1
|
Junaid M, Lee EJ, Lim SB. Single-cell and spatial omics: exploring hypothalamic heterogeneity. Neural Regen Res 2025; 20:1525-1540. [PMID: 38993130 PMCID: PMC11688568 DOI: 10.4103/nrr.nrr-d-24-00231] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2024] [Revised: 05/06/2024] [Accepted: 06/03/2024] [Indexed: 07/13/2024] Open
Abstract
Elucidating the complex dynamic cellular organization in the hypothalamus is critical for understanding its role in coordinating fundamental body functions. Over the past decade, single-cell and spatial omics technologies have significantly evolved, overcoming initial technical challenges in capturing and analyzing individual cells. These high-throughput omics technologies now offer a remarkable opportunity to comprehend the complex spatiotemporal patterns of transcriptional diversity and cell-type characteristics across the entire hypothalamus. Current single-cell and single-nucleus RNA sequencing methods comprehensively quantify gene expression by exploring distinct phenotypes across various subregions of the hypothalamus. However, single-cell/single-nucleus RNA sequencing requires isolating the cell/nuclei from the tissue, potentially resulting in the loss of spatial information concerning neuronal networks. Spatial transcriptomics methods, by bypassing the cell dissociation, can elucidate the intricate spatial organization of neural networks through their imaging and sequencing technologies. In this review, we highlight the applicative value of single-cell and spatial transcriptomics in exploring the complex molecular-genetic diversity of hypothalamic cell types, driven by recent high-throughput achievements.
Collapse
Affiliation(s)
- Muhammad Junaid
- Department of Biochemistry & Molecular Biology, Ajou University School of Medicine, Suwon, South Korea
- Department of Biomedical Sciences, Graduate School of Ajou University, Suwon, South Korea
| | - Eun Jeong Lee
- Department of Biomedical Sciences, Graduate School of Ajou University, Suwon, South Korea
- Department of Brain Science, Ajou University School of Medicine, Suwon, South Korea
| | - Su Bin Lim
- Department of Biochemistry & Molecular Biology, Ajou University School of Medicine, Suwon, South Korea
- Department of Biomedical Sciences, Graduate School of Ajou University, Suwon, South Korea
| |
Collapse
|
2
|
Zhu B, Gao S, Chen S, Wang Y, Yeung J, Bai Y, Huang AY, Yeo YY, Liao G, Mao S, Jiang ZG, Rodig SJ, Wong KC, Shalek AK, Nolan GP, Jiang S, Ma Z. CellLENS enables cross-domain information fusion for enhanced cell population delineation in single-cell spatial omics data. Nat Immunol 2025:10.1038/s41590-025-02163-1. [PMID: 40404817 DOI: 10.1038/s41590-025-02163-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2024] [Accepted: 04/14/2025] [Indexed: 05/24/2025]
Abstract
Delineating cell populations is crucial for understanding immune function in health and disease. Spatial omics technologies offer insights by capturing three complementary domains: single-cell molecular biomarker expression, cellular spatial relationships and tissue architecture. However, current computational methods often fail to fully integrate these multidimensional data, particularly for immune cell populations and intrinsic functional states. We introduce Cell Local Environment and Neighborhood Scan (CellLENS), a self-supervised computational method that learns cellular representations by fusing information across three spatial omics domains (expression, neighborhood and image). CellLENS markedly enhances de novo discovery of biologically relevant immune cell populations at fine granularity by integrating individual cells' molecular profiles with their neighborhood context and tissue localization. By applying CellLENS to diverse spatial proteomic and transcriptomic datasets across multiple tissue types and disease settings, we uncover unique immune cell populations functionally stratified according to their spatial contexts. Our work demonstrates the power of multi-domain data integration in spatial omics to reveal insights into immune cell heterogeneity and tissue-specific functions.
Collapse
Affiliation(s)
- Bokai Zhu
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Sheng Gao
- Department of Statistics and Data Science, The Wharton School, University of Pennsylvania, Philadelphia, PA, USA
| | - Shuxiao Chen
- Department of Statistics and Data Science, The Wharton School, University of Pennsylvania, Philadelphia, PA, USA
| | - Yuchen Wang
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
- Department of Computer Science, City University of Hong Kong, Hong Kong, People's Republic of China
| | - Jason Yeung
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Yunhao Bai
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Amy Y Huang
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Massachusetts Institute of Technology, Cambridge, MA, USA
- Dana-Farber Cancer Institute, Boston, MA, USA
| | - Yao Yu Yeo
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Guanrui Liao
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
- Center of Hepato-Pancreato-Biliary Surgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, People's Republic of China
| | - Shulin Mao
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
- Division of Genetics and Genomics, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
- Program in Biological and Biomedical Sciences, Harvard Medical School, Boston, MA, USA
| | - Zhenghui G Jiang
- Division of Gastroenterology/Liver Center, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Scott J Rodig
- Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Ka-Chun Wong
- Department of Computer Science, City University of Hong Kong, Hong Kong, People's Republic of China
| | - Alex K Shalek
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, MA, USA
- Department of Chemistry, Massachusetts Institute of Technology, Cambridge, MA, USA
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Garry P Nolan
- Department of Pathology, Stanford University, Stanford, CA, USA.
| | - Sizun Jiang
- Broad Institute of MIT and Harvard, Cambridge, MA, USA.
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA.
- Dana-Farber Cancer Institute, Boston, MA, USA.
- Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA.
| | - Zongming Ma
- Department of Statistics and Data Science, Yale University, New Haven, CT, USA.
| |
Collapse
|
3
|
Verpoort B, Amado L, Vandensteen J, Leysen E, Dascenco D, Vandenbempt J, Lemmens I, Wauman J, Vennekens K, Escamilla-Ayala A, Freitas ACN, Voets T, Munck S, Tavernier J, Wierda K, de Wit J. A postsynaptic GPR158-PLCXD2 complex controls spine apparatus abundance and dendritic spine maturation. Dev Cell 2025:S1534-5807(25)00263-1. [PMID: 40393451 DOI: 10.1016/j.devcel.2025.04.025] [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: 04/10/2024] [Revised: 11/08/2024] [Accepted: 04/28/2025] [Indexed: 05/22/2025]
Abstract
The spine apparatus (SA), an endoplasmic reticulum (ER)-related organelle present in a subset of dendritic spines, plays a key role in postsynaptic development and is implicated in various neurological disorders. The molecular mechanisms that dictate SA localization at selected synapses remain elusive. Here, we identify a postsynaptic signaling complex comprising the G protein-coupled receptor (GPCR)- GPR158 and a constitutively active phospholipase C (PLC) family member, PLC X-domain containing 2 (PLCXD2), that controls SA abundance. Sparse genetic manipulations of mouse cortical neurons in vivo demonstrate that, in the absence of GPR158, unrestrained PLCXD2 activity impedes postsynaptic SA incorporation and hampers structural and functional dendritic spine maturation. Extracellular heparan sulfate proteoglycan (HSPG) binding modulates the GPR158-PLCXD2 interaction, providing spatiotemporal control over GPR158 signaling. Together, our findings uncover a direct GPCR-like receptor-to-PLC signaling pathway that bypasses canonical PLC regulation via G proteins. This GPR158-PLCXD2 module regulates SA abundance, essential for proper postsynaptic structure and function.
Collapse
Affiliation(s)
- Ben Verpoort
- VIB-KU Leuven Center for Brain & Disease Research, Leuven 3000, Belgium; KU Leuven, Department of Neurosciences, Leuven Brain Institute, Leuven 3000, Belgium
| | - Luísa Amado
- VIB-KU Leuven Center for Brain & Disease Research, Leuven 3000, Belgium; KU Leuven, Department of Neurosciences, Leuven Brain Institute, Leuven 3000, Belgium
| | - Jeroen Vandensteen
- VIB-KU Leuven Center for Brain & Disease Research, Leuven 3000, Belgium; KU Leuven, Department of Neurosciences, Leuven Brain Institute, Leuven 3000, Belgium
| | - Elke Leysen
- VIB-KU Leuven Center for Brain & Disease Research, Leuven 3000, Belgium; KU Leuven, Department of Neurosciences, Leuven Brain Institute, Leuven 3000, Belgium
| | - Dan Dascenco
- VIB-KU Leuven Center for Brain & Disease Research, Leuven 3000, Belgium; KU Leuven, Department of Neurosciences, Leuven Brain Institute, Leuven 3000, Belgium
| | - Joris Vandenbempt
- VIB-KU Leuven Center for Brain & Disease Research, Leuven 3000, Belgium; KU Leuven, Department of Neurosciences, Leuven Brain Institute, Leuven 3000, Belgium
| | - Irma Lemmens
- VIB-UGent Center for Medical Biotechnology, Department of Biomolecular Medicine, Faculty of Medicine and Health Sciences, Ghent University, Ghent 9000, Belgium
| | - Joris Wauman
- VIB-UGent Center for Medical Biotechnology, Department of Biomolecular Medicine, Faculty of Medicine and Health Sciences, Ghent University, Ghent 9000, Belgium
| | - Kristel Vennekens
- VIB-KU Leuven Center for Brain & Disease Research, Leuven 3000, Belgium; KU Leuven, Department of Neurosciences, Leuven Brain Institute, Leuven 3000, Belgium
| | - Abril Escamilla-Ayala
- VIB-KU Leuven Center for Brain & Disease Research, Leuven 3000, Belgium; KU Leuven, Department of Neurosciences, Leuven Brain Institute, Leuven 3000, Belgium; VIB Bioimaging Core Leuven, Center for Brain and Disease Research, Leuven 3000, Belgium
| | - Ana Cristina Nogueira Freitas
- VIB-KU Leuven Center for Brain & Disease Research, Leuven 3000, Belgium; KU Leuven, Department of Cellular and Molecular Medicine, Leuven 3000, Belgium
| | - Thomas Voets
- VIB-KU Leuven Center for Brain & Disease Research, Leuven 3000, Belgium; KU Leuven, Department of Cellular and Molecular Medicine, Leuven 3000, Belgium
| | - Sebastian Munck
- VIB-KU Leuven Center for Brain & Disease Research, Leuven 3000, Belgium; KU Leuven, Department of Neurosciences, Leuven Brain Institute, Leuven 3000, Belgium; VIB Bioimaging Core Leuven, Center for Brain and Disease Research, Leuven 3000, Belgium
| | - Jan Tavernier
- VIB-UGent Center for Medical Biotechnology, Department of Biomolecular Medicine, Faculty of Medicine and Health Sciences, Ghent University, Ghent 9000, Belgium
| | - Keimpe Wierda
- VIB-KU Leuven Center for Brain & Disease Research, Leuven 3000, Belgium; KU Leuven, Department of Neurosciences, Leuven Brain Institute, Leuven 3000, Belgium; VIB-KU Leuven Center for Brain & Disease Research Technologies, Electrophysiology Unit, Leuven 3000, Belgium
| | - Joris de Wit
- VIB-KU Leuven Center for Brain & Disease Research, Leuven 3000, Belgium; KU Leuven, Department of Neurosciences, Leuven Brain Institute, Leuven 3000, Belgium.
| |
Collapse
|
4
|
Wang H, Cheng P, Wang J, Lv H, Han J, Hou Z, Xu R, Chen W. Advances in spatial transcriptomics and its application in the musculoskeletal system. Bone Res 2025; 13:54. [PMID: 40379648 DOI: 10.1038/s41413-025-00429-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2024] [Revised: 03/10/2025] [Accepted: 03/17/2025] [Indexed: 05/19/2025] Open
Abstract
While bulk RNA sequencing and single-cell RNA sequencing have shed light on cellular heterogeneity and potential molecular mechanisms in the musculoskeletal system in both physiological and various pathological states, the spatial localization of cells and molecules and intercellular interactions within the tissue context require further elucidation. Spatial transcriptomics has revolutionized biological research by simultaneously capturing gene expression profiles and in situ spatial information of tissues, gradually finding applications in musculoskeletal research. This review provides a summary of recent advances in spatial transcriptomics and its application to the musculoskeletal system. The classification and characteristics of data acquisition techniques in spatial transcriptomics are briefly outlined, with an emphasis on widely-adopted representative technologies and the latest technological breakthroughs, accompanied by a concise workflow for incorporating spatial transcriptomics into musculoskeletal system research. The role of spatial transcriptomics in revealing physiological mechanisms of the musculoskeletal system, particularly during developmental processes, is thoroughly summarized. Furthermore, recent discoveries and achievements of this emerging omics tool in addressing inflammatory, traumatic, degenerative, and tumorous diseases of the musculoskeletal system are compiled. Finally, challenges and potential future directions for spatial transcriptomics, both as a field and in its applications in the musculoskeletal system, are discussed.
Collapse
Affiliation(s)
- Haoyu Wang
- Department of Orthopedic Surgery, Hebei Medical University Third Hospital, Shijiazhuang, Hebei, China
- Key Laboratory of Biomechanics of Hebei Province, Shijiazhuang, Hebei, China
- NHC Key Laboratory of Intelligent Orthopedic Equipment, Shijiazhuang, Hebei, China
| | - Peng Cheng
- Department of Orthopedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Juan Wang
- Department of Orthopedic Surgery, Hebei Medical University Third Hospital, Shijiazhuang, Hebei, China
- Key Laboratory of Biomechanics of Hebei Province, Shijiazhuang, Hebei, China
- NHC Key Laboratory of Intelligent Orthopedic Equipment, Shijiazhuang, Hebei, China
| | - Hongzhi Lv
- Department of Orthopedic Surgery, Hebei Medical University Third Hospital, Shijiazhuang, Hebei, China
- Key Laboratory of Biomechanics of Hebei Province, Shijiazhuang, Hebei, China
- NHC Key Laboratory of Intelligent Orthopedic Equipment, Shijiazhuang, Hebei, China
| | - Jie Han
- State Key Laboratory of Cellular Stress Biology, Cancer Research Center, School of Medicine, Faculty of Medicine and Life Sciences, Xiamen University, Xiamen, Fujian, China
| | - Zhiyong Hou
- Department of Orthopedic Surgery, Hebei Medical University Third Hospital, Shijiazhuang, Hebei, China
- Key Laboratory of Biomechanics of Hebei Province, Shijiazhuang, Hebei, China
- NHC Key Laboratory of Intelligent Orthopedic Equipment, Shijiazhuang, Hebei, China
| | - Ren Xu
- The First Affiliated Hospital of Xiamen University-ICMRS Collaborating Center for Skeletal Stem Cells, State Key Laboratory of Cellular Stress Biology, School of Medicine, Faculty of Medicine and Life Sciences, Xiamen University, Xiamen, Fujian, China.
| | - Wei Chen
- Department of Orthopedic Surgery, Hebei Medical University Third Hospital, Shijiazhuang, Hebei, China.
