copenhagen University Hospital, Herlev, Denmark
| | - Jong Joo Kim
- Department of Biotechnology, Yeungnam University, Gyeongsan, Gyeongbuk 38541, Korea.
| |
Collapse
|
8
|
Li R, Chen X, Yang X. Navigating the landscapes of spatial transcriptomics: How computational methods guide the way. WILEY INTERDISCIPLINARY REVIEWS. RNA 2024; 15:e1839. [PMID: 38527900 DOI: 10.1002/wrna.1839] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/23/2023] [Revised: 02/24/2024] [Accepted: 03/04/2024] [Indexed: 03/27/2024]
Abstract
Spatially resolved transcriptomics has been dramatically transforming biological and medical research in various fields. It enables transcriptome profiling at single-cell, multi-cellular, or sub-cellular resolution, while retaining the information of geometric localizations of cells in complex tissues. The coupling of cell spatial information and its molecular characteristics generates a novel multi-modal high-throughput data source, which poses new challenges for the development of analytical methods for data-mining. Spatial transcriptomic data are often highly complex, noisy, and biased, presenting a series of difficulties, many unresolved, for data analysis and generation of biological insights. In addition, to keep pace with the ever-evolving spatial transcriptomic experimental technologies, the existing analytical theories and tools need to be updated and reformed accordingly. In this review, we provide an overview and discussion of the current computational approaches for mining of spatial transcriptomics data. Future directions and perspectives of methodology design are proposed to stimulate further discussions and advances in new analytical models and algorithms. This article is categorized under: RNA Methods > RNA Analyses in Cells RNA Evolution and Genomics > Computational Analyses of RNA RNA Export and Localization > RNA Localization.
Collapse
Affiliation(s)
- Runze Li
- MOE Key Laboratory of Bioinformatics, Center for Synthetic & Systems Biology, School of Life Sciences, Tsinghua University, Beijing, China
| | - Xu Chen
- MOE Key Laboratory of Bioinformatics, Center for Synthetic & Systems Biology, School of Life Sciences, Tsinghua University, Beijing, China
| | - Xuerui Yang
- MOE Key Laboratory of Bioinformatics, Center for Synthetic & Systems Biology, School of Life Sciences, Tsinghua University, Beijing, China
| |
Collapse
|
9
|
Vahid MR, Brown EL, Steen CB, Zhang W, Jeon HS, Kang M, Gentles AJ, Newman AM. High-resolution alignment of single-cell and spatial transcriptomes with CytoSPACE. Nat Biotechnol 2023; 41:1543-1548. [PMID: 36879008 PMCID: PMC10635828 DOI: 10.1038/s41587-023-01697-9] [Citation(s) in RCA: 70] [Impact Index Per Article: 35.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2022] [Accepted: 01/25/2023] [Indexed: 03/08/2023]
Abstract
Recent studies have emphasized the importance of single-cell spatial biology, yet available assays for spatial transcriptomics have limited gene recovery or low spatial resolution. Here we introduce CytoSPACE, an optimization method for mapping individual cells from a single-cell RNA sequencing atlas to spatial expression profiles. Across diverse platforms and tissue types, we show that CytoSPACE outperforms previous methods with respect to noise tolerance and accuracy, enabling tissue cartography at single-cell resolution.
Collapse
Affiliation(s)
- Milad R Vahid
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University, Stanford, CA, USA
- Department of Biomedical Data Science, Stanford University, Stanford, CA, USA
| | - Erin L Brown
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University, Stanford, CA, USA
- Department of Biomedical Data Science, Stanford University, Stanford, CA, USA
| | - Chloé B Steen
- Department of Biomedical Data Science, Stanford University, Stanford, CA, USA
- Department of Cancer Immunology, Institute for Cancer Research, Oslo University Hospital, Oslo, Norway
- Department of Medical Genetics, Oslo University Hospital, Oslo, Norway
| | - Wubing Zhang
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University, Stanford, CA, USA
- Department of Biomedical Data Science, Stanford University, Stanford, CA, USA
| | - Hyun Soo Jeon
- Department of Computer Science, Stanford University, Stanford, CA, USA
| | - Minji Kang
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University, Stanford, CA, USA
- Department of Biomedical Data Science, Stanford University, Stanford, CA, USA
| | - Andrew J Gentles
- Department of Biomedical Data Science, Stanford University, Stanford, CA, USA
- Stanford Cancer Institute, Stanford University, Stanford, CA, USA
- Department of Pathology, Stanford University, Stanford, CA, USA
- Department of Medicine, Stanford Center for Biomedical Informatics Research, Stanford University, Stanford, CA, USA
| | - Aaron M Newman
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University, Stanford, CA, USA.
- Department of Biomedical Data Science, Stanford University, Stanford, CA, USA.
- Stanford Cancer Institute, Stanford University, Stanford, CA, USA.
| |
Collapse
|
10
|
Erfanian N, Heydari AA, Feriz AM, Iañez P, Derakhshani A, Ghasemigol M, Farahpour M, Razavi SM, Nasseri S, Safarpour H, Sahebkar A. Deep learning applications in single-cell genomics and transcriptomics data analysis. Biomed Pharmacother 2023; 165:115077. [PMID: 37393865 DOI: 10.1016/j.biopha.2023.115077] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2023] [Revised: 06/22/2023] [Accepted: 06/23/2023] [Indexed: 07/04/2023] Open
Abstract
Traditional bulk sequencing methods are limited to measuring the average signal in a group of cells, potentially masking heterogeneity, and rare populations. The single-cell resolution, however, enhances our understanding of complex biological systems and diseases, such as cancer, the immune system, and chronic diseases. However, the single-cell technologies generate massive amounts of data that are often high-dimensional, sparse, and complex, thus making analysis with traditional computational approaches difficult and unfeasible. To tackle these challenges, many are turning to deep learning (DL) methods as potential alternatives to the conventional machine learning (ML) algorithms for single-cell studies. DL is a branch of ML capable of extracting high-level features from raw inputs in multiple stages. Compared to traditional ML, DL models have provided significant improvements across many domains and applications. In this work, we examine DL applications in genomics, transcriptomics, spatial transcriptomics, and multi-omics integration, and address whether DL techniques will prove to be advantageous or if the single-cell omics domain poses unique challenges. Through a systematic literature review, we have found that DL has not yet revolutionized the most pressing challenges of the single-cell omics field. However, using DL models for single-cell omics has shown promising results (in many cases outperforming the previous state-of-the-art models) in data preprocessing and downstream analysis. Although developments of DL algorithms for single-cell omics have generally been gradual, recent advances reveal that DL can offer valuable resources in fast-tracking and advancing research in single-cell.
