1
|
Wang W, Zheng S, Shin SC, Chávez-Fuentes JC, Yuan GC. ONTraC characterizes spatially continuous variations of tissue microenvironment through niche trajectory analysis. Genome Biol 2025; 26:117. [PMID: 40340854 PMCID: PMC12060293 DOI: 10.1186/s13059-025-03588-5] [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: 05/21/2024] [Accepted: 04/25/2025] [Indexed: 05/10/2025] Open
Abstract
Recent technological advances enable mapping of tissue spatial organization at single-cell resolution, but methods for analyzing spatially continuous microenvironments are still lacking. We introduce ONTraC, a graph neural network-based framework for constructing spatial trajectories at niche-level. Through benchmarking analyses using multiple simulated and real datasets, we show that ONTraC outperforms existing methods. ONTraC captures both normal anatomical structures and disease-associated tissue microenvironment changes. In addition, it identifies tissue microenvironment-dependent shifts in gene expression, regulatory network, and cell-cell interaction patterns. Taken together, ONTraC provides a useful framework for characterizing the structural and functional organization of tissue microenvironments.
Collapse
Affiliation(s)
- Wen Wang
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Shiwei Zheng
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Sujung Crystal Shin
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | | | - Guo-Cheng Yuan
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA.
| |
Collapse
|
2
|
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
|
3
|
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
|
4
|
Reynolds DE, Roh YH, Oh D, Vallapureddy P, Fan R, Ko J. Temporal and spatial omics technologies for 4D profiling. Nat Methods 2025:10.1038/s41592-025-02683-6. [PMID: 40263585 DOI: 10.1038/s41592-025-02683-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2023] [Accepted: 03/26/2025] [Indexed: 04/24/2025]
Abstract
Cells have distinct molecular repertoires on their surfaces and unique intracellular biomolecular profiles that play pivotal roles in orchestrating a myriad of biological responses in the context of growth, development and disease. A persistent challenge in the deep exploration of these cues has been in our inability to effectively and precisely capture the temporal and spatial characteristics of living cells. In this Perspective, we delve into techniques for temporal and two- and three-dimensional spatial omics analyses and underscore how their harmonious fusion promises to unlock insights into the dynamics and diversity of individual cells within biological systems such as tissues and organoids. We then explore four-dimensional profiling, a nascent but promising frontier that adds a temporal (fourth-dimension) component to three-dimensional omics; highlight the advancements, challenges and gaps in the field; and discuss potential strategies for further technological development.
Collapse
Affiliation(s)
- David E Reynolds
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, USA
| | - Yoon Ho Roh
- Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Department of Energy and Chemical Engineering, Incheon National University, Incheon, Republic of Korea
| | - Daniel Oh
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, USA
| | - Phoebe Vallapureddy
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, USA
| | - Rong Fan
- Department of Biomedical Engineering, Yale University, New Haven, CT, USA
- Yale Stem Cell Center and Yale Cancer Center, Yale 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 School of Medicine, New Haven, CT, USA
| | - Jina Ko
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, USA.
- Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, PA, USA.
| |
Collapse
|
5
|
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
|
6
|
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
|
7
|
Tan Y, Wang A, Wang Z, Lin W, Yan Y, Nie Q, Shi J. Transfer learning of multicellular organization via single-cell and spatial transcriptomics. PLoS Comput Biol 2025; 21:e1012991. [PMID: 40258090 PMCID: PMC12061427 DOI: 10.1371/journal.pcbi.1012991] [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: 11/02/2024] [Revised: 05/08/2025] [Accepted: 03/24/2025] [Indexed: 04/23/2025] Open
Abstract
Biological tissues exhibit complex gene expression and multicellular patterns that are valuable to dissect. Single-cell RNA sequencing (scRNA-seq) provides full coverages of genes, but lacks spatial information, whereas spatial transcriptomics (ST) measures spatial locations of individual or group of cells, with more restrictions on gene information. Here we show a transfer learning method named iSORT to decipher spatial organization of cells by integrating scRNA-seq and ST data. iSORT trains a neural network that maps gene expressions to spatial locations. iSORT can find spatial patterns at single-cell scale, identify spatial-organizing genes (SOGs) that drive the patterning, and infer pseudo-growth trajectories using a concept of SpaRNA velocity. Benchmarking on a range of biological systems, such as human cortex, mouse embryo, mouse brain, Drosophila embryo, and human developmental heart, demonstrates iSORT's accuracy and practicality in reconstructing multicellular organization. We further conducted scRNA-seq and ST sequencing from normal and atherosclerotic arteries, and the functional enrichment analysis shows that SOGs found by iSORT are strongly associated with vascular structural anomalies.
Collapse
Affiliation(s)
- Yecheng Tan
- Research Institute of Intelligent Complex Systems, Fudan University, Shanghai, China
- Institute of Science and Technology for Brain-Inspired Intelligence, Fudan University, Shanghai, China
| | - Ai Wang
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Zezhou Wang
- Research Institute of Intelligent Complex Systems, Fudan University, Shanghai, China
- Shanghai Center for Mathematical Sciences, Fudan University, Shanghai, China
| | - Wei Lin
- Research Institute of Intelligent Complex Systems, Fudan University, Shanghai, China
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Fudan University, Shanghai, China
- Shanghai Artificial Intelligence Laboratory, Shanghai, China
| | - Yan Yan
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Qing Nie
- Department of Mathematics, University of California Irvine, Irvine, California, United States of America
| | - Jifan Shi
- Research Institute of Intelligent Complex Systems, Fudan University, Shanghai, China
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Fudan University, Shanghai, China
- Shanghai Artificial Intelligence Laboratory, Shanghai, China
| |
Collapse
|
8
|
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
|
9
|
Cai J, Wu S, Cheng H, Zhong W, Yuan GC, Ma P. Protocol to boost the robustness and accuracy of spatial transcriptomics algorithms using ensemble techniques. STAR Protoc 2025; 6:103608. [PMID: 39879360 PMCID: PMC11803146 DOI: 10.1016/j.xpro.2025.103608] [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: 08/20/2024] [Revised: 11/14/2024] [Accepted: 01/08/2025] [Indexed: 01/31/2025] Open
Abstract
Spatial transcriptomics enhances our understanding of cellular organization by mapping gene expression data to precise tissue locations. Here, we present a protocol for using weighted ensemble method for spatial transcriptomics (WEST), which uses ensemble techniques to boost the robustness and accuracy of existing algorithms. We describe steps for preprocessing data, obtaining embeddings from individual algorithms, and ensemble integrating all embeddings as a similarity matrix. We then detail procedures for using the similarity matrix to identify spatial domains and obtain new embeddings. For complete details on the use and execution of this protocol, please refer to Cai et al.1.
Collapse
Affiliation(s)
- Jiazhang Cai
- Department of Statistics, University of Georgia, 310 Herty Drive, Athens, GA 30602, USA
| | - Shushan Wu
- Department of Statistics, University of Georgia, 310 Herty Drive, Athens, GA 30602, USA
| | - Huimin Cheng
- Department of Biostatistics, Boston University, 801 Massachusetts Avenue Crosstown Center, Boston, MA 02118, USA
| | - Wenxuan Zhong
- Department of Statistics, University of Georgia, 310 Herty Drive, Athens, GA 30602, USA.
| | - Guo-Cheng Yuan
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, 1468 Madison Avenue, New York, NY 10029, USA.
| | - Ping Ma
- Department of Statistics, University of Georgia, 310 Herty Drive, Athens, GA 30602, USA.
| |
Collapse
|
10
|
Bryan JP, Farhi SL, Cleary B. Accurate trajectory inference in time-series spatial transcriptomics with structurally-constrained optimal transport. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.03.19.644194. [PMID: 40166168 PMCID: PMC11957147 DOI: 10.1101/2025.03.19.644194] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/02/2025]
Abstract
New experimental and computational methods use genetic or gene expression observations with single cell resolution to study the relationship between single-cell molecular profiles and developmental trajectories. Most tissues contain spatially contiguous regions that develop as a unit, such as follicles in the ovary, or tubules and glomeruli in the kidney. We find that existing approaches designed to use time series spatial transcriptomics (ST) data produce biologically incoherent trajectories that fail to maintain these structural units over time. We present Spatiotemporal Optimal transport with Contiguous Structures (SOCS), an Optimal Transport-based trajectory inference method for time-series ST that produces trajectory inferences preserving the structural integrity of contiguous biologically meaningful units, along with gene expression similarity and global geometric structure. We show that SOCS produces more plausible trajectory estimates, maintaining the spatial coherence of biological structures across time, enabling more accurate trajectory inference and biological insight than other approaches.
Collapse
Affiliation(s)
- John P Bryan
- Department of Electrical Engineering and Computer Science, Massachusetts Institute of Technology, Cambridge, MA
- Spatial Technology Platform, Broad Institute of MIT and Harvard, Cambridge, MA
| | - Samouil L Farhi
- Spatial Technology Platform, Broad Institute of MIT and Harvard, Cambridge, MA
| | - Brian Cleary
- Faculty of Computing and Data Sciences, Boston University, Boston, MA; Department of Biology, Boston University, Boston, MA; Department of Biomedical Engineering, Boston University, Boston, MA; Program in Bioinformatics, Boston University, Boston, MA; Biological Design Center, Boston University, Boston, MA
| |
Collapse
|
11
|
Jing SY, Wang HQ, Lin P, Yuan J, Tang ZX, Li H. Quantifying and interpreting biologically meaningful spatial signatures within tumor microenvironments. NPJ Precis Oncol 2025; 9:68. [PMID: 40069556 PMCID: PMC11897387 DOI: 10.1038/s41698-025-00857-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2024] [Accepted: 02/25/2025] [Indexed: 03/15/2025] Open
Abstract
The tumor microenvironment (TME) plays a crucial role in orchestrating tumor cell behavior and cancer progression. Recent advances in spatial profiling technologies have uncovered novel spatial signatures, including univariate distribution patterns, bivariate spatial relationships, and higher-order structures. These signatures have the potential to revolutionize tumor mechanism and treatment. In this review, we summarize the current state of spatial signature research, highlighting computational methods to uncover spatially relevant biological significance. We discuss the impact of these advances on fundamental cancer biology and translational research, address current challenges and future research directions.
Collapse
Affiliation(s)
- Si-Yu Jing
- CAS Key Laboratory of Computational Biology, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, People's Republic of China
| | - He-Qi Wang
- CAS Key Laboratory of Computational Biology, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, People's Republic of 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, People's Republic of 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, People's Republic of China
| | - Zhi-Xuan Tang
- CAS Key Laboratory of Computational Biology, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, People's Republic of 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, People's Republic of China.
| |
Collapse
|
12
|
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
|
13
|
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
|
14
|
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
|
15
|
Su H, Wu Y, Chen B, Cui Y. STANCE: a unified statistical model to detect cell-type-specific spatially variable genes in spatial transcriptomics. Nat Commun 2025; 16:1793. [PMID: 39979358 PMCID: PMC11842841 DOI: 10.1038/s41467-025-57117-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2024] [Accepted: 02/10/2025] [Indexed: 02/22/2025] Open
Abstract
One of the major challenges in spatial transcriptomics is to detect spatially variable genes (SVGs), whose expression patterns are non-random across tissue locations. Many SVGs correlate with cell type compositions, introducing the concept of cell type-specific SVGs (ctSVGs). Existing ctSVG detection methods treat cell type-specific spatial effects as fixed effects, leading to tissue spatial rotation-dependent results. Moreover, SVGs may exhibit random spatial patterns within cell types, meaning an SVG is not always a ctSVG, and vice versa, further complicating detection. We propose STANCE, a unified statistical model for both SVGs and ctSVGs detection under a linear mixed-effect model framework that integrates gene expression, spatial location, and cell type composition information. STANCE ensures tissue rotation-invariant results, with a two-stage approach: initial SVG/ctSVG detection followed by ctSVG-specific testing. We demonstrate its performance through extensive simulations and analyses of public datasets. Downstream analyses reveal STANCE's potential in spatial transcriptomics analysis.