- Key Laboratory of Biomechanics of Hebei Province, Shijiazhuang, Hebei, China.
- NHC Key Laboratory of Intelligent Orthopedic Equipment, Shijiazhuang, Hebei, China.
| |
Collapse
|
5
|
Tavakoli MR, Lyudchik J, Januszewski M, Vistunou V, Agudelo Dueñas N, Vorlaufer J, Sommer C, Kreuzinger C, Oliveira B, Cenameri A, Novarino G, Jain V, Danzl JG. Light-microscopy-based connectomic reconstruction of mammalian brain tissue. Nature 2025:10.1038/s41586-025-08985-1. [PMID: 40335689 DOI: 10.1038/s41586-025-08985-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2024] [Accepted: 04/04/2025] [Indexed: 05/09/2025]
Abstract
The information-processing capability of the brain's cellular network depends on the physical wiring pattern between neurons and their molecular and functional characteristics. Mapping neurons and resolving their individual synaptic connections can be achieved by volumetric imaging at nanoscale resolution1,2 with dense cellular labelling. Light microscopy is uniquely positioned to visualize specific molecules, but dense, synapse-level circuit reconstruction by light microscopy has been out of reach, owing to limitations in resolution, contrast and volumetric imaging capability. Here we describe light-microscopy-based connectomics (LICONN). We integrated specifically engineered hydrogel embedding and expansion with comprehensive deep-learning-based segmentation and analysis of connectivity, thereby directly incorporating molecular information into synapse-level reconstructions of brain tissue. LICONN will allow synapse-level phenotyping of brain tissue in biological experiments in a readily adoptable manner.
Collapse
Affiliation(s)
| | - Julia Lyudchik
- Institute of Science and Technology Austria, Klosterneuburg, Austria
| | | | - Vitali Vistunou
- Institute of Science and Technology Austria, Klosterneuburg, Austria
| | | | - Jakob Vorlaufer
- Institute of Science and Technology Austria, Klosterneuburg, Austria
| | - Christoph Sommer
- Institute of Science and Technology Austria, Klosterneuburg, Austria
| | | | - Bárbara Oliveira
- Institute of Science and Technology Austria, Klosterneuburg, Austria
| | - Alban Cenameri
- Institute of Science and Technology Austria, Klosterneuburg, Austria
| | - Gaia Novarino
- Institute of Science and Technology Austria, Klosterneuburg, Austria
| | | | - Johann G Danzl
- Institute of Science and Technology Austria, Klosterneuburg, Austria.
| |
Collapse
|
6
|
Zhang W, Zhang Z, Yang H, Zhang T, Jiang S, Qiao N, Deng Z, Pan X, Shen HB, Yu DJ, Wang S. m2ST: dual multi-scale graph clustering for spatially resolved transcriptomics. Bioinformatics 2025; 41:btaf221. [PMID: 40272889 PMCID: PMC12085222 DOI: 10.1093/bioinformatics/btaf221] [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: 01/21/2025] [Revised: 03/20/2025] [Accepted: 04/22/2025] [Indexed: 04/26/2025] Open
Abstract
MOTIVATION Spatial clustering is a key analytical technique for exploring spatial transcriptomics data. Recent graph neural network-based methods have shown promise in spatial clustering but face notable challenges. One significant issue is that analyzing the functions and complex mechanisms of organisms from a single scale is difficult and most methods focus exclusively on the single-scale representation of transcriptomic data, potentially limiting the discriminative power of extracted features for spatial domain clustering. Furthermore, classical clustering algorithms are often applied directly to latent representation, making it a worthwhile endeavor to explore a tailored clustering method to further improve the accuracy of spatial domain annotation. RESULTS To address these limitations, we propose m2ST, a novel dual multi-scale graph clustering method. m2ST first uses a multi-scale masked graph autoencoder to extract representations across different scales from spatial transcriptomic data. To effectively compress and distill meaningful knowledge embedded in the data, m2ST introduces a random masking mechanism for node features and uses a scaled cosine error as the loss function. Additionally, we introduce a tailored multi-scale clustering framework that integrates scale-common and scale-specific information exploration into the clustering process, achieving more robust annotation performance. Shannon entropy is finally utilized to dynamically adjust the importance of different scales. Extensive experiments on multiple spatial transcriptomic datasets demonstrate the superior performance of m2ST compared to existing methods. AVAILABILITY AND IMPLEMENTATION https://github.com/BBKing49/m2ST.
Collapse
Affiliation(s)
- Wei Zhang
- The School of Artificial Intelligence and Computer Science, Nantong University, Nantong, 226019, China
- The School of Artificial Intelligence and Computer Science, Jiangnan University, Wuxi, 214122, China
| | - Ziqi Zhang
- The School of Artificial Intelligence and Computer Science, Jiangnan University, Wuxi, 214122, China
| | - Hailong Yang
- The School of Artificial Intelligence and Computer Science, Jiangnan University, Wuxi, 214122, China
| | - Te Zhang
- The Lab for Uncertainty in Data and Decision Making (LUCID), School of Computer Science, University of Nottingham, Nottingham, NG81BB, United Kingdom
| | - Shu Jiang
- The School of Artificial Intelligence and Computer Science, Nantong University, Nantong, 226019, China
| | - Ning Qiao
- The School of Artificial Intelligence and Computer Science, Jiangnan University, Wuxi, 214122, China
| | - Zhaohong Deng
- The School of Artificial Intelligence and Computer Science, Jiangnan University, Wuxi, 214122, China
| | - Xiaoyong Pan
- Department of Automation, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Hong-Bin Shen
- Department of Automation, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Dong-Jun Yu
- School of Computer Science and Engineering, Nanjing University of Science and Technology, Nanjing, 210094, China
| | - Shitong Wang
- The School of Artificial Intelligence and Computer Science, Jiangnan University, Wuxi, 214122, China
| |
Collapse
|
7
|
Jia BB, Sun BK, Lee EY, Ren B. Emerging Techniques in Spatial Multiomics: Fundamental Principles and Applications to Dermatology. J Invest Dermatol 2025; 145:1017-1032. [PMID: 39503694 DOI: 10.1016/j.jid.2024.09.006] [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: 02/04/2024] [Revised: 09/09/2024] [Accepted: 09/09/2024] [Indexed: 04/25/2025]
Abstract
Molecular pathology, such as high-throughput genomic and proteomic profiling, identifies precise disease targets from biopsies but require tissue dissociation, losing valuable histologic and spatial context. Emerging spatial multi-omic technologies now enable multiplexed visualization of genomic, proteomic, and epigenomic targets within a single tissue slice, eliminating the need for labeling multiple adjacent slices. Although early work focused on RNA (spatial transcriptomics), spatial technologies can now concurrently capture DNA, genome accessibility, histone modifications, and proteins with spatially-resolved single-cell resolution. This review outlines the principles, advantages, limitations, and potential for spatial technologies to advance dermatologic research. By jointly profiling multiple molecular channels, spatial multiomics enables novel studies of copy number variations, clonal heterogeneity, and enhancer dysregulation, replete with spatial context, illuminating the skin's complex heterogeneity.
Collapse
Affiliation(s)
- Bojing B Jia
- Bioinformatics and Systems Biology Graduate Program, University of California, San Diego, La Jolla, California, USA; Medical Scientist Training Program, University of California, San Diego, La Jolla, California, USA
| | - Bryan K Sun
- Department of Dermatology, University of California, Irvine, Irvine, California, USA
| | - Ernest Y Lee
- Department of Dermatology, University of California, San Francisco, San Francisco, California, USA
| | - Bing Ren
- Center for Epigenomics, Department of Cellular & Molecular Medicine, University of California, San Diego, La Jolla, California, USA; Institute of Genomic Medicine, Moores Cancer Center, School of Medicine, University of California, San Diego, La Jolla, California, USA.
| |
Collapse
|
8
|
Xu Y, Yu B, Chen X, Peng A, Tao Q, He Y, Wang Y, Li XM. DSCT: a novel deep-learning framework for rapid and accurate spatial transcriptomic cell typing. Natl Sci Rev 2025; 12:nwaf030. [PMID: 40313458 PMCID: PMC12045154 DOI: 10.1093/nsr/nwaf030] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2024] [Revised: 01/05/2025] [Accepted: 01/09/2025] [Indexed: 05/03/2025] Open
Abstract
Unraveling complex cell-type-composition and gene-expression patterns at the cellular spatial resolution is crucial for understanding intricate cell functions in the brain. In this study, we developed Deep Neural Network-based Spatial Cell Typing (DSCT)-an innovative framework for spatial cell typing within spatial transcriptomic data sets. This approach utilizes a synergistic integration of an enhanced gene-selection strategy and a lightweight deep neural network for data training, offering a more rapid and accurate solution for the analysis of spatial transcriptomic data. Based on comprehensive analysis, DSCT achieved exceptional accuracy in cell-type identification across various brain regions, species and spatial transcriptomic platforms. It also performed well in mapping finer cell types, thereby showcasing its versatility and adaptability across diverse data sets. Strikingly, DSCT exhibited high efficiency and remarkable processing speed, with fewer computational resource demands. As such, this novel approach opens new avenues for exploring the spatial organization of cell types and gene-expression patterns, advancing our understanding of biological functions and pathologies within the nervous system.
Collapse
Affiliation(s)
- Yiheng Xu
- Department of Neurology and Department of Psychiatry, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310058, China
- NHC and CAMS Key Laboratory of Medical Neurobiology, MOE Frontier Center of Brain Science and Brain-machine Integration, School of Brain Science and Brain Medicine, Zhejiang University, Hangzhou 310058, China
- College of Computer Science and Technology, Zhejiang University, Hangzhou 310000, China
| | - Bin Yu
- Key Laboratory of Novel Targets and Drug Study for Neural Repair of Zhejiang Province, Institute of brain and cognitive science, Hangzhou City University School of Medicine, Hangzhou 310015, China
| | - Xuan Chen
- NHC and CAMS Key Laboratory of Medical Neurobiology, MOE Frontier Center of Brain Science and Brain-machine Integration, School of Brain Science and Brain Medicine, Zhejiang University, Hangzhou 310058, China
- Department of Neurobiology, Affiliated Mental Health Center & Hangzhou Seventh People's Hospital, Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Aibing Peng
- NHC and CAMS Key Laboratory of Medical Neurobiology, MOE Frontier Center of Brain Science and Brain-machine Integration, School of Brain Science and Brain Medicine, Zhejiang University, Hangzhou 310058, China
| | | | - Youzhe He
- BGI Research, Hangzhou 310030, China
| | - Yueming Wang
- College of Computer Science and Technology, Zhejiang University, Hangzhou 310000, China
- Qiushi Academy for Advanced Studies, Zhejiang University, Hangzhou 310058, China
- The Nanhu Brain-computer Interface institute, Hangzhou 311100, China
| | - Xiao-Ming Li
- Department of Neurology and Department of Psychiatry, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310058, China
- NHC and CAMS Key Laboratory of Medical Neurobiology, MOE Frontier Center of Brain Science and Brain-machine Integration, School of Brain Science and Brain Medicine, Zhejiang University, Hangzhou 310058, China
| |
Collapse
|
9
|
VanInsberghe M, van Oudenaarden A. Sequencing technologies to measure translation in single cells. Nat Rev Mol Cell Biol 2025; 26:337-346. [PMID: 39833532 DOI: 10.1038/s41580-024-00822-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/09/2024] [Indexed: 01/22/2025]
Abstract
Translation is one of the most energy-intensive processes in a cell and, accordingly, is tightly regulated. Genome-wide methods to measure translation and the translatome and to study the complex regulation of protein synthesis have enabled unprecedented characterization of this crucial step of gene expression. However, technological limitations have hampered our understanding of translation control in multicellular tissues, rare cell types and dynamic cellular processes. Recent optimizations, adaptations and new techniques have enabled these measurements to be made at single-cell resolution. In this Progress, we discuss single-cell sequencing technologies to measure translation, including ribosome profiling, ribosome affinity purification and spatial translatome methods.
Collapse
Affiliation(s)
- Michael VanInsberghe
- Oncode Institute, Utrecht, the Netherlands.
- Hubrecht Institute-KNAW (Royal Netherlands Academy of Arts and Sciences), Utrecht, the Netherlands.
- University Medical Center Utrecht, Utrecht, the Netherlands.
| | - Alexander van Oudenaarden
- Oncode Institute, Utrecht, the Netherlands
- Hubrecht Institute-KNAW (Royal Netherlands Academy of Arts and Sciences), Utrecht, the Netherlands
- University Medical Center Utrecht, Utrecht, the Netherlands
| |
Collapse
|
10
|
Hui T, Zhou J, Yao M, Xie Y, Zeng H. Advances in Spatial Omics Technologies. SMALL METHODS 2025; 9:e2401171. [PMID: 40099571 DOI: 10.1002/smtd.202401171] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/29/2024] [Revised: 03/03/2025] [Indexed: 03/20/2025]
Abstract
Rapidly developing spatial omics technologies provide us with new approaches to deeply understanding the diversity and functions of cell types within organisms. Unlike traditional approaches, spatial omics technologies enable researchers to dissect the complex relationships between tissue structure and function at the cellular or even subcellular level. The application of spatial omics technologies provides new perspectives on key biological processes such as nervous system development, organ development, and tumor microenvironment. This review focuses on the advancements and strategies of spatial omics technologies, summarizes their applications in biomedical research, and highlights the power of spatial omics technologies in advancing the understanding of life sciences related to development and disease.
