1
|
Cui Y, Cui Y, Ding Y, Nakai K, Wei L, Le Y, Ye X, Sakurai T. OmniClust: A versatile clustering toolkit for single-cell and spatial transcriptomics data. Methods 2025; 238:84-94. [PMID: 40057293 DOI: 10.1016/j.ymeth.2025.03.007] [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: 01/31/2025] [Revised: 02/24/2025] [Accepted: 03/05/2025] [Indexed: 03/22/2025] Open
Abstract
In recent years, RNA transcriptome sequencing technology has been continuously evolving, ranging from single-cell transcriptomics to spatial transcriptomics. Although these technologies are all based on RNA sequencing, each sequencing technology has its own unique characteristics, and there is an urgent need to develop an algorithmic toolkit that integrates both sequencing techniques. To address this, we have developed OmniClust, a toolkit based on single-cell RNA sequencing (scRNA-seq) and spatial transcriptomics data. OmniClust employs deep learning algorithms for feature learning and clustering of spatial transcriptomics data, while utilizing machine learning algorithms for clustering scRNA-seq data. OmniClust was tested on 12 spatial transcriptomics benchmark datasets, demonstrating high clustering accuracy across multiple clustering evaluation metrics. It was also evaluated on four scRNA-seq benchmark datasets, achieving high clustering accuracy based on various clustering evaluation metrics. Furthermore, we applied OmniClust to downstream analyses of spatial transcriptomics and single-cell RNA breast cancer data, showcasing its potential to uncover and interpret the biological significance of cancer transcriptome data. In summary, OmniClust is a clustering tool designed for both single-cell transcriptomics and spatial transcriptomics data, demonstrating outstanding performance.
Collapse
Affiliation(s)
- Yaxuan Cui
- Department of Computer Science, University of Tsukuba, Tsukuba 3058577, Japan
| | - Yang Cui
- Department of Computational Biology and Medical Sciences, Graduate School of Frontier Sciences, University of Tokyo, Tokyo, Japan
| | - Yi Ding
- Department of Computational Biology and Medical Sciences, Graduate School of Frontier Sciences, University of Tokyo, Tokyo, Japan
| | - Kenta Nakai
- Department of Computational Biology and Medical Sciences, Graduate School of Frontier Sciences, University of Tokyo, Tokyo, Japan; Human Genome Center, The Institute of Medical Science, The University of Tokyo, Tokyo 108-8639, Japan
| | - Leyi Wei
- Centre for Artificial Intelligence Driven Drug Discovery, Faculty of Applied Science, Macao Polytechnic University, Macao SAR, China
| | - Yuyin Le
- Department of Radiation Oncology Fuzhou Pulmonary Hospital of Fujian Province , Teaching Hospital of Fujian Medical University, China.
| | - Xiucai Ye
- Department of Computer Science, University of Tsukuba, Tsukuba 3058577, Japan.
| | - Tetsuya Sakurai
- Department of Computer Science, University of Tsukuba, Tsukuba 3058577, Japan
| |
Collapse
|
2
|
Junaid M, Lee EJ, Lim SB. Single-cell and spatial omics: exploring hypothalamic heterogeneity. Neural Regen Res 2025; 20:1525-1540. [PMID: 38993130 PMCID: PMC11688568 DOI: 10.4103/nrr.nrr-d-24-00231] [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: 02/26/2024] [Revised: 05/06/2024] [Accepted: 06/03/2024] [Indexed: 07/13/2024] Open
Abstract
Elucidating the complex dynamic cellular organization in the hypothalamus is critical for understanding its role in coordinating fundamental body functions. Over the past decade, single-cell and spatial omics technologies have significantly evolved, overcoming initial technical challenges in capturing and analyzing individual cells. These high-throughput omics technologies now offer a remarkable opportunity to comprehend the complex spatiotemporal patterns of transcriptional diversity and cell-type characteristics across the entire hypothalamus. Current single-cell and single-nucleus RNA sequencing methods comprehensively quantify gene expression by exploring distinct phenotypes across various subregions of the hypothalamus. However, single-cell/single-nucleus RNA sequencing requires isolating the cell/nuclei from the tissue, potentially resulting in the loss of spatial information concerning neuronal networks. Spatial transcriptomics methods, by bypassing the cell dissociation, can elucidate the intricate spatial organization of neural networks through their imaging and sequencing technologies. In this review, we highlight the applicative value of single-cell and spatial transcriptomics in exploring the complex molecular-genetic diversity of hypothalamic cell types, driven by recent high-throughput achievements.
Collapse
Affiliation(s)
- Muhammad Junaid
- Department of Biochemistry & Molecular Biology, Ajou University School of Medicine, Suwon, South Korea
- Department of Biomedical Sciences, Graduate School of Ajou University, Suwon, South Korea
| | - Eun Jeong Lee
- Department of Biomedical Sciences, Graduate School of Ajou University, Suwon, South Korea
- Department of Brain Science, Ajou University School of Medicine, Suwon, South Korea
| | - Su Bin Lim
- Department of Biochemistry & Molecular Biology, Ajou University School of Medicine, Suwon, South Korea
- Department of Biomedical Sciences, Graduate School of Ajou University, Suwon, South Korea
| |
Collapse
|
3
|
Zeng X, Liu S, Liu B, Zhang W, Xu W, Toriumi F, Nakai K. Gene2role: a role-based gene embedding method for comparative analysis of signed gene regulatory networks. BMC Bioinformatics 2025; 26:134. [PMID: 40413377 DOI: 10.1186/s12859-025-06128-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: 09/15/2024] [Accepted: 04/01/2025] [Indexed: 05/27/2025] Open
Abstract
BACKGROUND Understanding the dynamics of gene regulatory networks (GRNs) across various cellular states is crucial for deciphering the underlying mechanisms governing cell behavior and functionality. However, current comparative analytical methods, which often focus on simple topological information such as the degree of genes, are limited in their ability to fully capture the similarities and differences among the complex GRNs. RESULTS We present Gene2role, a gene embedding approach that leverages multi-hop topological information from genes within signed GRNs. Initially, we demonstrated the effectiveness of Gene2role in capturing the intricate topological nuances of genes using GRNs inferred from four distinct data sources. Then, applying Gene2role to integrated GRNs allowed us to identify genes with significant topological changes across cell types or states, offering a fresh perspective beyond traditional differential gene expression analyses. Additionally, we quantified the stability of gene modules between two cellular states by measuring the changes in the gene embeddings within these modules. CONCLUSIONS Our method augments the existing toolkit for probing the dynamic regulatory landscape, thereby opening new avenues for understanding gene behavior and interaction patterns across cellular transitions.
Collapse
Affiliation(s)
- Xin Zeng
- Department of Computational Biology and Medical Sciences, The University of Tokyo, Kashiwa, 277-8563, Japan
- Human Genome Center, The Institute of Medical Science, The University of Tokyo, Tokyo, 108-8639, Japan
| | - Shu Liu
- Department of Systems Innovation, The University of Tokyo, Tokyo, 113-8654, Japan.
| | - Bowen Liu
- Department of Computational Biology and Medical Sciences, The University of Tokyo, Kashiwa, 277-8563, Japan
- Human Genome Center, The Institute of Medical Science, The University of Tokyo, Tokyo, 108-8639, Japan
| | - Weihang Zhang
- Department of Computational Biology and Medical Sciences, The University of Tokyo, Kashiwa, 277-8563, Japan
- Human Genome Center, The Institute of Medical Science, The University of Tokyo, Tokyo, 108-8639, Japan
| | - Wanzhe Xu
- Department of Computational Biology and Medical Sciences, The University of Tokyo, Kashiwa, 277-8563, Japan
- Human Genome Center, The Institute of Medical Science, The University of Tokyo, Tokyo, 108-8639, Japan
| | - Fujio Toriumi
- Department of Systems Innovation, The University of Tokyo, Tokyo, 113-8654, Japan
| | - Kenta Nakai
- Department of Computational Biology and Medical Sciences, The University of Tokyo, Kashiwa, 277-8563, Japan.
- Human Genome Center, The Institute of Medical Science, The University of Tokyo, Tokyo, 108-8639, Japan.
| |
Collapse
|
4
|
Jing J, Gao Y, Gao YL, Li F, Wang J, Liu JX. stDGAC: A novel identifying spatial domains method via graph attention contrastive network for spatial transcriptomics data. Comput Biol Med 2025; 193:110280. [PMID: 40403639 DOI: 10.1016/j.compbiomed.2025.110280] [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: 10/15/2023] [Revised: 11/26/2024] [Accepted: 04/24/2025] [Indexed: 05/24/2025]
Abstract
Complex biological tissues are composed of many cells in a highly coordinated manner and play a variety of biological functions. In recent years, many methods for spatial clustering have been developed. However, it remains a major challenge to effectively utilize these high-dimensional and noisy spatial transcriptomics data, with similar gene expression and histological features, to more accurately identify spatial domains. Here, a novel method of spatial domain identification, named stDGAC, was proposed by jointly using a denoising autoencoder and a graph attention contrastive network for subsequent analysis of spatial transcriptomics data. The pre-trained denoising autoencoder performed dimensionality reduction and denoising, and then the low-dimensional latent representation was learned through a graph attention contrastive network to aggregate the neighborhood information of the spatial context and acquire a more robust representation. Finally, the proposed method stDGAC was experimentally demonstrated to outperform other existing methods in downstream analysis, such as identifying spatial domains, trajectory inference, and gene expression data denoising.
Collapse
Affiliation(s)
- Jing Jing
- School of Computer Science, Qufu Normal University, Rizhao, 276826, Shandong, China
| | - Yue Gao
- School of Computer Science, Qufu Normal University, Rizhao, 276826, Shandong, China
| | - Ying-Lian Gao
- Qufu Normal University Library, Qufu Normal University, Rizhao, 276826, Shandong, China
| | - Feng Li
- School of Computer Science, Qufu Normal University, Rizhao, 276826, Shandong, China
| | - Juan Wang
- School of Computer Science, Qufu Normal University, Rizhao, 276826, Shandong, China
| | - Jin-Xing Liu
- School of Health and Life Sciences, University of Health and Rehabilitation Sciences, Qingdao, 266113, Shandong, China.
| |
Collapse
|
5
|
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
|
6
|
Li X, Xu H, Zhao H, Jin F, Zhang X, Zhou Q. Clustering-Triggered Emission of Carboxymethyl β-Cyclodextrin Powder and Tablet. Chempluschem 2025:e2500230. [PMID: 40356544 DOI: 10.1002/cplu.202500230] [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: 03/29/2025] [Revised: 04/29/2025] [Accepted: 05/08/2025] [Indexed: 05/15/2025]
Abstract
Nonconventional luminogens have great potential applications in fields like cell imaging and anti-counterfeiting encryption. But so far, the photoluminescence quantum yield (PLQY) of most these solids is still relatively low and the persistent room-temperature phosphorescence (p-RTP) emission is relatively weak. To improve their PLQY and p-RTP, pressing the powder into tablets is preliminarily proven to be an effective method, but the specific mechanism has not been fully elucidated yet. Herein, carboxymethyl β-cyclodextrin (CM-β-CD) is chosen as the representative to solve the problem. The results show that the PLQY and p-RTP lifetimes of the tablet of CM-β-CD are improved compared to the powder. By the mechanism of clustering-triggered emission and average packing density-promoted emission mechanism, it is demonstrated that the enhanced molecular interactions after compression is the key reason, resulting in the formation of cluster emission centers with stronger emission capabilities. CM-β-CD is proven to have excellent cell imaging and anti-counterfeiting functions.
Collapse
Affiliation(s)
- Xintong Li
- Engineering Research Center for Eco-Dyeing and Finishing of Textiles, Key Laboratory of Advanced Textile Materials and Manufacturing Technology, Ministry of Education, College of Textile Science and Engineering (International Institute of Silk), Zhejiang Sci-Tech University, Hangzhou, 310018, P. R. China
| | - Haiyan Xu
- Engineering Research Center for Eco-Dyeing and Finishing of Textiles, Key Laboratory of Advanced Textile Materials and Manufacturing Technology, Ministry of Education, College of Textile Science and Engineering (International Institute of Silk), Zhejiang Sci-Tech University, Hangzhou, 310018, P. R. China
| | - Hongyan Zhao
- Engineering Research Center for Eco-Dyeing and Finishing of Textiles, Key Laboratory of Advanced Textile Materials and Manufacturing Technology, Ministry of Education, College of Textile Science and Engineering (International Institute of Silk), Zhejiang Sci-Tech University, Hangzhou, 310018, P. R. China
| | - Feng Jin
- Engineering Research Center for Eco-Dyeing and Finishing of Textiles, Key Laboratory of Advanced Textile Materials and Manufacturing Technology, Ministry of Education, College of Textile Science and Engineering (International Institute of Silk), Zhejiang Sci-Tech University, Hangzhou, 310018, P. R. China
| | - Xiangxi Zhang
- Engineering Research Center for Eco-Dyeing and Finishing of Textiles, Key Laboratory of Advanced Textile Materials and Manufacturing Technology, Ministry of Education, College of Textile Science and Engineering (International Institute of Silk), Zhejiang Sci-Tech University, Hangzhou, 310018, P. R. China
| | - Qing Zhou
- Engineering Research Center for Eco-Dyeing and Finishing of Textiles, Key Laboratory of Advanced Textile Materials and Manufacturing Technology, Ministry of Education, College of Textile Science and Engineering (International Institute of Silk), Zhejiang Sci-Tech University, Hangzhou, 310018, P. R. China
- Zhejiang Sci-Tech University Shaoxing-Keqiao Research Institute, Building 8 Cross border E-commerce Park, Huashe Street, Keqiao District, Shaoxing City, Zhejiang, 312030, China
- Zhejiang Provincial Innovation Center of Advanced Textile Technology, Building 7 Cross border E-commerce Park, Huashe Street, Keqiao District, Shaoxing City, Zhejiang, 312030, China
| |
Collapse
|
7
|
Zuo C, Zhu J, Zou J, Chen L. Unravelling tumour spatiotemporal heterogeneity using spatial multimodal data. Clin Transl Med 2025; 15:e70331. [PMID: 40341789 PMCID: PMC12059211 DOI: 10.1002/ctm2.70331] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2024] [Revised: 04/07/2025] [Accepted: 04/24/2025] [Indexed: 05/11/2025] Open
Abstract
Analysing the genome, epigenome, transcriptome, proteome, and metabolome within the spatial context of cells has transformed our understanding of tumour spatiotemporal heterogeneity. Advances in spatial multi-omics technologies now reveal complex molecular interactions shaping cellular behaviour and tissue dynamics. This review highlights key technologies and computational methods that have advanced spatial domain identification and their pseudo-relations, as well as inference of intra- and inter-cellular molecular networks that drive disease progression. We also discuss strategies to address major challenges, including data sparsity, high-dimensionality, scalability, and heterogeneity. Furthermore, we outline how spatial multi-omics enables novel insights into disease mechanisms, advancing precision medicine and informing targeted therapies. KEY POINTS: Advancements in spatial multi-omics facilitate our understanding of tumour spatiotemporal heterogeneity. AI-driven multimodal models uncover complex molecular interactions that underlie cellular behaviours and tissue dynamics. Combining multi-omics technologies and AI-enabled bioinformatics tools helps predict critical disease stages, such as pre-cancer, advancing precision medicine, and informing targeted therapeutic strategies.