Collapse
Affiliation(s)
- Nafiseh Erfanian
- Student Research Committee, Birjand University of Medical Sciences, Birjand, Iran
| | - A Ali Heydari
- Department of Applied Mathematics, University of California, Merced, CA, USA; Health Sciences Research Institute, University of California, Merced, CA, USA
| | - Adib Miraki Feriz
- Student Research Committee, Birjand University of Medical Sciences, Birjand, Iran
| | - Pablo Iañez
- Cellular Systems Genomics Group, Josep Carreras Research Institute, Barcelona, Spain
| | - Afshin Derakhshani
- Department of Biochemistry and Molecular Biology, University of Calgary, Calgary, AB, Canada
| | | | - Mohsen Farahpour
- Department of Electronics, Faculty of Electrical and Computer Engineering, University of Birjand, Birjand, Iran
| | - Seyyed Mohammad Razavi
- Department of Electronics, Faculty of Electrical and Computer Engineering, University of Birjand, Birjand, Iran
| | - Saeed Nasseri
- Cellular and Molecular Research Center, Birjand University of Medical Sciences, Birjand, Iran
| | - Hossein Safarpour
- Cellular and Molecular Research Center, Birjand University of Medical Sciences, Birjand, Iran.
| | - Amirhossein Sahebkar
- Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran; Applied Biomedical Research Center, Mashhad University of Medical Sciences, Mashhad, Iran; Department of Biotechnology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
11
|
Zidane M, Makky A, Bruhns M, Rochwarger A, Babaei S, Claassen M, Schürch CM. A review on deep learning applications in highly multiplexed tissue imaging data analysis. FRONTIERS IN BIOINFORMATICS 2023; 3:1159381. [PMID: 37564726 PMCID: PMC10410935 DOI: 10.3389/fbinf.2023.1159381] [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: 02/05/2023] [Accepted: 07/12/2023] [Indexed: 08/12/2023] Open
Abstract
Since its introduction into the field of oncology, deep learning (DL) has impacted clinical discoveries and biomarker predictions. DL-driven discoveries and predictions in oncology are based on a variety of biological data such as genomics, proteomics, and imaging data. DL-based computational frameworks can predict genetic variant effects on gene expression, as well as protein structures based on amino acid sequences. Furthermore, DL algorithms can capture valuable mechanistic biological information from several spatial "omics" technologies, such as spatial transcriptomics and spatial proteomics. Here, we review the impact that the combination of artificial intelligence (AI) with spatial omics technologies has had on oncology, focusing on DL and its applications in biomedical image analysis, encompassing cell segmentation, cell phenotype identification, cancer prognostication, and therapy prediction. We highlight the advantages of using highly multiplexed images (spatial proteomics data) compared to single-stained, conventional histopathological ("simple") images, as the former can provide deep mechanistic insights that cannot be obtained by the latter, even with the aid of explainable AI. Furthermore, we provide the reader with the advantages/disadvantages of DL-based pipelines used in preprocessing highly multiplexed images (cell segmentation, cell type annotation). Therefore, this review also guides the reader to choose the DL-based pipeline that best fits their data. In conclusion, DL continues to be established as an essential tool in discovering novel biological mechanisms when combined with technologies such as highly multiplexed tissue imaging data. In balance with conventional medical data, its role in clinical routine will become more important, supporting diagnosis and prognosis in oncology, enhancing clinical decision-making, and improving the quality of care for patients. Since its introduction into the field of oncology, deep learning (DL) has impacted clinical discoveries and biomarker predictions. DL-driven discoveries and predictions in oncology are based on a variety of biological data such as genomics, proteomics, and imaging data. DL-based computational frameworks can predict genetic variant effects on gene expression, as well as protein structures based on amino acid sequences. Furthermore, DL algorithms can capture valuable mechanistic biological information from several spatial "omics" technologies, such as spatial transcriptomics and spatial proteomics. Here, we review the impact that the combination of artificial intelligence (AI) with spatial omics technologies has had on oncology, focusing on DL and its applications in biomedical image analysis, encompassing cell segmentation, cell phenotype identification, cancer prognostication, and therapy prediction. We highlight the advantages of using highly multiplexed images (spatial proteomics data) compared to single-stained, conventional histopathological ("simple") images, as the former can provide deep mechanistic insights that cannot be obtained by the latter, even with the aid of explainable AI. Furthermore, we provide the reader with the advantages/disadvantages of the DL-based pipelines used in preprocessing the highly multiplexed images (cell segmentation, cell type annotation). Therefore, this review also guides the reader to choose the DL-based pipeline that best fits their data. In conclusion, DL continues to be established as an essential tool in discovering novel biological mechanisms when combined with technologies such as highly multiplexed tissue imaging data. In balance with conventional medical data, its role in clinical routine will become more important, supporting diagnosis and prognosis in oncology, enhancing clinical decision-making, and improving the quality of care for patients.