Collapse
Affiliation(s)
- Tianxiao Hui
- State Key Laboratory of Gene Function and Modulation Research, College of Future Technology, Peking-Tsinghua Center for Life Sciences, Peking University, Beijing, 100871, China
| | - Jian Zhou
- Peking-Tsinghua Center for Life Sciences, Tsinghua University, Beijing, 100084, China
| | - Muchen Yao
- College of Biological Sciences, China Agricultural University, Beijing, 100193, China
| | - Yige Xie
- School of Nursing, Peking University, Beijing, 100871, China
| | - Hu Zeng
- State Key Laboratory of Gene Function and Modulation Research, College of Future Technology, Peking-Tsinghua Center for Life Sciences, Peking University, Beijing, 100871, China
- Beijing Advanced Center of RNA Biology (BEACON), Peking University, Beijing, 100871, China
| |
Collapse
|
11
|
Wang Q, Zhu H, Deng L, Xu S, Xie W, Li M, Wang R, Tie L, Zhan L, Yu G. Spatial Transcriptomics: Biotechnologies, Computational Tools, and Neuroscience Applications. SMALL METHODS 2025; 9:e2401107. [PMID: 39760243 DOI: 10.1002/smtd.202401107] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/19/2024] [Revised: 12/22/2024] [Indexed: 01/07/2025]
Abstract
Spatial transcriptomics (ST) represents a revolutionary approach in molecular biology, providing unprecedented insights into the spatial organization of gene expression within tissues. This review aims to elucidate advancements in ST technologies, their computational tools, and their pivotal applications in neuroscience. It is begun with a historical overview, tracing the evolution from early image-based techniques to contemporary sequence-based methods. Subsequently, the computational methods essential for ST data analysis, including preprocessing, cell type annotation, spatial clustering, detection of spatially variable genes, cell-cell interaction analysis, and 3D multi-slices integration are discussed. The central focus of this review is the application of ST in neuroscience, where it has significantly contributed to understanding the brain's complexity. Through ST, researchers advance brain atlas projects, gain insights into brain development, and explore neuroimmune dysfunctions, particularly in brain tumors. Additionally, ST enhances understanding of neuronal vulnerability in neurodegenerative diseases like Alzheimer's and neuropsychiatric disorders such as schizophrenia. In conclusion, while ST has already profoundly impacted neuroscience, challenges remain issues such as enhancing sequencing technologies and developing robust computational tools. This review underscores the transformative potential of ST in neuroscience, paving the way for new therapeutic insights and advancements in brain research.
Collapse
Affiliation(s)
- Qianwen Wang
- Department of Bioinformatics, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Hongyuan Zhu
- Department of Bioinformatics, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Lin Deng
- Department of Bioinformatics, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Shuangbin Xu
- Department of Bioinformatics, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Wenqin Xie
- Department of Bioinformatics, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Ming Li
- Department of Bioinformatics, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Rui Wang
- Department of Bioinformatics, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Liang Tie
- Department of Bioinformatics, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Li Zhan
- Department of Bioinformatics, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Guangchuang Yu
- Department of Bioinformatics, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, China
| |
Collapse
|
12
|
Wang J, Ye F, Chai H, Jiang Y, Wang T, Ran X, Xia Q, Xu Z, Fu Y, Zhang G, Wu H, Guo G, Guo H, Ruan Y, Wang Y, Xing D, Xu X, Zhang Z. Advances and applications in single-cell and spatial genomics. SCIENCE CHINA. LIFE SCIENCES 2025; 68:1226-1282. [PMID: 39792333 DOI: 10.1007/s11427-024-2770-x] [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: 08/20/2024] [Accepted: 10/10/2024] [Indexed: 01/12/2025]
Abstract
The applications of single-cell and spatial technologies in recent times have revolutionized the present understanding of cellular states and the cellular heterogeneity inherent in complex biological systems. These advancements offer unprecedented resolution in the examination of the functional genomics of individual cells and their spatial context within tissues. In this review, we have comprehensively discussed the historical development and recent progress in the field of single-cell and spatial genomics. We have reviewed the breakthroughs in single-cell multi-omics technologies, spatial genomics methods, and the computational strategies employed toward the analyses of single-cell atlas data. Furthermore, we have highlighted the advances made in constructing cellular atlases and their clinical applications, particularly in the context of disease. Finally, we have discussed the emerging trends, challenges, and opportunities in this rapidly evolving field.
Collapse
Affiliation(s)
- Jingjing Wang
- Bone Marrow Transplantation Center of the First Affiliated Hospital & Liangzhu Laboratory, Zhejiang University School of Medicine, Hangzhou, 310058, China
| | - Fang Ye
- Bone Marrow Transplantation Center of the First Affiliated Hospital & Liangzhu Laboratory, Zhejiang University School of Medicine, Hangzhou, 310058, China
| | - Haoxi Chai
- Life Sciences Institute and The Second Affiliated Hospital, Zhejiang University, Hangzhou, 310058, China
| | - Yujia Jiang
- BGI Research, Shenzhen, 518083, China
- BGI Research, Hangzhou, 310030, China
| | - Teng Wang
- Biomedical Pioneering Innovation Center (BIOPIC) and School of Life Sciences, Peking University, Beijing, 100871, China
- Academy for Advanced Interdisciplinary Studies, Peking University, Beijing, 100871, China
| | - Xia Ran
- Bone Marrow Transplantation Center of the First Affiliated Hospital & Liangzhu Laboratory, Zhejiang University School of Medicine, Hangzhou, 310058, China
- Institute of Hematology, Zhejiang University, Hangzhou, 310000, China
| | - Qimin Xia
- Biomedical Pioneering Innovation Center (BIOPIC) and School of Life Sciences, Peking University, Beijing, 100871, China
| | - Ziye Xu
- Department of Laboratory Medicine of The First Affiliated Hospital & Liangzhu Laboratory, Zhejiang University School of Medicine, Hangzhou, 310058, China
| | - Yuting Fu
- Bone Marrow Transplantation Center of the First Affiliated Hospital & Liangzhu Laboratory, Zhejiang University School of Medicine, Hangzhou, 310058, China
- Center for Stem Cell and Regenerative Medicine, Zhejiang University School of Medicine, Hangzhou, 310058, China
| | - Guodong Zhang
- Bone Marrow Transplantation Center of the First Affiliated Hospital & Liangzhu Laboratory, Zhejiang University School of Medicine, Hangzhou, 310058, China
- Center for Stem Cell and Regenerative Medicine, Zhejiang University School of Medicine, Hangzhou, 310058, China
| | - Hanyu Wu
- Bone Marrow Transplantation Center of the First Affiliated Hospital & Liangzhu Laboratory, Zhejiang University School of Medicine, Hangzhou, 310058, China
- Center for Stem Cell and Regenerative Medicine, Zhejiang University School of Medicine, Hangzhou, 310058, China
| | - Guoji Guo
- Bone Marrow Transplantation Center of the First Affiliated Hospital & Liangzhu Laboratory, Zhejiang University School of Medicine, Hangzhou, 310058, China.
- Center for Stem Cell and Regenerative Medicine, Zhejiang University School of Medicine, Hangzhou, 310058, China.
- Zhejiang Provincial Key Lab for Tissue Engineering and Regenerative Medicine, Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cell and Regenerative Medicine, Hangzhou, 310058, China.
- Institute of Hematology, Zhejiang University, Hangzhou, 310000, China.
| | - Hongshan Guo
- Bone Marrow Transplantation Center of the First Affiliated Hospital & Liangzhu Laboratory, Zhejiang University School of Medicine, Hangzhou, 310058, China.
- Institute of Hematology, Zhejiang University, Hangzhou, 310000, China.
| | - Yijun Ruan
- Life Sciences Institute and The Second Affiliated Hospital, Zhejiang University, Hangzhou, 310058, China.
| | - Yongcheng Wang
- Department of Laboratory Medicine of The First Affiliated Hospital & Liangzhu Laboratory, Zhejiang University School of Medicine, Hangzhou, 310058, China.
| | - Dong Xing
- Biomedical Pioneering Innovation Center (BIOPIC) and School of Life Sciences, Peking University, Beijing, 100871, China.
- Beijing Advanced Innovation Center for Genomics (ICG), Peking University, Beijing, 100871, China.
| | - Xun Xu
- BGI Research, Shenzhen, 518083, China.
- BGI Research, Hangzhou, 310030, China.
- Guangdong Provincial Key Laboratory of Genome Read and Write, BGI Research, Shenzhen, 518083, China.
| | - Zemin Zhang
- Biomedical Pioneering Innovation Center (BIOPIC) and School of Life Sciences, Peking University, Beijing, 100871, China.
| |
Collapse
|
13
|
Cheng Y, Hu M, Yang B, Jensen TB, Zhang Y, Yang T, Yu R, Ma Z, Radda JSD, Jin S, Zang C, Wang S. Perturb-tracing enables high-content screening of multi-scale 3D genome regulators. Nat Methods 2025; 22:950-961. [PMID: 40211002 PMCID: PMC12074983 DOI: 10.1038/s41592-025-02652-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2024] [Accepted: 03/07/2025] [Indexed: 04/12/2025]
Abstract
Three-dimensional (3D) genome organization becomes altered during development, aging and disease, but the factors regulating chromatin topology are incompletely understood and currently no technology can efficiently screen for new regulators of multi-scale chromatin organization. Here, we developed an image-based high-content screening platform (Perturb-tracing) that combines pooled CRISPR screens, a cellular barcode readout method (BARC-FISH) and chromatin tracing. We performed a loss-of-function screen in human cells, and visualized alterations to their 3D chromatin folding conformations, alongside perturbation-paired barcode readout in the same single cells. We discovered tens of new regulators of chromatin folding at different length scales, ranging from chromatin domains and compartments to chromosome territory. A subset of the regulators exhibited 3D genome effects associated with loop extrusion and A-B compartmentalization mechanisms, while others were largely unrelated to these known 3D genome mechanisms. Finally, we identified new regulators of nuclear architectures and found a functional link between chromatin compaction and nuclear shape. Altogether, our method enables scalable, high-content identification of chromatin and nuclear topology regulators that will stimulate new insights into the 3D genome.
Collapse
Affiliation(s)
- Yubao Cheng
- Department of Genetics, Yale School of Medicine, Yale University, New Haven, CT, USA
| | - Mengwei Hu
- Department of Genetics, Yale School of Medicine, Yale University, New Haven, CT, USA
| | - Bing Yang
- Department of Genetics, Yale School of Medicine, Yale University, New Haven, CT, USA
| | - Tyler B Jensen
- Department of Genetics, Yale School of Medicine, Yale University, New Haven, CT, USA
- M.D.-Ph.D. Program, Yale University, New Haven, CT, USA
| | - Yuan Zhang
- Department of Genetics, Yale School of Medicine, Yale University, New Haven, CT, USA
| | - Tianqi Yang
- Department of Genetics, Yale School of Medicine, Yale University, New Haven, CT, USA
| | - Ruihuan Yu
- Department of Genetics, Yale School of Medicine, Yale University, New Haven, CT, USA
| | - Zhaoxia Ma
- Department of Genome Sciences, University of Virginia, Charlottesville, VA, USA
- Department of Biochemistry and Molecular Genetics, University of Virginia, Charlottesville, VA, USA
| | - Jonathan S D Radda
- Department of Genetics, Yale School of Medicine, Yale University, New Haven, CT, USA
| | - Shengyan Jin
- Department of Genetics, Yale School of Medicine, Yale University, New Haven, CT, USA
| | - Chongzhi Zang
- Department of Genome Sciences, University of Virginia, Charlottesville, VA, USA
- Department of Biochemistry and Molecular Genetics, University of Virginia, Charlottesville, VA, USA
- Department of Biomedical Engineering, University of Virginia, Charlottesville, VA, USA
- UVA Comprehensive Cancer Center, University of Virginia, Charlottesville, VA, USA
| | - Siyuan Wang
- Department of Genetics, Yale School of Medicine, Yale University, New Haven, CT, USA.
- M.D.-Ph.D. Program, Yale University, New Haven, CT, USA.
- Department of Cell Biology, Yale School of Medicine, Yale University, New Haven, CT, USA.
- Yale Combined Program in the Biological and Biomedical Sciences, Yale University, New Haven, CT, USA.
- Molecular Cell Biology, Genetics and Development Program, Yale University, New Haven, CT, USA.
- Biochemistry, Quantitative Biology, Biophysics, and Structural Biology Program, Yale University, New Haven, CT, USA.
- Yale Center for RNA Science and Medicine, Yale University School of Medicine, New Haven, CT, USA.