Collapse
Affiliation(s)
- Chunman Zuo
- School of Life SciencesSun Yat‐sen UniversityGuangzhouChina
| | - Junchao Zhu
- Key Laboratory of Systems Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell ScienceChinese Academy of SciencesShanghaiChina
| | - Jiawei Zou
- Key Laboratory of Systems Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell ScienceChinese Academy of SciencesShanghaiChina
| | - Luonan Chen
- Key Laboratory of Systems Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell ScienceChinese Academy of SciencesShanghaiChina
- Key Laboratory of Systems Health Science of Zhejiang Province, School of Life Science, Hangzhou Institute for Advanced StudyUniversity of Chinese Academy of SciencesChinese Academy of SciencesHangzhouChina
- West China Biomedical Big Data Center, Med‐X Center for InformaticsWest China HospitalSichuan UniversityChengduChina
- School of Mathematical Sciences and School of AIShanghai Jiao Tong UniversityShanghaiChina
| |
Collapse
|
8
|
Guo B, Ling W, Kwon SH, Panwar P, Ghazanfar S, Martinowich K, Hicks SC. Integrating Spatially-Resolved Transcriptomics Data Across Tissues and Individuals: Challenges and Opportunities. SMALL METHODS 2025; 9:e2401194. [PMID: 39935130 PMCID: PMC12103234 DOI: 10.1002/smtd.202401194] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/01/2024] [Revised: 12/13/2024] [Indexed: 02/13/2025]
Abstract
Advances in spatially-resolved transcriptomics (SRT) technologies have propelled the development of new computational analysis methods to unlock biological insights. The lowering cost of SRT data generation presents an unprecedented opportunity to create large-scale spatial atlases and enable population-level investigation, integrating SRT data across multiple tissues, individuals, species, or phenotypes. Here, unique challenges are described in the SRT data integration, where the analytic impact of varying spatial and biological resolutions is characterized and explored. A succinct review of spatially-aware integration methods and computational strategies is provided. Exciting opportunities to advance computational algorithms amenable to atlas-scale datasets along with standardized preprocessing methods, leading to improved sensitivity and reproducibility in the future are further highlighted.
Collapse
Affiliation(s)
- Boyi Guo
- Department of BiostatisticsJohns Hopkins Bloomberg School of Public HealthBaltimoreMD21205USA
| | - Wodan Ling
- Division of BiostatisticsDepartment of Population Health SciencesWeill Cornell MedicineNew YorkNY10065USA
| | - Sang Ho Kwon
- Lieber Institute for Brain DevelopmentJohns Hopkins Medical CampusBaltimoreMD21205USA
- Solomon H. Snyder Department of NeuroscienceJohns Hopkins School of MedicineBaltimoreMD21205USA
- Biochemistry, Cellular, and Molecular Biology Graduate ProgramJohns Hopkins School of MedicineBaltimoreMD21205USA
| | - Pratibha Panwar
- School of Mathematics and StatisticsThe University of SydneyCamperdownNSW2006Australia
- Sydney Precision Data Science CentreUniversity of SydneyCamperdownNSW2006Australia
- Charles Perkins CentreThe University of SydneyCamperdownNSW2006Australia
| | - Shila Ghazanfar
- School of Mathematics and StatisticsThe University of SydneyCamperdownNSW2006Australia
- Sydney Precision Data Science CentreUniversity of SydneyCamperdownNSW2006Australia
- Charles Perkins CentreThe University of SydneyCamperdownNSW2006Australia
| | - Keri Martinowich
- Lieber Institute for Brain DevelopmentJohns Hopkins Medical CampusBaltimoreMD21205USA
- Solomon H. Snyder Department of NeuroscienceJohns Hopkins School of MedicineBaltimoreMD21205USA
- Department of Psychiatry and Behavioral SciencesJohns Hopkins School of MedicineBaltimoreMDUSA
- Johns Hopkins Kavli Neuroscience Discovery InstituteJohns Hopkins UniversityBaltimoreMD21218USA
- Department of Biomedical EngineeringJohns Hopkins UniversityBaltimoreMD21218USA
| | - Stephanie C. Hicks
- Department of BiostatisticsJohns Hopkins Bloomberg School of Public HealthBaltimoreMD21205USA
- Center for Computational BiologyJohns Hopkins UniversityBaltimoreMD21218USA
- Malone Center for Engineering in HealthcareJohns Hopkins UniversityBaltimoreMD21218USA
| |
Collapse
|
9
|
Wang Q, Zhu H, Deng L, Xu S, Xie W, Li M, Wang R, Tie L, Zhan L, Yu G. Spatial Transcriptomics: Biotechnologies, Computational Tools, and Neuroscience Applications. SMALL METHODS 2025; 9:e2401107. [PMID: 39760243 DOI: 10.1002/smtd.202401107] [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: 07/19/2024] [Revised: 12/22/2024] [Indexed: 01/07/2025]
Abstract
Spatial transcriptomics (ST) represents a revolutionary approach in molecular biology, providing unprecedented insights into the spatial organization of gene expression within tissues. This review aims to elucidate advancements in ST technologies, their computational tools, and their pivotal applications in neuroscience. It is begun with a historical overview, tracing the evolution from early image-based techniques to contemporary sequence-based methods. Subsequently, the computational methods essential for ST data analysis, including preprocessing, cell type annotation, spatial clustering, detection of spatially variable genes, cell-cell interaction analysis, and 3D multi-slices integration are discussed. The central focus of this review is the application of ST in neuroscience, where it has significantly contributed to understanding the brain's complexity. Through ST, researchers advance brain atlas projects, gain insights into brain development, and explore neuroimmune dysfunctions, particularly in brain tumors. Additionally, ST enhances understanding of neuronal vulnerability in neurodegenerative diseases like Alzheimer's and neuropsychiatric disorders such as schizophrenia. In conclusion, while ST has already profoundly impacted neuroscience, challenges remain issues such as enhancing sequencing technologies and developing robust computational tools. This review underscores the transformative potential of ST in neuroscience, paving the way for new therapeutic insights and advancements in brain research.
Collapse
Affiliation(s)
- Qianwen Wang
- Department of Bioinformatics, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Hongyuan Zhu
- Department of Bioinformatics, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Lin Deng
- Department of Bioinformatics, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Shuangbin Xu
- Department of Bioinformatics, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Wenqin Xie
- Department of Bioinformatics, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Ming Li
- Department of Bioinformatics, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Rui Wang
- Department of Bioinformatics, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Liang Tie
- Department of Bioinformatics, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Li Zhan
- Department of Bioinformatics, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Guangchuang Yu
- Department of Bioinformatics, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, China
| |
Collapse
|
10
|
Ren S, Liao X, Liu F, Li J, Gao X, Yu B. Exploring the Latent Information in Spatial Transcriptomics Data via Multi-View Graph Convolutional Network Based on Implicit Contrastive Learning. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025:e2413545. [PMID: 40304359 DOI: 10.1002/advs.202413545] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/23/2024] [Revised: 01/09/2025] [Indexed: 05/02/2025]
Abstract
Latest developments in spatial transcriptomics enable thoroughly profiling of gene expression while preserving tissue microenvironment. Connecting gene expression with spatial arrangement is key for precise spatial domain identification, enhancing the comprehension of tissue microenvironments and biological processes. However, accurately analyzing spatial domains with similar gene expression and histological features is still challenging. This study introduces STMIGCL, a novel framework that leverages a multi-view graph convolutional network and implicit contrastive learning. First, it creates neighbor graphs from gene expression and spatial coordinates, and then combines these with gene expression through multi-view learning to learn low-dimensional representations. To further refine the obtained low-dimensional representations, a graph contrastive learning method with contrastive enhancement in the latent space is employed, aiming to better capture critical information in the data and improve the accuracy and discriminative power of the embeddings. Finally, an attention mechanism is used to adaptively integrate different views, capturing the importance of spots in various views to obtain the final spot representation. Experimental data confirms that STMIGCL significantly enhances spatial domain recognition precision and outperforms all baseline methods in tasks such as trajectory inference and Spatially Variable Genes (SVGs) recognition.
Collapse
Affiliation(s)
- Sheng Ren
- School of Data Science, Qingdao University of Science and Technology, Qingdao, 266061, China
| | - Xingyu Liao
- School of Computer Science, Northwestern Polytechnical University, Xi'an, 710072, China
| | - Farong Liu
- College of Mathematics and Physics, Qingdao University of Science and Technology, Qingdao, 266061, China
| | - Jie Li
- School of Data Science, Qingdao University of Science and Technology, Qingdao, 266061, China
| | - Xin Gao
- Computational Bioscience Research Center (CBRC), Computer, Electrical and Mathematical Sciences and Engineering Division, King Abdullah University of Science and Technology (KAUST), Thuwal, 23955, Saudi Arabia
| | - Bin Yu
- School of Data Science, Qingdao University of Science and Technology, Qingdao, 266061, China
- School of Artificial Intelligence and Data Science, University of Science and Technology of China, Hefei, 230026, China
| |
Collapse
|
11
|
Gong Y, Yuan X, Jiao Q, Yu Z. Unveiling fine-scale spatial structures and amplifying gene expression signals in ultra-large ST slices with HERGAST. Nat Commun 2025; 16:3977. [PMID: 40295488 PMCID: PMC12037780 DOI: 10.1038/s41467-025-59139-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2024] [Accepted: 04/08/2025] [Indexed: 04/30/2025] Open
Abstract
We propose HERGAST, a system for spatial structure identification and signal amplification in ultra-large-scale and ultra-high-resolution spatial transcriptomics data. To handle ultra-large spatial transcriptomics (ST) data, we consider the divide and conquer strategy and devise a Divide-Iterate-Conquer framework especially for spatial transcriptomics data analysis, which can also be adopted by other computational methods for extending to ultra-large-scale ST data analysis. To tackle the potential over-smoothing problem arising from data splitting, we construct a heterogeneous graph network to incorporate both local and global spatial relationships. In simulations, HERGAST consistently outperforms other methods across all settings with more than a 10% increase in average adjusted rand index (ARI). In real-world datasets, HERGAST's high-precision spatial clustering identifies SPP1+ macrophages intermingled within colorectal tumors, while the enhanced gene expression signals reveal unique spatial expression patterns of key genes in breast cancer.
Collapse
Affiliation(s)
- Yuqiao Gong
- Department of Bioinformatics and Biostatistics, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai, China
| | - Xin Yuan
- SJTU-Yale Joint Center for Biostatistics and Data Science Organization, Shanghai Jiao Tong University, Shanghai, China
- Institute of Translational Medicine, Shanghai Jiao Tong University, Shanghai, China
- National Center for Translational Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Qiong Jiao
- Department of Pathology, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| | - Zhangsheng Yu
- Department of Bioinformatics and Biostatistics, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai, China.
- SJTU-Yale Joint Center for Biostatistics and Data Science Organization, Shanghai Jiao Tong University, Shanghai, China.
- Institute of Translational Medicine, Shanghai Jiao Tong University, Shanghai, China.
- Clinical Research Institute, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| |
Collapse
|
12
|
Fang S, Xu M, Cao L, Liu X, Bezulj M, Tan L, Yuan Z, Li Y, Xia T, Guo L, Kovacevic V, Hui J, Guo L, Liu C, Cheng M, Lin L, Wen Z, Josic B, Milicevic N, Qiu P, Lu Q, Li Y, Wang L, Hu L, Zhang C, Kang Q, Chen F, Deng Z, Li J, Li M, Li S, Zhao Y, Fan G, Zhang Y, Chen A, Li Y, Xu X. Stereopy: modeling comparative and spatiotemporal cellular heterogeneity via multi-sample spatial transcriptomics. Nat Commun 2025; 16:3741. [PMID: 40258830 PMCID: PMC12012134 DOI: 10.1038/s41467-025-58079-9] [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: 03/04/2024] [Accepted: 03/05/2025] [Indexed: 04/23/2025] Open
Abstract
Understanding complex biological systems requires tracing cellular dynamic changes across conditions, time, and space. However, integrating multi-sample data in a unified way to explore cellular heterogeneity remains challenging. Here, we present Stereopy, a flexible framework for modeling and dissecting comparative and spatiotemporal patterns in multi-sample spatial transcriptomics with interactive data visualization. To optimize this framework, we devise a universal container, a scope controller, and an integrative transformer tailored for multi-sample multimodal data storage, management, and processing. Stereopy showcases three representative applications: investigating specific cell communities and genes responsible for pathological changes, detecting spatiotemporal gene patterns by considering spatial and temporal features, and inferring three-dimensional niche-based cell-gene interaction network that bridges intercellular communications and intracellular regulations. Stereopy serves as both a comprehensive bioinformatics toolbox and an extensible framework that empowers researchers with enhanced data interpretation abilities and new perspectives for mining multi-sample spatial transcriptomics data.
Collapse
Affiliation(s)
| | - Mengyang Xu
- BGI Research, Shenzhen, China
- BGI Research, Qingdao, China
| | - Lei Cao
- BGI Research, Beijing, China
- BGI Research, Shenzhen, China
| | | | | | | | - Zhiyuan Yuan
- Institute of Science and Technology for Brain-Inspired Intelligence, MOE Key Laboratory of Computational Neuroscience and Brain-Inspired Intelligence, Fudan University, Shanghai, China
| | - Yao Li
- BGI Research, Qingdao, China
| | - Tianyi Xia
- BGI Research, Beijing, China
- BGI Research, Shenzhen, China
| | | | | | | | - Lidong Guo
- BGI Research, Qingdao, China
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, China
| | | | - Mengnan Cheng
- BGI Research, Shenzhen, China
- BGI Research, Hangzhou, China
| | | | | | | | | | | | - Qin Lu
- BGI Research, Shenzhen, China
| | | | | | - Luni Hu
- BGI Research, Beijing, China
- BGI Research, Shenzhen, China
| | | | | | | | | | - Junhua Li
- BGI Research, Shenzhen, China
- Shenzhen Key Laboratory of Unknown Pathogen Identification, BGI Research, Shenzhen, China
- BGI Research, Riga, Latvia
| | - Mei Li
- BGI Research, Shenzhen, China
| | | | - Yi Zhao
- Beijing Key Laboratory of Mobile Computing and Pervasive Device, Institute of Computing Technology, Chinese Academy of Sciences, Beijing, China.
| | - Guangyi Fan
- BGI Research, Shenzhen, China.
- BGI Research, Qingdao, China.
| | - Yong Zhang
- BGI Research, Shenzhen, China.
- BGI Research, Wuhan, China.
- Guangdong Bigdata Engineering Technology Research Center for Life Sciences, BGI research, Shenzhen, China.
| | - Ao Chen
- BGI Research, Shenzhen, China.
| | - Yuxiang Li
- BGI Research, Shenzhen, China.
- BGI Research, Wuhan, China.
- Guangdong Bigdata Engineering Technology Research Center for Life Sciences, BGI research, Shenzhen, China.
| | - Xun Xu
- BGI Research, Wuhan, China.
| |
Collapse
|
13
|
Liu F, Ren S, Li J, Lv H, Jiang F, Bin Yu. SGTB: A graph representation learning model combining transformer and BERT for optimizing gene expression analysis in spatial transcriptomics data. Comput Biol Chem 2025; 118:108482. [PMID: 40306096 DOI: 10.1016/j.compbiolchem.2025.108482] [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: 03/09/2025] [Revised: 04/05/2025] [Accepted: 04/17/2025] [Indexed: 05/02/2025]
Abstract
In recent years, spatial transcriptomics (ST) has emerged as an innovative technology that enables the simultaneous acquisition of gene expression information and its spatial distribution at the single-cell or regional level, providing deeper insights into cellular interactions and tissue organization, this technology provides a more holistic view of tissue organization and intercellular dynamics. However, existing methods still face certain limitations in data representation capabilities, making it challenging to fully capture complex spatial dependencies and global features. To address this, this paper proposes an innovative spatial multi-scale graph convolutional network (SGTB) based on large language models, integrating graph convolutional networks (GCN), Transformer, and BERT language models to optimize the representation of spatial transcriptomics data. The Graph Convolutional Network (GCN) employs a multi-layer architecture to extract features from gene expression matrices. Through iterative aggregation of neighborhood information, it captures spatial dependencies among cells and gene co-expression patterns, thereby constructing hierarchical cell embeddings. Subsequently, the model integrates an attention mechanism to assign weights to critical features and leverages Transformer layers to model global relationships, refining the ability of learned representations to reflect variations in spatial patterns. Finally, the model incorporates the BERT language model, mapping cell embeddings into textual inputs to exploit its deep semantic representation capabilities for high-dimensional feature extraction. These features are then fused with the embeddings generated by the Transformer, further optimizing feature learning for spatial transcriptomics data. This approach holds significant application value in improving the accuracy of tasks such as cell type classification and gene regulatory network construction, providing a novel computational framework for deep mining of spatial multi-scale biological data.