Collapse
Affiliation(s)
- Haohao Su
- Department of Statistics and Probability, Michigan State University, East Lansing, 48824, MI, USA
| | - Yuesong Wu
- Department of Statistics and Probability, Michigan State University, East Lansing, 48824, MI, USA
| | - Bin Chen
- Department of Pharmacology and Toxicology, Michigan State University, East Lansing, 48824, MI, USA
- Department of Computer Science and Engineering, Michigan State University, East Lansing, 48824, MI, USA
- Department of Pediatrics and Human Development, Michigan State University, Grand Rapids, 49503, MI, USA
| | - Yuehua Cui
- Department of Statistics and Probability, Michigan State University, East Lansing, 48824, MI, USA.
| |
Collapse
|
16
|
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
|
17
|
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
|
18
|
Zhou Y, Tang C, Xiao X, Zhan X, Wang T, Xiao G, Xu L. Dimensionality reduction for visualizing spatially resolved profiling data using SpaSNE. Gigascience 2025; 14:giaf002. [PMID: 39960663 PMCID: PMC11831803 DOI: 10.1093/gigascience/giaf002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2024] [Revised: 11/05/2024] [Accepted: 01/06/2025] [Indexed: 02/20/2025] Open
Abstract
BACKGROUND Spatially resolved profiling technologies to quantify transcriptomes, epigenomes, and proteomes have been emerging as groundbreaking methods for comprehensive molecular characterizations. Dimensionality reduction and visualization is an essential step to analyze and interpret spatially resolved profiling data. However, state-of-the-art dimensionality reduction methods for single-cell sequencing data, such as the t-distributed stochastic neighbor embedding (t-SNE) and uniform manifold approximation and projection (UMAP), were not tailored for spatially resolved profiling data. RESULTS Here we developed a spatially resolved t-SNE (SpaSNE) method to integrate both spatial and molecular information. We applied it to a variety of public spatially resolved profiling datasets that were generated from 3 experimental platforms and consisted of cells from different diseases, tissues, and cell types. To compare the performances of SpaSNE, t-SNE, and UMAP, we applied them to 4 spatially resolved profiling datasets obtained from 3 distinct experimental platforms (Visium, STARmap, and MERFISH) on both diseased and normal tissues. Comparisons between SpaSNE and these state-of-the-art approaches reveal that SpaSNE achieves more accurate and meaningful visualization that better elucidates the underlying spatial and molecular data structures. CONCLUSIONS This work demonstrates the broad application of SpaSNE for reliable and robust interpretation of cell types based on both molecular and spatial information, which can set the foundation for many subsequent analysis steps, such as differential gene expression and trajectory or pseudotime analysis on the spatially resolved profiling data.
Collapse
Affiliation(s)
- Yuansheng Zhou
- Quantitative Biomedical Research Center, Peter O'Donnell Jr. School of Public Health, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Chen Tang
- Quantitative Biomedical Research Center, Peter O'Donnell Jr. School of Public Health, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Xue Xiao
- Quantitative Biomedical Research Center, Peter O'Donnell Jr. School of Public Health, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Xiaowei Zhan
- Quantitative Biomedical Research Center, Peter O'Donnell Jr. School of Public Health, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
- Center for the Genetics of Host Defense, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Tao Wang
- Quantitative Biomedical Research Center, Peter O'Donnell Jr. School of Public Health, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
- Center for the Genetics of Host Defense, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Guanghua Xiao
- Quantitative Biomedical Research Center, Peter O'Donnell Jr. School of Public Health, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
- Department of Bioinformatics, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Lin Xu
- Quantitative Biomedical Research Center, Peter O'Donnell Jr. School of Public Health, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
- Department of Pediatrics, Division of Hematology/Oncology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| |
Collapse
|
19
|
Zhou Y, Xiao X, Dong L, Tang C, Xiao G, Xu L. Cooperative integration of spatially resolved multi-omics data with COSMOS. Nat Commun 2025; 16:27. [PMID: 39747840 PMCID: PMC11696235 DOI: 10.1038/s41467-024-55204-y] [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: 05/02/2024] [Accepted: 11/26/2024] [Indexed: 01/04/2025] Open
Abstract
Recent advancements in biological technologies have enabled the measurement of spatially resolved multi-omics data, yet computational algorithms for this purpose are scarce. Existing tools target either single omics or lack spatial integration. We generate a graph neural network algorithm named COSMOS to address this gap and demonstrated the superior performance of COSMOS in domain segmentation, visualization, and spatiotemporal map for spatially resolved multi-omics data integration tasks.
Collapse
Affiliation(s)
- Yuansheng Zhou
- Quantitative Biomedical Research Center, Peter O'Donnell Jr. School of Public Health, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
| | - Xue Xiao
- Quantitative Biomedical Research Center, Peter O'Donnell Jr. School of Public Health, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
| | - Lei Dong
- Quantitative Biomedical Research Center, Peter O'Donnell Jr. School of Public Health, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
| | - Chen Tang
- Quantitative Biomedical Research Center, Peter O'Donnell Jr. School of Public Health, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
| | - Guanghua Xiao
- Quantitative Biomedical Research Center, Peter O'Donnell Jr. School of Public Health, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA.
- Department of Bioinformatics, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA.
| | - Lin Xu
- Quantitative Biomedical Research Center, Peter O'Donnell Jr. School of Public Health, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA.
- Department of Pediatrics, Division of Hematology/Oncology, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA.
| |
Collapse
|
20
|
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
|
21
|
Yu Z, Yang Y, Chen X, Wong K, Zhang Z, Zhao Y, Li X. Accurate Spatial Heterogeneity Dissection and Gene Regulation Interpretation for Spatial Transcriptomics using Dual Graph Contrastive Learning. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2410081. [PMID: 39605202 PMCID: PMC11744562 DOI: 10.1002/advs.202410081] [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: 08/22/2024] [Revised: 10/31/2024] [Indexed: 11/29/2024]
Abstract
Recent advances in spatial transcriptomics have enabled simultaneous preservation of high-throughput gene expression profiles and the spatial context, enabling high-resolution exploration of distinct regional characterization in tissue. To effectively understand the underlying biological mechanisms within tissue microenvironments, there is a requisite for methods that can accurately capture external spatial heterogeneity and interpret internal gene regulation from spatial transcriptomics data. However, current methods for region identification often lack the simultaneous characterizing of spatial structure and gene regulation, thereby limiting the ability of spatial dissection and gene interpretation. Here, stDCL is developed, a dual graph contrastive learning method to identify spatial domains and interpret gene regulation in spatial transcriptomics data. stDCL adaptively incorporates gene expression data and spatial information via a graph embedding autoencoder, thereby preserving critical information within the latent embedding representations. In addition, dual graph contrastive learning is proposed to train the model, ensuring that the latent embedding representation closely resembles the actual spatial distribution and exhibits cluster similarity. Benchmarking stDCL against other state-of-the-art clustering methods using complex cortex datasets demonstrates its superior accuracy and effectiveness in identifying spatial domains. Our analysis of the imputation matrices generated by stDCL reveals its capability to reconstruct spatial hierarchical structures and refine differential expression assessment. Furthermore, it is demonstrated that the versatility of stDCL in interpretability of gene regulation, spatial heterogeneity at high resolution, and embryonic developmental patterns. In addition, it is also showed that stDCL can successfully annotate disease-associated astrocyte subtypes in Alzheimer's disease and unravel multiple relevant pathways and regulatory mechanisms.
Collapse
Affiliation(s)
- Zhuohan Yu
- School of Artificial IntelligenceJilin UniversityJilin130012China
| | - Yuning Yang
- Terrence Donnelly Centre for Cellular and Biomolecular ResearchUniversity of TorontoTorontoONM5S 3E1Canada
| | - Xingjian Chen
- Cutaneous Biology Research Center, Massachusetts General HospitalHarvard Medical SchoolBostonMA02115USA
| | - Ka‐Chun Wong
- Department of Computer ScienceCity University of Hong KongHong KongSAR999077Hong Kong
| | - Zhaolei Zhang
- Terrence Donnelly Centre for Cellular and Biomolecular ResearchUniversity of TorontoTorontoONM5S 3E1Canada
| | - Yuming Zhao
- College of Computer and Control EngineeringNortheast Forestry UniversityHarbin150040China
| | - Xiangtao Li
- School of Artificial IntelligenceJilin UniversityJilin130012China
- Department of Computer ScienceCity University of Hong KongHong KongSAR999077Hong Kong
| |
Collapse
|
22
|
Hua Y, Zhang Y, Guo Z, Bian S, Zhang Y. ImSpiRE: image feature-aided spatial resolution enhancement method. SCIENCE CHINA. LIFE SCIENCES 2025; 68:272-283. [PMID: 39327391 DOI: 10.1007/s11427-023-2636-9] [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: 04/09/2024] [Accepted: 05/31/2024] [Indexed: 09/28/2024]
Abstract
The resolution of most spatially resolved transcriptomic technologies usually cannot attain the single-cell level, limiting their applications in biological discoveries. Here, we introduce ImSpiRE, an image feature-aided spatial resolution enhancement method for in situ capturing spatial transcriptome. Taking the information stored in histological images, ImSpiRE solves an optimal transport problem to redistribute the expression profiles of spots to construct new transcriptional profiles with enhanced resolution, together with extending the gene expression profiles into unmeasured regions. Applications to multiple datasets confirm that ImSpiRE can enhance spatial resolution to the subspot level while contributing to the discovery of tissue domains, signaling communication patterns, and spatiotemporal characterization.
Collapse
Affiliation(s)
- Yuwei Hua
- State Key Laboratory of Cardiovascular Diseases and Medical Innovation Center, Institute for Regenerative Medicine, Department of Neurosurgery, Shanghai East Hospital, Shanghai Key Laboratory of Signaling and Disease Research, Frontier Science Center for Stem Cell Research, School of Life Sciences and Technology, Tongji University, Shanghai, 200092, China
| | - Yizhi Zhang
- State Key Laboratory of Cardiovascular Diseases and Medical Innovation Center, Institute for Regenerative Medicine, Department of Neurosurgery, Shanghai East Hospital, Shanghai Key Laboratory of Signaling and Disease Research, Frontier Science Center for Stem Cell Research, School of Life Sciences and Technology, Tongji University, Shanghai, 200092, China
| | - Zhenming Guo
- State Key Laboratory of Cardiovascular Diseases and Medical Innovation Center, Institute for Regenerative Medicine, Department of Neurosurgery, Shanghai East Hospital, Shanghai Key Laboratory of Signaling and Disease Research, Frontier Science Center for Stem Cell Research, School of Life Sciences and Technology, Tongji University, Shanghai, 200092, China
| | - Shan Bian
- State Key Laboratory of Cardiovascular Diseases and Medical Innovation Center, Institute for Regenerative Medicine, Department of Neurosurgery, Shanghai East Hospital, Shanghai Key Laboratory of Signaling and Disease Research, Frontier Science Center for Stem Cell Research, School of Life Sciences and Technology, Tongji University, Shanghai, 200092, China
| | - Yong Zhang
- State Key Laboratory of Cardiovascular Diseases and Medical Innovation Center, Institute for Regenerative Medicine, Department of Neurosurgery, Shanghai East Hospital, Shanghai Key Laboratory of Signaling and Disease Research, Frontier Science Center for Stem Cell Research, School of Life Sciences and Technology, Tongji University, Shanghai, 200092, China.
| |
Collapse
|
23
|
Zhang F, Shen Z, Huang S, Zhu Y, Yi M. SpaInGNN: Enhanced clustering and integration of spatial transcriptomics based on refined graph neural networks. Methods 2025; 233:42-51. [PMID: 39542070 DOI: 10.1016/j.ymeth.2024.11.006] [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: 09/02/2024] [Revised: 10/22/2024] [Accepted: 11/07/2024] [Indexed: 11/17/2024] Open
Abstract
Recent developments in spatial transcriptomics (ST) technology have markedly enhanced the proposed capacity to comprehensively characterize gene expression patterns within tissue microenvironments while crucially preserving spatial context. However, the identification of spatial domains at the single-cell level remains a significant challenge in elucidating biological processes. To address this, SpaInGNN was developed, a sophisticated graph neural network (GNN) framework that accurately delineates spatial domains by integrating spatial location data, histological information, and gene expression profiles into low-dimensional latent embeddings. Additionally, to fully leverage spatial coordinate data, spatial integration using graph neural network (SpaInGNN) refines the graph constructed for spatial locations by incorporating both tissue image distance and Euclidean distance, following a pre-clustering of gene expression profiles. This refined graph is then embedded using a self-supervised GNN, which minimizes self-reconfiguration loss. By applying SpaInGNN to refined graphs across multiple consecutive tissue slices, this study mitigates the impact of batch effects in data analysis. The proposed method demonstrates substantial improvements in the accuracy of spatial domain recognition, providing a more faithful representation of the tissue organization in both mouse olfactory bulb and human lateral prefrontal cortex samples.