Collapse
Affiliation(s)
- Mohammed Zidane
- Department of Pathology and Neuropathology, University Hospital and Comprehensive Cancer Center Tübingen, Tübingen, Germany
| | - Ahmad Makky
- Department of Pathology and Neuropathology, University Hospital and Comprehensive Cancer Center Tübingen, Tübingen, Germany
| | - Matthias Bruhns
- Department of Internal Medicine I, University Hospital Tübingen, Tübingen, Germany
- Department of Computer Science, University of Tübingen, Tübingen, Germany
| | - Alexander Rochwarger
- Department of Pathology and Neuropathology, University Hospital and Comprehensive Cancer Center Tübingen, Tübingen, Germany
| | - Sepideh Babaei
- Department of Internal Medicine I, University Hospital Tübingen, Tübingen, Germany
| | - Manfred Claassen
- Department of Internal Medicine I, University Hospital Tübingen, Tübingen, Germany
- Department of Computer Science, University of Tübingen, Tübingen, Germany
| | - Christian M. Schürch
- Department of Pathology and Neuropathology, University Hospital and Comprehensive Cancer Center Tübingen, Tübingen, Germany
| |
Collapse
|
12
|
Park HE, Jo SH, Lee RH, Macks CP, Ku T, Park J, Lee CW, Hur JK, Sohn CH. Spatial Transcriptomics: Technical Aspects of Recent Developments and Their Applications in Neuroscience and Cancer Research. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2206939. [PMID: 37026425 PMCID: PMC10238226 DOI: 10.1002/advs.202206939] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/25/2022] [Revised: 03/10/2023] [Indexed: 06/04/2023]
Abstract
Spatial transcriptomics is a newly emerging field that enables high-throughput investigation of the spatial localization of transcripts and related analyses in various applications for biological systems. By transitioning from conventional biological studies to "in situ" biology, spatial transcriptomics can provide transcriptome-scale spatial information. Currently, the ability to simultaneously characterize gene expression profiles of cells and relevant cellular environment is a paradigm shift for biological studies. In this review, recent progress in spatial transcriptomics and its applications in neuroscience and cancer studies are highlighted. Technical aspects of existing technologies and future directions of new developments (as of March 2023), computational analysis of spatial transcriptome data, application notes in neuroscience and cancer studies, and discussions regarding future directions of spatial multi-omics and their expanding roles in biomedical applications are emphasized.
Collapse
Affiliation(s)
- Han-Eol Park
- Center for Nanomedicine, Institute for Basic Science, Yonsei University, Seoul, 03722, Republic of Korea
- Graduate Program in Nanobiomedical Engineering, Advanced Science Institute, Yonsei University, Seoul, 03722, Republic of Korea
- School of Biological Sciences, Seoul National University, Seoul, 08826, Republic of Korea
| | - Song Hyun Jo
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, 34141, Republic of Korea
| | - Rosalind H Lee
- School of Life Sciences, Gwangju Institute of Science and Technology (GIST), Gwangju, 61005, Republic of Korea
| | - Christian P Macks
- Center for Nanomedicine, Institute for Basic Science, Yonsei University, Seoul, 03722, Republic of Korea
- Graduate Program in Nanobiomedical Engineering, Advanced Science Institute, Yonsei University, Seoul, 03722, Republic of Korea
| | - Taeyun Ku
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, 34141, Republic of Korea
| | - Jihwan Park
- School of Life Sciences, Gwangju Institute of Science and Technology (GIST), Gwangju, 61005, Republic of Korea
| | - Chung Whan Lee
- Department of Chemistry, Gachon University, Seongnam, Gyeonggi-do, 13120, Republic of Korea
| | - Junho K Hur
- Department of Genetics, College of Medicine, Hanyang University, Seoul, 04763, Republic of Korea
| | - Chang Ho Sohn
- Center for Nanomedicine, Institute for Basic Science, Yonsei University, Seoul, 03722, Republic of Korea
- Graduate Program in Nanobiomedical Engineering, Advanced Science Institute, Yonsei University, Seoul, 03722, Republic of Korea
| |
Collapse
|
13
|
Meng-Lin K, Ung CY, Zhang C, Weiskittel TM, Wisniewski P, Zhang Z, Tan SH, Yeo KS, Zhu S, Correia C, Li H. SPIN-AI: A Deep Learning Model That Identifies Spatially Predictive Genes. Biomolecules 2023; 13:895. [PMID: 37371475 PMCID: PMC10296445 DOI: 10.3390/biom13060895] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2023] [Revised: 05/23/2023] [Accepted: 05/23/2023] [Indexed: 06/29/2023] Open
Abstract
Spatially resolved sequencing technologies help us dissect how cells are organized in space. Several available computational approaches focus on the identification of spatially variable genes (SVGs), genes whose expression patterns vary in space. The detection of SVGs is analogous to the identification of differentially expressed genes and permits us to understand how genes and associated molecular processes are spatially distributed within cellular niches. However, the expression activities of SVGs fail to encode all information inherent in the spatial distribution of cells. Here, we devised a deep learning model, Spatially Informed Artificial Intelligence (SPIN-AI), to identify spatially predictive genes (SPGs), whose expression can predict how cells are organized in space. We used SPIN-AI on spatial transcriptomic data from squamous cell carcinoma (SCC) as a proof of concept. Our results demonstrate that SPGs not only recapitulate the biology of SCC but also identify genes distinct from SVGs. Moreover, we found a substantial number of ribosomal genes that were SPGs but not SVGs. Since SPGs possess the capability to predict spatial cellular organization, we reason that SPGs capture more biologically relevant information for a given cellular niche than SVGs. Thus, SPIN-AI has broad applications for detecting SPGs and uncovering which biological processes play important roles in governing cellular organization.