Collapse
Affiliation(s)
- Farong Liu
- College of Mathematics and Physics, Qingdao University of Science and Technology, Qingdao 266061, China; School of Data Science, Qingdao University of Science and Technology, Qingdao 266061, China
| | - Sheng Ren
- School of Data Science, Qingdao University of Science and Technology, Qingdao 266061, China; Artificial Intelligence and Biomedical Big Data Research Center, Qingdao University of Science and Technology, Qingdao 266061, China
| | - Jie Li
- School of Data Science, Qingdao University of Science and Technology, Qingdao 266061, China; Artificial Intelligence and Biomedical Big Data Research Center, Qingdao University of Science and Technology, Qingdao 266061, China
| | - Haoyang Lv
- School of Data Science, Qingdao University of Science and Technology, Qingdao 266061, China; Artificial Intelligence and Biomedical Big Data Research Center, Qingdao University of Science and Technology, Qingdao 266061, China
| | - Fenghui Jiang
- Editorial Office of Journal of Qingdao University of Science and Technology (Natural Science Edition), Qingdao University of Science and Technology, Qingdao 266061, China.
| | - Bin Yu
- School of Data Science, Qingdao University of Science and Technology, Qingdao 266061, China; Artificial Intelligence and Biomedical Big Data Research Center, Qingdao University of Science and Technology, Qingdao 266061, China.
| |
Collapse
|
14
|
Liu SS, Wang S, Chen Y, Rustgi AK, Yuan M, Hu J. TransST: Transfer Learning Embedded Spatial Factor Modeling of Spatial Transcriptomics Data. ARXIV 2025:arXiv:2504.12353v1. [PMID: 40321945 PMCID: PMC12047910] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 05/11/2025]
Abstract
Background Spatial transcriptomics have emerged as a powerful tool in biomedical research because of its ability to capture both the spatial contexts and abundance of the complete RNA transcript profile in organs of interest. However, limitations of the technology such as the relatively low resolution and comparatively insufficient sequencing depth make it difficult to reliably extract real biological signals from these data. To alleviate this challenge, we propose a novel transfer learning framework, referred to as TransST, to adaptively leverage the cell-labeled information from external sources in inferring cell-level heterogeneity of a target spatial transcriptomics data. Results Applications in several real studies as well as a number of simulation settings show that our approach significantly improves existing techniques. For example, in the breast cancer study, TransST successfully identifies five biologically meaningful cell clusters, including the two subgroups of cancer in situ and invasive cancer; in addition, only TransST is able to separate the adipose tissues from the connective issues among all the studied methods. Conclusions In summary, the proposed method TransST is both effective and robust in identifying cell subclusters and detecting corresponding driving biomarkers in spatial transcriptomics data.
Collapse
Affiliation(s)
- Shuo Shuo Liu
- Department of Biostatistics, Columbia University, 10032, NY, United States
| | - Shikun Wang
- Department of Biostatistics, Columbia University, 10032, NY, United States
| | - Yuxuan Chen
- Department of Biostatistics, Columbia University, 10032, NY, United States
| | - Anil K. Rustgi
- Department of Medicine, Columbia University, 10027, NY, United States
| | - Ming Yuan
- Department of Statistics, Columbia University, 10027, NY, United States
| | - Jianhua Hu
- Department of Biostatistics, Columbia University, 10032, NY, United States
| |
Collapse
|
15
|
Kang L, Zhang Q, Qian F, Liang J, Wu X. Benchmarking computational methods for detecting spatial domains and domain-specific spatially variable genes from spatial transcriptomics data. Nucleic Acids Res 2025; 53:gkaf303. [PMID: 40240000 PMCID: PMC12000868 DOI: 10.1093/nar/gkaf303] [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: 11/02/2024] [Revised: 03/21/2025] [Accepted: 04/03/2025] [Indexed: 04/18/2025] Open
Abstract
Advances in spatially resolved transcriptomics (SRT) have led to the emergence of numerous computational methods for identifying spatial domains and spatially variable genes (SVGs); however, a comprehensive assessment of existing methods is lacking. We comprehensively benchmarked 19 methods for detecting spatial domains and domain-specific SVGs from SRT data, using 30 real-world datasets covering six SRT technologies and 27 synthetic datasets. We first evaluated the performance of these methods on spatial domain identification in terms of accuracy, stability, generalizability, and scalability. Results reveal that there is no single method that works best for all datasets, and the optimal method depends on the data, especially the SRT platform. Further, we proposed a quantitative strategy to evaluate domain-specific SVG recognition results and assessed the impact of spatial domains on SVG detection. We found that SVG detection based on spatial domains identified by different GNN methods have high accuracy but low concordance. Generally, the more accurate the recognized spatial domains, the higher the number and accuracy of domain-specific SVGs detected. Moreover, integrating spatial clustering results from different methods can lead to more robust and better clustering and SVG results. Practical guidelines were provided for choosing appropriate methods for spatial domain and domain-specific SVG identification.
Collapse
Affiliation(s)
- Liping Kang
- Department of Hematology, Children's Hospital of Soochow University, Suzhou 215000, China
- Cancer Institute, Suzhou Medical College, Soochow University, Suzhou 215000, China
| | - Qinglong Zhang
- Cancer Institute, Suzhou Medical College, Soochow University, Suzhou 215000, China
| | | | - Junyao Liang
- Cancer Institute, Suzhou Medical College, Soochow University, Suzhou 215000, China
| | - Xiaohui Wu
- Department of Hematology, Children's Hospital of Soochow University, Suzhou 215000, China
- Cancer Institute, Suzhou Medical College, Soochow University, Suzhou 215000, China
- MOE Key Laboratory of Geriatric Diseases and Immunology, Suzhou Medical College, Soochow University, Suzhou 215000, China
- Jiangsu Key Laboratory of Infection and Immunity, Soochow University, Suzhou 215000, China
- Pediatric Hematology & Oncology Key Laboratory of Higher Education Institutions in Jiangsu Province, Suzhou 215000, China
| |
Collapse
|
16
|
Liu F, He R, Sheeley T, Scheiblin D, Lockett SJ, Ridnour LA, Wink DA, Jensen M, Cortner J, Zaki G. SPAC: a scalable, integrated enterprise platform for end-to-end single cell spatial analysis of multiplexed tissue imaging. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.04.02.646782. [PMID: 40291751 PMCID: PMC12026498 DOI: 10.1101/2025.04.02.646782] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/30/2025]
Abstract
Background Multiplexed tissue imaging enables the simultaneous detection of dozens of proteins at single-cell resolution, providing unprecedented insights into tissue organization and disease microenvironments. However, the resulting high-dimensional, gigabyte-scale datasets pose significant computational and methodological challenges. Existing analytical workflows, often fragmented between bespoke scripts and static visualizations, lack the scalability and user-friendly interfaces required for efficient, reproducible analysis. To overcome these limitations, we developed SPAC (analysis of SPAtial single-Cell datasets), a scalable, web-based ecosystem that integrates modular pipelines, high-performance computing (HPC) connectivity, and interactive visualization to democratize end-to-end single-cell spatial analysis applied to cellular positional data and protein expression levels. Results SPAC is built on a modular, layered architecture that leverages community-based and newly developed tools for single-cell and spatial proteomics analysis. A specialized Python package extends these functionalities with custom analysis routines and established software engineering practices. An Interactive Analysis Layer provides web-hosted pipelines for configuring and executing complex workflows, and scalability enhancements that support distributed or parallel execution on GPU-enabled clusters. A Real-Time Visualization Layer delivers dynamic dashboards for immediate data exploration and sharing. As a showcase of its capabilities, SPAC was applied to a 4T1 breast cancer model, analyzing a multiplex imaging dataset comprising 2.6 million cells. GPU acceleration reduced unsupervised clustering runtimes from several hours to under ten minutes, and real-time visualization enabled detailed spatial characterization of tumor subregions. Conclusions SPAC effectively overcomes key challenges in spatial single-cell analysis by streamlining high-throughput data processing and spatial profiling within an accessible and scalable framework. Its robust architecture, interactive interface and ease of access have the potential to accelerate biomedical research and clinical applications by converting complex imaging data into actionable biological and clinical insights.
Collapse
|
17
|
Bao X, Bai X, Liu X, Shi Q, Zhang C. Spatially informed graph transformers for spatially resolved transcriptomics. Commun Biol 2025; 8:574. [PMID: 40188303 PMCID: PMC11972348 DOI: 10.1038/s42003-025-08015-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2024] [Accepted: 03/28/2025] [Indexed: 04/07/2025] Open
Abstract
Spatially resolved transcriptomics (SRT) has emerged as a powerful technique for mapping gene expression landscapes within spatial contexts. However, significant challenges persist in effectively integrating gene expression with spatial information to elucidate the heterogeneity of biological tissues. Here, we present a Spatially informed Graph Transformers framework, SpaGT, which leverages both node and edge channels to model spatially aware graph representation for denoising gene expression and identifying spatial domains. Unlike conventional graph neural networks, which rely on static, localized convolutional aggregation, SpaGT employs a structure-reinforced self-attention mechanism that iteratively evolves topological structural information and transcriptional signal representation. By replacing graph convolution with global self-attention, SpaGT enables the integration of both global and spatially localized information, thereby improving the detection of fine-grained spatial domains. We demonstrate that SpaGT achieves superior performance in identifying spatial domains and denoising gene expression data across diverse platforms and species. Furthermore, SpaGT facilitates the discovery of spatially variable genes with significant prognostic potential in cancer tissues. These findings establish SpaGT as a powerful tool for unraveling the complexities of biological tissues.
Collapse
Affiliation(s)
- Xinyu Bao
- 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, China
| | - Xiaosheng Bai
- 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, 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, China.
| | - Qianqian Shi
- Hubei Engineering Technology Research Center of Agricultural Big Data, Huazhong Agricultural University, Wuhan, China.
- Hubei Key Laboratory of Agricultural Bioinformatics, College of Informatics, Huazhong Agricultural University, Wuhan, China.
| | - Chuanchao Zhang
- 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, China.
| |
Collapse
|
18
|
Leng J, Yu J, Wu LY, Chen H. Flexible integration of spatial and expression information for precise spot embedding via ZINB-based graph-enhanced autoencoder. Commun Biol 2025; 8:556. [PMID: 40186054 PMCID: PMC11971412 DOI: 10.1038/s42003-025-07965-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2024] [Accepted: 03/19/2025] [Indexed: 04/07/2025] Open
Abstract
Domain identification is a critical problem in spatially resolved transcriptomics data analysis, which aims to identify distinct spatial domains within a tissue that maintain both spatial continuity and expression consistency. The degree of coupling between expression data and spatial information in different datasets often varies significantly. Some regions have intact and clear boundaries, while others exhibit blurred boundaries with high intra-domain expression similarity. However, most domain identification methods do not adequately integrate expression and spatial information to flexibly identify different types of domains. To address these issues, we introduce Spot2vector, a computational framework that leverages a graph-enhanced autoencoder integrating zero-inflated negative binomial distribution modeling, combining both graph convolutional networks and graph attention networks to extract the latent embeddings of spots. Spot2vector encodes and integrates spatial and expression information, enabling effective identification of domains with diverse spatial patterns across spatially resolved transcriptomics data generated by different platforms. The decoders enable us to decipher the distribution and generation mechanisms of data while improving expression quality through denoising. Extensive validation and analyses demonstrate that Spot2vector excels in enhancing domain identification accuracy, effectively reducing data dimensionality, improving expression recovery and denoising, and precisely capturing spatial gene expression patterns.
Collapse
Affiliation(s)
| | - Jiating Yu
- School of Mathematics and Statistics, Nanjing University of Information Science & Technology, Nanjing, 210044, China
| | - Ling-Yun Wu
- IAM, MADIS, NCMIS, Academy of Mathematics and Systems Science, Chinese Academy of Sciences, Beijing, 100190, China
- School of Mathematical Sciences, University of Chinese Academy of Sciences, Beijing, 100049, China
| | | |
Collapse
|
19
|
Min W, Fang D, Chen J, Zhang S. SpaMask: Dual masking graph autoencoder with contrastive learning for spatial transcriptomics. PLoS Comput Biol 2025; 21:e1012881. [PMID: 40179332 PMCID: PMC11968113 DOI: 10.1371/journal.pcbi.1012881] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2024] [Accepted: 02/17/2025] [Indexed: 04/05/2025] Open
Abstract
Understanding the spatial locations of cell within tissues is crucial for unraveling the organization of cellular diversity. Recent advancements in spatial resolved transcriptomics (SRT) have enabled the analysis of gene expression while preserving the spatial context within tissues. Spatial domain characterization is a critical first step in SRT data analysis, providing the foundation for subsequent analyses and insights into biological implications. Graph neural networks (GNNs) have emerged as a common tool for addressing this challenge due to the structural nature of SRT data. However, current graph-based deep learning approaches often overlook the instability caused by the high sparsity of SRT data. Masking mechanisms, as an effective self-supervised learning strategy, can enhance the robustness of these models. To this end, we propose SpaMask, dual masking graph autoencoder with contrastive learning for SRT analysis. Unlike previous GNNs, SpaMask masks a portion of spot nodes and spot-to-spot edges to enhance its performance and robustness. SpaMask combines Masked Graph Autoencoders (MGAE) and Masked Graph Contrastive Learning (MGCL) modules, with MGAE using node masking to leverage spatial neighbors for improved clustering accuracy, while MGCL applies edge masking to create a contrastive loss framework that tightens embeddings of adjacent nodes based on spatial proximity and feature similarity. We conducted a comprehensive evaluation of SpaMask on eight datasets from five different platforms. Compared to existing methods, SpaMask achieves superior clustering accuracy and effective batch correction.
Collapse
Affiliation(s)
- Wenwen Min
- School of Information Science and Engineering, Yunnan University, Kunming, Yunnan, China
| | - Donghai Fang
- School of Information Science and Engineering, Yunnan University, Kunming, Yunnan, China
| | - Jinyu Chen
- School of Mathematics, Statistics and Mechanics, Beijing University of Technology, Beijing, China
| | - Shihua Zhang
- NCMIS, CEMS, RCSDS, Academy of Mathematics and Systems Science, Chinese Academy of Sciences, Beijing, China
- School of Mathematical Sciences, University of Chinese Academy of Sciences, Beijing, China
- 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, Zhejiang, China
| |
Collapse
|
20
|
Birk S, Bonafonte-Pardàs I, Feriz AM, Boxall A, Agirre E, Memi F, Maguza A, Yadav A, Armingol E, Fan R, Castelo-Branco G, Theis FJ, Bayraktar OA, Talavera-López C, Lotfollahi M. Quantitative characterization of cell niches in spatially resolved omics data. Nat Genet 2025; 57:897-909. [PMID: 40102688 PMCID: PMC11985353 DOI: 10.1038/s41588-025-02120-6] [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: 03/21/2024] [Accepted: 02/05/2025] [Indexed: 03/20/2025]
Abstract
Spatial omics enable the characterization of colocalized cell communities that coordinate specific functions within tissues. These communities, or niches, are shaped by interactions between neighboring cells, yet existing computational methods rarely leverage such interactions for their identification and characterization. To address this gap, here we introduce NicheCompass, a graph deep-learning method that models cellular communication to learn interpretable cell embeddings that encode signaling events, enabling the identification of niches and their underlying processes. Unlike existing methods, NicheCompass quantitatively characterizes niches based on communication pathways and consistently outperforms alternatives. We show its versatility by mapping tissue architecture during mouse embryonic development and delineating tumor niches in human cancers, including a spatial reference mapping application. Finally, we extend its capabilities to spatial multi-omics, demonstrate cross-technology integration with datasets from different sequencing platforms and construct a whole mouse brain spatial atlas comprising 8.4 million cells, highlighting NicheCompass' scalability. Overall, NicheCompass provides a scalable framework for identifying and analyzing niches through signaling events.