Collapse
Affiliation(s)
- Fangqin Zhang
- Shool of Mathematics and Physics, China University of Geosciences, Wuhan 430074, China
| | - Zhan Shen
- Shool of Mathematics and Physics, China University of Geosciences, Wuhan 430074, China
| | - Siyi Huang
- Shool of Mathematics and Physics, China University of Geosciences, Wuhan 430074, China
| | - Yuan Zhu
- Shool of Mathematics and Physics, China University of Geosciences, Wuhan 430074, China
| | - Ming Yi
- Shool of Mathematics and Physics, China University of Geosciences, Wuhan 430074, China.
| |
Collapse
|
24
|
Gulati GS, D'Silva JP, Liu Y, Wang L, Newman AM. Profiling cell identity and tissue architecture with single-cell and spatial transcriptomics. Nat Rev Mol Cell Biol 2025; 26:11-31. [PMID: 39169166 DOI: 10.1038/s41580-024-00768-2] [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] [Accepted: 07/16/2024] [Indexed: 08/23/2024]
Abstract
Single-cell transcriptomics has broadened our understanding of cellular diversity and gene expression dynamics in healthy and diseased tissues. Recently, spatial transcriptomics has emerged as a tool to contextualize single cells in multicellular neighbourhoods and to identify spatially recurrent phenotypes, or ecotypes. These technologies have generated vast datasets with targeted-transcriptome and whole-transcriptome profiles of hundreds to millions of cells. Such data have provided new insights into developmental hierarchies, cellular plasticity and diverse tissue microenvironments, and spurred a burst of innovation in computational methods for single-cell analysis. In this Review, we discuss recent advancements, ongoing challenges and prospects in identifying and characterizing cell states and multicellular neighbourhoods. We discuss recent progress in sample processing, data integration, identification of subtle cell states, trajectory modelling, deconvolution and spatial analysis. Furthermore, we discuss the increasing application of deep learning, including foundation models, in analysing single-cell and spatial transcriptomics data. Finally, we discuss recent applications of these tools in the fields of stem cell biology, immunology, and tumour biology, and the future of single-cell and spatial transcriptomics in biological research and its translation to the clinic.
Collapse
Affiliation(s)
- Gunsagar S Gulati
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | | | - Yunhe Liu
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Linghua Wang
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
- The University of Texas MD Anderson Cancer Center UTHealth Houston Graduate School of Biomedical Sciences, Houston, TX, USA
| | - Aaron M Newman
- Department of Biomedical Data Science, Stanford University, Stanford, CA, USA.
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University, Stanford, CA, USA.
- Stanford Cancer Institute, Stanford University, Stanford, CA, USA.
- Chan Zuckerberg Biohub - San Francisco, San Francisco, CA, USA.
| |
Collapse
|
25
|
Sun X, Zhang W, Li W, Yu N, Zhang D, Zou Q, Dong Q, Zhang X, Liu Z, Yuan Z, Gao R. SpaGRA: Graph augmentation facilitates domain identification for spatially resolved transcriptomics. J Genet Genomics 2025; 52:93-104. [PMID: 39362628 DOI: 10.1016/j.jgg.2024.09.015] [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: 06/23/2024] [Revised: 09/07/2024] [Accepted: 09/22/2024] [Indexed: 10/05/2024]
Abstract
Recent advances in spatially resolved transcriptomics (SRT) have provided new opportunities for characterizing spatial structures of various tissues. Graph-based geometric deep learning has gained widespread adoption for spatial domain identification tasks. Currently, most methods define adjacency relation between cells or spots by their spatial distance in SRT data, which overlooks key biological interactions like gene expression similarities, and leads to inaccuracies in spatial domain identification. To tackle this challenge, we propose a novel method, SpaGRA (https://github.com/sunxue-yy/SpaGRA), for automatic multi-relationship construction based on graph augmentation. SpaGRA uses spatial distance as prior knowledge and dynamically adjusts edge weights with multi-head graph attention networks (GATs). This helps SpaGRA to uncover diverse node relationships and enhance message passing in geometric contrastive learning. Additionally, SpaGRA uses these multi-view relationships to construct negative samples, addressing sampling bias posed by random selection. Experimental results show that SpaGRA presents superior domain identification performance on multiple datasets generated from different protocols. Using SpaGRA, we analyze the functional regions in the mouse hypothalamus, identify key genes related to heart development in mouse embryos, and observe cancer-associated fibroblasts enveloping cancer cells in the latest Visium HD data. Overall, SpaGRA can effectively characterize spatial structures across diverse SRT datasets.
Collapse
Affiliation(s)
- Xue Sun
- Center of Intelligent Medicine, School of Control Science and Engineering, Shandong University, Jinan, Shandong 250061, China
| | - Wei Zhang
- Center of Intelligent Medicine, School of Control Science and Engineering, Shandong University, Jinan, Shandong 250061, China
| | - Wenrui Li
- MOE Key Lab of Bioinformatics and Bioinformatics Division of BNRIST, Department of Automation, Tsinghua University, Beijing 100084, China
| | - Na Yu
- Center of Intelligent Medicine, School of Control Science and Engineering, Shandong University, Jinan, Shandong 250061, China
| | - Daoliang Zhang
- Center of Intelligent Medicine, School of Control Science and Engineering, Shandong University, Jinan, Shandong 250061, China
| | - Qi Zou
- Center of Intelligent Medicine, School of Control Science and Engineering, Shandong University, Jinan, Shandong 250061, China
| | - Qiongye Dong
- Institute of Precision Medicine, Peking University Shenzhen Hospital, Shenzhen, Guangdong 518036, China
| | - Xianglin Zhang
- Department of Clinical Laboratory, The Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250033, China
| | - Zhiping Liu
- Center of Intelligent Medicine, School of Control Science and Engineering, Shandong University, Jinan, Shandong 250061, China
| | - Zhiyuan Yuan
- Institute of Science and Technology for Brain-Inspired Intelligence, Center for Medical Research and Innovation, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Fudan University, Shanghai 200433, China.
| | - Rui Gao
- Center of Intelligent Medicine, School of Control Science and Engineering, Shandong University, Jinan, Shandong 250061, China.
| |
Collapse
|
26
|
Schaub DP, Yousefi B, Kaiser N, Khatri R, Puelles VG, Krebs CF, Panzer U, Bonn S. PCA-based spatial domain identification with state-of-the-art performance. Bioinformatics 2024; 41:btaf005. [PMID: 39775801 PMCID: PMC11761416 DOI: 10.1093/bioinformatics/btaf005] [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: 08/26/2024] [Revised: 11/25/2024] [Accepted: 01/06/2025] [Indexed: 01/11/2025] Open
Abstract
MOTIVATION The identification of biologically meaningful domains is a central step in the analysis of spatial transcriptomic data. RESULTS Following Occam's razor, we show that a simple PCA-based algorithm for unsupervised spatial domain identification rivals the performance of ten competing state-of-the-art methods across six single-cell spatial transcriptomic datasets. Our reductionist approach, NichePCA, provides researchers with intuitive domain interpretation and excels in execution speed, robustness, and scalability. AVAILABILITY AND IMPLEMENTATION The code is available at https://github.com/imsb-uke/nichepca.
Collapse
Affiliation(s)
- Darius P Schaub
- Institute of Medical Systems Bioinformatics, Center for Biomedical AI (bAIome), Center for Molecular Neurobiology (ZMNH), University Medical Center Hamburg-Eppendorf, Hamburg 20246, Germany
- III Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg 20246, Germany
| | - Behnam Yousefi
- Institute of Medical Systems Bioinformatics, Center for Biomedical AI (bAIome), Center for Molecular Neurobiology (ZMNH), University Medical Center Hamburg-Eppendorf, Hamburg 20246, Germany
- German Center for Child and Adolescent Health (DZKJ), Partner Site Hamburg, University Medical Center Hamburg-Eppendorf, Hamburg 20246, Germany
| | - Nico Kaiser
- Institute of Medical Systems Bioinformatics, Center for Biomedical AI (bAIome), Center for Molecular Neurobiology (ZMNH), University Medical Center Hamburg-Eppendorf, Hamburg 20246, Germany
- III Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg 20246, Germany
| | - Robin Khatri
- Institute of Medical Systems Bioinformatics, Center for Biomedical AI (bAIome), Center for Molecular Neurobiology (ZMNH), University Medical Center Hamburg-Eppendorf, Hamburg 20246, Germany
| | - Victor G Puelles
- III Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg 20246, Germany
- Hamburg Center for Kidney Health (HCKH), University Medical Center Hamburg-Eppendorf, Hamburg 20246, Germany
- Department of Clinical Medicine, Aarhus University, Aarhus 8200, Denmark
- Department of Pathology, Aarhus University Hospital, Aarhus 8200, Denmark
| | - Christian F Krebs
- III Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg 20246, Germany
- Hamburg Center for Kidney Health (HCKH), University Medical Center Hamburg-Eppendorf, Hamburg 20246, Germany
- Hamburg Center for Translational Immunology (HCTI), University Medical Center Hamburg-Eppendorf, Hamburg 20246, Germany
| | - Ulf Panzer
- III Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg 20246, Germany
- Hamburg Center for Kidney Health (HCKH), University Medical Center Hamburg-Eppendorf, Hamburg 20246, Germany
- Hamburg Center for Translational Immunology (HCTI), University Medical Center Hamburg-Eppendorf, Hamburg 20246, Germany
| | - Stefan Bonn
- Institute of Medical Systems Bioinformatics, Center for Biomedical AI (bAIome), Center for Molecular Neurobiology (ZMNH), University Medical Center Hamburg-Eppendorf, Hamburg 20246, Germany
- German Center for Child and Adolescent Health (DZKJ), Partner Site Hamburg, University Medical Center Hamburg-Eppendorf, Hamburg 20246, Germany
- Hamburg Center for Kidney Health (HCKH), University Medical Center Hamburg-Eppendorf, Hamburg 20246, Germany
- Hamburg Center for Translational Immunology (HCTI), University Medical Center Hamburg-Eppendorf, Hamburg 20246, Germany
| |
Collapse
|
27
|
Wang Z, Geng A, Duan H, Cui F, Zou Q, Zhang Z. A comprehensive review of approaches for spatial domain recognition of spatial transcriptomes. Brief Funct Genomics 2024; 23:702-712. [PMID: 39426802 DOI: 10.1093/bfgp/elae040] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2024] [Revised: 10/02/2024] [Accepted: 10/07/2024] [Indexed: 10/21/2024] Open
Abstract
In current bioinformatics research, spatial transcriptomics (ST) as a rapidly evolving technology is gradually receiving widespread attention from researchers. Spatial domains are regions where gene expression and histology are consistent in space, and detecting spatial domains can better understand the organization and functional distribution of tissues. Spatial domain recognition is a fundamental step in the process of ST data interpretation, which is also a major challenge in ST analysis. Therefore, developing more accurate, efficient, and general spatial domain recognition methods has become an important and urgent research direction. This article aims to review the current status and progress of spatial domain recognition research, explore the advantages and limitations of existing methods, and provide suggestions and directions for future tool development.
Collapse
Affiliation(s)
- Ziyi Wang
- School of Computer Science and Technology, Hainan University, Haikou 570228, China
| | - Aoyun Geng
- School of Computer Science and Technology, Hainan University, Haikou 570228, China
| | - Hao Duan
- School of Computer Science and Technology, Hainan University, Haikou 570228, China
| | - Feifei Cui
- School of Computer Science and Technology, Hainan University, Haikou 570228, China
| | - Quan Zou
- Institute of Fundamental and Frontier Sciences, University of Electronic Science and Technology of China, Chengdu 610054, China
- Yangtze Delta Region Institute (Quzhou), University of Electronic Science and Technology of China, Quzhou 324000, China
| | - Zilong Zhang
- School of Computer Science and Technology, Hainan University, Haikou 570228, China
| |
Collapse
|
28
|
Liu T, Fang ZY, Zhang Z, Yu Y, Li M, Yin MZ. A comprehensive overview of graph neural network-based approaches to clustering for spatial transcriptomics. Comput Struct Biotechnol J 2024; 23:106-128. [PMID: 38089467 PMCID: PMC10714345 DOI: 10.1016/j.csbj.2023.11.055] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2023] [Revised: 11/24/2023] [Accepted: 11/27/2023] [Indexed: 10/16/2024] Open
Abstract
Spatial transcriptomics technologies enable researchers to accurately quantify and localize messenger ribonucleic acid (mRNA) transcripts at a high resolution while preserving their spatial context. The identification of spatial domains, or the task of spatial clustering, plays a crucial role in investigating data on spatial transcriptomes. One promising approach for classifying spatial domains involves the use of graph neural networks (GNNs) by leveraging gene expressions, spatial locations, and histological images. This study provided a comprehensive overview of the most recent GNN-based methods of spatial clustering methods for the analysis of data on spatial transcriptomics. We extensively evaluated the performance of current methods on prevalent datasets of spatial transcriptomics by considering their accuracy of clustering, robustness, data stabilization, relevant requirements, computational efficiency, and memory use. To this end, we explored 60 clustering scenarios by extending the essential frameworks of spatial clustering for the selection of the GNNs, algorithms of downstream clustering, principal component analysis (PCA)-based reduction, and refined methods of correction. We comparatively analyzed the performance of the methods in terms of spatial clustering to identify their limitations and outline future directions of research in the field. Our survey yielded novel insights, and provided motivation for further investigating spatial transcriptomics.