- Yale Liver Center, Yale University School of Medicine, New Haven, CT, USA.
| |
Collapse
|
14
|
Wang L, Bai X, Zhang C, Shi Q, Chen L. Spatially Aware Domain Adaptation Enables Cell Type Deconvolution from Multi-Modal Spatially Resolved Transcriptomics. SMALL METHODS 2025; 9:e2401163. [PMID: 39623794 DOI: 10.1002/smtd.202401163] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/28/2024] [Revised: 11/16/2024] [Indexed: 05/26/2025]
Abstract
Spatially Resolved Transcriptomics (SRT) offers unprecedented opportunities to elucidate the cellular arrangements within tissues. Nevertheless, the absence of deconvolution methods that simultaneously model multi-modal features has impeded progress in understanding cellular heterogeneity in spatial contexts. To address this issue, SpaDA is developed, a novel spatially aware domain adaptation method that integrates multi-modal data (i.e., transcriptomics, histological images, and spatial locations) from SRT to accurately estimate the spatial distribution of cell types. SpaDA utilizes a self-expressive variational autoencoder, coupled with deep spatial distribution alignment, to learn and align spatial and graph representations from spatial multi-modal SRT data and single-cell RNA sequencing (scRNA-seq) data. This strategy facilitates the transfer of cell type annotation information across these two similarity graphs, thereby enhancing the prediction accuracy of cell type composition. The results demonstrate that SpaDA surpasses existing methods in cell type deconvolution and the identification of cell types and spatial domains across diverse platforms. Moreover, SpaDA excels in identifying spatially colocalized cell types and key marker genes in regions of low-quality measurements, exemplified by high-resolution mouse cerebellum SRT data. In conclusion, SpaDA offers a powerful and flexible framework for the analysis of multi-modal SRT datasets, advancing the understanding of complex biological systems.
Collapse
Affiliation(s)
- Lequn Wang
- Key Laboratory of Systems Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, Shanghai, 200031, China
- Key Laboratory of Systems Health Science of Zhejiang Province, School of Life Science, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou, 310024, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Xiaosheng Bai
- Key Laboratory of Systems Health Science of Zhejiang Province, School of Life Science, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou, 310024, China
| | - Chuanchao Zhang
- Key Laboratory of Systems Health Science of Zhejiang Province, School of Life Science, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou, 310024, China
| | - Qianqian Shi
- Hubei Engineering Technology Research Center of Agricultural Big Data, Huazhong Agricultural University, Wuhan, 430070, China
- Hubei Key Laboratory of Agricultural Bioinformatics, College of Informatics, Huazhong Agricultural University, Wuhan, 430070, China
| | - Luonan Chen
- Key Laboratory of Systems Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, Shanghai, 200031, China
- Key Laboratory of Systems Health Science of Zhejiang Province, School of Life Science, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou, 310024, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| |
Collapse
|
15
|
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
|
16
|
Zhang Z, Sun Y, Peng Q, Li T, Zhou P. Integrating Dynamical Systems Modeling with Spatiotemporal scRNA-Seq Data Analysis. ENTROPY (BASEL, SWITZERLAND) 2025; 27:453. [PMID: 40422408 DOI: 10.3390/e27050453] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/14/2025] [Revised: 04/18/2025] [Accepted: 04/19/2025] [Indexed: 05/28/2025]
Abstract
Understanding the dynamic nature of biological systems is fundamental to deciphering cellular behavior, developmental processes, and disease progression. Single-cell RNA sequencing (scRNA-seq) has provided static snapshots of gene expression, offering valuable insights into cellular states at a single time point. Recent advancements in temporally resolved scRNA-seq, spatial transcriptomics (ST), and time-series spatial transcriptomics (temporal-ST) have further revolutionized our ability to study the spatiotemporal dynamics of individual cells. These technologies, when combined with computational frameworks such as Markov chains, stochastic differential equations (SDEs), and generative models like optimal transport and Schrödinger bridges, enable the reconstruction of dynamic cellular trajectories and cell fate decisions. This review discusses how these dynamical system approaches offer new opportunities to model and infer cellular dynamics from a systematic perspective.
Collapse
Affiliation(s)
- Zhenyi Zhang
- School of Mathematical Sciences, Peking University, Beijing 100871, China
| | - Yuhao Sun
- Center for Machine Learning Research, Peking University, Beijing 100871, China
| | - Qiangwei Peng
- School of Mathematical Sciences, Peking University, Beijing 100871, China
| | - Tiejun Li
- School of Mathematical Sciences, Peking University, Beijing 100871, China
- Center for Machine Learning Research, Peking University, Beijing 100871, China
- Laboratory of Mathematics and Its Applications (LMAM), Peking University, Beijing 100871, China
| | - Peijie Zhou
- Center for Machine Learning Research, Peking University, Beijing 100871, China
- Center for Quantitative Biology, Peking University, Beijing 100871, China
- National Engineering Laboratory for Big Data Analysis and Applications, Peking University, Beijing 100871, China
- AI for Science Institute, Beijing 100080, China
| |
Collapse
|
17
|
Lee Y, Lee M, Shin Y, Kim K, Kim T. Spatial Omics in Clinical Research: A Comprehensive Review of Technologies and Guidelines for Applications. Int J Mol Sci 2025; 26:3949. [PMID: 40362187 PMCID: PMC12071594 DOI: 10.3390/ijms26093949] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2025] [Revised: 04/17/2025] [Accepted: 04/17/2025] [Indexed: 05/15/2025] Open
Abstract
Spatial omics integrates molecular profiling with spatial tissue context, enabling high-resolution analysis of gene expression, protein interactions, and epigenetic modifications. This approach provides critical insights into disease mechanisms and therapeutic responses, with applications in cancer, neurology, and immunology. Spatial omics technologies, including spatial transcriptomics, proteomics, and epigenomics, facilitate the study of cellular heterogeneity, tissue organization, and cell-cell interactions within their native environments. Despite challenges in data complexity and integration, advancements in multi-omics pipelines and computational tools are enhancing data accuracy and biological interpretation. This review provides a comprehensive overview of key spatial omics technologies, their analytical methods, validation strategies, and clinical applications. By integrating spatially resolved molecular data with traditional omics, spatial omics is transforming precision medicine, biomarker discovery, and personalized therapy. Future research should focus on improving standardization, reproducibility, and multimodal data integration to fully realize the potential of spatial omics in clinical and translational research.
Collapse
Affiliation(s)
- Yoonji Lee
- College of Medicine, The Catholic University of Korea, 222 Banpo-daero, Seocho-gu, Seoul 06591, Republic of Korea; (Y.L.); (M.L.); (Y.S.)
| | - Mingyu Lee
- College of Medicine, The Catholic University of Korea, 222 Banpo-daero, Seocho-gu, Seoul 06591, Republic of Korea; (Y.L.); (M.L.); (Y.S.)
| | - Yoojin Shin
- College of Medicine, The Catholic University of Korea, 222 Banpo-daero, Seocho-gu, Seoul 06591, Republic of Korea; (Y.L.); (M.L.); (Y.S.)
| | - Kyuri Kim
- College of Medicine, Ewha Womans University, 25 Magokdong-ro 2-gil, Gangseo-gu, Seoul 07804, Republic of Korea;
| | - Taejung Kim
- Department of Hospital Pathology, Yeouido St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, 10, 63-ro, Yeongdeungpo-gu, Seoul 07345, Republic of Korea
| |
Collapse
|
18
|
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
|
19
|
Fang S, Xu M, Cao L, Liu X, Bezulj M, Tan L, Yuan Z, Li Y, Xia T, Guo L, Kovacevic V, Hui J, Guo L, Liu C, Cheng M, Lin L, Wen Z, Josic B, Milicevic N, Qiu P, Lu Q, Li Y, Wang L, Hu L, Zhang C, Kang Q, Chen F, Deng Z, Li J, Li M, Li S, Zhao Y, Fan G, Zhang Y, Chen A, Li Y, Xu X. Stereopy: modeling comparative and spatiotemporal cellular heterogeneity via multi-sample spatial transcriptomics. Nat Commun 2025; 16:3741. [PMID: 40258830 PMCID: PMC12012134 DOI: 10.1038/s41467-025-58079-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2024] [Accepted: 03/05/2025] [Indexed: 04/23/2025] Open
Abstract
Understanding complex biological systems requires tracing cellular dynamic changes across conditions, time, and space. However, integrating multi-sample data in a unified way to explore cellular heterogeneity remains challenging. Here, we present Stereopy, a flexible framework for modeling and dissecting comparative and spatiotemporal patterns in multi-sample spatial transcriptomics with interactive data visualization. To optimize this framework, we devise a universal container, a scope controller, and an integrative transformer tailored for multi-sample multimodal data storage, management, and processing. Stereopy showcases three representative applications: investigating specific cell communities and genes responsible for pathological changes, detecting spatiotemporal gene patterns by considering spatial and temporal features, and inferring three-dimensional niche-based cell-gene interaction network that bridges intercellular communications and intracellular regulations. Stereopy serves as both a comprehensive bioinformatics toolbox and an extensible framework that empowers researchers with enhanced data interpretation abilities and new perspectives for mining multi-sample spatial transcriptomics data.
Collapse
Affiliation(s)
| | - Mengyang Xu
- BGI Research, Shenzhen, China
- BGI Research, Qingdao, China
| | - Lei Cao
- BGI Research, Beijing, China
- BGI Research, Shenzhen, China
| | | | | | | | - Zhiyuan Yuan
- Institute of Science and Technology for Brain-Inspired Intelligence, MOE Key Laboratory of Computational Neuroscience and Brain-Inspired Intelligence, Fudan University, Shanghai, China
| | - Yao Li
- BGI Research, Qingdao, China
| | - Tianyi Xia
- BGI Research, Beijing, China
- BGI Research, Shenzhen, China
| | | | | | | | - Lidong Guo
- BGI Research, Qingdao, China
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, China
| | | | - Mengnan Cheng
- BGI Research, Shenzhen, China
- BGI Research, Hangzhou, China
| | | | | | | | | | | | - Qin Lu
- BGI Research, Shenzhen, China
| | | | | | - Luni Hu
- BGI Research, Beijing, China
- BGI Research, Shenzhen, China
| | | | | | | | | | - Junhua Li
- BGI Research, Shenzhen, China
- Shenzhen Key Laboratory of Unknown Pathogen Identification, BGI Research, Shenzhen, China
- BGI Research, Riga, Latvia
| | - Mei Li
- BGI Research, Shenzhen, China
| | | | - Yi Zhao
- Beijing Key Laboratory of Mobile Computing and Pervasive Device, Institute of Computing Technology, Chinese Academy of Sciences, Beijing, China.
| | - Guangyi Fan
- BGI Research, Shenzhen, China.
- BGI Research, Qingdao, China.
| | - Yong Zhang
- BGI Research, Shenzhen, China.
- BGI Research, Wuhan, China.
- Guangdong Bigdata Engineering Technology Research Center for Life Sciences, BGI research, Shenzhen, China.
| | - Ao Chen
- BGI Research, Shenzhen, China.
| | - Yuxiang Li
- BGI Research, Shenzhen, China.
- BGI Research, Wuhan, China.
- Guangdong Bigdata Engineering Technology Research Center for Life Sciences, BGI research, Shenzhen, China.
| | - Xun Xu
- BGI Research, Wuhan, China.
| |
Collapse
|
20
|
Liu F, Ren S, Li J, Lv H, Jiang F, Bin Yu. SGTB: A graph representation learning model combining transformer and BERT for optimizing gene expression analysis in spatial transcriptomics data. Comput Biol Chem 2025; 118:108482. [PMID: 40306096 DOI: 10.1016/j.compbiolchem.2025.108482] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2025] [Revised: 04/05/2025] [Accepted: 04/17/2025] [Indexed: 05/02/2025]
Abstract
In recent years, spatial transcriptomics (ST) has emerged as an innovative technology that enables the simultaneous acquisition of gene expression information and its spatial distribution at the single-cell or regional level, providing deeper insights into cellular interactions and tissue organization, this technology provides a more holistic view of tissue organization and intercellular dynamics. However, existing methods still face certain limitations in data representation capabilities, making it challenging to fully capture complex spatial dependencies and global features. To address this, this paper proposes an innovative spatial multi-scale graph convolutional network (SGTB) based on large language models, integrating graph convolutional networks (GCN), Transformer, and BERT language models to optimize the representation of spatial transcriptomics data. The Graph Convolutional Network (GCN) employs a multi-layer architecture to extract features from gene expression matrices. Through iterative aggregation of neighborhood information, it captures spatial dependencies among cells and gene co-expression patterns, thereby constructing hierarchical cell embeddings. Subsequently, the model integrates an attention mechanism to assign weights to critical features and leverages Transformer layers to model global relationships, refining the ability of learned representations to reflect variations in spatial patterns. Finally, the model incorporates the BERT language model, mapping cell embeddings into textual inputs to exploit its deep semantic representation capabilities for high-dimensional feature extraction. These features are then fused with the embeddings generated by the Transformer, further optimizing feature learning for spatial transcriptomics data. This approach holds significant application value in improving the accuracy of tasks such as cell type classification and gene regulatory network construction, providing a novel computational framework for deep mining of spatial multi-scale biological data.