Collapse
Affiliation(s)
- Kevin Meng-Lin
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, MN 55905, USA; (K.M.-L.); (C.-Y.U.); (C.Z.); (T.M.W.); (P.W.); (Z.Z.); (S.-H.T.)
| | - Choong-Yong Ung
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, MN 55905, USA; (K.M.-L.); (C.-Y.U.); (C.Z.); (T.M.W.); (P.W.); (Z.Z.); (S.-H.T.)
| | - Cheng Zhang
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, MN 55905, USA; (K.M.-L.); (C.-Y.U.); (C.Z.); (T.M.W.); (P.W.); (Z.Z.); (S.-H.T.)
| | - Taylor M. Weiskittel
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, MN 55905, USA; (K.M.-L.); (C.-Y.U.); (C.Z.); (T.M.W.); (P.W.); (Z.Z.); (S.-H.T.)
| | - Philip Wisniewski
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, MN 55905, USA; (K.M.-L.); (C.-Y.U.); (C.Z.); (T.M.W.); (P.W.); (Z.Z.); (S.-H.T.)
| | - Zhuofei Zhang
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, MN 55905, USA; (K.M.-L.); (C.-Y.U.); (C.Z.); (T.M.W.); (P.W.); (Z.Z.); (S.-H.T.)
| | - Shyang-Hong Tan
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, MN 55905, USA; (K.M.-L.); (C.-Y.U.); (C.Z.); (T.M.W.); (P.W.); (Z.Z.); (S.-H.T.)
| | - Kok-Siong Yeo
- Department of Biochemistry and Molecular Biology, Mayo Clinic College of Medicine and Science, Rochester, MN 55905, USA; (K.-S.Y.); (S.Z.)
| | - Shizhen Zhu
- Department of Biochemistry and Molecular Biology, Mayo Clinic College of Medicine and Science, Rochester, MN 55905, USA; (K.-S.Y.); (S.Z.)
| | - Cristina Correia
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, MN 55905, USA; (K.M.-L.); (C.-Y.U.); (C.Z.); (T.M.W.); (P.W.); (Z.Z.); (S.-H.T.)
| | - Hu Li
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, MN 55905, USA; (K.M.-L.); (C.-Y.U.); (C.Z.); (T.M.W.); (P.W.); (Z.Z.); (S.-H.T.)
| |
Collapse
|
14
|
Li X, Xiao C, Qi J, Xue W, Xu X, Mu Z, Zhang J, Li CY, Ding W. STellaris: a web server for accurate spatial mapping of single cells based on spatial transcriptomics data. Nucleic Acids Res 2023:7177883. [PMID: 37224539 DOI: 10.1093/nar/gkad419] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2023] [Revised: 04/23/2023] [Accepted: 05/08/2023] [Indexed: 05/26/2023] Open
Abstract
Single-cell RNA sequencing (scRNA-seq) provides insights into gene expression heterogeneities in diverse cell types underlying homeostasis, development and pathological states. However, the loss of spatial information hinders its applications in deciphering spatially related features, such as cell-cell interactions in a spatial context. Here, we present STellaris (https://spatial.rhesusbase.com), a web server aimed to rapidly assign spatial information to scRNA-seq data based on their transcriptomic similarity with public spatial transcriptomics (ST) data. STellaris is founded on 101 manually curated ST datasets comprising 823 sections across different organs, developmental stages and pathological states from humans and mice. STellaris accepts raw count matrix and cell type annotation of scRNA-seq data as the input, and maps single cells to spatial locations in the tissue architecture of properly matched ST section. Spatially resolved information for intercellular communications, such as spatial distance and ligand-receptor interactions (LRIs), are further characterized between annotated cell types. Moreover, we also expanded the application of STellaris in spatial annotation of multiple regulatory levels with single-cell multiomics data, using the transcriptome as a bridge. STellaris was applied to several case studies to showcase its utility of adding value to the ever-growing scRNA-seq data from a spatial perspective.
Collapse
Affiliation(s)
- Xiangshang Li
- Laboratory of Bioinformatics and Genomic Medicine, Institute of Molecular Medicine, College of Future Technology, Peking University, Beijing 100871, China
| | - Chunfu Xiao
- Laboratory of Bioinformatics and Genomic Medicine, Institute of Molecular Medicine, College of Future Technology, Peking University, Beijing 100871, China
| | - Juntian Qi
- Laboratory of Bioinformatics and Genomic Medicine, Institute of Molecular Medicine, College of Future Technology, Peking University, Beijing 100871, China
| | | | - Xinwei Xu
- Laboratory of Bioinformatics and Genomic Medicine, Institute of Molecular Medicine, College of Future Technology, Peking University, Beijing 100871, China
- College of Life Science, Peking University, Beijing 100871, China
| | - Zelin Mu
- Laboratory of Bioinformatics and Genomic Medicine, Institute of Molecular Medicine, College of Future Technology, Peking University, Beijing 100871, China
- School of Basic Medical Sciences, Peking University Health Science Center, Beijing 100191, China
| | - Jie Zhang
- Laboratory of Bioinformatics and Genomic Medicine, Institute of Molecular Medicine, College of Future Technology, Peking University, Beijing 100871, China
| | - Chuan-Yun Li
- Laboratory of Bioinformatics and Genomic Medicine, Institute of Molecular Medicine, College of Future Technology, Peking University, Beijing 100871, China
- Chinese Institute for Brain Research, Beijing 102206, China
| | - Wanqiu Ding
- Laboratory of Bioinformatics and Genomic Medicine, Institute of Molecular Medicine, College of Future Technology, Peking University, Beijing 100871, China
- Bioinformatics Core, Institute of Molecular Medicine, College of Future Technology, Peking University, Beijing 100871, China
| |
Collapse
|
15
|
Zhu M, Li C, Lv K, Guo H, Hou R, Tian G, Yang J. MLSpatial: A machine-learning method to reconstruct the spatial distribution of cells from scRNA-seq by extracting spatial features. Comput Biol Med 2023; 159:106873. [PMID: 37105115 DOI: 10.1016/j.compbiomed.2023.106873] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2023] [Revised: 03/03/2023] [Accepted: 03/30/2023] [Indexed: 04/29/2023]