Collapse
Affiliation(s)
- Teng Liu
- Clinical Research Center (CRC), Clinical Pathology Center (CPC), Cancer Early Detection and Treatment Center (CEDTC) and Translational Medicine Research Center (TMRC), Chongqing University Three Gorges Hospital, Chongqing University, Wanzhou, Chongqing, China
- Chongqing Technical Innovation Center for Quality Evaluation and Identification of Authentic Medicinal Herbs, Chongqing, China
| | - Zhao-Yu Fang
- School of Computer Science and Engineering, Central South University, Changsha 410083, China
| | - Zongbo Zhang
- Clinical Research Center (CRC), Clinical Pathology Center (CPC), Cancer Early Detection and Treatment Center (CEDTC) and Translational Medicine Research Center (TMRC), Chongqing University Three Gorges Hospital, Chongqing University, Wanzhou, Chongqing, China
| | - Yongxiang Yu
- Clinical Research Center (CRC), Clinical Pathology Center (CPC), Cancer Early Detection and Treatment Center (CEDTC) and Translational Medicine Research Center (TMRC), Chongqing University Three Gorges Hospital, Chongqing University, Wanzhou, Chongqing, China
| | - Min Li
- School of Computer Science and Engineering, Central South University, Changsha 410083, China
- Hunan Provincial Engineering Research Center of Intelligent Computing in Biology and Medicine, Central South University, Changsha 410083, China
| | - Ming-Zhu Yin
- Clinical Research Center (CRC), Clinical Pathology Center (CPC), Cancer Early Detection and Treatment Center (CEDTC) and Translational Medicine Research Center (TMRC), Chongqing University Three Gorges Hospital, Chongqing University, Wanzhou, Chongqing, China
- Chongqing Technical Innovation Center for Quality Evaluation and Identification of Authentic Medicinal Herbs, Chongqing, China
| |
Collapse
|
29
|
Gui Y, Li C, Xu Y. Spatial domains identification in spatial transcriptomics using modality-aware and subspace-enhanced graph contrastive learning. Comput Struct Biotechnol J 2024; 23:3703-3713. [PMID: 39507820 PMCID: PMC11539238 DOI: 10.1016/j.csbj.2024.10.029] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2024] [Revised: 10/18/2024] [Accepted: 10/18/2024] [Indexed: 11/08/2024] Open
Abstract
Spatial transcriptomics (ST) technologies have emerged as an effective tool to identify the spatial architecture of tissues, facilitating a comprehensive understanding of organ function and the tissue microenvironment. Spatial domain identification is the first and most critical step in ST data analysis, which requires thoughtful utilization of tissue microenvironment and morphological priors. Here, we propose a graph contrastive learning framework, GRAS4T, which combines contrastive learning and a subspace analysis model to accurately distinguish different spatial domains by capturing the tissue microenvironment through self-expressiveness of spots within the same domain. To uncover the pertinent features for spatial domain identification, GRAS4T employs a graph augmentation based on histological image priors, preserving structural information crucial for the clustering task. Experimental results on eight ST datasets from five different platforms show that GRAS4T outperforms five state-of-the-art competing methods. Significantly, GRAS4T excels at separating distinct tissue structures and unveiling more detailed spatial domains. GRAS4T combines the advantages of subspace analysis and graph representation learning with extensibility, making it an ideal framework for ST domain identification.
Collapse
Affiliation(s)
- Yang Gui
- School of Mathematics and Physics, University of Science and Technology Beijing, Beijing, 100083, China
| | - Chao Li
- School of Statistics and Applied Mathematics, Anhui University of Finance and Economics, Bengbu, 233041, China
| | - Yan Xu
- School of Mathematics and Physics, University of Science and Technology Beijing, Beijing, 100083, China
| |
Collapse
|
30
|
Zhang D, Yu N, Sun X, Li H, Zhang W, Qiao X, Zhang W, Gao R. Deciphering spatial domains from spatially resolved transcriptomics through spatially regularized deep graph networks. BMC Genomics 2024; 25:1160. [PMID: 39614161 DOI: 10.1186/s12864-024-11072-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: 09/17/2024] [Accepted: 11/21/2024] [Indexed: 12/01/2024] Open
Abstract
BACKGROUND Recent advancements in spatially resolved transcriptomics (SRT) have opened up unprecedented opportunities to explore gene expression patterns within spatial contexts. Deciphering spatial domains is a critical task in spatial transcriptomic data analysis, aiding in the elucidation of tissue structural heterogeneity and biological functions. However, existing spatial domain detection methods ignore the consistency of expression patterns and spatial arrangements between spots, as well as the severe gene dropout phenomenon present in SRT data, resulting in suboptimal performance in identifying tissue spatial heterogeneity. RESULTS In this paper, we introduce a novel framework, spatially regularized deep graph networks (SR-DGN), which integrates gene expression profiles with spatial information to learn spatially-consistent and informative spot representations. Specifically, SR-DGN employs graph attention networks (GAT) to adaptively aggregate gene expression information from neighboring spots, considering local expression patterns between spots. In addition, the spatial regularization constraint ensures the consistency of neighborhood relationships between physical and embedded spaces in an end-to-end manner. SR-DGN also employs cross-entropy (CE) loss to model gene expression states, effectively mitigating the impact of noisy gene dropouts. CONCLUSIONS Experimental results demonstrate that SR-DGN outperforms state-of-the-art methods in spatial domain identification across SRT data from different sequencing platforms. Moreover, SR-DGN is capable of recovering known microanatomical structures, yielding clearer low-dimensional visualizations and more accurate spatial trajectory inferences.
Collapse
Affiliation(s)
- Daoliang Zhang
- Center of Intelligent Medicine, School of Control Science and Engineering, Shandong University, Jinan, Shandong, 250061, China
| | - Na Yu
- Center of Intelligent Medicine, School of Control Science and Engineering, Shandong University, Jinan, Shandong, 250061, China
| | - Xue Sun
- Center of Intelligent Medicine, School of Control Science and Engineering, Shandong University, Jinan, Shandong, 250061, China
| | - Haoyang Li
- Center of Intelligent Medicine, School of Control Science and Engineering, Shandong University, Jinan, Shandong, 250061, China
| | - Wenjing Zhang
- Center of Intelligent Medicine, School of Control Science and Engineering, Shandong University, Jinan, Shandong, 250061, China
| | - Xu Qiao
- Center of Intelligent Medicine, School of Control Science and Engineering, Shandong University, Jinan, Shandong, 250061, China.
| | - Wei Zhang
- Center of Intelligent Medicine, School of Control Science and Engineering, Shandong University, Jinan, Shandong, 250061, China.
| | - Rui Gao
- Center of Intelligent Medicine, School of Control Science and Engineering, Shandong University, Jinan, Shandong, 250061, China.
| |
Collapse
|
31
|
Yang Y, Zhang C, Liu Z, Aihara K, Zhang C, Chen L, Wei W. MCGAE: unraveling tumor invasion through integrated multimodal spatial transcriptomics. Brief Bioinform 2024; 26:bbae608. [PMID: 39576225 PMCID: PMC11583448 DOI: 10.1093/bib/bbae608] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Revised: 10/16/2024] [Accepted: 11/07/2024] [Indexed: 11/24/2024] Open
Abstract
Spatially Resolved Transcriptomics (SRT) serves as a cornerstone in biomedical research, revealing the heterogeneity of tissue microenvironments. Integrating multimodal data including gene expression, spatial coordinates, and morphological information poses significant challenges for accurate spatial domain identification. Herein, we present the Multi-view Contrastive Graph Autoencoder (MCGAE), a cutting-edge deep computational framework specifically designed for the intricate analysis of spatial transcriptomics (ST) data. MCGAE advances the field by creating multi-view representations from gene expression and spatial adjacency matrices. Utilizing modular modeling, contrastive graph convolutional networks, and attention mechanisms, it generates modality-specific spatial representations and integrates them into a unified embedding. This integration process is further enriched by the inclusion of morphological image features, markedly enhancing the framework's capability to process multimodal data. Applied to both simulated and real SRT datasets, MCGAE demonstrates superior performance in spatial domain detection, data denoising, trajectory inference, and 3D feature extraction, outperforming existing methods. Specifically, in colorectal cancer liver metastases, MCGAE integrates histological and gene expression data to identify tumor invasion regions and characterize cellular molecular regulation. This breakthrough extends ST analysis and offers new tools for cancer and complex disease research.
Collapse
Affiliation(s)
- Yiwen Yang
- Lingang Laboratory, Building 8, 319 Yueyang Road, Xuhui District, Shanghai 200031, China
| | - Chengming Zhang
- International Research Center for Neurointelligence, The University of Tokyo Institutes for Advanced Study, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-0033, Japan
| | - Zhaonan Liu
- Department of Biliary-Pancreatic Surgery, Renji Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, 160 Pujian Road, Pudong District, Shanghai 200127, China
| | - Kazuyuki Aihara
- International Research Center for Neurointelligence, The University of Tokyo Institutes for Advanced Study, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-0033, Japan
| | - Chuanchao Zhang
- Key Laboratory of Systems Health Science of Zhejiang Province, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Chinese Academy of Sciences, 1 Sub-Lane Xiangshan Road, West Lake District, Hangzhou 310024, China
- Guangdong Institute of Intelligence Science and Technology, Hengqin, Zhuhai, Guangdong 519031, China
| | - Luonan Chen
- Key Laboratory of Systems Health Science of Zhejiang Province, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Chinese Academy of Sciences, 1 Sub-Lane Xiangshan Road, West Lake District, Hangzhou 310024, China
- Guangdong Institute of Intelligence Science and Technology, Hengqin, Zhuhai, Guangdong 519031, China
- Key Laboratory of Systems Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, 320 Yueyang Road, Xuhui District, Shanghai 200031, China
| | - Wu Wei
- Lingang Laboratory, Building 8, 319 Yueyang Road, Xuhui District, Shanghai 200031, China
| |
Collapse
|
32
|
Mo Y, Liu J, Zhang L. Deconvolution of spatial transcriptomics data via graph contrastive learning and partial least square regression. Brief Bioinform 2024; 26:bbaf052. [PMID: 39924717 PMCID: PMC11807730 DOI: 10.1093/bib/bbaf052] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2024] [Revised: 12/19/2024] [Accepted: 01/24/2025] [Indexed: 02/11/2025] Open
Abstract
Deciphering the cellular abundance in spatial transcriptomics (ST) is crucial for revealing the spatial architecture of cellular heterogeneity within tissues. However, some of the current spatial sequencing technologies are in low resolutions, leading to each spot having multiple heterogeneous cells. Additionally, current spatial deconvolution methods lack the ability to utilize multi-modality information such as gene expression and chromatin accessibility from single-cell multi-omics data. In this study, we introduce a graph Contrastive Learning and Partial Least Squares regression-based method, CLPLS, to deconvolute ST data. CLPLS is a flexible method that it can be extended to integrate ST data and single-cell multi-omics data, enabling the exploration of the spatially epigenomic heterogeneity. We applied CLPLS to both simulated and real datasets coming from different platforms. Benchmark analyses with other methods on these datasets show the superior performance of CLPLS in deconvoluting spots in single cell level.