Collapse
Affiliation(s)
- Farong Liu
- College of Mathematics and Physics, Qingdao University of Science and Technology, Qingdao 266061, China; School of Data Science, Qingdao University of Science and Technology, Qingdao 266061, China
| | - Sheng Ren
- School of Data Science, Qingdao University of Science and Technology, Qingdao 266061, China; Artificial Intelligence and Biomedical Big Data Research Center, Qingdao University of Science and Technology, Qingdao 266061, China
| | - Jie Li
- School of Data Science, Qingdao University of Science and Technology, Qingdao 266061, China; Artificial Intelligence and Biomedical Big Data Research Center, Qingdao University of Science and Technology, Qingdao 266061, China
| | - Haoyang Lv
- School of Data Science, Qingdao University of Science and Technology, Qingdao 266061, China; Artificial Intelligence and Biomedical Big Data Research Center, Qingdao University of Science and Technology, Qingdao 266061, China
| | - Fenghui Jiang
- Editorial Office of Journal of Qingdao University of Science and Technology (Natural Science Edition), Qingdao University of Science and Technology, Qingdao 266061, China.
| | - Bin Yu
- School of Data Science, Qingdao University of Science and Technology, Qingdao 266061, China; Artificial Intelligence and Biomedical Big Data Research Center, Qingdao University of Science and Technology, Qingdao 266061, China.
| |
Collapse
|
21
|
Gandin V, Kim J, Yang LZ, Lian Y, Kawase T, Hu A, Rokicki K, Fleishman G, Tillberg P, Castrejon AA, Stringer C, Preibisch S, Liu ZJ. Deep-tissue transcriptomics and subcellular imaging at high spatial resolution. Science 2025; 388:eadq2084. [PMID: 39977545 PMCID: PMC12005972 DOI: 10.1126/science.adq2084] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2024] [Revised: 11/11/2024] [Accepted: 02/07/2025] [Indexed: 02/22/2025]
Abstract
Limited color channels in fluorescence microscopy have long constrained spatial analysis in biological specimens. We introduce cycle hybridization chain reaction (cycleHCR), a method that integrates multicycle DNA barcoding with HCR to overcome this limitation. cycleHCR enables highly multiplexed imaging of RNA and proteins using a unified barcode system. Whole-embryo transcriptomics imaging achieved precise three-dimensional gene expression and cell fate mapping across a specimen depth of ~310 μm. When combined with expansion microscopy, cycleHCR revealed an intricate network of 10 subcellular structures in mouse embryonic fibroblasts. In mouse hippocampal slices, multiplex RNA and protein imaging uncovered complex gene expression gradients and cell-type-specific nuclear structural variations. cycleHCR provides a quantitative framework for elucidating spatial regulation in deep tissue contexts for research and has potential diagnostic applications.
Collapse
Affiliation(s)
- Valentina Gandin
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, VA 20147, USA
| | - Jun Kim
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, VA 20147, USA
| | - Liang-Zhong Yang
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, VA 20147, USA
| | - Yumin Lian
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, VA 20147, USA
| | - Takashi Kawase
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, VA 20147, USA
| | - Amy Hu
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, VA 20147, USA
| | - Konrad Rokicki
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, VA 20147, USA
| | - Greg Fleishman
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, VA 20147, USA
| | - Paul Tillberg
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, VA 20147, USA
| | | | - Carsen Stringer
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, VA 20147, USA
| | - Stephan Preibisch
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, VA 20147, USA
| | - Zhe J. Liu
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, VA 20147, USA
| |
Collapse
|
22
|
Aggarwal B, Sinha S. CellSP: Module discovery and visualization for subcellular spatial transcriptomics data. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.01.12.632553. [PMID: 39868198 PMCID: PMC11761418 DOI: 10.1101/2025.01.12.632553] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 01/28/2025]
Abstract
Spatial transcriptomics has enabled the study of mRNA distributions within cells, a key aspect of cellular function. However, there is a dearth of tools that can identify and interpret functionally relevant spatial patterns of subcellular transcript distribution. To address this, we present CellSP, a computational framework for identifying, visualizing, and characterizing consistent subcellular spatial patterns of mRNA. CellSP introduces the concept of "gene-cell modules", which are gene sets with coordinated subcellular transcript distributions in many cells. It provides intuitive visualizations of the captured patterns and offers functional insights into each discovered module. We demonstrate that CellSP reliably identifies functionally significant modules across diverse tissues and technologies. We use the tool to discover subcellular spatial phenomena related to myelination, axonogenesis and synapse formation in the mouse brain. We find immune response-related modules that change between kidney cancer and healthy samples, and myelination-related modules specific to mouse models of Alzheimer's Disease.
Collapse
|
23
|
Hsieh HC, Han Q, Brenes D, Bishop KW, Wang R, Wang Y, Poudel C, Glaser AK, Freedman BS, Vaughan JC, Allbritton NL, Liu JTC. Imaging 3D cell cultures with optical microscopy. Nat Methods 2025:10.1038/s41592-025-02647-w. [PMID: 40247123 DOI: 10.1038/s41592-025-02647-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2024] [Accepted: 01/16/2025] [Indexed: 04/19/2025]
Abstract
Three-dimensional (3D) cell cultures have gained popularity in recent years due to their ability to represent complex tissues or organs more faithfully than conventional two-dimensional (2D) cell culture. This article reviews the application of both 2D and 3D microscopy approaches for monitoring and studying 3D cell cultures. We first summarize the most popular optical microscopy methods that have been used with 3D cell cultures. We then discuss the general advantages and disadvantages of various microscopy techniques for several broad categories of investigation involving 3D cell cultures. Finally, we provide perspectives on key areas of technical need in which there are clear opportunities for innovation. Our goal is to guide microscope engineers and biomedical end users toward optimal imaging methods for specific investigational scenarios and to identify use cases in which additional innovations in high-resolution imaging could be helpful.
Collapse
Affiliation(s)
- Huai-Ching Hsieh
- Department of Bioengineering, University of Washington, Seattle, WA, USA
- Department of Mechanical Engineering, University of Washington, Seattle, WA, USA
| | - Qinghua Han
- Department of Bioengineering, University of Washington, Seattle, WA, USA
- Department of Mechanical Engineering, University of Washington, Seattle, WA, USA
| | - David Brenes
- Department of Mechanical Engineering, University of Washington, Seattle, WA, USA
| | - Kevin W Bishop
- Department of Bioengineering, University of Washington, Seattle, WA, USA
- Department of Mechanical Engineering, University of Washington, Seattle, WA, USA
| | - Rui Wang
- Department of Mechanical Engineering, University of Washington, Seattle, WA, USA
| | - Yuli Wang
- Department of Bioengineering, University of Washington, Seattle, WA, USA
| | - Chetan Poudel
- Department of Chemistry, University of Washington, Seattle, WA, USA
| | - Adam K Glaser
- Allen Institute for Neural Dynamics, Seattle, WA, USA
| | - Benjamin S Freedman
- Department of Bioengineering, University of Washington, Seattle, WA, USA
- Department of Medicine, Division of Nephrology, Kidney Research Institute and Institute for Stem Cell and Regenerative Medicine, Seattle, WA, USA
- Plurexa LLC, Seattle, WA, USA
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA, USA
| | - Joshua C Vaughan
- Department of Chemistry, University of Washington, Seattle, WA, USA
- Department of Physiology and Biophysics, University of Washington, Seattle, WA, USA
| | - Nancy L Allbritton
- Department of Bioengineering, University of Washington, Seattle, WA, USA
| | - Jonathan T C Liu
- Department of Bioengineering, University of Washington, Seattle, WA, USA.
- Department of Mechanical Engineering, University of Washington, Seattle, WA, USA.
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA, USA.
| |
Collapse
|
24
|
Binan L, Jiang A, Danquah SA, Valakh V, Simonton B, Bezney J, Manguso RT, Yates KB, Nehme R, Cleary B, Farhi SL. Simultaneous CRISPR screening and spatial transcriptomics reveal intracellular, intercellular, and functional transcriptional circuits. Cell 2025; 188:2141-2158.e18. [PMID: 40081369 DOI: 10.1016/j.cell.2025.02.012] [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: 11/30/2023] [Revised: 10/24/2024] [Accepted: 02/14/2025] [Indexed: 03/16/2025]
Abstract
Pooled optical screens have enabled the study of cellular interactions, morphology, or dynamics at massive scale, but they have not yet leveraged the power of highly plexed single-cell resolved transcriptomic readouts to inform molecular pathways. Here, we present a combination of imaging spatial transcriptomics with parallel optical detection of in situ amplified guide RNAs (Perturb-FISH). Perturb-FISH recovers intracellular effects that are consistent with single-cell RNA-sequencing-based readouts of perturbation effects (Perturb-seq) in a screen of lipopolysaccharide response in cultured monocytes, and it uncovers intercellular and density-dependent regulation of the innate immune response. Similarly, in three-dimensional xenograft models, Perturb-FISH identifies tumor-immune interactions altered by genetic knockout. When paired with a functional readout in a separate screen of autism spectrum disorder risk genes in human-induced pluripotent stem cell (hIPSC) astrocytes, Perturb-FISH shows common calcium activity phenotypes and their associated genetic interactions and dysregulated molecular pathways. Perturb-FISH is thus a general method for studying the genetic and molecular associations of spatial and functional biology at single-cell resolution.
Collapse
Affiliation(s)
- Loϊc Binan
- Spatial Technology Platform, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA; Klarman Cell Observatory, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Aiping Jiang
- Cancer Program, Broad Institute of Harvard and MIT, Cambridge, MA 02142, USA; Krantz Family Center for Cancer Research, Massachusetts General Hospital, Boston, MA 02144, USA; Department of Medicine, Harvard Medical School, Boston, MA 02115, USA
| | - Serwah A Danquah
- Spatial Technology Platform, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA; Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Vera Valakh
- Spatial Technology Platform, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA; Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Brooke Simonton
- Klarman Cell Observatory, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Jon Bezney
- Klarman Cell Observatory, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Robert T Manguso
- Cancer Program, Broad Institute of Harvard and MIT, Cambridge, MA 02142, USA; Krantz Family Center for Cancer Research, Massachusetts General Hospital, Boston, MA 02144, USA; Department of Medicine, Harvard Medical School, Boston, MA 02115, USA
| | - Kathleen B Yates
- Cancer Program, Broad Institute of Harvard and MIT, Cambridge, MA 02142, USA; Krantz Family Center for Cancer Research, Massachusetts General Hospital, Boston, MA 02144, USA; Department of Medicine, Harvard Medical School, Boston, MA 02115, USA
| | - Ralda Nehme
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Brian Cleary
- Faculty of Computing and Data Sciences, Boston University, Boston, MA 02215, USA; Department of Biology, Boston University, Boston, MA 02215, USA; Department of Biomedical Engineering, Boston University, Boston, MA 02215, USA; Program in Bioinformatics, Boston University, Boston, MA 02215, USA; Biological Design Center, Boston University, Boston, MA 02215, USA.
| | - Samouil L Farhi
- Spatial Technology Platform, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA.
| |
Collapse
|
25
|
Li Y, Liu X, Guo L, Han K, Fang S, Wan X, Wang D, Xu X, Jiang L, Fan G, Xu M. SpaGRN: Investigating spatially informed regulatory paths for spatially resolved transcriptomics data. Cell Syst 2025; 16:101243. [PMID: 40179878 DOI: 10.1016/j.cels.2025.101243] [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: 11/27/2023] [Revised: 08/30/2024] [Accepted: 03/07/2025] [Indexed: 04/05/2025]
Abstract
Cells spatially organize into distinct cell types or functional domains through localized gene regulatory networks. However, current spatially resolved transcriptomics analyses fail to integrate spatial constraints and proximal cell influences, limiting the mechanistic understanding of tissue organization. Here, we introduce SpaGRN, a statistical framework that reconstructs cell-type- or functional-domain-specific, dynamic, and spatial regulons by coupling intracellular spatial regulatory causality with extracellular signaling path information. Benchmarking across synthetic and real datasets demonstrates SpaGRN's superior precision over state-of-the-art tools in identifying context-dependent regulons. Applied to diverse spatially resolved transcriptomics platforms (Stereo-seq, STARmap, MERFISH, CosMx, Slide-seq, and 10x Visium), complex cancerous samples, and 3D datasets of developing Drosophila embryos and larvae, SpaGRN not only provides a versatile toolkit for decoding receptor-mediated spatial regulons but also reveals spatiotemporal regulatory mechanisms underlying organogenesis and inflammation.
Collapse
Affiliation(s)
- Yao Li
- BGI Research, Sanya 572025, China; BGI Research, Qingdao 266555, China
| | | | - Lidong Guo
- BGI Research, Qingdao 266555, China; College of Life Sciences, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Kai Han
- BGI Research, Qingdao 266555, China
| | - Shuangsang Fang
- BGI Research, Beijing 102601, China; BGI Research, Shenzhen 518083, China
| | - Xinjiang Wan
- CAS Key Laboratory of Marine Ecology and Environmental Sciences, and Center of Deep Sea Research Institute of Oceanology, Chinese Academy of Sciences, Qingdao 266071, China
| | | | - Xun Xu
- BGI Research, Wuhan 430074, China; State Key Laboratory of Genome and Multi-omics Technologies, BGI Research, Shenzhen 518083, China
| | - Ling Jiang
- State Key Laboratory of Materials-Oriented Chemical Engineering, Nanjing Tech University, Nanjing 211816, China.
| | - Guangyi Fan
- BGI Research, Sanya 572025, China; BGI Research, Qingdao 266555, China; BGI Research, Shenzhen 518083, China; State Key Laboratory of Genome and Multi-omics Technologies, BGI Research, Shenzhen 518083, China.
| | - Mengyang Xu
- BGI Research, Sanya 572025, China; BGI Research, Qingdao 266555, China; BGI Research, Shenzhen 518083, China; State Key Laboratory of Genome and Multi-omics Technologies, BGI Research, Shenzhen 518083, China.
| |
Collapse
|
26
|
Yusoh NA, Gill MR, Tian X. Advancing super-resolution microscopy with metal complexes: functional imaging agents for nanoscale visualization. Chem Soc Rev 2025; 54:3616-3646. [PMID: 39981712 DOI: 10.1039/d4cs01193g] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/22/2025]
Abstract
Super-resolution microscopy (SRM) has transformed biological imaging by overcoming the diffraction limit, offering nanoscale visualization of cellular structures and processes. However, the widespread use of organic fluorescent probes is often hindered by limitations such as photobleaching, short photostability, and inadequate performance in deep-tissue imaging. Metal complexes, with their superior photophysical properties, including exceptional photostability, tuneable luminescence, and extended excited-state lifetimes, address these challenges, enabling precise subcellular targeting and long-term imaging. Beyond imaging, their theranostic potential unlocks real-time diagnostics and treatments for diseases such as cancer and bacterial infections. This review explores recent advancements in applying metal complexes for SRM, focusing on their utility in visualizing intricate subcellular structures, capturing temporal dynamics in live cells and elucidating in vivo spatial organization. We emphasize how rational design strategies optimize biocompatibility, organelle specificity, and deep-tissue penetration, expanding their applicability in multiplexed imaging. Furthermore, we discuss the design of various metal nanoparticles (NPs) for SRM, along with emerging hybrid nanoscale probes that integrate metal complexes with gold (Au) scaffolds, offering promising avenues for overcoming current limitations. By highlighting both established successes and potential frontiers, this review provides a roadmap for leveraging metal complexes as versatile tools in advancing SRM applications.