Collapse
Affiliation(s)
- Sebastian Birk
- Institute of AI for Health, Helmholtz Center Munich-German Research Center for Environmental Health, Neuherberg, Germany
- School of Computation, Information and Technology, Technical University of Munich, Munich, Germany
- Würzburg Institute of Systems Immunology (WüSI), University of Würzburg, Würzburg, Germany
- Wellcome Sanger Institute, Wellcome Genome Campus, Cambridge, UK
| | - Irene Bonafonte-Pardàs
- Institute of Computational Biology, Helmholtz Center Munich-German Research Center for Environmental Health, Neuherberg, Germany
- Biomedical Center (BMC), Physiological Chemistry, Faculty of Medicine, Ludwig Maximilian University of Munich, Planegg-Martinsried, Germany
| | | | - Adam Boxall
- Wellcome Sanger Institute, Wellcome Genome Campus, Cambridge, UK
| | - Eneritz Agirre
- Laboratory of Molecular Neurobiology, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden
| | - Fani Memi
- Wellcome Sanger Institute, Wellcome Genome Campus, Cambridge, UK
| | - Anna Maguza
- Würzburg Institute of Systems Immunology (WüSI), University of Würzburg, Würzburg, Germany
- Faculty of Medicine, University of Würzburg, Würzburg, Germany
| | - Anamika Yadav
- Wellcome Sanger Institute, Wellcome Genome Campus, Cambridge, UK
| | - Erick Armingol
- Wellcome Sanger Institute, Wellcome Genome Campus, Cambridge, UK
| | - Rong Fan
- Department of Biomedical Engineering, Yale University, New Haven, CT, USA
- Yale Stem Cell Center and Yale Cancer Center, Yale University School of Medicine, New Haven, CT, USA
- Department of Pathology, Yale University School of Medicine, New Haven, CT, USA
- Human and Translational Immunology Program, Yale University School of Medicine, New Haven, CT, USA
| | - Gonçalo Castelo-Branco
- Laboratory of Molecular Neurobiology, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden
- Ming Wai Lau Centre for Reparative Medicine, Stockholm Node, Karolinska Institutet, Stockholm, Sweden
| | - Fabian J Theis
- School of Computation, Information and Technology, Technical University of Munich, Munich, Germany
- Wellcome Sanger Institute, Wellcome Genome Campus, Cambridge, UK
- Institute of Computational Biology, Helmholtz Center Munich-German Research Center for Environmental Health, Neuherberg, Germany
- School of Life Sciences Weihenstephan, Technical University of Munich, Munich, Germany
| | | | - Carlos Talavera-López
- Würzburg Institute of Systems Immunology (WüSI), University of Würzburg, Würzburg, Germany.
- Faculty of Medicine, University of Würzburg, Würzburg, Germany.
| | - Mohammad Lotfollahi
- Wellcome Sanger Institute, Wellcome Genome Campus, Cambridge, UK.
- Institute of Computational Biology, Helmholtz Center Munich-German Research Center for Environmental Health, Neuherberg, Germany.
| |
Collapse
|
21
|
Tu W, Zhang L. Integrating multiple spatial transcriptomics data using community-enhanced graph contrastive learning. PLoS Comput Biol 2025; 21:e1012948. [PMID: 40179111 PMCID: PMC11990772 DOI: 10.1371/journal.pcbi.1012948] [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: 11/10/2024] [Revised: 04/11/2025] [Accepted: 03/10/2025] [Indexed: 04/05/2025] Open
Abstract
Due to the rapid development of spatial sequencing technologies, large amounts of spatial transcriptomic datasets have been generated across various technological platforms or different biological conditions (e.g., control vs. treatment). Spatial transcriptomics data coming from different platforms usually has different resolutions. Moreover, current methods do not consider the heterogeneity of spatial structures within and across slices when modeling spatial transcriptomics data with graph-based methods. In this study, we propose a community-enhanced graph contrastive learning-based method named Tacos to integrate multiple spatial transcriptomics data. We applied Tacos to several real datasets coming from different platforms under different scenarios. Systematic benchmark analyses demonstrate Tacos's superior performance in integrating different slices. Furthermore, Tacos can accurately denoise the spatially resolved transcriptomics data.
Collapse
Affiliation(s)
- Wenqian Tu
- School of Artificial Intelligence, School of Computer Science, Wuhan University, Wuhan, China
| | - Lihua Zhang
- School of Artificial Intelligence, School of Computer Science, Wuhan University, Wuhan, China
| |
Collapse
|
22
|
Chen Y, Zhen C, Mo Y, Liu J, Zhang L. Multiscale Dissection of Spatial Heterogeneity by Integrating Multi-Slice Spatial and Single-Cell Transcriptomics. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2413124. [PMID: 39999288 PMCID: PMC12005799 DOI: 10.1002/advs.202413124] [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: 10/17/2024] [Revised: 02/05/2025] [Indexed: 02/27/2025]
Abstract
The spatial structure of cells is highly organized at multiscale levels from global spatial domains to local cell type heterogeneity. Existing methods for analyzing spatially resolved transcriptomics (SRT) are separately designed for either domain alignment across multiple slices or deconvoluting cell type compositions within a single slice. To this end, a novel deep learning method, SMILE, is proposed which combines graph contrastive autoencoder and multilayer perceptron with local constraints to learn multiscale and informative spot representations. By comparing SMILE with the state-of-the-art methods on simulation and real datasets, the superior performance of SMILE is demonstrated on spatial alignment, domain identification, and cell type deconvolution. The results show SMILE's capability not only in simultaneously dissecting spatial variations at different scales but also in unraveling altered cellular microenvironments in diseased conditions. Moreover, SMILE can utilize prior domain annotation information of one slice to further enhance the performance.
Collapse
Affiliation(s)
- Yuqi Chen
- School of Computer ScienceWuhan UniversityWuchang DistrictWuhanHubei430072China
| | - Caiwei Zhen
- School of Computer ScienceWuhan UniversityWuchang DistrictWuhanHubei430072China
| | - Yuanyuan Mo
- School of Computer ScienceWuhan UniversityWuchang DistrictWuhanHubei430072China
| | - Juan Liu
- School of Computer ScienceWuhan UniversityWuchang DistrictWuhanHubei430072China
| | - Lihua Zhang
- School of Computer ScienceWuhan UniversityWuchang DistrictWuhanHubei430072China
| |
Collapse
|
23
|
Wang H, Li J, Jing S, Lin P, Qiu Y, Yan X, Yuan J, Tang Z, Li Y, Zhang H, Chen Y, Wang Z, Li H. SOAPy: a Python package to dissect spatial architecture, dynamics, and communication. Genome Biol 2025; 26:80. [PMID: 40158115 PMCID: PMC11954224 DOI: 10.1186/s13059-025-03550-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2024] [Accepted: 03/18/2025] [Indexed: 04/01/2025] Open
Abstract
Advances in spatial omics enable deeper insights into tissue microenvironments while posing computational challenges. Therefore, we developed SOAPy, a comprehensive tool for analyzing spatial omics data, which offers methods for spatial domain identification, spatial expression tendency, spatiotemporal expression pattern, cellular co-localization, multi-cellular niches, cell-cell communication, and so on. SOAPy can be applied to diverse spatial omics technologies and multiple areas in physiological and pathological contexts, such as tumor biology and developmental biology. Its versatility and robust performance make it a universal platform for spatial omics analysis, providing diverse insights into the dynamics and architecture of tissue microenvironments.
Collapse
Affiliation(s)
- Heqi Wang
- CAS Key Laboratory of Computational Biology, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, 200031, China
| | - Jiarong Li
- CAS Key Laboratory of Computational Biology, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, 200031, China
| | - Siyu Jing
- CAS Key Laboratory of Computational Biology, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, 200031, China
| | - Ping Lin
- CAS Key Laboratory of Computational Biology, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, 200031, China
| | - Yiling Qiu
- CAS Key Laboratory of Computational Biology, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, 200031, China
| | - Xi Yan
- CAS Key Laboratory of Computational Biology, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, 200031, China
| | - Jiao Yuan
- CAS Key Laboratory of Computational Biology, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, 200031, China
| | - ZhiXuan Tang
- CAS Key Laboratory of Computational Biology, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, 200031, China
| | - Yu Li
- CAS Key Laboratory of Nutrition, Metabolism and Food Safety, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, 200031, China
| | - Haibing Zhang
- CAS Key Laboratory of Nutrition, Metabolism and Food Safety, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, 200031, China
| | - Yujie Chen
- CAS Key Laboratory of Computational Biology, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, 200031, China
| | - Zhen Wang
- CAS Key Laboratory of Computational Biology, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, 200031, China
| | - Hong Li
- CAS Key Laboratory of Computational Biology, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, 200031, China.
| |
Collapse
|
24
|
Chen X, Ran Q, Tang J, Chen Z, Huang S, Shi X, Xi R. Benchmarking algorithms for spatially variable gene identification in spatial transcriptomics. Bioinformatics 2025; 41:btaf131. [PMID: 40139667 PMCID: PMC12036962 DOI: 10.1093/bioinformatics/btaf131] [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: 08/13/2024] [Revised: 03/15/2025] [Accepted: 03/27/2025] [Indexed: 03/29/2025] Open
Abstract
MOTIVATION The rapid development of spatial transcriptomics has underscored the importance of identifying spatially variable genes. As a fundamental task in spatial transcriptomic data analysis, spatially variable gene identification has been extensively studied. However, the lack of comprehensive benchmark makes it difficult to validate the effectiveness of various algorithms scattered across a large number of studies with real-world datasets. RESULTS In response, this article proposes a benchmark framework to evaluate algorithms for identifying spatially variable genes through the analysis of 30 synthesized and 74 real-world datasets, aiming to identify the best algorithms and their corresponding application scenarios. This framework can assist medical and life scientists in selecting suitable algorithms for their research, while also aid bioinformatics scientists in developing more powerful and efficient computational methods in spatial transcriptomic research. AVAILABILITY AND IMPLEMENTATION The source code of this benchmarking framework is available at both Github (https://github.com/XiDsLab/svg-benchmark) and Zenodo (https://doi.org/10.5281/zenodo.15031083). In addition, all real and synthetic datasets considered in this study are also publicly available at Zenodo (https://doi.org/10.5281/zenodo.7227771).
Collapse
Affiliation(s)
- Xuanwei Chen
- School of Mathematical Sciences, Peking University, Beijing 100871, China
| | - Qinghua Ran
- Academy for Advanced Interdisciplinary Studies, Peking University, Beijing 100871, China
| | - Junjie Tang
- Center for Statistical Science, Peking University, Beijing 100871, China
| | - Zihao Chen
- School of Mathematical Sciences, Peking University, Beijing 100871, China
| | - Siyuan Huang
- Academy for Advanced Interdisciplinary Studies, Peking University, Beijing 100871, China
| | - Xingjie Shi
- KLATASDS-MOE, Academy of Statistics and Interdisciplinary Sciences, School of Statistics, East China Normal University, Shanghai 200062, China
| | - Ruibin Xi
- School of Mathematical Sciences, Peking University, Beijing 100871, China
- Academy for Advanced Interdisciplinary Studies, Peking University, Beijing 100871, China
- Center for Statistical Science, Peking University, Beijing 100871, China
| |
Collapse
|
25
|
Fu Y, Nan M, Ren Q, Chen X, Gao J. A multi-view graph convolutional network framework based on adaptive adjacency matrix and multi-strategy fusion mechanism for identifying spatial domains. Bioinformatics 2025; 41:btaf172. [PMID: 40233119 PMCID: PMC12041416 DOI: 10.1093/bioinformatics/btaf172] [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: 12/23/2024] [Revised: 03/31/2025] [Accepted: 04/11/2025] [Indexed: 04/17/2025] Open
Abstract
MOTIVATION Spatial transcriptomics (ST) addresses the loss of spatial context in single-cell RNA-sequencing by simultaneously capturing gene expression and spatial location information. A critical task of ST is the identification of spatial domains. However, challenges such as high noise levels and data sparsity make the identification process more difficult. RESULTS To tackle these challenges, STMGAMF, a multi-view graph convolutional network model that employs an adaptive adjacency matrix and a multi-strategy fusion mechanism is proposed. STMGAMF dynamically adjusts the edge weights to capture complex spatial structures during training by implementing the adaptive adjacency matrix and optimizes the embedded features through the multi-strategy fusion mechanism. STMGAMF is evaluated on multiple ST datasets and outperforms existing algorithms in tasks like spatial domain identification, visualization, and spatial trajectory inference. Its robust performance in spatial domain identification and strong generalization capability position STMGAMF as a valuable tool for unraveling the complexity of tissue structures and underlying biological processes. AVAILABILITY AND IMPLEMENTATION Source code is available at Github (https://github.com/Fuyh0628/STMGAMF) and Zenodo (https://zenodo.org/records/15103358).
Collapse
Affiliation(s)
- Yuhan Fu
- School of Science, Jiangnan University, Wuxi, Jiangsu 214122, China
| | - Mengdi Nan
- School of Science, Jiangnan University, Wuxi, Jiangsu 214122, China
| | - Qing Ren
- School of Science, Jiangnan University, Wuxi, Jiangsu 214122, China
| | - Xiang Chen
- School of Science, Jiangnan University, Wuxi, Jiangsu 214122, China
| | - Jie Gao
- School of Science, Jiangnan University, Wuxi, Jiangsu 214122, China
| |
Collapse
|
26
|
Dong M, Su DG, Kluger H, Fan R, Kluger Y. SIMVI disentangles intrinsic and spatial-induced cellular states in spatial omics data. Nat Commun 2025; 16:2990. [PMID: 40148341 PMCID: PMC11950362 DOI: 10.1038/s41467-025-58089-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2025] [Accepted: 03/05/2025] [Indexed: 03/29/2025] Open
Abstract
Spatial omics technologies enable analysis of gene expression and interaction dynamics in relation to tissue structure and function. However, existing computational methods may not properly distinguish cellular intrinsic variability and intercellular interactions, and may thus fail to reliably capture spatial regulations. Here, we present Spatial Interaction Modeling using Variational Inference (SIMVI), an annotation-free deep learning framework that disentangles cell intrinsic and spatial-induced latent variables in spatial omics data with rigorous theoretical support. By this disentanglement, SIMVI enables estimation of spatial effects at a single-cell resolution, and empowers various downstream analyses. We demonstrate the superior performance of SIMVI across datasets from diverse platforms and tissues. SIMVI illuminates the cyclical spatial dynamics of germinal center B cells in human tonsil. Applying SIMVI to multiome melanoma data reveals potential tumor epigenetic reprogramming states. On our newly-collected cohort-level CosMx melanoma data, SIMVI uncovers space-and-outcome-dependent macrophage states and cellular communication machinery in tumor microenvironments.
Collapse
Affiliation(s)
- Mingze Dong
- Interdepartmental Program in Computational Biology & Bioinformatics, Yale University, New Haven, CT, USA
- Department of Pathology, Yale School of Medicine, New Haven, CT, USA
- Department of Biomedical Engineering, Yale University, New Haven, CT, USA
| | - David G Su
- Department of Medicine, Yale School of Medicine, New Haven, CT, USA
- Yale Cancer Center, Yale School of Medicine, New Haven, CT, USA
- Yale Center for Immuno-Oncology, Yale School of Medicine, New Haven, CT, USA
- Department of Surgery, Yale School of Medicine, New Haven, CT, USA
| | - Harriet Kluger
- Department of Medicine, Yale School of Medicine, New Haven, CT, USA
- Yale Cancer Center, Yale School of Medicine, New Haven, CT, USA
- Yale Center for Immuno-Oncology, Yale School of Medicine, New Haven, CT, USA
| | - Rong Fan
- Department of Pathology, Yale School of Medicine, New Haven, CT, USA
- Department of Biomedical Engineering, Yale University, New Haven, CT, USA
- Yale Cancer Center, Yale School of Medicine, New Haven, CT, USA
| | - Yuval Kluger
- Interdepartmental Program in Computational Biology & Bioinformatics, Yale University, New Haven, CT, USA.
- Department of Pathology, Yale School of Medicine, New Haven, CT, USA.