Collapse
Affiliation(s)
- Yuanyuan Mo
- School of Artificial Intelligence, School of Computer Science, Wuhan University, Wuhan 430072, China
| | - Juan Liu
- School of Artificial Intelligence, School of Computer Science, Wuhan University, Wuhan 430072, China
| | - Lihua Zhang
- School of Artificial Intelligence, School of Computer Science, Wuhan University, Wuhan 430072, China
| |
Collapse
|
33
|
Lin L, Wang H, Chen Y, Wang Y, Xu Y, Chen Z, Yang Y, Liu K, Ma X. STMGraph: spatial-context-aware of transcriptomes via a dual-remasked dynamic graph attention model. Brief Bioinform 2024; 26:bbae685. [PMID: 39764614 PMCID: PMC11704419 DOI: 10.1093/bib/bbae685] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2024] [Revised: 10/20/2024] [Accepted: 12/19/2024] [Indexed: 01/11/2025] Open
Abstract
Spatial transcriptomics (ST) technologies enable dissecting the tissue architecture in spatial context. To perceive the global contextual information of gene expression patterns in tissue, the spatial dependence of cells must be fully considered by integrating both local and non-local features by means of spatial-context-aware. However, the current ST integration algorithm ignores for ST dropouts, which impedes the spatial-aware of ST features, resulting in challenges in the accuracy and robustness of microenvironmental heterogeneity detecting, spatial domain clustering, and batch-effects correction. Here, we developed an STMGraph, a universal dual-view dynamic deep learning framework that combines dual-remask (MASK-REMASK) with dynamic graph attention model (DGAT) to exploit ST data outperforming pre-existing tools. The dual-remask mechanism masks the embeddings before encoding and decoding, establishing dual-decoding-view to share features mutually. DGAT leverages self-supervision to update graph linkage relationships from two distinct perspectives, thereby generating a comprehensive representation for each node. Systematic benchmarking against 10 state-of-the-art tools revealed that the STMGraph has the optimal performance with high accuracy and robustness on spatial domain clustering for the datasets of diverse ST platforms from multi- to sub-cellular resolutions. Furthermore, STMGraph aggregates ST information cross regions by dual-remask to realize the batch-effects correction implicitly, allowing for spatial domain clustering of ST multi-slices. STMGraph is platform independent and superior in spatial-context-aware to achieve microenvironmental heterogeneity detection, spatial domain clustering, batch-effects correction, and new biological discovery, and is therefore a desirable novel tool for diverse ST studies.
Collapse
Affiliation(s)
- Lixian Lin
- Center for Genomics and Biotechnology, Fujian Provincial Key Laboratory of Haixia Applied Plant Systems Biology, Haixia Institute of Science and Technology, Fujian Agriculture and Forestry University, No. 15 Shangxiadian Road, Cangshan District, Fuzhou 350002, China
- College of Life Science, Fujian Agriculture and Forestry University, No. 15 Shangxiadian Road, Cangshan District, Fuzhou 350002, China
| | - Haoyu Wang
- Center for Genomics and Biotechnology, Fujian Provincial Key Laboratory of Haixia Applied Plant Systems Biology, Haixia Institute of Science and Technology, Fujian Agriculture and Forestry University, No. 15 Shangxiadian Road, Cangshan District, Fuzhou 350002, China
| | - Yuxiao Chen
- Center for Genomics and Biotechnology, Fujian Provincial Key Laboratory of Haixia Applied Plant Systems Biology, Haixia Institute of Science and Technology, Fujian Agriculture and Forestry University, No. 15 Shangxiadian Road, Cangshan District, Fuzhou 350002, China
| | - Yuanyuan Wang
- Center for Genomics and Biotechnology, Fujian Provincial Key Laboratory of Haixia Applied Plant Systems Biology, Haixia Institute of Science and Technology, Fujian Agriculture and Forestry University, No. 15 Shangxiadian Road, Cangshan District, Fuzhou 350002, China
| | - Yujie Xu
- Center for Genomics and Biotechnology, Fujian Provincial Key Laboratory of Haixia Applied Plant Systems Biology, Haixia Institute of Science and Technology, Fujian Agriculture and Forestry University, No. 15 Shangxiadian Road, Cangshan District, Fuzhou 350002, China
| | - Zhenglin Chen
- Center for Genomics and Biotechnology, Fujian Provincial Key Laboratory of Haixia Applied Plant Systems Biology, Haixia Institute of Science and Technology, Fujian Agriculture and Forestry University, No. 15 Shangxiadian Road, Cangshan District, Fuzhou 350002, China
| | - Yuemin Yang
- Center for Genomics and Biotechnology, Fujian Provincial Key Laboratory of Haixia Applied Plant Systems Biology, Haixia Institute of Science and Technology, Fujian Agriculture and Forestry University, No. 15 Shangxiadian Road, Cangshan District, Fuzhou 350002, China
- College of Life Science, Fujian Agriculture and Forestry University, No. 15 Shangxiadian Road, Cangshan District, Fuzhou 350002, China
| | - Kunpeng Liu
- Center for Genomics and Biotechnology, Fujian Provincial Key Laboratory of Haixia Applied Plant Systems Biology, Haixia Institute of Science and Technology, Fujian Agriculture and Forestry University, No. 15 Shangxiadian Road, Cangshan District, Fuzhou 350002, China
| | - Xiaokai Ma
- Center for Genomics and Biotechnology, Fujian Provincial Key Laboratory of Haixia Applied Plant Systems Biology, Haixia Institute of Science and Technology, Fujian Agriculture and Forestry University, No. 15 Shangxiadian Road, Cangshan District, Fuzhou 350002, China
- Key Laboratory of Orchid Conservation and Utilization of National Forestry and Grassland Administration, Fujian Agriculture and Forestry University, No. 15 Shangxiadian Road, Cangshan District, Fuzhou 350002, China
| |
Collapse
|
34
|
Cai J, Cheng H, Wu S, Zhong W, Yuan GC, Ma P. WEST is an ensemble method for spatial transcriptomics analysis. CELL REPORTS METHODS 2024; 4:100886. [PMID: 39515332 PMCID: PMC11705770 DOI: 10.1016/j.crmeth.2024.100886] [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: 01/02/2024] [Revised: 06/29/2024] [Accepted: 10/15/2024] [Indexed: 11/16/2024]
Abstract
Spatial transcriptomics is a groundbreaking technology, enabling simultaneous profiling of gene expression and spatial orientation within biological tissues. Yet when analyzing spatial transcriptomics data, effective integration of expression and spatial information poses considerable analytical challenges. Although many methods have been developed to address this issue, many are platform specific and lack the general applicability to analyze diverse datasets. In this article, we propose a method called the weighted ensemble method for spatial transcriptomics (WEST) that utilizes ensemble techniques to improve the performance and robustness of spatial transcriptomics data analytics. We compare the performance of WEST with six methods on both synthetic and real-world datasets. WEST represents a significant advance in detecting spatial domains, offering improved accuracy and flexibility compared to existing methods, making it a valuable tool for spatial transcriptomics data analytics.
Collapse
Affiliation(s)
- Jiazhang Cai
- Department of Statistics, University of Georgia, 310 Herty Drive, Athens, GA 30602, USA
| | - Huimin Cheng
- Department of Biostatistics, Boston University, 801 Massachusetts Avenue, Crosstown Center, Boston, MA 02118, USA
| | - Shushan Wu
- Department of Statistics, University of Georgia, 310 Herty Drive, Athens, GA 30602, USA
| | - Wenxuan Zhong
- Department of Statistics, University of Georgia, 310 Herty Drive, Athens, GA 30602, USA.
| | - Guo-Cheng Yuan
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, 1468 Madison Avenue, New York, NY 10029, USA.
| | - Ping Ma
- Department of Statistics, University of Georgia, 310 Herty Drive, Athens, GA 30602, USA.
| |
Collapse
|
35
|
Sun R, Cao W, Li S, Jiang J, Shi Y, Zhang B. scGRN-Entropy: Inferring cell differentiation trajectories using single-cell data and gene regulation network-based transfer entropy. PLoS Comput Biol 2024; 20:e1012638. [PMID: 39585902 PMCID: PMC11627384 DOI: 10.1371/journal.pcbi.1012638] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2024] [Revised: 12/09/2024] [Accepted: 11/12/2024] [Indexed: 11/27/2024] Open
Abstract
Research on cell differentiation facilitates a deeper understanding of the fundamental processes of life, elucidates the intrinsic mechanisms underlying diseases such as cancer, and advances the development of therapeutics and precision medicine. Existing methods for inferring cell differentiation trajectories from single-cell RNA sequencing (scRNA-seq) data primarily rely on static gene expression data to measure distances between cells and subsequently infer pseudotime trajectories. In this work, we introduce a novel method, scGRN-Entropy, for inferring cell differentiation trajectories and pseudotime from scRNA-seq data. Unlike existing approaches, scGRN-Entropy improves inference accuracy by incorporating dynamic changes in gene regulatory networks (GRN). In scGRN-Entropy, an undirected graph representing state transitions between cells is constructed by integrating both static relationships in gene expression space and dynamic relationships in the GRN space. The edges of the undirected graph are then refined using pseudotime inferred based on cell entropy in the GRN space. Finally, the Minimum Spanning Tree (MST) algorithm is applied to derive the cell differentiation trajectory. We validate the accuracy of scGRN-Entropy on eight different real scRNA-seq datasets, demonstrating its superior performance in inferring cell differentiation trajectories through comparative analysis with existing state-of-the-art methods.
Collapse
Affiliation(s)
- Rui Sun
- School of Mathematical & Physical Sciences, Wuhan Textile University, Wuhan, Hubei, China
- Center for Applied Mathematics and Interdisciplinary Studies, Wuhan Textile University, Wuhan, Hubei, China
| | - Wenjie Cao
- School of Mathematics, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - ShengXuan Li
- School of Mathematical & Physical Sciences, Wuhan Textile University, Wuhan, Hubei, China
- Center for Applied Mathematics and Interdisciplinary Studies, Wuhan Textile University, Wuhan, Hubei, China
| | - Jian Jiang
- School of Mathematical & Physical Sciences, Wuhan Textile University, Wuhan, Hubei, China
- Center for Applied Mathematics and Interdisciplinary Studies, Wuhan Textile University, Wuhan, Hubei, China
| | - Yazhou Shi
- School of Mathematical & Physical Sciences, Wuhan Textile University, Wuhan, Hubei, China
- Center for Applied Mathematics and Interdisciplinary Studies, Wuhan Textile University, Wuhan, Hubei, China
| | - Bengong Zhang
- School of Mathematical & Physical Sciences, Wuhan Textile University, Wuhan, Hubei, China
- Center for Applied Mathematics and Interdisciplinary Studies, Wuhan Textile University, Wuhan, Hubei, China
| |
Collapse
|
36
|
Zhang W, Zhao L, Zheng T, Fan L, Wang K, Li G. Comprehensive multi-omics integration uncovers mitochondrial gene signatures for prognosis and personalized therapy in lung adenocarcinoma. J Transl Med 2024; 22:952. [PMID: 39434164 PMCID: PMC11492473 DOI: 10.1186/s12967-024-05754-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: 04/02/2024] [Accepted: 10/08/2024] [Indexed: 10/23/2024] Open
Abstract
The therapeutic efficacy of lung adenocarcinoma (LUAD), the most prevalent histological subtype of primary lung cancer, remains inadequate, with accurate prognostic assessment posing significant challenges. This study sought to elucidate the prognostic significance of mitochondrial-related genes in LUAD through an integrative multi-omics approach, aimed at developing personalized therapeutic strategies. Utilizing transcriptomic and single-cell RNA sequencing (scRNA-seq) data, alongside clinical information from publicly available databases, we first applied dimensionality reduction and clustering techniques to the LUAD single-cell dataset, focusing on the subclassification of fibroblasts, epithelial cells, and T cells. Mitochondrial-related prognostic genes were subsequently identified using TCGA-LUAD data, and LUAD cases were stratified into distinct molecular subtypes through consensus clustering, allowing for the exploration of gene expression profiles and clinical feature distributions across subtypes. By leveraging an ensemble of machine learning algorithms, we developed an Artificial Intelligence-Derived Prognostic Signature (AIDPS) model based on mitochondrial-related genes and validated its prognostic accuracy across multiple independent datasets. The AIDPS model demonstrated robust predictive power for LUAD patient outcomes, revealing significant differences in responses to immunotherapy and chemotherapy, as well as survival outcomes between risk groups. Furthermore, we conducted comprehensive analyses of tumor mutation burden (TMB), immune microenvironment characteristics, and genome-wide association study (GWAS) data, providing additional insights into the mechanistic roles of mitochondrial-related genes in LUAD pathogenesis. This study not only offers a novel approach to improving prognostic assessments in LUAD but also establishes a strong foundation for the development of personalized therapeutic interventions.