Collapse
Affiliation(s)
- Nur Aininie Yusoh
- Department of Radiology, Huaxi MR Research Center (HMRRC), Institution of Radiology and Medical Imaging, West China Hospital of Sichuan University, Sichuan University, Chengdu, Sichuan, China.
| | - Martin R Gill
- Department of Chemistry, Faculty of Science and Engineering, Swansea University, Swansea, UK.
| | - Xiaohe Tian
- Department of Radiology, Huaxi MR Research Center (HMRRC), Institution of Radiology and Medical Imaging, West China Hospital of Sichuan University, Sichuan University, Chengdu, Sichuan, China.
| |
Collapse
|
27
|
Barmukh R, Garg V, Liu H, Chitikineni A, Xin L, Henry R, Varshney RK. Spatial omics for accelerating plant research and crop improvement. Trends Biotechnol 2025:S0167-7799(25)00092-7. [PMID: 40221306 DOI: 10.1016/j.tibtech.2025.03.007] [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: 09/06/2024] [Revised: 03/10/2025] [Accepted: 03/11/2025] [Indexed: 04/14/2025]
Abstract
Plant cells communicate information to regulate developmental processes and respond to environmental stresses. This communication spans various 'omics' layers within a cell and operates through intricate regulatory networks. The emergence of spatial omics presents a promising approach to thoroughly analyze cells, allowing the combined analysis of diverse modalities either in parallel or on the same tissue section. Here, we provide an overview of recent advancements in spatial omics and delineate scientific discoveries in plant research enabled by these technologies. We delve into experimental and computational challenges and outline strategies to navigate these challenges for advancing breeding efforts. With ongoing insightful discoveries and improved accessibility, spatial omics stands on the brink of playing a crucial role in designing future crops.
Collapse
Affiliation(s)
- Rutwik Barmukh
- WA State Agricultural Biotechnology Centre, Centre for Crop and Food Innovation, Food Futures Institute, Murdoch University, Murdoch 6150, Western Australia, Australia
| | - Vanika Garg
- WA State Agricultural Biotechnology Centre, Centre for Crop and Food Innovation, Food Futures Institute, Murdoch University, Murdoch 6150, Western Australia, Australia
| | - Hao Liu
- Guangdong Provincial Key Laboratory of Crop Genetic Improvement, South China Peanut Sub-Center of National Center of Oilseed Crops Improvement, Crops Research Institute, Guangdong Academy of Agricultural Sciences, Guangzhou, Guangdong Province, 510640, China
| | - Annapurna Chitikineni
- WA State Agricultural Biotechnology Centre, Centre for Crop and Food Innovation, Food Futures Institute, Murdoch University, Murdoch 6150, Western Australia, Australia
| | - Liu Xin
- WA State Agricultural Biotechnology Centre, Centre for Crop and Food Innovation, Food Futures Institute, Murdoch University, Murdoch 6150, Western Australia, Australia; BGI-Shenzhen, Shenzhen, 518083, China
| | - Robert Henry
- Queensland Alliance for Agriculture & Food Innovation, Queensland Biosciences Precinct, The University of Queensland, St Lucia, QLD 4072, Australia
| | - Rajeev K Varshney
- WA State Agricultural Biotechnology Centre, Centre for Crop and Food Innovation, Food Futures Institute, Murdoch University, Murdoch 6150, Western Australia, Australia.
| |
Collapse
|
28
|
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
|
29
|
Lin Z, Wang W, Marin-Llobet A, Li Q, Pollock SD, Sui X, Aljovic A, Lee J, Baek J, Liang N, Zhang X, Wang CK, Huang J, Liu M, Gao Z, Sheng H, Du J, Lee SJ, Wang B, He Y, Ding J, Wang X, Alvarez-Dominguez JR, Liu J. Spatial transcriptomics AI agent charts hPSC-pancreas maturation in vivo. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.04.01.646731. [PMID: 40236029 PMCID: PMC11996516 DOI: 10.1101/2025.04.01.646731] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/17/2025]
Abstract
Spatial transcriptomics has revolutionized our understanding of tissue organization by simultaneously capturing gene expression and spatial localization within intact tissues. However, analyzing these increasingly complex datasets requires specialized expertise across computational biology, statistics, and biological context. To address this challenge, we introduce the Spatial Transcriptomics AI Agent (STAgent), an autonomous multimodal agentic AI that integrates multimodal large language models (LLMs) with specialized computational tools to transform weeks-long analysis tasks into minutes of automated processing. Unlike conventional machine learning approaches that are limited to narrow, predefined tasks, STAgent leverages the emergent capabilities of multimodal LLMs - such as flexible reasoning, contextual understanding, and cross-modal integration - which allow it to adapt to novel data, execute multi-step analyses, and generate biologically meaningful insights with minimal human input. STAgent enables autonomous deep research through integrated capabilities, including dynamic code generation for complex analytical workflows, visual reasoning for interpreting spatial patterns, real-time retrieval of relevant peer-reviewd scientific literature, and synthesis of comprehensive, actionable reports. We applied STAgent to investigate the in vivo maturation of human stem cell-derived pancreatic cells (SC-pancreas) transplanted into immunodeficient mice. We generated single-cell spatial transcriptomics data spanning multiple developmental timepoints. STAgent autonomously (1) identified the maturation of initially scattered endocrine cells into well-defined islet-like structures, with predominantly peripheral α-cells surrounding β-cell cores supported by an expanding mesenchymal network; (2) revealed strengthening endocrine-endocrine cell interactions over time and, through context-aware gene set analysis, uncovered spatially resolved biological processes driving maturation; (3) unlike traditional analytical approaches, STAgent offers mechanistic explanations of spatial patterns, contextualizing findings with relevant literatures and developing cohesive insights into human pancreatic development. This agentic approach establishes a new paradigm in spatial transcriptomics analysis by substantially lowering the expertise barrier and reducing analysis time, accelerating biological and biomedical discovery.
Collapse
|
30
|
Hu C, Borji M, Marrero GJ, Kumar V, Weir JA, Kammula SV, Macosko EZ, Chen F. Scalable spatial transcriptomics through computational array reconstruction. Nat Biotechnol 2025:10.1038/s41587-025-02612-0. [PMID: 40181168 DOI: 10.1038/s41587-025-02612-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2024] [Accepted: 02/21/2025] [Indexed: 04/05/2025]
Abstract
Spatial transcriptomics enables gene expression mapping within tissues but is often limited by imaging constraints. We present an imaging-free approach that reconstructs spatial barcode locations using molecular diffusion and dimensionality reduction. Validated against ground truth imaging, our method achieves high fidelity and scales to centimeter-sized tissues. This approach enhances spatial transcriptomics' accessibility and throughput, enabling large-scale studies without specialized imaging equipment.
Collapse
Affiliation(s)
- Chenlei Hu
- Broad Institute of Harvard and MIT, Cambridge, MA, USA
- Department of Molecular and Cellular Biology, Harvard University, Cambridge, MA, USA
| | - Mehdi Borji
- Broad Institute of Harvard and MIT, Cambridge, MA, USA
| | | | - Vipin Kumar
- Broad Institute of Harvard and MIT, Cambridge, MA, USA
| | - Jackson A Weir
- Broad Institute of Harvard and MIT, Cambridge, MA, USA
- Biological and Biomedical Sciences Program, Harvard University, Cambridge, MA, USA
| | | | - Evan Z Macosko
- Broad Institute of Harvard and MIT, Cambridge, MA, USA
- Department of Psychiatry, Massachusetts General Hospital, Boston, MA, USA
| | - Fei Chen
- Broad Institute of Harvard and MIT, Cambridge, MA, USA.
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA, USA.
| |
Collapse
|
31
|
Li X, Nguyen J, Korkut A. Recurrent Composite Markers of Cell Types and States. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2023.07.17.549344. [PMID: 37503180 PMCID: PMC10370072 DOI: 10.1101/2023.07.17.549344] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/29/2023]
Abstract
Biological function is mediated by the hierarchical organization of cell types and states within tissue ecosystems. Identifying interpretable composite marker sets that both define and distinguish hierarchical cell identities is essential for decoding biological complexity, yet remains a major challenge. Here, we present RECOMBINE, an algorithm that identifies recurrent composite marker sets to define hierarchical cell identities. Validation using both simulated and biological datasets demonstrates that RECOMBINE achieves higher accuracy in identifying discriminative markers compared to existing approaches, including differential gene expression analysis. When applied to single-cell data and validated with spatial transcriptomics data from the mouse visual cortex, RECOMBINE identified key cell type markers and generated a robust gene panel for targeted spatial profiling. It also uncovered markers of CD8+; T cell states, including GZMK+;HAVCR2-; effector memory cells associated with anti-PD-1 therapy response, and revealed a rare intestinal subpopulation with composite markers in mice. Finally, using data from the Tabula Sapiens project, RECOMBINE identified composite marker sets across a broad range of human tissues. Together, these results highlight RECOMBINE as a robust, data-driven framework for optimized marker selection, enabling the discovery and validation of hierarchical cell identities across diverse tissue contexts.
Collapse
|
32
|
Yuan C, Patel K, Shi H, Wang HLV, Wang F, Li R, Li Y, Corces VG, Shi H, Das S, Yu J, Jin P, Yao B, Hu J. mcDETECT: Decoding 3D Spatial Synaptic Transcriptomes with Subcellular-Resolution Spatial Transcriptomics. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.03.27.645744. [PMID: 40236251 PMCID: PMC11996425 DOI: 10.1101/2025.03.27.645744] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/17/2025]
Abstract
Spatial transcriptomics (ST) has shown great potential for unraveling the molecular mechanisms of neurodegenerative diseases. However, most existing analyses of ST data focus on bulk or single-cell resolution, overlooking subcellular compartments such as synapses, which are fundamental structures of the brain's neural network. Here we present mcDETECT, a novel framework that integrates machine learning algorithms and in situ ST (iST) with targeted gene panels to study synapses. mcDETECT identifies individual synapses based on the aggregation of synaptic mRNAs in three-dimensional (3D) space, allowing for the construction of single-synapse spatial transcriptome profiles. By benchmarking the synapse density measured by volume electron microscopy and genetic labeling, we demonstrate that mcDETECT can faithfully and accurately recover the spatial location of single synapses using iST data from multiple platforms, including Xenium, Xenium 5K, MERSCOPE, and CosMx. Based on the subsequent transcriptome profiling, we further stratify total synapses into various subtypes and explore their pathogenic dysregulation associated with Alzheimer's disease (AD) progression, which provides potential targets for synapse-specific therapies in AD progression.
Collapse
|
33
|
Lok J, Harris JM, Carey I, Agarwal K, McKeating JA. Assessing the virological response to direct-acting antiviral therapies in the HBV cure programme. Virology 2025; 605:110458. [PMID: 40022943 DOI: 10.1016/j.virol.2025.110458] [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: 11/28/2024] [Revised: 01/16/2025] [Accepted: 02/20/2025] [Indexed: 03/04/2025]
Abstract
Hepatitis B virus (HBV) is a global health problem with over 250 million people affected worldwide. Nucleos(t)ide analogues remain the standard of care and suppress production of progeny virions; however, they have limited effect on the viral transcriptome and long-term treatment is associated with off-target toxicities. Promising results are emerging from clinical trials and several drug classes have been evaluated, including capsid assembly modulators and RNA interfering agents. Whilst peripheral biomarkers are used to monitor responses and define treatment endpoints, they fail to reflect the full reservoir of infected hepatocytes. Given these limitations, consideration should be given to the merits of sampling liver tissue, especially in the context of clinical trials. In this review article, we will discuss methods for profiling HBV in liver tissue and their value to the HBV cure programme.
Collapse
Affiliation(s)
- James Lok
- Institute of Liver Studies, King's College Hospital, London, SE5 9RS, United Kingdom.
| | - James M Harris
- Nuffield Department of Medicine, University of Oxford, OX3 7FZ, United Kingdom
| | - Ivana Carey
- Institute of Liver Studies, King's College Hospital, London, SE5 9RS, United Kingdom
| | - Kosh Agarwal
- Institute of Liver Studies, King's College Hospital, London, SE5 9RS, United Kingdom
| | - Jane A McKeating
- Nuffield Department of Medicine, University of Oxford, OX3 7FZ, United Kingdom; Chinese Academy of Medical Sciences Oxford Institute, University of Oxford, Oxford, United Kingdom
| |
Collapse
|
34
|
Min W, Fang D, Chen J, Zhang S. SpaMask: Dual masking graph autoencoder with contrastive learning for spatial transcriptomics. PLoS Comput Biol 2025; 21:e1012881. [PMID: 40179332 PMCID: PMC11968113 DOI: 10.1371/journal.pcbi.1012881] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2024] [Accepted: 02/17/2025] [Indexed: 04/05/2025] Open
Abstract
Understanding the spatial locations of cell within tissues is crucial for unraveling the organization of cellular diversity. Recent advancements in spatial resolved transcriptomics (SRT) have enabled the analysis of gene expression while preserving the spatial context within tissues. Spatial domain characterization is a critical first step in SRT data analysis, providing the foundation for subsequent analyses and insights into biological implications. Graph neural networks (GNNs) have emerged as a common tool for addressing this challenge due to the structural nature of SRT data. However, current graph-based deep learning approaches often overlook the instability caused by the high sparsity of SRT data. Masking mechanisms, as an effective self-supervised learning strategy, can enhance the robustness of these models. To this end, we propose SpaMask, dual masking graph autoencoder with contrastive learning for SRT analysis. Unlike previous GNNs, SpaMask masks a portion of spot nodes and spot-to-spot edges to enhance its performance and robustness. SpaMask combines Masked Graph Autoencoders (MGAE) and Masked Graph Contrastive Learning (MGCL) modules, with MGAE using node masking to leverage spatial neighbors for improved clustering accuracy, while MGCL applies edge masking to create a contrastive loss framework that tightens embeddings of adjacent nodes based on spatial proximity and feature similarity. We conducted a comprehensive evaluation of SpaMask on eight datasets from five different platforms. Compared to existing methods, SpaMask achieves superior clustering accuracy and effective batch correction.