- Applied Mathematics Program, Yale University, New Haven, CT, USA.
| |
Collapse
|
27
|
Wang Y, Liu Z, Ma X. MuCST: restoring and integrating heterogeneous morphology images and spatial transcriptomics data with contrastive learning. Genome Med 2025; 17:21. [PMID: 40082941 PMCID: PMC11907906 DOI: 10.1186/s13073-025-01449-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2024] [Accepted: 03/07/2025] [Indexed: 03/16/2025] Open
Abstract
Spatially resolved transcriptomics (SRT) simultaneously measure spatial location, histology images, and transcriptional profiles of cells or regions in undissociated tissues. Integrative analysis of multi-modal SRT data holds immense potential for understanding biological mechanisms. Here, we present a flexible multi-modal contrastive learning for the integration of SRT data (MuCST), which joins denoising, heterogeneity elimination, and compatible feature learning. MuCST accurately identifies spatial domains and is applicable to diverse datasets platforms. Overall, MuCST provides an alternative for integrative analysis of multi-modal SRT data ( https://github.com/xkmaxidian/MuCST ).
Collapse
Affiliation(s)
- Yu Wang
- School of Computer Science and Technology, Xidian University, No.2 South Taibai Road, Xi'an, 710071, Shaanxi, China
- Key Laboratory of Smart Human-Computer Interaction and Wearable Technology of Shaanxi Province, Xidian University, No.2 South Taibai Road, Xi'an, 710071, Shaanxi, China
| | - Zaiyi Liu
- Department of Radiology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, 106 Zhongshan Er Road, Guangzhou, 510080, Guangdong, China
- Guangdong Provincial Key Laboratory of Artificial Intelligence in Medical Image Analysis and Application, 106 Zhongshan Er Road, Guangzhou, 510080, Guangdong, China
| | - Xiaoke Ma
- School of Computer Science and Technology, Xidian University, No.2 South Taibai Road, Xi'an, 710071, Shaanxi, China.
- Key Laboratory of Smart Human-Computer Interaction and Wearable Technology of Shaanxi Province, Xidian University, No.2 South Taibai Road, Xi'an, 710071, Shaanxi, China.
| |
Collapse
|
28
|
Fang Z, Krusen K, Priest H, Wang M, Kim S, Sriram A, Yellanki A, Singh A, Horwitz E, Coskun AF. Graph-Based 3-Dimensional Spatial Gene Neighborhood Networks of Single Cells in Gels and Tissues. BME FRONTIERS 2025; 6:0110. [PMID: 40084126 PMCID: PMC11906096 DOI: 10.34133/bmef.0110] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2024] [Revised: 02/12/2025] [Accepted: 02/13/2025] [Indexed: 03/16/2025] Open
Abstract
Objective: We developed 3-dimensional spatially resolved gene neighborhood network embedding (3D-spaGNN-E) to find subcellular gene proximity relationships and identify key subcellular motifs in cell-cell communication (CCC). Impact Statement: The pipeline combines 3D imaging-based spatial transcriptomics and graph-based deep learning to identify subcellular motifs. Introduction: Advancements in imaging and experimental technology allow the study of 3D spatially resolved transcriptomics and capture better spatial context than approximating the samples as 2D. However, the third spatial dimension increases the data complexity and requires new analyses. Methods: 3D-spaGNN-E detects single transcripts in 3D cell culture samples and identifies subcellular gene proximity relationships. Then, a graph autoencoder projects the gene proximity relationships into a latent space. We then applied explainability analysis to identify subcellular CCC motifs. Results: We first applied the pipeline to mesenchymal stem cells (MSCs) cultured in hydrogel. After clustering the cells based on the RNA count, we identified cells belonging to the same cluster as homotypic and those belonging to different clusters as heterotypic. We identified changes in local gene proximity near the border between homotypic and heterotypic cells. When applying the pipeline to the MSC-peripheral blood mononuclear cell (PBMC) coculture system, we identified CD4+ and CD8+ T cells. Local gene proximity and autoencoder embedding changes can distinguish strong and weak suppression of different immune cells. Lastly, we compared astrocyte-neuron CCC in mouse hypothalamus and cortex by analyzing 3D multiplexed-error-robust fluorescence in situ hybridization (MERFISH) data and identified regional gene proximity differences. Conclusion: 3D-spaGNN-E distinguished distinct CCCs in cell culture and tissue by examining subcellular motifs.
Collapse
Affiliation(s)
- Zhou Fang
- Wallace H. Coulter Department of Biomedical Engineering,
Georgia Institute of Technology and Emory University, Atlanta, GA, USA
- Machine Learning Graduate Program,
Georgia Institute of Technology, Atlanta, GA, USA
| | - Kelsey Krusen
- Wallace H. Coulter Department of Biomedical Engineering,
Georgia Institute of Technology and Emory University, Atlanta, GA, USA
| | - Hannah Priest
- Wallace H. Coulter Department of Biomedical Engineering,
Georgia Institute of Technology and Emory University, Atlanta, GA, USA
| | - Mingshuang Wang
- Wallace H. Coulter Department of Biomedical Engineering,
Georgia Institute of Technology and Emory University, Atlanta, GA, USA
| | - Sungwoong Kim
- Wallace H. Coulter Department of Biomedical Engineering,
Georgia Institute of Technology and Emory University, Atlanta, GA, USA
| | - Anirudh Sriram
- Wallace H. Coulter Department of Biomedical Engineering,
Georgia Institute of Technology and Emory University, Atlanta, GA, USA
| | - Ashritha Yellanki
- Wallace H. Coulter Department of Biomedical Engineering,
Georgia Institute of Technology and Emory University, Atlanta, GA, USA
| | - Ankur Singh
- Wallace H. Coulter Department of Biomedical Engineering,
Georgia Institute of Technology and Emory University, Atlanta, GA, USA
- Woodruff School of Mechanical Engineering,
Georgia Institute of Technology, Atlanta, GA, USA
- Interdisciplinary Bioengineering Graduate Program,
Georgia Institute of Technology, Atlanta, GA, USA
- Parker H. Petit Institute for Bioengineering and Bioscience, GeorgiaInstitute of Technology, Atlanta, GA 30332, USA
| | - Edwin Horwitz
- Department of Pediatrics,
Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Ahmet F. Coskun
- Wallace H. Coulter Department of Biomedical Engineering,
Georgia Institute of Technology and Emory University, Atlanta, GA, USA
- Interdisciplinary Bioengineering Graduate Program,
Georgia Institute of Technology, Atlanta, GA, USA
- Parker H. Petit Institute for Bioengineering and Bioscience, GeorgiaInstitute of Technology, Atlanta, GA 30332, USA
| |
Collapse
|
29
|
Zhang J, Wang J, Zhou Q, Chen Z, Zhuang J, Zhao X, Gan Z, Wang Y, Wang C, Molday RS, Yang YT, Li X, Zhao XM. Spatiotemporally resolved transcriptomics reveals the cellular dynamics of human retinal development. Nat Commun 2025; 16:2307. [PMID: 40055379 PMCID: PMC11889126 DOI: 10.1038/s41467-025-57625-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2024] [Accepted: 02/24/2025] [Indexed: 05/13/2025] Open
Abstract
The morphogenesis and cellular interactions in developing retina are incompletely characterized. The full understanding needs a precise mapping of the gene expression with a single-cell spatial resolution. Here, we present a spatial transcriptomic (ST) resource for the developing human retina at six developmental stages. Combining the spatial and single-cell transcriptomic data enables characterization of the cell-type-specific expression profiles at distinct anatomical regions at each developmental stage, highlighting the spatiotemporal dynamics of cellular composition during retinal development. All the ST spots are catalogued into consensus spatial domains, which are further associated to their specific expression signatures and biological functions associated with neuron and eye development. We prioritize a set of critical regulatory genes for the transitions of spatial domains during retinal development. Differentially expressed genes from different spatial domains are associated with distinct retinal diseases, indicating the biological relevance and clinical significance of the spatially defined gene expression. Finally, we reconstruct the spatial cellular communication networks, and highlight critical ligand-receptor interactions during retinal development. Overall, our study reports the spatiotemporal dynamics of gene expression and cellular profiles during retinal development, and provides a rich resource for the future studies on retinogenesis.
Collapse
Affiliation(s)
- Jinglong Zhang
- Department of Neurology, Zhongshan Hospital and Institute of Science and Technology for Brain-Inspired Intelligence, Fudan University, Shanghai, China
- MOE Key Laboratory of Computational Neuroscience and Brain-Inspired Intelligence, and MOE Frontiers Center for Brain Science, Fudan University, Shanghai, China
| | - Jiao Wang
- School of Life Sciences, Shanghai University, Shanghai, China
| | - Qiongjie Zhou
- Department of Obstetrics, Obstetrics and Gynecology Hospital of Fudan University, Shanghai, China
| | - Zixin Chen
- School of Life Sciences, Shanghai University, Shanghai, China
| | - Junyi Zhuang
- School of Life Sciences, Shanghai University, Shanghai, China
| | - Xingzhong Zhao
- Department of Neurology, Zhongshan Hospital and Institute of Science and Technology for Brain-Inspired Intelligence, Fudan University, Shanghai, China
- MOE Key Laboratory of Computational Neuroscience and Brain-Inspired Intelligence, and MOE Frontiers Center for Brain Science, Fudan University, Shanghai, China
| | - Ziquan Gan
- Department of Neurology, Zhongshan Hospital and Institute of Science and Technology for Brain-Inspired Intelligence, Fudan University, Shanghai, China
- MOE Key Laboratory of Computational Neuroscience and Brain-Inspired Intelligence, and MOE Frontiers Center for Brain Science, Fudan University, Shanghai, China
| | - Yinan Wang
- Department of Obstetrics, Obstetrics and Gynecology Hospital of Fudan University, Shanghai, China
| | - Chunxiu Wang
- Department of Obstetrics, Obstetrics and Gynecology Hospital of Fudan University, Shanghai, China
| | - Robert S Molday
- Department of Biochemistry & Molecular Biology, University of British Columbia, Vancouver, BC, Canada
- Department of Ophthalmology & Visual Sciences, University of British Columbia, Vancouver, BC, Canada
| | - Yucheng T Yang
- Department of Neurology, Zhongshan Hospital and Institute of Science and Technology for Brain-Inspired Intelligence, Fudan University, Shanghai, China.
- MOE Key Laboratory of Computational Neuroscience and Brain-Inspired Intelligence, and MOE Frontiers Center for Brain Science, Fudan University, Shanghai, China.
| | - Xiaotian Li
- Department of Obstetrics, Obstetrics and Gynecology Hospital of Fudan University, Shanghai, China.
- Department of Obstetrics, Shenzhen Maternity and Child Healthcare Hospital, Southern Medical University, Shenzhen, Guangdong, China.
| | - Xing-Ming Zhao
- Department of Neurology, Zhongshan Hospital and Institute of Science and Technology for Brain-Inspired Intelligence, Fudan University, Shanghai, China.
- MOE Key Laboratory of Computational Neuroscience and Brain-Inspired Intelligence, and MOE Frontiers Center for Brain Science, Fudan University, Shanghai, China.
- Huzhou Central Hospital, Affiliated Central Hospital Huzhou University, Huzhou, Zhejiang, China.
- State Key Laboratory of Medical Neurobiology, Institutes of Brain Science, Fudan University, Shanghai, China.
| |
Collapse
|
30
|
Li S, Hua H, Chen S. Graph neural networks for single-cell omics data: a review of approaches and applications. Brief Bioinform 2025; 26:bbaf109. [PMID: 40091193 PMCID: PMC11911123 DOI: 10.1093/bib/bbaf109] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2024] [Revised: 02/09/2025] [Accepted: 02/25/2025] [Indexed: 03/19/2025] Open
Abstract
Rapid advancement of sequencing technologies now allows for the utilization of precise signals at single-cell resolution in various omics studies. However, the massive volume, ultra-high dimensionality, and high sparsity nature of single-cell data have introduced substantial difficulties to traditional computational methods. The intricate non-Euclidean networks of intracellular and intercellular signaling molecules within single-cell datasets, coupled with the complex, multimodal structures arising from multi-omics joint analysis, pose significant challenges to conventional deep learning operations reliant on Euclidean geometries. Graph neural networks (GNNs) have extended deep learning to non-Euclidean data, allowing cells and their features in single-cell datasets to be modeled as nodes within a graph structure. GNNs have been successfully applied across a broad range of tasks in single-cell data analysis. In this survey, we systematically review 107 successful applications of GNNs and their six variants in various single-cell omics tasks. We begin by outlining the fundamental principles of GNNs and their six variants, followed by a systematic review of GNN-based models applied in single-cell epigenomics, transcriptomics, spatial transcriptomics, proteomics, and multi-omics. In each section dedicated to a specific omics type, we have summarized the publicly available single-cell datasets commonly utilized in the articles reviewed in that section, totaling 77 datasets. Finally, we summarize the potential shortcomings of current research and explore directions for future studies. We anticipate that this review will serve as a guiding resource for researchers to deepen the application of GNNs in single-cell omics.
Collapse
Affiliation(s)
- Sijie Li
- School of Mathematical Sciences and The Key Laboratory of Pure Mathematics and Combinatorics, Ministry of Education (LPMC), Nankai University, No. 94 Weijin Road, Nankai District, Tianjin 300071, China
| | - Heyang Hua
- School of Mathematical Sciences and The Key Laboratory of Pure Mathematics and Combinatorics, Ministry of Education (LPMC), Nankai University, No. 94 Weijin Road, Nankai District, Tianjin 300071, China
| | - Shengquan Chen
- School of Mathematical Sciences and The Key Laboratory of Pure Mathematics and Combinatorics, Ministry of Education (LPMC), Nankai University, No. 94 Weijin Road, Nankai District, Tianjin 300071, China
| |
Collapse
|
31
|
Pang Y, Wang C, Zhang YZ, Wang Z, Imoto S, Lee TY. STForte: tissue context-specific encoding and consistency-aware spatial imputation for spatially resolved transcriptomics. Brief Bioinform 2025; 26:bbaf174. [PMID: 40254832 PMCID: PMC12009714 DOI: 10.1093/bib/bbaf174] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2025] [Revised: 03/06/2025] [Accepted: 03/17/2025] [Indexed: 04/22/2025] Open
Abstract
Encoding spatially resolved transcriptomics (SRT) data serves to identify the biological semantics of RNA expression within the tissue while preserving spatial characteristics. Depending on the analytical scenario, one may focus on different contextual structures of tissues. For instance, anatomical regions reveal consistent patterns by focusing on spatial homogeneity, while elucidating complex tumor micro-environments requires more expression heterogeneity. However, current spatial encoding methods lack consideration of the tissue context. Meanwhile, most developed SRT technologies are still limited in providing exact patterns of intact tissues due to limitations such as low resolution or missed measurements. Here, we propose STForte, a novel pairwise graph autoencoder-based approach with cross-reconstruction and adversarial distribution matching, to model the spatial homogeneity and expression heterogeneity of SRT data. STForte extracts interpretable latent encodings, enabling downstream analysis by accurately portraying various tissue contexts. Moreover, STForte allows spatial imputation using only spatial consistency to restore the biological patterns of unobserved locations or low-quality cells, thereby providing fine-grained views to enhance the SRT analysis. Extensive evaluations of datasets under different scenarios and SRT platforms demonstrate that STForte is a scalable and versatile tool for providing enhanced insights into spatial data analysis.