Abstract
MOTIVATION Single-cell RNA sequencing (scRNA-seq) technologies allow us to interrogate the state of an individual cell within its microenvironment. However, prior to sequencing, cells should be dissociated first, making it difficult to obtain their spatial information. Since the spatial distribution of cells is critical in a few circumstances such as cancer immunotherapy, we present MLSpatial, a novel computational method to learn the relationship between gene expression patterns and spatial locations of cells, and then predict cell-to-cell distance distribution based on scRNA-seq data alone. RESULTS We collected the drosophila embryo dataset, which contains both the fluorescence in situ hybridization (FISH) data and single cell RNA-seq (scRNA-seq) data of drosophila embryo. The FISH data provided the spatial position of 3039 cells and the expression of 84 genes for each cell. The scRNA-seq data contains the expressions of 8924 genes in 1297 high-quality cells with cell location unknown. For a comparison, we also collected the MERFISH data of 645 osteosarcoma cells with cell location and the expression status of 10,050 genes known. For each data, the cells were randomly divided into a training set and a test set, in the ratio of 7:3. The cell-to-cell distances our model extracted had a higher correspondence (i.e., correlation coefficient 0.99) with those of the real situation than those of existing methods in the FISH data of drosophila embryo. However, in the osteosarcoma data, our model captured the spatial relationship between cells, with a correlation of 0.514 to that of the real situation. We also applied the model trained using the FISH data of drosophila embryo into the single cell data of drosophila embryo, for which the real location of cells are unknown. The reconstructed pseudo drosophila embryo and the real embryo (as shown by the FISH data) had a high similarity in the spatial distribution of gene expression. CONCLUSION MLSpatial can accurately restore the relative position of cells from scRNA-seq data; however, the performance depends on the type of cells. The trained model might be useful in reconstructing the spatial distributions of single cells with only scRNA-seq data, provided that the scRNA-seq data and the FISH data are under similar background (i.e., the same tissue with similar disease background).
Collapse
Affiliation(s)
- Mengbo Zhu
- Department of Mathematics, Ocean University of China, Qingdao, 266100, China; Geneis Beijing Co., Ltd., Beijing, 100102, China
| | - Changjun Li
- Department of Mathematics, Ocean University of China, Qingdao, 266100, China.
| | - Kebo Lv
- Department of Mathematics, Ocean University of China, Qingdao, 266100, China
| | - Hongzhe Guo
- Geneis Beijing Co., Ltd., Beijing, 100102, China.
| | - Rui Hou
- Geneis Beijing Co., Ltd., Beijing, 100102, China; Qingdao Geneis Institute of Big Data Mining and Precision Medicine, Qingdao, 266000, China
| | - Geng Tian
- Geneis Beijing Co., Ltd., Beijing, 100102, China; Qingdao Geneis Institute of Big Data Mining and Precision Medicine, Qingdao, 266000, China
| | - Jialiang Yang
- Geneis Beijing Co., Ltd., Beijing, 100102, China; Qingdao Geneis Institute of Big Data Mining and Precision Medicine, Qingdao, 266000, China; Chifeng Municipal Hospital, Chifeng, Inner Mongolia, 024000, China; Academician Workstation, Changsha Medical University, Changsha, 410219, China.
| |
Collapse
|
16
|
Heydari AA, Sindi SS. Deep learning in spatial transcriptomics: Learning from the next next-generation sequencing. BIOPHYSICS REVIEWS 2023; 4:011306. [PMID: 38505815 PMCID: PMC10903438 DOI: 10.1063/5.0091135] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/11/2022] [Accepted: 12/19/2022] [Indexed: 03/21/2024]
Abstract
Spatial transcriptomics (ST) technologies are rapidly becoming the extension of single-cell RNA sequencing (scRNAseq), holding the potential of profiling gene expression at a single-cell resolution while maintaining cellular compositions within a tissue. Having both expression profiles and tissue organization enables researchers to better understand cellular interactions and heterogeneity, providing insight into complex biological processes that would not be possible with traditional sequencing technologies. Data generated by ST technologies are inherently noisy, high-dimensional, sparse, and multi-modal (including histological images, count matrices, etc.), thus requiring specialized computational tools for accurate and robust analysis. However, many ST studies currently utilize traditional scRNAseq tools, which are inadequate for analyzing complex ST datasets. On the other hand, many of the existing ST-specific methods are built upon traditional statistical or machine learning frameworks, which have shown to be sub-optimal in many applications due to the scale, multi-modality, and limitations of spatially resolved data (such as spatial resolution, sensitivity, and gene coverage). Given these intricacies, researchers have developed deep learning (DL)-based models to alleviate ST-specific challenges. These methods include new state-of-the-art models in alignment, spatial reconstruction, and spatial clustering, among others. However, DL models for ST analysis are nascent and remain largely underexplored. In this review, we provide an overview of existing state-of-the-art tools for analyzing spatially resolved transcriptomics while delving deeper into the DL-based approaches. We discuss the new frontiers and the open questions in this field and highlight domains in which we anticipate transformational DL applications.