Collapse
Affiliation(s)
- Wenwen Min
- School of Information Science and Engineering, Yunnan University, Kunming, Yunnan, China
| | - Donghai Fang
- School of Information Science and Engineering, Yunnan University, Kunming, Yunnan, China
| | - Jinyu Chen
- School of Mathematics, Statistics and Mechanics, Beijing University of Technology, Beijing, China
| | - Shihua Zhang
- NCMIS, CEMS, RCSDS, Academy of Mathematics and Systems Science, Chinese Academy of Sciences, Beijing, China
- School of Mathematical Sciences, University of Chinese Academy of Sciences, Beijing, China
- Key Laboratory of Systems Health Science of Zhejiang Province, School of Life Science, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou, Zhejiang, China
| |
Collapse
|
35
|
Bae S, Lee H, Na KJ, Lee DS, Choi H, Kim YT. STopover captures spatial colocalization and interaction in the tumor microenvironment using topological analysis in spatial transcriptomics data. Genome Med 2025; 17:33. [PMID: 40170080 PMCID: PMC11963362 DOI: 10.1186/s13073-025-01457-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2024] [Accepted: 03/11/2025] [Indexed: 04/03/2025] Open
Abstract
Unraveling the spatial configuration of the tumor microenvironment (TME) is crucial for elucidating tumor-immune interactions based on immuno-oncology. We present STopover, a novel approach utilizing spatially resolved transcriptomics (SRT) data and topological analysis to investigate the TME. By gradually lowering the feature threshold, connected components (CCs) are extracted based on spatial distance and persistence, with Jaccard indices quantifying their spatial overlap, and transcriptomic profiles are permutated to assess statistical significance. Applied to lung and breast cancer SRT, STopover revealed immune and stromal cell infiltration patterns, predicted key cell-cell communication, and identified relevant regions, shedding light on cancer pathophysiology (URL: https://github.com/bsungwoo/STopover ).
Collapse
Affiliation(s)
- Sungwoo Bae
- Institute of Radiation Medicine, Medical Research Center, Seoul National University, Seoul, Republic of Korea
- Portrai, Inc., Seoul, Republic of Korea
| | - Hyekyoung Lee
- Department of Nuclear Medicine, Seoul National University Hospital, Seoul, Republic of Korea
| | - Kwon Joong Na
- Portrai, Inc., Seoul, Republic of Korea
- Department of Thoracic and Cardiovascular Surgery, Seoul National University Hospital, 101 Daehak-Ro, Jongno-Gu, Seoul, Republic of Korea, 03080
| | - Dong Soo Lee
- Medical Science and Engineering, School of Convergence Science and Technology, POSTECH, Pohang, Republic of Korea
- Department of Nuclear Medicine, Seoul National University College of Medicine, Daehak-Ro, Jongno-Gu, 101, Seoul03080, , Republic of Korea
| | - Hongyoon Choi
- Portrai, Inc., Seoul, Republic of Korea.
- Department of Nuclear Medicine, Seoul National University Hospital, Seoul, Republic of Korea.
- Department of Nuclear Medicine, Seoul National University College of Medicine, Daehak-Ro, Jongno-Gu, 101, Seoul03080, , Republic of Korea.
| | - Young Tae Kim
- Department of Thoracic and Cardiovascular Surgery, Seoul National University Hospital, 101 Daehak-Ro, Jongno-Gu, Seoul, Republic of Korea, 03080.
| |
Collapse
|
36
|
Ding DY, Tang Z, Zhu B, Ren H, Shalek AK, Tibshirani R, Nolan GP. Quantitative characterization of tissue states using multiomics and ecological spatial analysis. Nat Genet 2025; 57:910-921. [PMID: 40169791 PMCID: PMC11985343 DOI: 10.1038/s41588-025-02119-z] [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: 06/24/2024] [Accepted: 02/05/2025] [Indexed: 04/03/2025]
Abstract
The spatial organization of cells in tissues underlies biological function, and recent advances in spatial profiling technologies have enhanced our ability to analyze such arrangements to study biological processes and disease progression. We propose MESA (multiomics and ecological spatial analysis), a framework drawing inspiration from ecological concepts to delineate functional and spatial shifts across tissue states. MESA introduces metrics to systematically quantify spatial diversity and identify hot spots, linking spatial patterns to phenotypic outcomes, including disease progression. Furthermore, MESA integrates spatial and single-cell multiomics data to facilitate an in-depth, molecular understanding of cellular neighborhoods and their spatial interactions within tissue microenvironments. Applying MESA to diverse datasets demonstrates additional insights it brings over prior methods, including newly identified spatial structures and key cell populations linked to disease states. Available as a Python package, MESA offers a versatile framework for quantitative decoding of tissue architectures in spatial omics across health and disease.
Collapse
Affiliation(s)
- Daisy Yi Ding
- Department of Biomedical Data Science, Stanford University, Stanford, CA, USA
| | - Zeyu Tang
- Weill Cornell Graduate School of Medical Sciences, Weill Cornell Medicine, New York, NY, USA
| | - Bokai Zhu
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Hongyu Ren
- Department of Computer Science, Stanford University, Stanford, CA, USA
| | - Alex K Shalek
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Institute for Medical Engineering and Science, MIT, Cambridge, MA, USA
- Department of Chemistry, MIT, Cambridge, MA, USA
- Koch Institute for Integrative Cancer Research, MIT, Cambridge, MA, USA
| | - Robert Tibshirani
- Department of Biomedical Data Science, Stanford University, Stanford, CA, USA
- Department of Statistics, Stanford University, Stanford, CA, USA
| | - Garry P Nolan
- Department of Pathology, Stanford University, Stanford, CA, USA.
| |
Collapse
|
37
|
Liao K, Yan B, Ding Z, Huang J, Fan X, Wu S, Chen C, Li H. X-scPAE: An explainable deep learning model for embryonic lineage allocation prediction based on single-cell transcriptomics revealing key genes in embryonic cell development. Comput Biol Med 2025; 188:109787. [PMID: 39946788 DOI: 10.1016/j.compbiomed.2025.109787] [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: 09/21/2024] [Revised: 01/08/2025] [Accepted: 01/30/2025] [Indexed: 03/05/2025]
Abstract
In single-cell transcriptomics research, accurately predicting cell lineage allocation and identifying differences between lineages are crucial for understanding cell differentiation processes and reducing early pregnancy miscarriages in humans. This paper introduces an explainable PCA-based deep learning attention autoencoder model, X-scPAE (eXplained Single Cell PCA - Attention Auto Encoder), which is built on the Counterfactual Gradient Attribution (CGA) algorithm. The model is designed to predict lineage allocation in human and mouse single-cell transcriptomic data, while identifying and interpreting gene expression differences across lineages to extract key genes. It first reduces dimensionality using Principal Component Analysis (PCA) and ranks the importance of principal components. An autoencoder is then employed for feature extraction, integrating an attention mechanism to capture interactions between features. Finally, the Counterfactual Gradient Attribution algorithm calculates the importance of each feature. The model achieved an accuracy of 0.945 on the test set and 0.977 on the validation set, with other metrics such as F1-score, Precision, and Recall all reaching 0.94. It significantly outperformed both baseline algorithms (XGBoost, SVM, RF, and LR) and advanced approaches like F-Score-SVM, CV2-LR, scChrBin, and TripletCell. Notably, the explainability analysis uncovered key lineage predictor genes for both humans and mice and identified crucial genes distinguishing between developmental stages and lineages. A logistic regression model built using the extracted key genes still achieved an AUROC of 0.92, surpassing the performance of other feature extraction methods, including F-Score, CV2, PCA, random feature selection, and the interpretability method Shapley. Lastly, ablation studies demonstrated the effectiveness of each model component.
Collapse
Affiliation(s)
- Kai Liao
- The Central Laboratory of Birth Defects Prevention and Control, The Affiliated Women and Children's Hospital of Ningbo University, Ningbo, 315021, China; Ningbo Key Laboratory for the Prevention and Treatment of Embryogenic Diseases, The Affiliated Women and Children's Hospital of Ningbo University, Ningbo, 315021, China; Ningbo Key Laboratory of Genomic Medicine and Birth Defects Prevention, The Affiliated Women and Children's Hospital of Ningbo University, Ningbo, 315021, China; MOE Engineering Research Center of Gene Technology, School of Life Sciences, Fudan University, Shanghai, 200433, China
| | - Bowei Yan
- Institutes of Biomedical Sciences, Fudan University, Shanghai, 200433, China.
| | - Ziyin Ding
- Center for Reproductive Medicine, The Affiliated Women and Children's Hospital of Ningbo University, Ningbo, 315021, China
| | - Jian Huang
- College of Computer Science, Chongqing University, Chongqing, 400044, China
| | - Xiaodan Fan
- Center for Reproductive Medicine, The Affiliated Women and Children's Hospital of Ningbo University, Ningbo, 315021, China
| | - Shanshan Wu
- Ningbo Key Laboratory of Genomic Medicine and Birth Defects Prevention, The Affiliated Women and Children's Hospital of Ningbo University, Ningbo, 315021, China
| | - Changshui Chen
- Ningbo Key Laboratory for the Prevention and Treatment of Embryogenic Diseases, The Affiliated Women and Children's Hospital of Ningbo University, Ningbo, 315021, China
| | - Haibo Li
- The Central Laboratory of Birth Defects Prevention and Control, The Affiliated Women and Children's Hospital of Ningbo University, Ningbo, 315021, China; Ningbo Key Laboratory for the Prevention and Treatment of Embryogenic Diseases, The Affiliated Women and Children's Hospital of Ningbo University, Ningbo, 315021, China; Ningbo Key Laboratory of Genomic Medicine and Birth Defects Prevention, The Affiliated Women and Children's Hospital of Ningbo University, Ningbo, 315021, China.
| |
Collapse
|
38
|
Cheng Y, Dang S, Zhang Y, Chen Y, Yu R, Liu M, Jin S, Han A, Katz S, Wang S. Sequencing-free whole genome spatial transcriptomics at molecular resolution in intact tissue. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.03.06.641951. [PMID: 40161724 PMCID: PMC11952344 DOI: 10.1101/2025.03.06.641951] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 04/02/2025]
Abstract
Recent breakthroughs in spatial transcriptomics technologies have enhanced our understanding of diverse cellular identities, compositions, interactions, spatial organizations, and functions. Yet existing spatial transcriptomics tools are still limited in either transcriptomic coverage or spatial resolution. Leading spatial-capture or spatial-tagging transcriptomics techniques that rely on in-vitro sequencing offer whole-transcriptome coverage, in principle, but at the cost of lower spatial resolution compared to image-based techniques. In contrast, high-performance image-based spatial transcriptomics techniques, which rely on in situ hybridization or in situ sequencing, achieve single-molecule spatial resolution and retain sub-cellular morphologies, but are limited by probe libraries that target only a subset of the transcriptome, typically covering several hundred to a few thousand transcript species. Together, these limitations hinder unbiased, hypothesis-free transcriptomic analyses at high spatial resolution. Here we develop a new image-based spatial transcriptomics technology termed Reverse-padlock Amplicon Encoding FISH (RAEFISH) with whole-genome level coverage while retaining single-molecule spatial resolution in intact tissues. We demonstrate image-based spatial transcriptomics targeting 23,000 human transcript species or 22,000 mouse transcript species, including nearly the entire protein-coding transcriptome and several thousand long-noncoding RNAs, in single cells in cultures and in tissue sections. Our analyses reveal differential subcellular localizations of diverse transcripts, cell-type-specific and cell-type-invariant tissue zonation dependent transcriptome, and gene expression programs underlying preferential cell-cell interactions. Finally, we further develop our technology for direct spatial readout of gRNAs in an image-based high-content CRISPR screen. Overall, these developments provide the research community with a broadly applicable technology that enables high-coverage, high-resolution spatial profiling of both long and short, native and engineered RNA species in many biomedical contexts.