Collapse
Affiliation(s)
- Yuxuan Pang
- Division of Health Medical Intelligence, Human Genome Center, The Institute of Medical Science, The University of Tokyo, 4-6-1, Shirokanedai, Minato-ku, Tokyo, 108-8639, Japan
| | - Chunxuan Wang
- School of Data Science, The Chinese University of Hong Kong, Shenzhen (CUHK-Shenzhen), 2001 Longxiang Road, Longgang, Shenzhen, 518172, China
| | - Yao-zhong Zhang
- Division of Health Medical Intelligence, Human Genome Center, The Institute of Medical Science, The University of Tokyo, 4-6-1, Shirokanedai, Minato-ku, Tokyo, 108-8639, Japan
| | - Zhuo Wang
- Warshel Institute for Computational Biology, School of Medicine, The Chinese University of Hong Kong, Shenzhen (CUHK-Shenzhen), 2001 Longxiang Road, Longgang, Shenzhen, 518172, China
- School of Medicine, The Chinese University of Hong Kong, Shenzhen (CUHK-Shenzhen), 2001 Longxiang Road, Longgang, Shenzhen, 518172, China
| | - Seiya Imoto
- Division of Health Medical Intelligence, Human Genome Center, The Institute of Medical Science, The University of Tokyo, 4-6-1, Shirokanedai, Minato-ku, Tokyo, 108-8639, Japan
| | - Tzong-Yi Lee
- Institute of Bioinformatics and Systems Biology, National Yang Ming Chiao Tung University, No. 75 Bo-Ai Street, Hsinchu 300, Taiwan
| |
Collapse
|
32
|
Wei X, Chen T, Wang X, Shen W, Liu C, Wu S, Wong HS. COME: contrastive mapping learning for spatial reconstruction of single-cell RNA sequencing data. Bioinformatics 2025; 41:btaf083. [PMID: 39992219 PMCID: PMC11897431 DOI: 10.1093/bioinformatics/btaf083] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2024] [Revised: 02/12/2025] [Accepted: 02/20/2025] [Indexed: 02/25/2025] Open
Abstract
MOTIVATION Single-cell RNA sequencing (scRNA-seq) enables high-throughput transcriptomic profiling at single-cell resolution. The inherent spatial location is crucial for understanding how single cells orchestrate multicellular functions and drive diseases. However, spatial information is often lost during tissue dissociation. Spatial transcriptomic (ST) technologies can provide precise spatial gene expression atlas, while their practicality is constrained by the number of genes they can assay or the associated costs at a larger scale and the fine-grained cell-type annotation. By transferring knowledge between scRNA-seq and ST data through cell correspondence learning, it is possible to recover the spatial properties inherent in scRNA-seq datasets. RESULTS In this study, we introduce COME, a COntrastive Mapping lEarning approach that learns mapping between ST and scRNA-seq data to recover the spatial information of scRNA-seq data. Extensive experiments demonstrate that the proposed COME method effectively captures precise cell-spot relationships and outperforms previous methods in recovering spatial location for scRNA-seq data. More importantly, our method is capable of precisely identifying biologically meaningful information within the data, such as the spatial structure of missing genes, spatial hierarchical patterns, and the cell-type compositions for each spot. These results indicate that the proposed COME method can help to understand the heterogeneity and activities among cells within tissue environments. AVAILABILITY AND IMPLEMENTATION The COME is freely available in GitHub (https://github.com/cindyway/COME).
Collapse
Affiliation(s)
- Xindian Wei
- Department of Computer Science, City University of Hong Kong, Kowloon 999077, Hong Kong
| | - Tianyi Chen
- Department of Computer Science, City University of Hong Kong, Kowloon 999077, Hong Kong
| | - Xibiao Wang
- Department of Computer Science, Shantou University, Shantou 515063, China
| | - Wenjun Shen
- Department of Bioinformatics, Shantou University Medical College, Shantou 515041, China
| | - Cheng Liu
- Department of Computer Science, Shantou University, Shantou 515063, China
| | - Si Wu
- Department of Computer Science and Engineering, South China University of Technology, Guangzhou, Guangdong 510006, China
| | - Hau-San Wong
- Department of Computer Science, City University of Hong Kong, Kowloon 999077, Hong Kong
| |
Collapse
|
33
|
Zhu P, Shu H, Wang Y, Wang X, Zhao Y, Hu J, Peng J, Shang X, Tian Z, Chen J, Wang T. MAEST: accurately spatial domain detection in spatial transcriptomics with graph masked autoencoder. Brief Bioinform 2025; 26:bbaf086. [PMID: 40052440 PMCID: PMC11886571 DOI: 10.1093/bib/bbaf086] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2024] [Revised: 01/27/2025] [Accepted: 02/17/2025] [Indexed: 03/10/2025] Open
Abstract
Spatial transcriptomics (ST) technology provides gene expression profiles with spatial context, offering critical insights into cellular interactions and tissue architecture. A core task in ST is spatial domain identification, which involves detecting coherent regions with similar spatial expression patterns. However, existing methods often fail to fully exploit spatial information, leading to limited representational capacity and suboptimal clustering accuracy. Here, we introduce MAEST, a novel graph neural network model designed to address these limitations in ST data. MAEST leverages graph masked autoencoders to denoise and refine representations while incorporating graph contrastive learning to prevent feature collapse and enhance model robustness. By integrating one-hop and multi-hop representations, MAEST effectively captures both local and global spatial relationships, improving clustering precision. Extensive experiments across diverse datasets, including the human brain, mouse hippocampus, olfactory bulb, brain, and embryo, demonstrate that MAEST outperforms seven state-of-the-art methods in spatial domain identification. Furthermore, MAEST showcases its ability to integrate multi-slice data, identifying joint domains across horizontal tissue sections with high accuracy. These results highlight MAEST's versatility and effectiveness in unraveling the spatial organization of complex tissues. The source code of MAEST can be obtained at https://github.com/clearlove2333/MAEST.
Collapse
Affiliation(s)
- Pengfei Zhu
- School of Computer Science, Northwestern Polytechnical University, 1 Dongxiang Road, Xi’an 710072, China
- Key Laboratory of Big Data Storage and Management, Ministry of Industry and Information Technology, Northwestern Polytechnical University, 1 Dongxiang Road, Xi’an 710072, China
| | - Han Shu
- School of Computer Science, Northwestern Polytechnical University, 1 Dongxiang Road, Xi’an 710072, China
- Key Laboratory of Big Data Storage and Management, Ministry of Industry and Information Technology, Northwestern Polytechnical University, 1 Dongxiang Road, Xi’an 710072, China
| | - Yongtian Wang
- School of Computer Science, Northwestern Polytechnical University, 1 Dongxiang Road, Xi’an 710072, China
- Key Laboratory of Big Data Storage and Management, Ministry of Industry and Information Technology, Northwestern Polytechnical University, 1 Dongxiang Road, Xi’an 710072, China
| | - Xiaofeng Wang
- General Surgery Department, The Affiliated Hospital of Northwest University: Xi’an No 3 Hospital, Xi’an 710018, China
| | - Yuan Zhao
- School of Computer Science, Northwestern Polytechnical University, 1 Dongxiang Road, Xi’an 710072, China
- Key Laboratory of Big Data Storage and Management, Ministry of Industry and Information Technology, Northwestern Polytechnical University, 1 Dongxiang Road, Xi’an 710072, China
| | - Jialu Hu
- School of Computer Science, Northwestern Polytechnical University, 1 Dongxiang Road, Xi’an 710072, China
- Key Laboratory of Big Data Storage and Management, Ministry of Industry and Information Technology, Northwestern Polytechnical University, 1 Dongxiang Road, Xi’an 710072, China
| | - Jiajie Peng
- School of Computer Science, Northwestern Polytechnical University, 1 Dongxiang Road, Xi’an 710072, China
- Key Laboratory of Big Data Storage and Management, Ministry of Industry and Information Technology, Northwestern Polytechnical University, 1 Dongxiang Road, Xi’an 710072, China
| | - Xuequn Shang
- School of Computer Science, Northwestern Polytechnical University, 1 Dongxiang Road, Xi’an 710072, China
- Key Laboratory of Big Data Storage and Management, Ministry of Industry and Information Technology, Northwestern Polytechnical University, 1 Dongxiang Road, Xi’an 710072, China
| | - Zhen Tian
- School of Computer Science and Artificial Intelligence, Zhengzhou University, No. 100 Science Avenue, Zhengzhou 450001, China
| | - Jing Chen
- School of Computer Science and Engineering, Xi’an University of Technology, No. 5 South Jinhua Road, Xi’an 710048, China
| | - Tao Wang
- School of Computer Science, Northwestern Polytechnical University, 1 Dongxiang Road, Xi’an 710072, China
- Key Laboratory of Big Data Storage and Management, Ministry of Industry and Information Technology, Northwestern Polytechnical University, 1 Dongxiang Road, Xi’an 710072, China
| |
Collapse
|
34
|
Li Y, Hu Q, Han S, Wang-Sattler R, Du W. Multi-Manifolds fusing hyperbolic graph network balanced by pareto optimization for identifying spatial domains of spatial transcriptomics. Brief Bioinform 2025; 26:bbaf162. [PMID: 40220278 PMCID: PMC11992958 DOI: 10.1093/bib/bbaf162] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2024] [Revised: 02/28/2025] [Accepted: 03/19/2025] [Indexed: 04/14/2025] Open
Abstract
Identifying spatial domains for spatial transcriptomics is crucial for achieving comprehensive insights into the pathogenesis of gene expression. Increasingly, computational methods based on graph neural networks are being developed for spatial transcriptomics. However, previous methods have solely focused on the Euclidean manifold. To effectively exploit and explore the informative and deeper topological structures of inherent manifolds, we presented a Multi-Manifolds fusing hyperbolic graph network, balanced by Pareto optimization, for identifying spatial domains in Spatial Transcriptomics (MManiST). First, we developed multi-manifolds encoders for distinct manifolds using the hyperbolic neural network. Features from different manifolds were then combined using an attention mechanism, with multiple reconstruction losses balanced by Pareto optimization. Extensive experiments on commonly used benchmark datasets show that our method consistently outperforms seven state-of-the-art methods. Additionally, we investigated the validity of each component and the impact of fusion methods in ablation experiments.
Collapse
Affiliation(s)
- Ying Li
- Key Laboratory of Symbol Computation and Knowledge Engineering of Ministry of Education, College of Computer Science and Technology, Jilin University, Qianjin Street 2699, Changchun 130012, Jilin, China
| | - Qifeng Hu
- Key Laboratory of Symbol Computation and Knowledge Engineering of Ministry of Education, College of Computer Science and Technology, Jilin University, Qianjin Street 2699, Changchun 130012, Jilin, China
| | - Siyu Han
- TUM School of Medicine, Technical University of Munich, Ismaninger Straße 22, D-81675 Munich, Bavaria, Germany
| | - Rui Wang-Sattler
- Institute of Translational Genomics, Helmholtz Zentrum Munchen, Ingolstadter Landstraße 1, D-85764 Neuherberg, Bavaria, Germany
| | - Wei Du
- Key Laboratory of Symbol Computation and Knowledge Engineering of Ministry of Education, College of Computer Science and Technology, Jilin University, Qianjin Street 2699, Changchun 130012, Jilin, China
| |
Collapse
|
35
|
Xu X, Su J, Zhu R, Li K, Zhao X, Fan J, Mao F. From morphology to single-cell molecules: high-resolution 3D histology in biomedicine. Mol Cancer 2025; 24:63. [PMID: 40033282 PMCID: PMC11874780 DOI: 10.1186/s12943-025-02240-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2024] [Accepted: 01/18/2025] [Indexed: 03/05/2025] Open
Abstract
High-resolution three-dimensional (3D) tissue analysis has emerged as a transformative innovation in the life sciences, providing detailed insights into the spatial organization and molecular composition of biological tissues. This review begins by tracing the historical milestones that have shaped the development of high-resolution 3D histology, highlighting key breakthroughs that have facilitated the advancement of current technologies. We then systematically categorize the various families of high-resolution 3D histology techniques, discussing their core principles, capabilities, and inherent limitations. These 3D histology techniques include microscopy imaging, tomographic approaches, single-cell and spatial omics, computational methods and 3D tissue reconstruction (e.g. 3D cultures and spheroids). Additionally, we explore a wide range of applications for single-cell 3D histology, demonstrating how single-cell and spatial technologies are being utilized in the fields such as oncology, cardiology, neuroscience, immunology, developmental biology and regenerative medicine. Despite the remarkable progress made in recent years, the field still faces significant challenges, including high barriers to entry, issues with data robustness, ambiguous best practices for experimental design, and a lack of standardization across methodologies. This review offers a thorough analysis of these challenges and presents recommendations to surmount them, with the overarching goal of nurturing ongoing innovation and broader integration of cellular 3D tissue analysis in both biology research and clinical practice.
Collapse
Affiliation(s)
- Xintian Xu
- Institute of Medical Innovation and Research, Peking University Third Hospital, Beijing, China
- Cancer Center, Peking University Third Hospital, Beijing, China
- Department of Biochemistry and Molecular Biology, Beijing, Key Laboratory of Protein Posttranslational Modifications and Cell Function, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China
| | - 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
| | - Rongyi Zhu
- Department of Biochemistry and Molecular Biology, Beijing, Key Laboratory of Protein Posttranslational Modifications and Cell Function, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China
| | - Kailong Li
- Department of Biochemistry and Molecular Biology, Beijing, Key Laboratory of Protein Posttranslational Modifications and Cell Function, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China
| | - Xiaolu Zhao
- State Key Laboratory of Female Fertility Promotion, Center for Reproductive Medicine, Department of Obstetrics and GynecologyNational Clinical Research Center for Obstetrics and Gynecology (Peking University Third Hospital)Key Laboratory of Assisted Reproduction (Peking University), Ministry of EducationBeijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive Technology, Peking University Third Hospital, Beijing, China.
| | - Jibiao Fan
- College of Animal Science and Technology, Yangzhou University, Yangzhou, Jiangsu, China.
| | - Fengbiao Mao
- Institute of Medical Innovation and Research, Peking University Third Hospital, Beijing, China.
- Cancer Center, Peking University Third Hospital, Beijing, China.
- Beijing Key Laboratory for Interdisciplinary Research in Gastrointestinal Oncology (BLGO), Beijing, China.
| |
Collapse
|
36
|
Zhan Y, Zhang Y, Hu Z, Wang Y, Zhu Z, Du S, Yan X, Li X. LETSmix: a spatially informed and learning-based domain adaptation method for cell-type deconvolution in spatial transcriptomics. Genome Med 2025; 17:16. [PMID: 40022231 PMCID: PMC11869467 DOI: 10.1186/s13073-025-01442-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2024] [Accepted: 02/17/2025] [Indexed: 03/03/2025] Open
Abstract
Spatial transcriptomics (ST) enables the study of gene expression in spatial context, but many ST technologies face challenges due to limited resolution, leading to cell mixtures at each spot. We present LETSmix to deconvolve cell types by integrating spatial correlations through a tailored LETS filter, which leverages layer annotations, expression similarities, image texture features, and spatial coordinates to refine ST data. Additionally, LETSmix employs a mixup-augmented domain adaptation strategy to address discrepancies between ST and reference single-cell RNA sequencing data. Comprehensive evaluations across diverse ST platforms and tissue types demonstrate its high accuracy in estimating cell-type proportions and spatial patterns, surpassing existing methods (URL: https://github.com/ZhanYangen/LETSmix ).
Collapse
Affiliation(s)
- Yangen Zhan
- Division of Information Science and Technology, Tsinghua Shenzhen International Graduate School, Tsinghua University, Shenzhen, 518052, China
- School of Computer Science and Technology, Harbin Institute of Technology (Shenzhen), Shenzhen, 518055, China
| | - Yongbing Zhang
- School of Computer Science and Technology, Harbin Institute of Technology (Shenzhen), Shenzhen, 518055, China.
| | - Zheqi Hu
- School of Computer Science and Technology, Harbin Institute of Technology (Shenzhen), Shenzhen, 518055, China
| | - Yifeng Wang
- School of Computer Science and Technology, Harbin Institute of Technology (Shenzhen), Shenzhen, 518055, China
| | - Zirui Zhu
- School of Computer Science and Technology, Harbin Institute of Technology (Shenzhen), Shenzhen, 518055, China
| | - Sijing Du
- School of Computer Science and Technology, Harbin Institute of Technology (Shenzhen), Shenzhen, 518055, China
| | - Xiangming Yan
- School of Computer Science and Technology, Harbin Institute of Technology (Shenzhen), Shenzhen, 518055, China
| | - Xiu Li
- Division of Information Science and Technology, Tsinghua Shenzhen International Graduate School, Tsinghua University, Shenzhen, 518052, China.
| |
Collapse
|
37
|
Zhang P, Gao C, Zhang Z, Yuan Z, Zhang Q, Zhang P, Du S, Zhou W, Li Y, Li S. Systematic inference of super-resolution cell spatial profiles from histology images. Nat Commun 2025; 16:1838. [PMID: 39984438 PMCID: PMC11845739 DOI: 10.1038/s41467-025-57072-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2024] [Accepted: 02/07/2025] [Indexed: 02/23/2025] Open
Abstract
Inferring cell spatial profiles from histology images is critical for cancer diagnosis and treatment in clinical settings. In this study, we report a weakly-supervised deep-learning method, HistoCell, to directly infer super-resolution cell spatial profiles consisting of cell types, cell states and their spatial network from histology images at the single-nucleus-level. Benchmark analysis demonstrates that HistoCell robustly achieves state-of-the-art performance in terms of cell type/states prediction solely from histology images across multiple cancer tissues. HistoCell can significantly enhance the deconvolution accuracy for the spatial transcriptomics data and enable accurate annotation of subtle cancer tissue architectures. Moreover, HistoCell is applied to de novo discovery of clinically relevant spatial organization indicators, including prognosis and drug response biomarkers, across diverse cancer types. HistoCell also enable image-based screening of cell populations that drives phenotype of interest, and is applied to discover the cell population and corresponding spatial organization indicators associated with gastric malignant transformation risk. Overall, HistoCell emerges as a powerful and versatile tool for cancer studies in histology image-only cohorts.