Collapse
Affiliation(s)
- Wenjia Zhang
- Department of Respiratory Medicine, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, 200072, China
| | - Lei Zhao
- Shanghai YangZhi Rehabilitation Hospital(Shanghai Sunshine Rehabilitation Center), School of Medicine, Tongji University, Shanghai, China
| | - Tiansheng Zheng
- Department of Respiratory Medicine, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, 200072, China
| | - Lihong Fan
- Department of Respiratory Medicine, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, 200072, China.
| | - Kai Wang
- Shanghai YangZhi Rehabilitation Hospital(Shanghai Sunshine Rehabilitation Center), School of Medicine, Tongji University, Shanghai, China.
| | - Guoshu Li
- Shanghai YangZhi Rehabilitation Hospital(Shanghai Sunshine Rehabilitation Center), School of Medicine, Tongji University, Shanghai, China.
| |
Collapse
|
37
|
Wang T, Zhu H, Zhou Y, Ding W, Ding W, Han L, Zhang X. Graph attention automatic encoder based on contrastive learning for domain recognition of spatial transcriptomics. Commun Biol 2024; 7:1351. [PMID: 39424696 PMCID: PMC11489439 DOI: 10.1038/s42003-024-07037-0] [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: 01/06/2024] [Accepted: 10/09/2024] [Indexed: 10/21/2024] Open
Abstract
Spatial transcriptomics is an emerging technology that enables the profiling of gene expression in tissues while preserving spatial location information. This innovative approach is anticipated to provide a comprehensive understanding of the spatial distribution of different cells within tissues and facilitate in-depth analysis of tissue structure. To accurately recognize spatial domains from spatial transcriptomics, we have introduced a generalized deep learning method called GAAEST (Graph Attention-based Autoencoder for Spatial Transcriptomics). Our proposed approach effectively integrates both spatial location information and gene expression data from spatial transcriptomics. Specifically, it leverages spatial location details to construct a neighborhood graph and employs a graph attention network-based encoder to embed gene expression information into a spatially informed space. At the same time, to further optimize the learned potential embedding, self-supervised contrastive learning is introduced to capture spatial information at three levels: local, global and contextual feature of spots. Finally, the decoder reconstructs gene expressions, which are then clustered to identify spatial domains with similar expression patterns and spatial proximity. Based on our experiments conducted on multiple datasets, GAAEST consistently outperforms existing state-of-the-art methods. The proposed GAAEST demonstrates excellent capabilities in spatial domain recognition, positioning it as an ideal tool for advancing spatial transcriptomics research.
Collapse
Affiliation(s)
- Tianqi Wang
- School of Information Science and Engineering, East China University of Science and Technology, Shanghai, China
| | - Huitong Zhu
- School of Information Science and Engineering, East China University of Science and Technology, Shanghai, China
| | - Yunlan Zhou
- Department of Clinical Laboratory, Xinhua Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Weihong Ding
- Huashan Hospital Affiliated to Fudan University, Shanghai, China
| | - Weichao Ding
- School of Information Science and Engineering, East China University of Science and Technology, Shanghai, China.
| | - Liangxiu Han
- School of Computing, Mathematics and Digital Technology, Manchester Metropolitan University, Manchester, UK
| | - Xueqin Zhang
- School of Information Science and Engineering, East China University of Science and Technology, Shanghai, China.
- Shanghai Key Laboratory of Computer Software Evaluating and Testing, Shanghai, China.
| |
Collapse
|
38
|
Liang X, Liu P, Xue L, Chen B, Liu W, Shi W, Wang Y, Chen X, Luo J. A multi-modality and multi-granularity collaborative learning framework for identifying spatial domains and spatially variable genes. Bioinformatics 2024; 40:btae607. [PMID: 39418177 PMCID: PMC11513014 DOI: 10.1093/bioinformatics/btae607] [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: 08/05/2024] [Revised: 09/19/2024] [Accepted: 10/16/2024] [Indexed: 10/19/2024] Open
Abstract
MOTIVATION Recent advances in spatial transcriptomics technologies have provided multi-modality data integrating gene expression, spatial context, and histological images. Accurately identifying spatial domains and spatially variable genes is crucial for understanding tissue structures and biological functions. However, effectively combining multi-modality data to identify spatial domains and determining SVGs closely related to these spatial domains remains a challenge. RESULTS In this study, we propose spatial transcriptomics multi-modality and multi-granularity collaborative learning (spaMMCL). For detecting spatial domains, spaMMCL mitigates the adverse effects of modality bias by masking portions of gene expression data, integrates gene and image features using a shared graph convolutional network, and employs graph self-supervised learning to deal with noise from feature fusion. Simultaneously, based on the identified spatial domains, spaMMCL integrates various strategies to detect potential SVGs at different granularities, enhancing their reliability and biological significance. Experimental results demonstrate that spaMMCL substantially improves the identification of spatial domains and SVGs. AVAILABILITY AND IMPLEMENTATION The code and data of spaMMCL are available on Github: Https://github.com/liangxiao-cs/spaMMCL.
Collapse
Affiliation(s)
- Xiao Liang
- College of Computer Science and Electronic Engineering, Hunan University, Changsha 410082, China
| | - Pei Liu
- College of Computer Science and Electronic Engineering, Hunan University, Changsha 410082, China
| | - Li Xue
- College of Computer Science and Electronic Engineering, Hunan University, Changsha 410082, China
| | - Baiyun Chen
- Computer Science, Tuskegee University, State of Alabama 36088, United States
| | - Wei Liu
- College of Computer Science and Electronic Engineering, Hunan University, Changsha 410082, China
| | - Wanwan Shi
- College of Computer Science and Electronic Engineering, Hunan University, Changsha 410082, China
| | - Yongwang Wang
- College of Computer Science and Electronic Engineering, Hunan University, Changsha 410082, China
| | - Xiangtao Chen
- College of Computer Science and Electronic Engineering, Hunan University, Changsha 410082, China
| | - Jiawei Luo
- College of Computer Science and Electronic Engineering, Hunan University, Changsha 410082, China
| |
Collapse
|
39
|
Liu L, Chen A, Li Y, Mulder J, Heyn H, Xu X. Spatiotemporal omics for biology and medicine. Cell 2024; 187:4488-4519. [PMID: 39178830 DOI: 10.1016/j.cell.2024.07.040] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2024] [Revised: 07/05/2024] [Accepted: 07/23/2024] [Indexed: 08/26/2024]
Abstract
The completion of the Human Genome Project has provided a foundational blueprint for understanding human life. Nonetheless, understanding the intricate mechanisms through which our genetic blueprint is involved in disease or orchestrates development across temporal and spatial dimensions remains a profound scientific challenge. Recent breakthroughs in cellular omics technologies have paved new pathways for understanding the regulation of genomic elements and the relationship between gene expression, cellular functions, and cell fate determination. The advent of spatial omics technologies, encompassing both imaging and sequencing-based methodologies, has enabled a comprehensive understanding of biological processes from a cellular ecosystem perspective. This review offers an updated overview of how spatial omics has advanced our understanding of the translation of genetic information into cellular heterogeneity and tissue structural organization and their dynamic changes over time. It emphasizes the discovery of various biological phenomena, related to organ functionality, embryogenesis, species evolution, and the pathogenesis of diseases.
Collapse
Affiliation(s)
| | - Ao Chen
- BGI Research, Shenzhen 518083, China
| | | | - Jan Mulder
- Department of Neuroscience, Karolinska Institute, Stockholm, Sweden
| | - Holger Heyn
- Centro Nacional de Análisis Genómico (CNAG), Barcelona, Spain
| | - Xun Xu
- BGI Research, Hangzhou 310030, China; BGI Research, Shenzhen 518083, China.
| |
Collapse
|
40
|
Hu Y, Xie M, Li Y, Rao M, Shen W, Luo C, Qin H, Baek J, Zhou XM. Benchmarking clustering, alignment, and integration methods for spatial transcriptomics. Genome Biol 2024; 25:212. [PMID: 39123269 PMCID: PMC11312151 DOI: 10.1186/s13059-024-03361-0] [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: 03/12/2024] [Accepted: 07/30/2024] [Indexed: 08/12/2024] Open
Abstract
BACKGROUND Spatial transcriptomics (ST) is advancing our understanding of complex tissues and organisms. However, building a robust clustering algorithm to define spatially coherent regions in a single tissue slice and aligning or integrating multiple tissue slices originating from diverse sources for essential downstream analyses remains challenging. Numerous clustering, alignment, and integration methods have been specifically designed for ST data by leveraging its spatial information. The absence of comprehensive benchmark studies complicates the selection of methods and future method development. RESULTS In this study, we systematically benchmark a variety of state-of-the-art algorithms with a wide range of real and simulated datasets of varying sizes, technologies, species, and complexity. We analyze the strengths and weaknesses of each method using diverse quantitative and qualitative metrics and analyses, including eight metrics for spatial clustering accuracy and contiguity, uniform manifold approximation and projection visualization, layer-wise and spot-to-spot alignment accuracy, and 3D reconstruction, which are designed to assess method performance as well as data quality. The code used for evaluation is available on our GitHub. Additionally, we provide online notebook tutorials and documentation to facilitate the reproduction of all benchmarking results and to support the study of new methods and new datasets. CONCLUSIONS Our analyses lead to comprehensive recommendations that cover multiple aspects, helping users to select optimal tools for their specific needs and guide future method development.
Collapse
Affiliation(s)
- Yunfei Hu
- Department of Computer Science, Vanderbilt University, 37235, Nashville, USA
| | - Manfei Xie
- Department of Biomedical Engineering, Vanderbilt University, 37235, Nashville, USA
| | - Yikang Li
- Department of Biomedical Engineering, Vanderbilt University, 37235, Nashville, USA
| | - Mingxing Rao
- Department of Computer Science, Vanderbilt University, 37235, Nashville, USA
| | - Wenjun Shen
- Department of Bioinformatics, Shantou University Medical College, 515041, Shantou, China
| | - Can Luo
- Department of Biomedical Engineering, Vanderbilt University, 37235, Nashville, USA
| | - Haoran Qin
- Department of Computer Science, Vanderbilt University, 37235, Nashville, USA
| | - Jihoon Baek
- Department of Computer Science, Vanderbilt University, 37235, Nashville, USA
| | - Xin Maizie Zhou
- Department of Computer Science, Vanderbilt University, 37235, Nashville, USA.
- Department of Biomedical Engineering, Vanderbilt University, 37235, Nashville, USA.
| |
Collapse
|
41
|
Chang Z, Xu Y, Dong X, Gao Y, Wang C. Single-cell and spatial multiomic inference of gene regulatory networks using SCRIPro. Bioinformatics 2024; 40:btae466. [PMID: 39024032 PMCID: PMC11288411 DOI: 10.1093/bioinformatics/btae466] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2024] [Revised: 06/05/2024] [Accepted: 07/17/2024] [Indexed: 07/20/2024] Open
Abstract
MOTIVATION The burgeoning generation of single-cell or spatial multiomic data allows for the characterization of gene regulation networks (GRNs) at an unprecedented resolution. However, the accurate reconstruction of GRNs from sparse and noisy single-cell or spatial multiomic data remains challenging. RESULTS Here, we present SCRIPro, a comprehensive computational framework that robustly infers GRNs for both single-cell and spatial multi-omics data. SCRIPro first improves sample coverage through a density clustering approach based on multiomic and spatial similarities. Additionally, SCRIPro scans transcriptional regulator (TR) importance by performing chromatin reconstruction and in silico deletion analyses using a comprehensive reference covering 1,292 human and 994 mouse TRs. Finally, SCRIPro combines TR-target importance scores derived from multiomic data with TR-target expression levels to ensure precise GRN reconstruction. We benchmarked SCRIPro on various datasets, including single-cell multiomic data from human B-cell lymphoma, mouse hair follicle development, Stereo-seq of mouse embryos, and Spatial-ATAC-RNA from mouse brain. SCRIPro outperforms existing motif-based methods and accurately reconstructs cell type-specific, stage-specific, and region-specific GRNs. Overall, SCRIPro emerges as a streamlined and fast method capable of reconstructing TR activities and GRNs for both single-cell and spatial multi-omic data. AVAILABILITY SCRIPro is available at https://github.com/wanglabtongji/SCRIPro. SUPPLEMENTARY INFORMATION Supplementary data are available at Bioinformatics online.