Collapse
|
17
|
Duan H, Cheng T, Cheng H. Spatially resolved transcriptomics: advances and applications. BLOOD SCIENCE 2023; 5:1-14. [PMID: 36742187 PMCID: PMC9891446 DOI: 10.1097/bs9.0000000000000141] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2022] [Accepted: 10/19/2022] [Indexed: 11/06/2022] Open
Abstract
Spatial transcriptomics, which is capable of both measuring all gene activity in a tissue sample and mapping where this activity occurs, is vastly improving our understanding of biological processes and disease. The field has expanded rapidly in recent years, and the development of several new technologies has resulted in spatially resolved transcriptomics (SRT) becoming highly multiplexed, high-resolution, and high-throughput. Here, we summarize and compare the major methods of SRT, including imaging-based methods, sequencing-based methods, and in situ sequencing methods. We also highlight some typical applications of SRT in neuroscience, cancer biology, developmental biology, and hematology. Finally, we discuss future possibilities for improving spatially resolved transcriptomic methods and the expected applications of such methods, especially in the adult bone marrow, anticipating that new developments will unlock the full potential of spatially resolved multi-omics in both biological research and the clinic.
Collapse
Affiliation(s)
- Honglin Duan
- State Key Laboratory of Experimental Hematology, Haihe Laboratory of Cell Ecosystem, National Clinical Research Center for Blood Diseases, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China
| | - Tao Cheng
- State Key Laboratory of Experimental Hematology, Haihe Laboratory of Cell Ecosystem, National Clinical Research Center for Blood Diseases, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China
- Center for Stem Cell Medicine, Chinese Academy of Medical Sciences, Tianjin, China
- Department of Stem Cell & Regenerative Medicine, Peking Union Medical College, Tianjin, China
| | - Hui Cheng
- State Key Laboratory of Experimental Hematology, Haihe Laboratory of Cell Ecosystem, National Clinical Research Center for Blood Diseases, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China
- Center for Stem Cell Medicine, Chinese Academy of Medical Sciences, Tianjin, China
- Department of Stem Cell & Regenerative Medicine, Peking Union Medical College, Tianjin, China
| |
Collapse
|
18
|
Spatially resolved transcriptomics and the kidney: Many opportunities. Kidney Int 2022; 102:482-491. [PMID: 35788360 DOI: 10.1016/j.kint.2022.06.011] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2022] [Revised: 06/10/2022] [Accepted: 06/20/2022] [Indexed: 01/31/2023]
Abstract
Defining changes in gene expression during health and disease is critical for the understanding of human physiology. In recent years, single cell/nuclei RNA sequencing (sc/snRNAseq) has revolutionized the definition and discovery of cell types and states, as well as the interpretation of organ and cell type specific signaling pathways. However, these advances require tissue dissociation to the level of the single cell or single nuclei level. Spatially resolved transcriptomics (SrT) now provides a platform to overcome this barrier in understanding the physiological contexts of gene expression and cellular microenvironment changes in development and disease. Some of these transcriptomic tools allow for high resolution mapping of hundreds of genes simultaneously in cellular and subcellular compartments. Other tools offer genome depth mapping, but at lower resolution. Here, we will review advances in SrT, considerations for using SrT in your own research, and applications for kidney biology.
Collapse
|
19
|
Li Y, Stanojevic S, Garmire LX. Emerging artificial intelligence applications in Spatial Transcriptomics analysis. Comput Struct Biotechnol J 2022; 20:2895-2908. [PMID: 35765645 PMCID: PMC9201012 DOI: 10.1016/j.csbj.2022.05.056] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2022] [Revised: 05/28/2022] [Accepted: 05/28/2022] [Indexed: 11/19/2022] Open
Abstract
Spatial transcriptomics (ST) has advanced significantly in the last few years. Such advancement comes with the urgent need for novel computational methods to handle the unique challenges of ST data analysis. Many artificial intelligence (AI) methods have been developed to utilize various machine learning and deep learning techniques for computational ST analysis. This review provides a comprehensive and up-to-date survey of current AI methods for ST analysis.
Collapse
Affiliation(s)
- Yijun Li
- Department of Biostatistics, University of Michigan, Ann Arbor, MI, USA
| | - Stefan Stanojevic
- Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, MI, USA
| | - Lana X. Garmire
- Department of Biostatistics, University of Michigan, Ann Arbor, MI, USA
- Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, MI, USA
| |
Collapse
|
20
|
Affiliation(s)
- Jongwon Lee
- Department of Biomedical Sciences, Korea University College of Medicine, Seoul 02841, Korea
- Brain Korea 21 Plus Project for Biomedical Science, Korea University College of Medicine, Seoul 02841, Korea
| | - Minsu Yoo
- Department of Biomedical Sciences, Korea University College of Medicine, Seoul 02841, Korea
| | - Jungmin Choi
- Department of Biomedical Sciences, Korea University College of Medicine, Seoul 02841, Korea
- Department of Genetics, Yale University School of Medicine, New Haven, CT 06510, USA
| |
Collapse
|
21
|
Zeng Z, Li Y, Li Y, Luo Y. Statistical and machine learning methods for spatially resolved transcriptomics data analysis. Genome Biol 2022; 23:83. [PMID: 35337374 PMCID: PMC8951701 DOI: 10.1186/s13059-022-02653-7] [Citation(s) in RCA: 74] [Impact Index Per Article: 24.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2021] [Accepted: 03/15/2022] [Indexed: 01/28/2023] Open
Abstract
The recent advancement in spatial transcriptomics technology has enabled multiplexed profiling of cellular transcriptomes and spatial locations. As the capacity and efficiency of the experimental technologies continue to improve, there is an emerging need for the development of analytical approaches. Furthermore, with the continuous evolution of sequencing protocols, the underlying assumptions of current analytical methods need to be re-evaluated and adjusted to harness the increasing data complexity. To motivate and aid future model development, we herein review the recent development of statistical and machine learning methods in spatial transcriptomics, summarize useful resources, and highlight the challenges and opportunities ahead.