Collapse
Affiliation(s)
- Peng Zhang
- Institute of TCM-X/MOE Key Laboratory of Bioinformatics, Bioinformatics Division, BNRist/Department of Automation, Tsinghua University, Beijing, China
| | - Chaofei Gao
- Institute of TCM-X/MOE Key Laboratory of Bioinformatics, Bioinformatics Division, BNRist/Department of Automation, Tsinghua University, Beijing, China
| | - Zhuoyu Zhang
- Institute of TCM-X/MOE Key Laboratory of Bioinformatics, Bioinformatics Division, BNRist/Department of Automation, Tsinghua University, Beijing, China
| | - Zhiyuan Yuan
- Institute of Science and Technology for Brain-Inspired Intelligence; MOE Key Laboratory of Computational Neuroscience and Brain-Inspired Intelligence; MOE Frontiers Center for Brain Science, Fudan University, Shanghai, China
| | - Qian Zhang
- Institute of TCM-X/MOE Key Laboratory of Bioinformatics, Bioinformatics Division, BNRist/Department of Automation, Tsinghua University, Beijing, China
| | - Ping Zhang
- Department of Pathology, Wangjing Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Shiyu Du
- Department of Gastroenterology, China-Japan Friendship Hospital, Beijing, China
| | - Weixun Zhou
- Department of Pathology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yan Li
- Department of Traditional Chinese Medicine, the First Affiliated Hospital of Wannan Medical College, Wuhu, China
| | - Shao Li
- Institute of TCM-X/MOE Key Laboratory of Bioinformatics, Bioinformatics Division, BNRist/Department of Automation, Tsinghua University, Beijing, China.
| |
Collapse
|
38
|
Gingerich IK, Goods BA, Frost HR. Randomized Spatial PCA (RASP): a computationally efficient method for dimensionality reduction of high-resolution spatial transcriptomics data. RESEARCH SQUARE 2025:rs.3.rs-6050441. [PMID: 40034439 PMCID: PMC11875318 DOI: 10.21203/rs.3.rs-6050441/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/05/2025]
Abstract
Spatial transcriptomics (ST) provides critical insights into the spatial organization of gene expression, enabling researchers to unravel the intricate relationship between cellular environments and biological function. Identifying spatial domains within tissues is key to understanding tissue architecture and mechanisms underlying development and disease progression. Here, we present Randomized Spatial PCA (RASP), a novel spatially-aware dimensionality reduction method for ST data. RASP is designed to be orders-of-magnitude faster than existing techniques, scale to datasets with 100, 000+ locations, support flexible integration of non-transcriptomic covariates, and reconstruct de-noised, spatially-smoothed gene expression values. It employs a randomized two-stage PCA framework with sparse matrix operations and configurable spatial smoothing. RASP was compared to BASS, GraphST, SEDR, SpatialPCA, and STAGATE using diverse ST datasets (10x Visium, Stereo-Seq, MERFISH, 10x Xenium) on human and mouse tissues. RASP demonstrates comparable or superior tissue domain detection with substantial improvements in computational speed, enhancing exploration of high-resolution subcellular datasets.
Collapse
Affiliation(s)
- Ian K. Gingerich
- Department of Biomedical Data Science, Geisel School of Medicine, Dartmouth College, Hanover, NH, USA
- Thayer School of Engineering, Dartmouth College, Hanover, NH, USA
| | | | - H. Robert Frost
- Department of Biomedical Data Science, Geisel School of Medicine, Dartmouth College, Hanover, NH, USA
| |
Collapse
|
39
|
Xu K, Xu Y, Wang Z, Zhou XM, Zhang L. stDyer enables spatial domain clustering with dynamic graph embedding. Genome Biol 2025; 26:34. [PMID: 39980033 PMCID: PMC11843776 DOI: 10.1186/s13059-025-03503-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2024] [Accepted: 02/12/2025] [Indexed: 02/22/2025] Open
Abstract
Spatially resolved transcriptomics (SRT) data provide critical insights into gene expression patterns within tissue contexts, necessitating effective methods for identifying spatial domains. We introduce stDyer, an end-to-end deep learning framework for spatial domain clustering in SRT data. stDyer combines Gaussian Mixture Variational AutoEncoder with graph attention networks to learn embeddings and perform clustering. Its dynamic graphs adaptively link units based on Gaussian Mixture assignments, improving clustering and producing smoother domain boundaries. stDyer's mini-batch strategy and multi-GPU support facilitate scalability to large datasets. Benchmarking against state-of-the-art tools, stDyer demonstrates superior performance in spatial domain clustering, multi-slice analysis, and large-scale dataset handling.
Collapse
Affiliation(s)
- Ke Xu
- Department of Computer Science, Hong Kong Baptist University, Hong Kong, China
| | - Yu Xu
- Department of Computer Science, Hong Kong Baptist University, Hong Kong, China
| | - Zirui Wang
- Department of Computer Science, Hong Kong Baptist University, Hong Kong, China
| | - Xin Maizie Zhou
- Department of Biomedical Engineering, Vanderbilt University, 2301 Vanderbilt Place, 37235, Nashville, Tennessee, USA.
| | - Lu Zhang
- Department of Computer Science, Hong Kong Baptist University, Hong Kong, China.
- Institute of Systems Medicine and Health Sciences, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong, China.
| |
Collapse
|
40
|
Mou L, Wang TB, Chen Y, Luo Z, Wang X, Pu Z. Single-cell genomics and spatial transcriptomics in islet transplantation for diabetes treatment: advancing towards personalized therapies. Front Immunol 2025; 16:1554876. [PMID: 40051625 PMCID: PMC11882877 DOI: 10.3389/fimmu.2025.1554876] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2025] [Accepted: 01/21/2025] [Indexed: 03/09/2025] Open
Abstract
Diabetes mellitus (DM) is a global health crisis affecting millions, with islet transplantation emerging as a promising treatment strategy to restore insulin production. This review synthesizes the current research on single-cell and spatial transcriptomics in the context of islet transplantation, highlighting their potential to revolutionize DM management. Single-cell RNA sequencing, offers a detailed look into the diversity and functionality within islet grafts, identifying specific cell types and states that influence graft acceptance and function. Spatial transcriptomics complements this by mapping gene expression within the tissue's spatial context, crucial for understanding the microenvironment surrounding transplanted islets and their interactions with host tissues. The integration of these technologies offers a comprehensive view of cellular interactions and microenvironments, elucidating mechanisms underlying islet function, survival, and rejection. This understanding is instrumental in developing targeted therapies to enhance graft performance and patient outcomes. The review emphasizes the significance of these research avenues in informing clinical practices and improving outcomes for patients with DM through more effective islet transplantation strategies. Future research directions include the application of these technologies in personalized medicine, developmental biology, and regenerative medicine, with the potential to predict disease progression and treatment responses. Addressing ethical and technical challenges will be crucial for the successful implementation of these integrated approaches in research and clinical practice, ultimately enhancing our ability to manage DM and improve patient quality of life.
Collapse
Affiliation(s)
- Lisha Mou
- Department of Endocrinology, Institute of Translational Medicine, Health Science Center, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People’s Hospital, Shenzhen, Guangdong, China
- MetaLife Lab, Shenzhen Institute of Translational Medicine, Shenzhen, Guangdong, China
| | - Tony Bowei Wang
- Imaging Department, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People’s Hospital, Shenzhen, Guangdong, China
| | - Yuxian Chen
- Department of Endocrinology, Institute of Translational Medicine, Health Science Center, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People’s Hospital, Shenzhen, Guangdong, China
| | - Ziqi Luo
- Department of Endocrinology, Institute of Translational Medicine, Health Science Center, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People’s Hospital, Shenzhen, Guangdong, China
| | - Xinyu Wang
- Department of Endocrinology, Institute of Translational Medicine, Health Science Center, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People’s Hospital, Shenzhen, Guangdong, China
| | - Zuhui Pu
- MetaLife Lab, Shenzhen Institute of Translational Medicine, Shenzhen, Guangdong, China
- Imaging Department, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People’s Hospital, Shenzhen, Guangdong, China
| |
Collapse
|
41
|
Kordowski A, Mulay O, Tan X, Vo T, Baumgartner U, Maybury MK, Hassall T, Harris L, Nguyen Q, Day BW. Spatial analysis of a complete DIPG-infiltrated brainstem reveals novel ligand-receptor mediators of tumour-to-TME crosstalk. Acta Neuropathol Commun 2025; 13:35. [PMID: 39972389 PMCID: PMC11837654 DOI: 10.1186/s40478-025-01952-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: 10/31/2024] [Accepted: 02/09/2025] [Indexed: 02/21/2025] Open
Abstract
Previous studies have highlighted the capacity of brain cancer cells to functionally interact with the tumour microenvironment (TME). This TME-cancer crosstalk crucially contributes to tumour cell invasion and disease progression. In this study, we performed spatial transcriptomic sequencing analysis of a complete annotated tumour-infiltrated brainstem from a single diffuse intrinsic pontine glioma (DIPG) patient. Gene signatures from ten sequential tumour regions were analysed to assess mechanisms of disease progression and oncogenic interactions with the TME. We identified four distinct tumour subpopulations and assessed respective ligand-receptor pairs that actively promote DIPG tumour progression via crosstalk with endothelial, neuronal and immune cell communities. Our analysis found potential targetable mediators of tumour-to-TME communication, including members of the complement component system and the neuropeptide/GPCR ligand-receptor pair ADCYAP1-ADCYAP1R1. These interactions could influence DIPG tumour progression and represent novel therapeutic targets.
Collapse
Affiliation(s)
- Anja Kordowski
- Queensland Institute for Medical Research, Brisbane, QLD, 4006, Australia.
| | - Onkar Mulay
- Queensland Institute for Medical Research, Brisbane, QLD, 4006, Australia
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, QLD, 4067, Australia
| | - Xiao Tan
- Queensland Institute for Medical Research, Brisbane, QLD, 4006, Australia
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, QLD, 4067, Australia
| | - Tuan Vo
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, QLD, 4067, Australia
- Hunter Medical Research Institute, Precision Medicine Research Program, Newcastle, NSW, 2305, Australia
- School of Biomedical Sciences and Pharmacy, The University of Newcastle, Newcastle, NSW, 2308, Australia
| | - Ulrich Baumgartner
- School of Biomedical Sciences, The University of Queensland, Brisbane, QLD, 4067, Australia
| | - Mellissa K Maybury
- Child Health Research Centre, The University of Queensland, Brisbane, QLD, 4101, Australia
| | - Timothy Hassall
- Children's Brain Cancer Centre, UQ Frazer Institute, Brisbane, QLD, 4102, Australia
- Queensland Children's Hospital, Brisbane, QLD, 4101, Australia
| | - Lachlan Harris
- Queensland Institute for Medical Research, Brisbane, QLD, 4006, Australia
- School of Biomedical Sciences, The University of Queensland, Brisbane, QLD, 4067, Australia
- School of Biomedical Sciences, Queensland University of Technology, Brisbane, QLD, 4059, Australia
| | - Quan Nguyen
- Queensland Institute for Medical Research, Brisbane, QLD, 4006, Australia
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, QLD, 4067, Australia
| | - Bryan W Day
- Queensland Institute for Medical Research, Brisbane, QLD, 4006, Australia.
- School of Biomedical Sciences, The University of Queensland, Brisbane, QLD, 4067, Australia.
- Children's Brain Cancer Centre, UQ Frazer Institute, Brisbane, QLD, 4102, Australia.
| |
Collapse
|
42
|
Qian J, Shao X, Bao H, Fang Y, Guo W, Li C, Li A, Hua H, Fan X. Identification and characterization of cell niches in tissue from spatial omics data at single-cell resolution. Nat Commun 2025; 16:1693. [PMID: 39956823 PMCID: PMC11830827 DOI: 10.1038/s41467-025-57029-9] [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: 08/12/2024] [Accepted: 02/03/2025] [Indexed: 02/18/2025] Open
Abstract
Deciphering the features, structure, and functions of the cell niche in tissues remains a major challenge. Here, we present scNiche, a computational framework to identify and characterize cell niches from spatial omics data at single-cell resolution. We benchmark scNiche with both simulated and biological datasets, and demonstrate that scNiche can effectively and robustly identify cell niches while outperforming other existing methods. In spatial proteomics data from human triple-negative breast cancer, scNiche reveals the influence of the microenvironment on cellular phenotypes, and further dissects patient-specific niches with distinct cellular compositions or phenotypic characteristics. By analyzing mouse liver spatial transcriptomics data across normal and early-onset liver failure donors, scNiche uncovers disease-specific liver injury niches, and further delineates the niche remodeling from normal liver to liver failure. Overall, scNiche enables decoding the cellular microenvironment in tissues from single-cell spatial omics data.
Collapse
Affiliation(s)
- Jingyang Qian
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China
- State Key Laboratory of Chinese Medicine Modernization, Innovation Center of Yangtze River Delta, Zhejiang University, Jiaxing, 314102, China
| | - Xin Shao
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China.
- State Key Laboratory of Chinese Medicine Modernization, Innovation Center of Yangtze River Delta, Zhejiang University, Jiaxing, 314102, China.
- Zhejiang Key Laboratory of Chinese Medicine Modernization, Innovation Center of Yangtze River Delta, Zhejiang University, Jiaxing, 314100, China.
| | - Hudong Bao
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China
| | - Yin Fang
- College of Computer Science and Technology, Zhejiang University, Hangzhou, 310013, China
| | - Wenbo Guo
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China
- State Key Laboratory of Chinese Medicine Modernization, Innovation Center of Yangtze River Delta, Zhejiang University, Jiaxing, 314102, China
- Zhejiang Key Laboratory of Chinese Medicine Modernization, Innovation Center of Yangtze River Delta, Zhejiang University, Jiaxing, 314100, China
| | - Chengyu Li
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China
- State Key Laboratory of Chinese Medicine Modernization, Innovation Center of Yangtze River Delta, Zhejiang University, Jiaxing, 314102, China
| | - Anyao Li
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China
- State Key Laboratory of Chinese Medicine Modernization, Innovation Center of Yangtze River Delta, Zhejiang University, Jiaxing, 314102, China
| | - Hua Hua
- Translational Chinese Medicine Key Laboratory of Sichuan Province, SiChuan Institute for Translational Chinese Medicine, Chengdu, 610041, China.
| | - Xiaohui Fan
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China.
- State Key Laboratory of Chinese Medicine Modernization, Innovation Center of Yangtze River Delta, Zhejiang University, Jiaxing, 314102, China.
- Zhejiang Key Laboratory of Chinese Medicine Modernization, Innovation Center of Yangtze River Delta, Zhejiang University, Jiaxing, 314100, China.