Collapse
Affiliation(s)
- Zhanhe Chang
- Key Laboratory of Spine and Spinal Cord Injury Repair and Regeneration of Ministry of Education, Department of Orthopedics, Tongji Hospital, School of Life Sciences and Technology, Tongji University, Shanghai 200092, China
- Frontier Science Center for Stem Cell Research, Tongji University, Shanghai, China
- Institute for Regenerative Medicine, Shanghai East Hospital, Shanghai Key Laboratory of Signaling and Disease Research, School of Life Sciences and Technology, Tongji University, Shanghai, China
| | - Yunfan Xu
- Key Laboratory of Spine and Spinal Cord Injury Repair and Regeneration of Ministry of Education, Department of Orthopedics, Tongji Hospital, School of Life Sciences and Technology, Tongji University, Shanghai 200092, China
- Frontier Science Center for Stem Cell Research, Tongji University, Shanghai, China
| | - Xin Dong
- Key Laboratory of Spine and Spinal Cord Injury Repair and Regeneration of Ministry of Education, Department of Orthopedics, Tongji Hospital, School of Life Sciences and Technology, Tongji University, Shanghai 200092, China
- Frontier Science Center for Stem Cell Research, Tongji University, Shanghai, China
| | - Yawei Gao
- Frontier Science Center for Stem Cell Research, Tongji University, Shanghai, China
- Institute for Regenerative Medicine, Shanghai East Hospital, Shanghai Key Laboratory of Signaling and Disease Research, School of Life Sciences and Technology, Tongji University, Shanghai, China
| | - Chenfei Wang
- Key Laboratory of Spine and Spinal Cord Injury Repair and Regeneration of Ministry of Education, Department of Orthopedics, Tongji Hospital, School of Life Sciences and Technology, Tongji University, Shanghai 200092, China
- Frontier Science Center for Stem Cell Research, Tongji University, Shanghai, China
- National Key Laboratory of Autonomous Intelligent Unmanned Systems, Tongji University, Shanghai 200120, China
- Frontier Science Center for Intelligent Autonomous Systems, Tongji University, Shanghai 200120, China
| |
Collapse
|
42
|
Zhang Y, Yu Z, Wong KC, Li X. Unraveling Spatial Domain Characterization in Spatially Resolved Transcriptomics with Robust Graph Contrastive Clustering. Bioinformatics 2024; 40:btae451. [PMID: 39012523 PMCID: PMC11272174 DOI: 10.1093/bioinformatics/btae451] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2023] [Revised: 06/12/2024] [Accepted: 07/12/2024] [Indexed: 07/17/2024] Open
Abstract
MOTIVATION Spatial transcriptomics can quantify gene expression and its spatial distribution in tissues, thus revealing molecular mechanisms of cellular interactions underlying tissue heterogeneity, tissue regeneration, and spatially localized disease mechanisms. However, existing spatial clustering methods often fail to exploit the full potential of spatial information, resulting in inaccurate identification of spatial domains. RESULTS In this paper, we develop a deep graph contrastive clustering framework, stDGCC, that accurately uncovers underlying spatial domains via explicitly modeling spatial information and gene expression profiles from spatial transcriptomics data. The stDGCC framework proposes a spatially informed graph node embedding model to preserve the topological information of spots and to learn the informative and discriminative characterization of spatial transcriptomics data through self-supervised contrastive learning. By simultaneously optimizing the contrastive learning loss, reconstruction loss, and Kullback-Leibler (KL) divergence loss, stDGCC achieves joint optimization of feature learning and topology structure preservation in an end-to-end manner. We validate the effectiveness of stDGCC on various spatial transcriptomics datasets acquired from different platforms, each with varying spatial resolutions. Our extensive experiments demonstrate the superiority of stDGCC over various state-of-the-art clustering methods in accurately identifying cellular-level biological structures. AVAILABILITY Code and data are available from https://github.com/TimE9527/stDGCC and https://figshare.com/projects/stDGCC/186525. SUPPLEMENTARY INFORMATION Supplementary data are available at Bioinformatics online.
Collapse
Affiliation(s)
- Yingxi Zhang
- School of Artificial Intelligence, Jilin University, Changchun 130012, China
| | - Zhuohan Yu
- School of Artificial Intelligence, Jilin University, Changchun 130012, China
| | - Ka-Chun Wong
- Department of Computer Science, City University of Hong Kong, Hong Kong 999077, Hong Kong SAR
| | - Xiangtao Li
- School of Artificial Intelligence, Jilin University, Changchun 130012, China
| |
Collapse
|
43
|
Li Y, Zhang J, Gao X, Zhang QC. Tissue module discovery in single-cell-resolution spatial transcriptomics data via cell-cell interaction-aware cell embedding. Cell Syst 2024; 15:578-592.e7. [PMID: 38823396 DOI: 10.1016/j.cels.2024.05.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2023] [Revised: 01/08/2024] [Accepted: 05/07/2024] [Indexed: 06/03/2024]
Abstract
Computational methods are desired for single-cell-resolution spatial transcriptomics (ST) data analysis to uncover spatial organization principles for how individual cells exert tissue-specific functions. Here, we present ST data analysis via interaction-aware cell embedding (SPACE), a deep-learning method for cell-type identification and tissue module discovery from single-cell-resolution ST data by learning a cell representation that captures its gene expression profile and interactions with its spatial neighbors. SPACE identified spatially informed cell subtypes defined by their special spatial distribution patterns and distinct proximal-interacting cell types. SPACE also automatically discovered "cell communities"-tissue modules with discernible boundaries and a uniform spatial distribution of constituent cell types. For each cell community, SPACE outputs a characteristic proximal cell-cell interaction network associated with physiological processes, which can be used to refine ligand-receptor-based intercellular signaling analyses. We envision that SPACE can be used in large-scale ST projects to understand how proximal cell-cell interactions contribute to emergent biological functions within cell communities. A record of this paper's transparent peer review process is included in the supplemental information.
Collapse
Affiliation(s)
- Yuzhe Li
- MOE Key Laboratory of Bioinformatics, Beijing Advanced Innovation Center for Structural Biology & Frontier Research Center for Biological Structure, Center for Synthetic and Systems Biology, School of Life Sciences, Tsinghua University, Beijing 100084, China; Academy for Advanced Interdisciplinary Studies, Peking University, Beijing 100871, China
| | - Jinsong Zhang
- MOE Key Laboratory of Bioinformatics, Beijing Advanced Innovation Center for Structural Biology & Frontier Research Center for Biological Structure, Center for Synthetic and Systems Biology, School of Life Sciences, Tsinghua University, Beijing 100084, China; Tsinghua-Peking Center for Life Sciences, Beijing 100084, China; Shanghai Qi Zhi Institute, Shanghai 200030, China
| | - Xin Gao
- Computer Science Program, Computer, Electrical and Mathematical Sciences and Engineering (CEMSE) Division, King Abdullah University of Science and Technology (KAUST), Thuwal 23955-6900, Kingdom of Saudi Arabia; KAUST Computational Bioscience Research Center (CBRC), King Abdullah University of Science and Technology (KAUST), Thuwal 23955-6900, Kingdom of Saudi Arabia; BioMap, Beijing 100086, China.
| | - Qiangfeng Cliff Zhang
- MOE Key Laboratory of Bioinformatics, Beijing Advanced Innovation Center for Structural Biology & Frontier Research Center for Biological Structure, Center for Synthetic and Systems Biology, School of Life Sciences, Tsinghua University, Beijing 100084, China; Tsinghua-Peking Center for Life Sciences, Beijing 100084, China.
| |
Collapse
|
44
|
Lin S, Cui Y, Zhao F, Yang Z, Song J, Yao J, Zhao Y, Qian BZ, Zhao Y, Yuan Z. Complete spatially resolved gene expression is not necessary for identifying spatial domains. CELL GENOMICS 2024; 4:100565. [PMID: 38781966 PMCID: PMC11228956 DOI: 10.1016/j.xgen.2024.100565] [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: 11/13/2023] [Revised: 02/29/2024] [Accepted: 04/30/2024] [Indexed: 05/25/2024]
Abstract
Spatially resolved transcriptomics (SRT) technologies have revolutionized the study of tissue organization. We introduce a graph convolutional network with an attention and positive emphasis mechanism, termed BINARY, relying exclusively on binarized SRT data to accurately delineate spatial domains. BINARY outperforms existing methods across various SRT data types while using significantly less input information. Our study suggests that precise gene expression quantification may not always be essential, inspiring further exploration of the broader applications of spatially resolved binarized gene expression data.
Collapse
Affiliation(s)
- Senlin Lin
- Institute of Computing Technology, Chinese Academy of Sciences, Beijing, China; University of Chinese Academy of Sciences, Beijing, China
| | - Yan Cui
- 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; Center for Medical Research and Innovation, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Fudan University, Shanghai, China
| | - Fangyuan Zhao
- Institute of Computing Technology, Chinese Academy of Sciences, Beijing, China; University of Chinese Academy of Sciences, Beijing, China
| | - Zhidong Yang
- Institute of Computing Technology, Chinese Academy of Sciences, Beijing, China; University of Chinese Academy of Sciences, Beijing, China
| | - Jiangning Song
- Biomedicine Discovery Institute and Department of Biochemistry and Molecular Biology, Monash University, Clayton, VIC 3800, Australia
| | | | - Yu Zhao
- AI Lab, Tencent, Shenzhen, China
| | - Bin-Zhi Qian
- Fudan University Shanghai Cancer Center, Department of Oncology, Shanghai Medical College, The Human Phenome Institute, Zhangjiang-Fudan International Innovation Center, Fudan University, Shanghai, China
| | - Yi Zhao
- Institute of Computing Technology, Chinese Academy of Sciences, Beijing, China; University of Chinese Academy of Sciences, 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; Center for Medical Research and Innovation, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Fudan University, Shanghai, China.
| |
Collapse
|
45
|
Yu Y, He Y, Xie Z. Accurate Identification of Spatial Domain by Incorporating Global Spatial Proximity and Local Expression Proximity. Biomolecules 2024; 14:674. [PMID: 38927077 PMCID: PMC11201407 DOI: 10.3390/biom14060674] [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: 05/03/2024] [Revised: 06/01/2024] [Accepted: 06/07/2024] [Indexed: 06/28/2024] Open
Abstract
Accurate identification of spatial domains is essential in the analysis of spatial transcriptomics data in order to elucidate tissue microenvironments and biological functions. However, existing methods only perform domain segmentation based on local or global spatial relationships between spots, resulting in an underutilization of spatial information. To this end, we propose SECE, a deep learning-based method that captures both local and global relationships among spots and aggregates their information using expression similarity and spatial similarity. We benchmarked SECE against eight state-of-the-art methods on six real spatial transcriptomics datasets spanning four different platforms. SECE consistently outperformed other methods in spatial domain identification accuracy. Moreover, SECE produced spatial embeddings that exhibited clearer patterns in low-dimensional visualizations and facilitated a more accurate trajectory inference.
Collapse
Affiliation(s)
- Yuanyuan Yu
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou 510060, China;
| | - Yao He
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou 510060, China;
| | - Zhi Xie
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou 510060, China;
- Center for Precision Medicine, Sun Yat-sen University, Guangzhou 510080, China
| |
Collapse
|
46
|
Dao L, You Z, Lu L, Xu T, Sarkar AK, Zhu H, Liu M, Calandrelli R, Yoshida G, Lin P, Miao Y, Mierke S, Kalva S, Zhu H, Gu M, Vadivelu S, Zhong S, Huang LF, Guo Z. Modeling blood-brain barrier formation and cerebral cavernous malformations in human PSC-derived organoids. Cell Stem Cell 2024; 31:818-833.e11. [PMID: 38754427 PMCID: PMC11162335 DOI: 10.1016/j.stem.2024.04.019] [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: 06/30/2023] [Revised: 02/24/2024] [Accepted: 04/22/2024] [Indexed: 05/18/2024]
Abstract
The human blood-brain barrier (hBBB) is a highly specialized structure that regulates passage across blood and central nervous system (CNS) compartments. Despite its critical physiological role, there are no reliable in vitro models that can mimic hBBB development and function. Here, we constructed hBBB assembloids from brain and blood vessel organoids derived from human pluripotent stem cells. We validated the acquisition of blood-brain barrier (BBB)-specific molecular, cellular, transcriptomic, and functional characteristics and uncovered an extensive neuro-vascular crosstalk with a spatial pattern within hBBB assembloids. When we used patient-derived hBBB assembloids to model cerebral cavernous malformations (CCMs), we found that these assembloids recapitulated the cavernoma anatomy and BBB breakdown observed in patients. Upon comparison of phenotypes and transcriptome between patient-derived hBBB assembloids and primary human cavernoma tissues, we uncovered CCM-related molecular and cellular alterations. Taken together, we report hBBB assembloids that mimic the core properties of the hBBB and identify a potentially underlying cause of CCMs.