Collapse
Affiliation(s)
- Zexian Zeng
- Center for Quantitative Biology, Academy for Advanced Interdisciplinary Studies, Peking University, Beijing, 100084, China
- Peking-Tsinghua Center for Life Sciences, Academy for Advanced Interdisciplinary Studies, Peking University, Beijing, 100084, China
- Department of Data Sciences, Dana Farber Cancer Institute, Harvard T.H. Chan School of Public Health, Boston, MA, 02215, USA
| | - Yawei Li
- Division of Health and Biomedical Informatics, Department of Preventive Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, 60611, USA
| | - Yiming Li
- Division of Health and Biomedical Informatics, Department of Preventive Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, 60611, USA
| | - Yuan Luo
- Division of Health and Biomedical Informatics, Department of Preventive Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, 60611, USA.
- Northwestern University Clinical and Translational Sciences Institute, Chicago, IL, 60611, USA.
- Institute for Augmented Intelligence in Medicine, Northwestern University, Chicago, IL, 60611, USA.
- Center for Health Information Partnerships, Northwestern University, Chicago, IL, 60611, USA.
| |
Collapse
|
22
|
Walker BL, Cang Z, Ren H, Bourgain-Chang E, Nie Q. Deciphering tissue structure and function using spatial transcriptomics. Commun Biol 2022; 5:220. [PMID: 35273328 PMCID: PMC8913632 DOI: 10.1038/s42003-022-03175-5] [Citation(s) in RCA: 55] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Accepted: 02/16/2022] [Indexed: 01/31/2023] Open
Abstract
The rapid development of spatial transcriptomics (ST) techniques has allowed the measurement of transcriptional levels across many genes together with the spatial positions of cells. This has led to an explosion of interest in computational methods and techniques for harnessing both spatial and transcriptional information in analysis of ST datasets. The wide diversity of approaches in aim, methodology and technology for ST provides great challenges in dissecting cellular functions in spatial contexts. Here, we synthesize and review the key problems in analysis of ST data and methods that are currently applied, while also expanding on open questions and areas of future development.
Collapse
Affiliation(s)
- Benjamin L Walker
- The NSF-Simons Center for Multiscale Cell Fate Research, University of California Irvine, Irvine, CA, USA
- Department of Mathematics, University of California Irvine, Irvine, CA, USA
| | - Zixuan Cang
- The NSF-Simons Center for Multiscale Cell Fate Research, University of California Irvine, Irvine, CA, USA
- Department of Mathematics, University of California Irvine, Irvine, CA, USA
| | - Honglei Ren
- The NSF-Simons Center for Multiscale Cell Fate Research, University of California Irvine, Irvine, CA, USA
- Department of Mathematics, University of California Irvine, Irvine, CA, USA
| | | | - Qing Nie
- The NSF-Simons Center for Multiscale Cell Fate Research, University of California Irvine, Irvine, CA, USA.
- Department of Mathematics, University of California Irvine, Irvine, CA, USA.
- Department of Developmental and Cell Biology, University of California Irvine, Irvine, CA, USA.
| |
Collapse
|
23
|
Lee J, Yoo M, Choi J. Recent advances in spatially resolved transcriptomics: challenges and opportunities. BMB Rep 2022; 55:113-124. [PMID: 35168703 PMCID: PMC8972138] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2021] [Revised: 02/07/2022] [Accepted: 02/11/2022] [Indexed: 03/09/2024] Open
Abstract
Single-cell RNA sequencing (scRNA-seq) has greatly advanced our understanding of cellular heterogeneity by profiling individual cell transcriptomes. However, cell dissociation from the tissue structure causes a loss of spatial information, which hinders the identification of intercellular communication networks and global transcriptional patterns present in the tissue architecture. To overcome this limitation, novel transcriptomic platforms that preserve spatial information have been actively developed. Significant achievements in imaging technologies have enabled in situ targeted transcriptomic profiling in single cells at singlemolecule resolution. In addition, technologies based on mRNA capture followed by sequencing have made possible profiling of the genome-wide transcriptome at the 55-100 μm resolution. Unfortunately, neither imaging-based technology nor capturebased method elucidates a complete picture of the spatial transcriptome in a tissue. Therefore, addressing specific biological questions requires balancing experimental throughput and spatial resolution, mandating the efforts to develop computational algorithms that are pivotal to circumvent technology-specific limitations. In this review, we focus on the current state-of-the-art spatially resolved transcriptomic technologies, describe their applications in a variety of biological domains, and explore recent discoveries demonstrating their enormous potential in biomedical research. We further highlight novel integrative computational methodologies with other data modalities that provide a framework to derive biological insight into heterogeneous and complex tissue organization. [BMB Reports 2022; 55(3): 113-124].