- Zhejiang Key Laboratory of Precision Diagnosis and Therapy for Major Gynecological Diseases, Women's Hospital, Zhejiang University School of Medicine, 310006, Hangzhou, China.
| |
Collapse
|
43
|
Wang Z, Liu Y, Chang X, Liu X. Deconvolution and inference of spatial communication through optimization algorithm for spatial transcriptomics. Commun Biol 2025; 8:235. [PMID: 39948133 PMCID: PMC11825862 DOI: 10.1038/s42003-025-07625-8] [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: 08/08/2024] [Accepted: 01/29/2025] [Indexed: 02/16/2025] Open
Abstract
Spatial transcriptomics technologies can capture gene expression at spatial loci. However, at certain resolutions, the obtained gene expression reflects the sum of either a heterogeneous or homogeneous set of cells, rather than individual cell. This limitation gives rise to the deconvolution algorithm to make cell-type inferences at each location. Yet, the vast majority of deconvolution methods that have been developed ignore the spatial information of the tissue and the communications between the cells or spots. To overcome these afflictions, we proposed a deconvolution method, non-negative least squares-based and optimization search-based deconvolution (NODE), that combines cell-type-specific information from single-cell RNA sequencing (scRNA-seq) and intercellular communications in tissue. NODE deconvolution algorithm, incorporating the spatial information of the tissue, allows us to quantify intercellular communications at the same instant. NODE can not only utilize optimization method to infer the deconvolution results of spatial transcriptomics data and reduce the probability of overfitting situations, but also make reasonable inferences for spatial communications. Subsequently, we applied NODE to four datasets to validate the correctness of the NODE deconvolution results and compare them with existing deconvolution algorithms. NODE also inferred spatial communications and validated them in tissue development of human heart.
Collapse
Affiliation(s)
- Zedong Wang
- 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
| | - Yi Liu
- 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
|
44
|
Jiang S, Mantri M, Maymi V, Leddon SA, Schweitzer P, Bhandari S, Holdener C, Ntekas I, Vollmers C, Flyak AI, Fowell DJ, Rudd BD, De Vlaminck I. A Temporal and Spatial Atlas of Adaptive Immune Responses in the Lymph Node Following Viral Infection. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.01.31.635509. [PMID: 39975238 PMCID: PMC11838507 DOI: 10.1101/2025.01.31.635509] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 02/21/2025]
Abstract
The spatial organization of adaptive immune cells within lymph nodes is critical for understanding immune responses during infection and disease. Here, we introduce AIR-SPACE, an integrative approach that combines high-resolution spatial transcriptomics with paired, high-fidelity long-read sequencing of T and B cell receptors. This method enables the simultaneous analysis of cellular transcriptomes and adaptive immune receptor (AIR) repertoires within their native spatial context. We applied AIR-SPACE to mouse popliteal lymph nodes at five distinct time points after Vaccinia virus footpad infection and constructed a comprehensive map of the developing adaptive immune response. Our analysis revealed heterogeneous activation niches, characterized by Interferon-gamma (IFN-γ) production, during the early stages of infection. At later stages, we delineated sub-anatomical structures within the germinal center (GC) and observed evidence that antibody-producing plasma cells differentiate and exit the GC through the dark zone. Furthermore, by combining clonotype data with spatial lineage tracing, we demonstrate that B cell clones are shared among multiple GCs within the same lymph node, reinforcing the concept of a dynamic, interconnected network of GCs. Overall, our study demonstrates how AIR-SPACE can be used to gain insight into the spatial dynamics of infection responses within lymphoid organs.
Collapse
Affiliation(s)
- Shaowen Jiang
- Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY, USA
- Department of Computational Biology, Cornell University, Ithaca, NY, USA
| | - Madhav Mantri
- Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY, USA
- Department of Computational Biology, Cornell University, Ithaca, NY, USA
| | - Viviana Maymi
- Department of Microbiology and Immunology, Cornell University, Ithaca, NY, USA
| | - Scott A Leddon
- Department of Microbiology and Immunology, Cornell University, Ithaca, NY, USA
| | - Peter Schweitzer
- Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY, USA
| | - Subash Bhandari
- Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY, USA
- Department of Microbiology and Immunology, Cornell University, Ithaca, NY, USA
| | - Chase Holdener
- Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY, USA
- Department of Computational Biology, Cornell University, Ithaca, NY, USA
| | - Ioannis Ntekas
- Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY, USA
| | - Christopher Vollmers
- Department of Biomolecular Engineering, University of California Santa Cruz, Santa Cruz, CA, USA
| | - Andrew I Flyak
- Department of Microbiology and Immunology, Cornell University, Ithaca, NY, USA
| | - Deborah J Fowell
- Department of Microbiology and Immunology, Cornell University, Ithaca, NY, USA
| | - Brian D Rudd
- Department of Microbiology and Immunology, Cornell University, Ithaca, NY, USA
| | - Iwijn De Vlaminck
- Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY, USA
| |
Collapse
|
45
|
Cooper RA, Thomas E, Sozanska AM, Pescia C, Royston DJ. Spatial transcriptomic approaches for characterising the bone marrow landscape: pitfalls and potential. Leukemia 2025; 39:291-295. [PMID: 39609595 PMCID: PMC11794127 DOI: 10.1038/s41375-024-02480-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2024] [Revised: 11/08/2024] [Accepted: 11/19/2024] [Indexed: 11/30/2024]
Affiliation(s)
- Rosalin A Cooper
- Nuffield Division of Clinical Laboratory Sciences, Radcliffe Department of Medicine, University of Oxford, Oxford, UK.
- Oxford University Hospitals NHS Foundation Trust, Oxford, UK.
| | - Emily Thomas
- Institute of Biomedical Engineering, University of Oxford, Oxford, UK
| | - Anna M Sozanska
- Nuffield Division of Clinical Laboratory Sciences, Radcliffe Department of Medicine, University of Oxford, Oxford, UK
| | - Carlo Pescia
- Nuffield Division of Clinical Laboratory Sciences, Radcliffe Department of Medicine, University of Oxford, Oxford, UK
- University of Milan, Milan, Italy
| | - Daniel J Royston
- Nuffield Division of Clinical Laboratory Sciences, Radcliffe Department of Medicine, University of Oxford, Oxford, UK
- Oxford University Hospitals NHS Foundation Trust, Oxford, UK
| |
Collapse
|
46
|
Chitra U, Arnold BJ, Sarkar H, Sanno K, Ma C, Lopez-Darwin S, Raphael BJ. Mapping the topography of spatial gene expression with interpretable deep learning. Nat Methods 2025; 22:298-309. [PMID: 39849132 DOI: 10.1038/s41592-024-02503-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2023] [Accepted: 10/14/2024] [Indexed: 01/25/2025]
Abstract
Spatially resolved transcriptomics technologies provide high-throughput measurements of gene expression in a tissue slice, but the sparsity of these data complicates analysis of spatial gene expression patterns. We address this issue by deriving a topographic map of a tissue slice-analogous to a map of elevation in a landscape-using a quantity called the isodepth. Contours of constant isodepths enclose domains with distinct cell type composition, while gradients of the isodepth indicate spatial directions of maximum change in expression. We develop GASTON (gradient analysis of spatial transcriptomics organization with neural networks), an unsupervised and interpretable deep learning algorithm that simultaneously learns the isodepth, spatial gradients and piecewise linear expression functions that model both continuous gradients and discontinuous variation in gene expression. We show that GASTON accurately identifies spatial domains and marker genes across several tissues, gradients of neuronal differentiation and firing in the brain, and gradients of metabolism and immune activity in the tumor microenvironment.
Collapse
Affiliation(s)
- Uthsav Chitra
- Department of Computer Science, Princeton University, Princeton, NJ, USA
| | - Brian J Arnold
- Department of Computer Science, Princeton University, Princeton, NJ, USA
- Center for Statistics and Machine Learning, Princeton University, Princeton, NJ, USA
| | - Hirak Sarkar
- Department of Computer Science, Princeton University, Princeton, NJ, USA
- Ludwig Cancer Institute, Princeton Branch, Princeton University, Princeton, NJ, USA
| | - Kohei Sanno
- Department of Computer Science, Princeton University, Princeton, NJ, USA
| | - Cong Ma
- Department of Computer Science, Princeton University, Princeton, NJ, USA
| | - Sereno Lopez-Darwin
- Lewis-Sigler Institute for Integrative Genomics, Princeton University, Princeton, NJ, USA
| | - Benjamin J Raphael
- Department of Computer Science, Princeton University, Princeton, NJ, USA.
| |
Collapse
|
47
|
Liu Y, Li Z, Chen X, Cui X, Gao Z, Jiang R. INSTINCT: Multi-sample integration of spatial chromatin accessibility sequencing data via stochastic domain translation. Nat Commun 2025; 16:1247. [PMID: 39893190 PMCID: PMC11787322 DOI: 10.1038/s41467-025-56535-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2024] [Accepted: 01/13/2025] [Indexed: 02/04/2025] Open
Abstract
Recent advances in spatial epigenomic techniques have given rise to spatial assay for transposase-accessible chromatin using sequencing (spATAC-seq) data, enabling the characterization of epigenomic heterogeneity and spatial information simultaneously. Integrative analysis of multiple spATAC-seq samples, for which no method has been developed, allows for effective identification and elimination of unwanted non-biological factors within the data, enabling comprehensive exploration of tissue structures and providing a holistic epigenomic landscape, thereby facilitating the discovery of biological implications and the study of regulatory processes. In this article, we present INSTINCT, a method for multi-sample INtegration of Spatial chromaTIN accessibility sequencing data via stochastiC domain Translation. INSTINCT can efficiently handle the high dimensionality of spATAC-seq data and eliminate the complex noise and batch effects of samples through a stochastic domain translation procedure. We demonstrate the superiority and robustness of INSTINCT in integrating spATAC-seq data across multiple simulated scenarios and real datasets. Additionally, we highlight the advantages of INSTINCT in spatial domain identification, visualization, spot-type annotation, and various downstream analyses, including motif enrichment analysis, expression enrichment analysis, and partitioned heritability analysis.
Collapse
Affiliation(s)
- Yuyao Liu
- Ministry of Education Key Laboratory of Bioinformatics, Bioinformatics Division at the Beijing National Research Center for Information Science and Technology, Center for Synthetic and Systems Biology, Department of Automation, Tsinghua University, Beijing, 100084, China
| | - Zhen Li
- Ministry of Education Key Laboratory of Bioinformatics, Bioinformatics Division at the Beijing National Research Center for Information Science and Technology, Center for Synthetic and Systems Biology, Department of Automation, Tsinghua University, Beijing, 100084, China
| | - Xiaoyang Chen
- Ministry of Education Key Laboratory of Bioinformatics, Bioinformatics Division at the Beijing National Research Center for Information Science and Technology, Center for Synthetic and Systems Biology, Department of Automation, Tsinghua University, Beijing, 100084, China
| | - Xuejian Cui
- Ministry of Education Key Laboratory of Bioinformatics, Bioinformatics Division at the Beijing National Research Center for Information Science and Technology, Center for Synthetic and Systems Biology, Department of Automation, Tsinghua University, Beijing, 100084, China
| | - Zijing Gao
- Ministry of Education Key Laboratory of Bioinformatics, Bioinformatics Division at the Beijing National Research Center for Information Science and Technology, Center for Synthetic and Systems Biology, Department of Automation, Tsinghua University, Beijing, 100084, China
| | - Rui Jiang
- Ministry of Education Key Laboratory of Bioinformatics, Bioinformatics Division at the Beijing National Research Center for Information Science and Technology, Center for Synthetic and Systems Biology, Department of Automation, Tsinghua University, Beijing, 100084, China.
| |
Collapse
|
48
|
Yang Y, Cui Y, Zeng X, Zhang Y, Loza M, Park SJ, Nakai K. STAIG: Spatial transcriptomics analysis via image-aided graph contrastive learning for domain exploration and alignment-free integration. Nat Commun 2025; 16:1067. [PMID: 39870633 PMCID: PMC11772580 DOI: 10.1038/s41467-025-56276-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2024] [Accepted: 01/06/2025] [Indexed: 01/29/2025] Open
Abstract
Spatial transcriptomics is an essential application for investigating cellular structures and interactions and requires multimodal information to precisely study spatial domains. Here, we propose STAIG, a deep-learning model that integrates gene expression, spatial coordinates, and histological images using graph-contrastive learning coupled with high-performance feature extraction. STAIG can integrate tissue slices without prealignment and remove batch effects. Moreover, it is designed to accept data acquired from various platforms, with or without histological images. By performing extensive benchmarks, we demonstrate the capability of STAIG to recognize spatial regions with high precision and uncover new insights into tumor microenvironments, highlighting its promising potential in deciphering spatial biological intricates.
Collapse
Affiliation(s)
- Yitao Yang
- Department of Computational Biology and Medical Science, Graduate School of Frontier Sciences, the University of Tokyo, Tokyo, Japan
| | - Yang Cui
- Department of Computational Biology and Medical Science, Graduate School of Frontier Sciences, the University of Tokyo, Tokyo, Japan
| | - Xin Zeng
- Department of Computational Biology and Medical Science, Graduate School of Frontier Sciences, the University of Tokyo, Tokyo, Japan
| | - Yubo Zhang
- Department of Computational Biology and Medical Science, Graduate School of Frontier Sciences, the University of Tokyo, Tokyo, Japan
| | - Martin Loza
- Human Genome Center, the Institute of Medical Science, the University of Tokyo, Tokyo, Japan
| | - Sung-Joon Park
- Human Genome Center, the Institute of Medical Science, the University of Tokyo, Tokyo, Japan
| | - Kenta Nakai
- Department of Computational Biology and Medical Science, Graduate School of Frontier Sciences, the University of Tokyo, Tokyo, Japan.
- Human Genome Center, the Institute of Medical Science, the University of Tokyo, Tokyo, Japan.
| |
Collapse
|
49
|
Zhou L, Peng X, Chen M, He X, Tian G, Yang J, Peng L. Unveiling patterns in spatial transcriptomics data: a novel approach utilizing graph attention autoencoder and multiscale deep subspace clustering network. Gigascience 2025; 14:giae103. [PMID: 39804726 PMCID: PMC11727722 DOI: 10.1093/gigascience/giae103] [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: 02/07/2024] [Revised: 07/06/2024] [Accepted: 11/21/2024] [Indexed: 01/16/2025] Open
Abstract
BACKGROUND The accurate deciphering of spatial domains, along with the identification of differentially expressed genes and the inference of cellular trajectory based on spatial transcriptomic (ST) data, holds significant potential for enhancing our understanding of tissue organization and biological functions. However, most of spatial clustering methods can neither decipher complex structures in ST data nor entirely employ features embedded in different layers. RESULTS This article introduces STMSGAL, a novel framework for analyzing ST data by incorporating graph attention autoencoder and multiscale deep subspace clustering. First, STMSGAL constructs ctaSNN, a cell type-aware shared nearest neighbor graph, using Louvian clustering exclusively based on gene expression profiles. Subsequently, it integrates expression profiles and ctaSNN to generate spot latent representations using a graph attention autoencoder and multiscale deep subspace clustering. Lastly, STMSGAL implements spatial clustering, differential expression analysis, and trajectory inference, providing comprehensive capabilities for thorough data exploration and interpretation. STMSGAL was evaluated against 7 methods, including SCANPY, SEDR, CCST, DeepST, GraphST, STAGATE, and SiGra, using four 10x Genomics Visium datasets, 1 mouse visual cortex STARmap dataset, and 2 Stereo-seq mouse embryo datasets. The comparison showcased STMSGAL's remarkable performance across Davies-Bouldin, Calinski-Harabasz, S_Dbw, and ARI values. STMSGAL significantly enhanced the identification of layer structures across ST data with different spatial resolutions and accurately delineated spatial domains in 2 breast cancer tissues, adult mouse brain (FFPE), and mouse embryos. CONCLUSIONS STMSGAL can serve as an essential tool for bridging the analysis of cellular spatial organization and disease pathology, offering valuable insights for researchers in the field.
Collapse
Affiliation(s)
- Liqian Zhou
- School of Computer Science, Hunan University of Technology, Zhuzhou 412007, Hunan, China
| | - Xinhuai Peng
- School of Computer Science, Hunan University of Technology, Zhuzhou 412007, Hunan, China
| | - Min Chen
- School of Computer Science, Hunan Institute of Technology, Hengyang 421002, Hunan, China
| | - Xianzhi He
- School of Computer Science, Hunan University of Technology, Zhuzhou 412007, Hunan, China
| | - Geng Tian
- Geneis (Beijing) Co. Ltd., Beijing 100102, China
| | | | - Lihong Peng
- School of Computer Science, Hunan University of Technology, Zhuzhou 412007, Hunan, China
- College of Life Science and Chemistry, Hunan University of Technology, Zhuzhou 412007, Hunan, China
| |
Collapse
|
50
|
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
|