Collapse
Affiliation(s)
- Lan Dao
- Center for Stem Cell and Organoid Medicine, Division of Developmental Biology, Department of Pediatrics, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA
| | - Zhen You
- Department of Pediatric and Adolescent Medicine, Department of Biochemistry and Molecular Biology, Mayo Clinic College of Medicine and Science, Rochester, MN 55905, USA
| | - Lu Lu
- Center for Stem Cell and Organoid Medicine, Division of Developmental Biology, Department of Pediatrics, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA
| | - Tianyang Xu
- Shu Chien-Gene Lay Department of Bioengineering, University of California, San Diego, La Jolla, CA 92093, USA
| | - Avijite Kumer Sarkar
- Center for Stem Cell and Organoid Medicine, Division of Developmental Biology, Department of Pediatrics, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA
| | - Hui Zhu
- Center for Stem Cell and Organoid Medicine, Division of Developmental Biology, Department of Pediatrics, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA
| | - Miao Liu
- Department of Pediatric and Adolescent Medicine, Department of Biochemistry and Molecular Biology, Mayo Clinic College of Medicine and Science, Rochester, MN 55905, USA
| | - Riccardo Calandrelli
- Shu Chien-Gene Lay Department of Bioengineering, University of California, San Diego, La Jolla, CA 92093, USA
| | - George Yoshida
- Center for Stem Cell and Organoid Medicine, Division of Developmental Biology, Department of Pediatrics, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA
| | - Pei Lin
- Shu Chien-Gene Lay Department of Bioengineering, University of California, San Diego, La Jolla, CA 92093, USA
| | - Yifei Miao
- Center for Stem Cell and Organoid Medicine, Division of Pulmonary Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA
| | - Sarah Mierke
- Divisions of Pediatric Neurosurgery and Interventional Neuroradiology, Cincinnati Children's Hospital, Cincinnati, OH 45229, USA
| | - Srijan Kalva
- Center for Stem Cell and Organoid Medicine, Division of Developmental Biology, Department of Pediatrics, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA
| | - Haining Zhu
- Department of Pharmacology and Toxicology, R. Ken Coit College of Pharmacy, University of Arizona, Tucson, AZ 85721, USA
| | - Mingxia Gu
- Center for Stem Cell and Organoid Medicine, Division of Pulmonary Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA
| | - Sudhakar Vadivelu
- Divisions of Pediatric Neurosurgery and Interventional Neuroradiology, Cincinnati Children's Hospital, Cincinnati, OH 45229, USA
| | - Sheng Zhong
- Shu Chien-Gene Lay Department of Bioengineering, University of California, San Diego, La Jolla, CA 92093, USA.
| | - L Frank Huang
- Department of Pediatric and Adolescent Medicine, Department of Biochemistry and Molecular Biology, Mayo Clinic College of Medicine and Science, Rochester, MN 55905, USA.
| | - Ziyuan Guo
- Center for Stem Cell and Organoid Medicine, Division of Developmental Biology, Department of Pediatrics, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA.
| |
Collapse
|
47
|
Ma Y, Liu L, Zhao Y, Hang B, Zhang Y. HyperGCN: an effective deep representation learning framework for the integrative analysis of spatial transcriptomics data. BMC Genomics 2024; 25:566. [PMID: 38840049 PMCID: PMC11155133 DOI: 10.1186/s12864-024-10469-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: 02/07/2024] [Accepted: 05/29/2024] [Indexed: 06/07/2024] Open
Abstract
BACKGROUND Advances of spatial transcriptomics technologies enabled simultaneously profiling gene expression and spatial locations of cells from the same tissue. Computational tools and approaches for integration of transcriptomics data and spatial context information are urgently needed to comprehensively explore the underlying structure patterns. In this manuscript, we propose HyperGCN for the integrative analysis of gene expression and spatial information profiled from the same tissue. HyperGCN enables data visualization and clustering, and facilitates downstream analysis, including domain segmentation, the characterization of marker genes for the specific domain structure and GO enrichment analysis. RESULTS Extensive experiments are implemented on four real datasets from different tissues (including human dorsolateral prefrontal cortex, human positive breast tumors, mouse brain, mouse olfactory bulb tissue and Zabrafish melanoma) and technologies (including 10X visium, osmFISH, seqFISH+, 10X Xenium and Stereo-seq) with different spatial resolutions. The results show that HyperGCN achieves superior clustering performance and produces good domain segmentation effects while identifies biologically meaningful spatial expression patterns. This study provides a flexible framework to analyze spatial transcriptomics data with high geometric complexity. CONCLUSIONS HyperGCN is an unsupervised method based on hypergraph induced graph convolutional network, where it assumes that there existed disjoint tissues with high geometric complexity, and models the semantic relationship of cells through hypergraph, which better tackles the high-order interactions of cells and levels of noise in spatial transcriptomics data.
Collapse
Affiliation(s)
- Yuanyuan Ma
- School of Computer Engineering, Hubei University of Arts and Science, Xiangyang, China.
- Hubei Key Laboratory of Power System Design and Test for Electrical Vehicle, Hubei University of Arts and Science, Xiangyang, China.
| | - Lifang Liu
- School of Physics and Electronic Engineering, Hubei University of Arts and Science, Xiangyang, China
| | - Yongbiao Zhao
- School of Computer Engineering, Hubei University of Arts and Science, Xiangyang, China
- School of Computer, Central China Normal University, Wuhan, China
| | - Bo Hang
- School of Computer Engineering, Hubei University of Arts and Science, Xiangyang, China
| | - Yanduo Zhang
- School of Computer Engineering, Hubei University of Arts and Science, Xiangyang, China
| |
Collapse
|
48
|
Zhou P, Bocci F, Li T, Nie Q. Spatial transition tensor of single cells. Nat Methods 2024; 21:1053-1062. [PMID: 38755322 PMCID: PMC11166574 DOI: 10.1038/s41592-024-02266-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: 07/03/2023] [Accepted: 04/02/2024] [Indexed: 05/18/2024]
Abstract
Spatial transcriptomics and messenger RNA splicing encode extensive spatiotemporal information for cell states and transitions. The current lineage-inference methods either lack spatial dynamics for state transition or cannot capture different dynamics associated with multiple cell states and transition paths. Here we present spatial transition tensor (STT), a method that uses messenger RNA splicing and spatial transcriptomes through a multiscale dynamical model to characterize multistability in space. By learning a four-dimensional transition tensor and spatial-constrained random walk, STT reconstructs cell-state-specific dynamics and spatial state transitions via both short-time local tensor streamlines between cells and long-time transition paths among attractors. Benchmarking and applications of STT on several transcriptome datasets via multiple technologies on epithelial-mesenchymal transitions, blood development, spatially resolved mouse brain and chicken heart development, indicate STT's capability in recovering cell-state-specific dynamics and their associated genes not seen using existing methods. Overall, STT provides a consistent multiscale description of single-cell transcriptome data across multiple spatiotemporal scales.
Collapse
Affiliation(s)
- Peijie Zhou
- Department of Mathematics, University of California, Irvine, Irvine, CA, USA
- Center for Machine Learning Research, Peking University, Beijing, China
- AI for Science Institute, Beijing, China
- National Engineering Laboratory for Big Data Analysis and Applications, Beijing, China
| | - Federico Bocci
- Department of Mathematics, University of California, Irvine, Irvine, CA, USA
| | - Tiejun Li
- LMAM and School of Mathematical Sciences, Peking University, Beijing, China
| | - Qing Nie
- Department of Mathematics, University of California, Irvine, Irvine, CA, USA.
- Department of Cell and Developmental Biology, University of California, Irvine, Irvine, CA, USA.
| |
Collapse
|
49
|
Condello V, Paulsson JO, Zedenius J, Näsman A, Juhlin CC. Spatial Transcriptomics in a Case of Follicular Thyroid Carcinoma Reveals Clone-Specific Dysregulation of Genes Regulating Extracellular Matrix in the Invading Front. Endocr Pathol 2024; 35:122-133. [PMID: 38280140 PMCID: PMC11176252 DOI: 10.1007/s12022-024-09798-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 01/12/2024] [Indexed: 01/29/2024]
Abstract
Follicular thyroid carcinoma (FTC) is recognized by its ability to invade the tumor capsule and blood vessels, although the exact molecular signals orchestrating this phenotype remain elusive. In this study, the spatial transcriptional landscape of an FTC is detailed with comparisons between the invasive front and histologically indolent central core tumor areas. The Visium spatial gene expression platform allowed us to interrogate and visualize the whole transcriptome in 2D across formalin-fixated paraffin-embedded (FFPE) tissue sections. Four different 6 × 6 mm areas of an FTC were scrutinized, including regions with capsular and vascular invasion, capsule-near area without invasion, and a central core area of the tumor. Following successful capturing and sequencing, several expressional clusters were identified with regional variation. Most notably, invasive tumor cell clusters were significantly over-expressing genes associated with pathways interacting with the extracellular matrix (ECM) remodeling and epithelial-to-mesenchymal transition (EMT). Subsets of these genes (POSTN and DPYSL3) were additionally validated using immunohistochemistry in an independent cohort of follicular thyroid tumors showing a clear gradient pattern from the core to the periphery of the tumor. Moreover, the reconstruction of the evolutionary tree identified the invasive clones as late events in follicular thyroid tumorigenesis. To our knowledge, this is one of the first 2D global transcriptional mappings of FTC using this platform to date. Invasive FTC clones develop in a stepwise fashion and display significant dysregulation of genes associated with the ECM and EMT - thus highlighting important molecular crosstalk for further investigations.
Collapse
Affiliation(s)
- Vincenzo Condello
- Department of Oncology-Pathology, Karolinska Institutet, Stockholm, Sweden.
| | - Johan O Paulsson
- Department of Oncology-Pathology, Karolinska Institutet, Stockholm, Sweden
- Department of Trauma and Emergency Surgery, Karolinska University Hospital, Stockholm, Sweden
| | - Jan Zedenius
- Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden
- Department of Breast, Endocrine Tumors, and Sarcoma, Karolinska University Hospital, Stockholm, Sweden
| | - Anders Näsman
- Department of Oncology-Pathology, Karolinska Institutet, Stockholm, Sweden
- Department of Pathology and Cancer Diagnostics, Karolinska University Hospital, Stockholm, Sweden
| | - C Christofer Juhlin
- Department of Oncology-Pathology, Karolinska Institutet, Stockholm, Sweden.
- Department of Pathology and Cancer Diagnostics, Karolinska University Hospital, Stockholm, Sweden.
| |
Collapse
|
50
|
Guo N, Vargas J, Reynoso S, Fritz D, Krishna R, Wang C, Zhang F. Uncover spatially informed variations for single-cell spatial transcriptomics with STew. BIOINFORMATICS ADVANCES 2024; 4:vbae064. [PMID: 38827413 PMCID: PMC11142628 DOI: 10.1093/bioadv/vbae064] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 01/02/2024] [Revised: 03/06/2024] [Accepted: 05/01/2024] [Indexed: 06/04/2024]
Abstract
Motivation The recent spatial transcriptomics (ST) technologies have enabled characterization of gene expression patterns and spatial information, advancing our understanding of cell lineages within diseased tissues. Several analytical approaches have been proposed for ST data, but effectively utilizing spatial information to unveil the shared variation with gene expression remains a challenge. Results We introduce STew, a Spatial Transcriptomic multi-viEW representation learning method, to jointly analyze spatial information and gene expression in a scalable manner, followed by a data-driven statistical framework to measure the goodness of model fit. Through benchmarking using human dorsolateral prefrontal cortex and mouse main olfactory bulb data with true manual annotations, STew achieved superior performance in both clustering accuracy and continuity of identified spatial domains compared with other methods. STew is also robust to generate consistent results insensitive to model parameters, including sparsity constraints. We next applied STew to various ST data acquired from 10× Visium, Slide-seqV2, and 10× Xenium, encompassing single-cell and multi-cellular resolution ST technologies, which revealed spatially informed cell type clusters and biologically meaningful axes. In particular, we identified a proinflammatory fibroblast spatial niche using ST data from psoriatic skins. Moreover, STew scales almost linearly with the number of spatial locations, guaranteeing its applicability to datasets with thousands of spatial locations to capture disease-relevant niches in complex tissues. Availability and implementation Source code and the R software tool STew are available from github.com/fanzhanglab/STew.
Collapse
Affiliation(s)
- Nanxi Guo
- Biostatistics and Informatics PhD Program, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, United States
- Department of Biomedical Informatics, Center for Health Artificial Intelligence, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, United States
| | - Juan Vargas
- Department of Biomedical Informatics, Center for Health Artificial Intelligence, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, United States
- MPH Biostatistics, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, United States
| | - Samantha Reynoso
- Department of Biomedical Informatics, Center for Health Artificial Intelligence, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, United States
- Computational Bioscience PhD Program, University of Colorado School of Medicine, Aurora, CO 80045, United States
| | - Douglas Fritz
- Department of Biomedical Informatics, Center for Health Artificial Intelligence, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, United States
- Medical Scientist Training Program, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, United States
| | - Revanth Krishna
- Department of Biomedical Informatics, Center for Health Artificial Intelligence, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, United States
- Division of Rheumatology, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, United States
| | - Chuangqi Wang
- Department of Immunology and Microbiology, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, United States
| | - Fan Zhang
- Department of Biomedical Informatics, Center for Health Artificial Intelligence, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, United States
- Division of Rheumatology, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, United States
| |
Collapse
|