Collapse
Affiliation(s)
- Jongwon Lee
- Department of Biomedical Sciences, Korea University College of Medicine, Seoul 02841, USA
- Brain Korea 21 Plus Project for Biomedical Science, Korea University College of Medicine, Seoul 02841, Korea, CT 06510, USA
| | - Minsu Yoo
- Department of Biomedical Sciences, Korea University College of Medicine, Seoul 02841, USA
| | - Jungmin Choi
- Department of Biomedical Sciences, Korea University College of Medicine, Seoul 02841, USA
- Department of Genetics, Yale University School of Medicine, New Haven, CT 06510, USA
| |
Collapse
|
24
|
Flores M, Liu Z, Zhang T, Hasib MM, Chiu YC, Ye Z, Paniagua K, Jo S, Zhang J, Gao SJ, Jin YF, Chen Y, Huang Y. Deep learning tackles single-cell analysis-a survey of deep learning for scRNA-seq analysis. Brief Bioinform 2022; 23:bbab531. [PMID: 34929734 PMCID: PMC8769926 DOI: 10.1093/bib/bbab531] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2021] [Revised: 11/15/2021] [Accepted: 11/16/2021] [Indexed: 12/17/2022] Open
Abstract
Since its selection as the method of the year in 2013, single-cell technologies have become mature enough to provide answers to complex research questions. With the growth of single-cell profiling technologies, there has also been a significant increase in data collected from single-cell profilings, resulting in computational challenges to process these massive and complicated datasets. To address these challenges, deep learning (DL) is positioned as a competitive alternative for single-cell analyses besides the traditional machine learning approaches. Here, we survey a total of 25 DL algorithms and their applicability for a specific step in the single cell RNA-seq processing pipeline. Specifically, we establish a unified mathematical representation of variational autoencoder, autoencoder, generative adversarial network and supervised DL models, compare the training strategies and loss functions for these models, and relate the loss functions of these models to specific objectives of the data processing step. Such a presentation will allow readers to choose suitable algorithms for their particular objective at each step in the pipeline. We envision that this survey will serve as an important information portal for learning the application of DL for scRNA-seq analysis and inspire innovative uses of DL to address a broader range of new challenges in emerging multi-omics and spatial single-cell sequencing.
Collapse
Affiliation(s)
- Mario Flores
- Department of Electrical and Computer Engineering, the University of Texas at San Antonio, San Antonio, TX 78249, USA
| | - Zhentao Liu
- Department of Electrical and Computer Engineering, the University of Texas at San Antonio, San Antonio, TX 78249, USA
| | - Tinghe Zhang
- Department of Electrical and Computer Engineering, the University of Texas at San Antonio, San Antonio, TX 78249, USA
| | - Md Musaddaqui Hasib
- Department of Electrical and Computer Engineering, the University of Texas at San Antonio, San Antonio, TX 78249, USA
| | - Yu-Chiao Chiu
- Greehey Children’s Cancer Research Institute, University of Texas Health San Antonio, San Antonio, TX 78229, USA
| | - Zhenqing Ye
- Greehey Children’s Cancer Research Institute, University of Texas Health San Antonio, San Antonio, TX 78229, USA
- Department of Population Health Sciences, University of Texas Health San Antonio, San Antonio, TX 78229, USA
| | - Karla Paniagua
- Department of Electrical and Computer Engineering, the University of Texas at San Antonio, San Antonio, TX 78249, USA
| | - Sumin Jo
- Department of Electrical and Computer Engineering, the University of Texas at San Antonio, San Antonio, TX 78249, USA
| | - Jianqiu Zhang
- Department of Electrical and Computer Engineering, the University of Texas at San Antonio, San Antonio, TX 78249, USA
| | - Shou-Jiang Gao
- Department of Microbiology and Molecular Genetics, University of Pittsburgh, Pittsburgh, Pennsylvania, PA 15232, USA
- UPMC Hillman Cancer Center, University of Pittsburgh, PA 15232, USA
| | - Yu-Fang Jin
- Department of Electrical and Computer Engineering, the University of Texas at San Antonio, San Antonio, TX 78249, USA
| | - Yidong Chen
- Greehey Children’s Cancer Research Institute, University of Texas Health San Antonio, San Antonio, TX 78229, USA
- Department of Population Health Sciences, University of Texas Health San Antonio, San Antonio, TX 78229, USA
| | - Yufei Huang
- Department of Medicine, School of Medicine, University of Pittsburgh, PA 15232, USA
- UPMC Hillman Cancer Center, University of Pittsburgh, PA 15232, USA
| |
Collapse
|
25
|
Abstract
Spatial transcriptomics is a rapidly growing field that promises to comprehensively characterize tissue organization and architecture at the single-cell or subcellular resolution. Such information provides a solid foundation for mechanistic understanding of many biological processes in both health and disease that cannot be obtained by using traditional technologies. The development of computational methods plays important roles in extracting biological signals from raw data. Various approaches have been developed to overcome technology-specific limitations such as spatial resolution, gene coverage, sensitivity, and technical biases. Downstream analysis tools formulate spatial organization and cell-cell communications as quantifiable properties, and provide algorithms to derive such properties. Integrative pipelines further assemble multiple tools in one package, allowing biologists to conveniently analyze data from beginning to end. In this review, we summarize the state of the art of spatial transcriptomic data analysis methods and pipelines, and discuss how they operate on different technological platforms.
Collapse
Affiliation(s)
- Ruben Dries
- Department of Medicine, Boston University School of Medicine, Boston, Massachusetts 02118, USA
- Bioinformatics Graduate Program, Boston University, Boston, Massachusetts 02215, USA
- Section of Computational Biomedicine, Boston University School of Medicine, Boston, Massachusetts 02118, USA
| | - Jiaji Chen
- Department of Medicine, Boston University School of Medicine, Boston, Massachusetts 02118, USA
| | - Natalie Del Rossi
- Department of Genetics and Genomic Sciences, Charles Bronfman Institute for Personalized Medicine, Icahn School of Medicine at Mount Sinai, New York, New York 10029, USA
| | - Mohammed Muzamil Khan
- Department of Medicine, Boston University School of Medicine, Boston, Massachusetts 02118, USA
- Bioinformatics Graduate Program, Boston University, Boston, Massachusetts 02215, USA
- Section of Computational Biomedicine, Boston University School of Medicine, Boston, Massachusetts 02118, USA
| | - Adriana Sistig
- Department of Genetics and Genomic Sciences, Charles Bronfman Institute for Personalized Medicine, Icahn School of Medicine at Mount Sinai, New York, New York 10029, USA
| | - Guo-Cheng Yuan
- Department of Genetics and Genomic Sciences, Charles Bronfman Institute for Personalized Medicine, Icahn School of Medicine at Mount Sinai, New York, New York 10029, USA
- Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, New York 10029, USA
| |
Collapse
|