Collapse
Affiliation(s)
- Yubao Cheng
- Department of Genetics, Yale University School of Medicine, New Haven, CT 06510, USA
| | - Shengyuan Dang
- Department of Genetics, Yale University School of Medicine, New Haven, CT 06510, USA
- These authors contributed equally to this work
| | - Yuan Zhang
- Department of Genetics, Yale University School of Medicine, New Haven, CT 06510, USA
- These authors contributed equally to this work
| | - Yanbo Chen
- Department of Genetics, Yale University School of Medicine, New Haven, CT 06510, USA
| | - Ruihuan Yu
- Department of Genetics, Yale University School of Medicine, New Haven, CT 06510, USA
- Present Address: Department of Biological Sciences, Columbia University, New York, NY 10027, USA
| | - Miao Liu
- Department of Genetics, Yale University School of Medicine, New Haven, CT 06510, USA
| | - Shengyan Jin
- Department of Genetics, Yale University School of Medicine, New Haven, CT 06510, USA
| | - Ailin Han
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT 06520, USA
| | - Samuel Katz
- Department of Pathology, Yale University School of Medicine, New Haven, CT 06520, USA
| | - Siyuan Wang
- Department of Genetics, Yale University School of Medicine, New Haven, CT 06510, USA
- M.D.-Ph.D. Program, Yale University, New Haven, CT 06510, USA
- Yale Combined Program in the Biological and Biomedical Sciences, Yale University, New Haven, CT 06510, USA
- Molecular Cell Biology, Genetics and Development Program, Yale University, New Haven, CT 06510, USA
- Department of Cell Biology, Yale University School of Medicine, New Haven, CT 06510, USA
- Biochemistry, Quantitative Biology, Biophysics, and Structural Biology Program, Yale University, New Haven, CT 06510, USA
- Yale Center for RNA Science and Medicine, Yale University School of Medicine, New Haven, CT 06510, USA
- Yale Liver Center, Yale University School of Medicine, New Haven, CT 06510, USA
- Lead contact
| |
Collapse
|
39
|
Chen X, Ran Q, Tang J, Chen Z, Huang S, Shi X, Xi R. Benchmarking algorithms for spatially variable gene identification in spatial transcriptomics. Bioinformatics 2025; 41:btaf131. [PMID: 40139667 PMCID: PMC12036962 DOI: 10.1093/bioinformatics/btaf131] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2024] [Revised: 03/15/2025] [Accepted: 03/27/2025] [Indexed: 03/29/2025] Open
Abstract
MOTIVATION The rapid development of spatial transcriptomics has underscored the importance of identifying spatially variable genes. As a fundamental task in spatial transcriptomic data analysis, spatially variable gene identification has been extensively studied. However, the lack of comprehensive benchmark makes it difficult to validate the effectiveness of various algorithms scattered across a large number of studies with real-world datasets. RESULTS In response, this article proposes a benchmark framework to evaluate algorithms for identifying spatially variable genes through the analysis of 30 synthesized and 74 real-world datasets, aiming to identify the best algorithms and their corresponding application scenarios. This framework can assist medical and life scientists in selecting suitable algorithms for their research, while also aid bioinformatics scientists in developing more powerful and efficient computational methods in spatial transcriptomic research. AVAILABILITY AND IMPLEMENTATION The source code of this benchmarking framework is available at both Github (https://github.com/XiDsLab/svg-benchmark) and Zenodo (https://doi.org/10.5281/zenodo.15031083). In addition, all real and synthetic datasets considered in this study are also publicly available at Zenodo (https://doi.org/10.5281/zenodo.7227771).
Collapse
Affiliation(s)
- Xuanwei Chen
- School of Mathematical Sciences, Peking University, Beijing 100871, China
| | - Qinghua Ran
- Academy for Advanced Interdisciplinary Studies, Peking University, Beijing 100871, China
| | - Junjie Tang
- Center for Statistical Science, Peking University, Beijing 100871, China
| | - Zihao Chen
- School of Mathematical Sciences, Peking University, Beijing 100871, China
| | - Siyuan Huang
- Academy for Advanced Interdisciplinary Studies, Peking University, Beijing 100871, China
| | - Xingjie Shi
- KLATASDS-MOE, Academy of Statistics and Interdisciplinary Sciences, School of Statistics, East China Normal University, Shanghai 200062, China
| | - Ruibin Xi
- School of Mathematical Sciences, Peking University, Beijing 100871, China
- Academy for Advanced Interdisciplinary Studies, Peking University, Beijing 100871, China
- Center for Statistical Science, Peking University, Beijing 100871, China
| |
Collapse
|
40
|
Du MRM, Wang C, Law CW, Amann-Zalcenstein D, Anttila CJA, Ling L, Hickey PF, Sargeant CJ, Chen Y, Ioannidis LJ, Rajasekhar P, Yip RKH, Rogers KL, Hansen DS, Bowden R, Ritchie ME. Benchmarking spatial transcriptomics technologies with the multi-sample SpatialBenchVisium dataset. Genome Biol 2025; 26:77. [PMID: 40156041 PMCID: PMC11954323 DOI: 10.1186/s13059-025-03543-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2024] [Accepted: 03/13/2025] [Indexed: 04/01/2025] Open
Abstract
BACKGROUND Spatial transcriptomics allows gene expression to be measured within complex tissue contexts. Among the array of spatial capture technologies available is 10x Genomics' Visium platform, a popular method which enables transcriptome-wide profiling of tissue sections. Visium offers a range of sample handling and library construction methods which introduces a need for benchmarking to compare data quality and assess how well the technology can recover expected tissue features and biological signatures. RESULTS Here we present SpatialBenchVisium, a unique reference dataset generated from spleen tissue of mice responding to malaria infection spanning several tissue preparation protocols (both fresh frozen and FFPE, with either manual or CytAssist tissue placement). We note better quality control metrics in reference samples prepared using probe-based capture methods, particularly those processed with CytAssist, validating the improvement in data quality produced with the platform. Our analysis of replicate samples extends to explore spatially variable gene detection, the outcomes of clustering and cell deconvolution using matched single-cell RNA-sequencing data and publicly available reference data to identify cell types and tissue regions expected in the spleen. Multi-sample differential expression analysis recovered known gene signatures related to biological sex or gene knockout.
Collapse
Affiliation(s)
- Mei R M Du
- The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, 3052, Australia
| | - Changqing Wang
- The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, 3052, Australia
- Department of Medical Biology, The University of Melbourne, Parkville, VIC, 3010, Australia
| | - Charity W Law
- The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, 3052, Australia
- Department of Medical Biology, The University of Melbourne, Parkville, VIC, 3010, Australia
| | - Daniela Amann-Zalcenstein
- The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, 3052, Australia
- Department of Medical Biology, The University of Melbourne, Parkville, VIC, 3010, Australia
| | - Casey J A Anttila
- The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, 3052, Australia
| | - Ling Ling
- The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, 3052, Australia
| | - Peter F Hickey
- The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, 3052, Australia
- Department of Medical Biology, The University of Melbourne, Parkville, VIC, 3010, Australia
| | - Callum J Sargeant
- The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, 3052, Australia
| | - Yunshun Chen
- The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, 3052, Australia
- Department of Medical Biology, The University of Melbourne, Parkville, VIC, 3010, Australia
| | - Lisa J Ioannidis
- The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, 3052, Australia
- Department of Medical Biology, The University of Melbourne, Parkville, VIC, 3010, Australia
| | - Pradeep Rajasekhar
- The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, 3052, Australia
- Department of Medical Biology, The University of Melbourne, Parkville, VIC, 3010, Australia
| | - Raymond K H Yip
- The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, 3052, Australia
- Department of Medical Biology, The University of Melbourne, Parkville, VIC, 3010, Australia
| | - Kelly L Rogers
- The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, 3052, Australia
- Department of Medical Biology, The University of Melbourne, Parkville, VIC, 3010, Australia
| | - Diana S Hansen
- Department of Microbiology, Monash Biomedicine Discovery Institute, Monash University, Clayton, VIC, 3800, Australia
| | - Rory Bowden
- The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, 3052, Australia
- Department of Medical Biology, The University of Melbourne, Parkville, VIC, 3010, Australia
| | - Matthew E Ritchie
- The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, 3052, Australia.
- Department of Medical Biology, The University of Melbourne, Parkville, VIC, 3010, Australia.
| |
Collapse
|
41
|
Qian N, Weinstein JA. Spatial transcriptomic imaging of an intact organism using volumetric DNA microscopy. Nat Biotechnol 2025:10.1038/s41587-025-02613-z. [PMID: 40148595 DOI: 10.1038/s41587-025-02613-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2023] [Revised: 02/21/2025] [Accepted: 02/24/2025] [Indexed: 03/29/2025]
Abstract
Lymphatic, nervous and tumor tissues exhibit complex physiology arising from three-dimensional interactions within genetically unique microenvironments. Here we develop a technology capable of volumetrically imaging transcriptomes, genotypes and morphologies in a single measurement, without relying on prior knowledge of spatial organization or genetic sequences. Our method extends DNA microscopy into three dimensions at scales involving 107 molecules by forming a distributed intermolecular network of proximal unique DNA barcodes tagging complementary DNA molecules inside the specimen. After sequencing the DNA-encoded network, an image of molecular positions is inferred using geodesic spectral embeddings, a dimensionality reduction approach that we show to be especially suitable for this data-inverse problem. Applying whole-transcriptome volumetric DNA microscopy to intact zebrafish embryos, we demonstrate that three-dimensional image inference recapitulates zebrafish morphology and known gene expression patterns, capturing the spatial organization of gene sequences. Our extension of spatial genetic measurements to three dimensions, independent of prior templates, opens the door to detailed joint resolution of genomics and morphology in biological tissues.
Collapse
Affiliation(s)
- Nianchao Qian
- Department of Medicine, Section of Genetic Medicine, University of Chicago, Chicago, IL, USA
| | - Joshua A Weinstein
- Department of Medicine, Section of Genetic Medicine, University of Chicago, Chicago, IL, USA.
- Pritzker School of Molecular Engineering, University of Chicago, Chicago, IL, USA.
| |
Collapse
|
42
|
Jana S, Glabman RA, Koehne AL. Bridging the gap between histopathology and genomics: Spotlighting spatial omics. Vet Pathol 2025:3009858251322729. [PMID: 40138497 DOI: 10.1177/03009858251322729] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/29/2025]
Abstract
Spatial biology has emerged as a transformative field, offering insights into cellular interactions and organization within tissues. The field has evolved rapidly since the coining of the term "spatial omics." Now, the ability to spatially resolve proteins, RNA, chromatin, and lipids is becoming widespread, and the technologies are continually refined. Reagents to support the analysis of veterinary species are available and more are emerging. These new tools will allow pathologists and scientists to unravel the intricate interplay between tissue architecture and diverse cellular phenotypes. By integrating histological observations with spatially resolved genomic data, spatial biology holds immense potential for advancing diagnostic and therapeutic strategies in veterinary medicine. These tools will undoubtedly equip veterinary pathologists to better decipher complex disease processes and identify novel therapeutic targets.
Collapse
|
43
|
Al-Mansour FSH, Almasoudi HH, Albarrati A. Mapping molecular landscapes in triple-negative breast cancer: insights from spatial transcriptomics. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2025:10.1007/s00210-025-04057-3. [PMID: 40119898 DOI: 10.1007/s00210-025-04057-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/05/2024] [Accepted: 03/13/2025] [Indexed: 03/25/2025]
Abstract
The tumor microenvironment (TME) of triple-negative breast cancer (TNBC) is a highly heterogeneous and very aggressive form of the disease that has few suitable treatment options; however, spatial transcriptomics (ST) is a powerful tool for elucidation of the TME in TNBC. Because of its spatial context preservation, ST has a unique capability to map tumor-stroma interactions, immune infiltration, and therapy resistance mechanisms (which are key to understanding TNBC progression), compared with conventional transcriptomics. This review shows the use of ST in TNBC, its utilization in spatial biomarker identification, intratumoral heterogeneity definition, molecular subtyping refinement, and prediction of immunotherapy responses. Recent insight from ST-driven insights has explained the key spatial patterns on immune evasion, chemotherapy resistance, racial disparities of TNBC, and aspects for patient stratification and therapeutic decision. With the integration of ST with the subjects of proteomics and imaging mass cytometry, this approach has been enlarged and is now applied in precision medicine and biomarker discovery. Recently, advancements in AI-based spatial analysis for tumor classification, identification of biomarkers, and creation of therapy prediction models have occurred. However, continued developments in ST technologies, computational tools, and partnerships amongst multiple centers to facilitate the integration of ST into clinical routine practice are needed to unlock novel therapeutic targets.
Collapse
Affiliation(s)
- Fares Saeed H Al-Mansour
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, Najran University, Najran, Saudi Arabia
| | - Hassan H Almasoudi
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, Najran University, Najran, Saudi Arabia
| | - Ali Albarrati
- Rehabilitation Sciences Department, College of Applied Medical Sciences, King Saud University, 11451, Riyadh, Saudi Arabia.
| |
Collapse
|
44
|
Han L, Liu Z, Jing Z, Liu Y, Peng Y, Chang H, Lei J, Wang K, Xu Y, Liu W, Wu Z, Li Q, Shi X, Zheng M, Wang H, Deng J, Zhong Y, Pan H, Lin J, Zhang R, Chen Y, Wu J, Xu M, Ren B, Cheng M, Yu Q, Song X, Lu Y, Tang Y, Yuan N, Sun S, An Y, Ding W, Sun X, Wei Y, Zhang S, Dou Y, Zhao Y, Han L, Zhu Q, Xu J, Wang S, Wang D, Bai Y, Liang Y, Liu Y, Chen M, Xie C, Bo B, Li M, Zhang X, Ting W, Chen Z, Fang J, Li S, Jiang Y, Tan X, Zuo G, Xie Y, Li H, Tao Q, Li Y, Liu J, Liu Y, Hao M, Wang J, Wen H, Liu J, Yan Y, Zhang H, Sheng Y, Yu S, Liao X, Jiang X, Wang G, Liu H, Wang C, Feng N, Liu X, Ma K, Xu X, Han T, Cao H, Zheng H, Chen Y, Lu H, Yu Z, Zhang J, Wang B, Wang Z, Xie Q, Pan S, Liu C, Xu C, Cui L, Li Y, Liu S, Liao S, Chen A, Wu QF, et alHan L, Liu Z, Jing Z, Liu Y, Peng Y, Chang H, Lei J, Wang K, Xu Y, Liu W, Wu Z, Li Q, Shi X, Zheng M, Wang H, Deng J, Zhong Y, Pan H, Lin J, Zhang R, Chen Y, Wu J, Xu M, Ren B, Cheng M, Yu Q, Song X, Lu Y, Tang Y, Yuan N, Sun S, An Y, Ding W, Sun X, Wei Y, Zhang S, Dou Y, Zhao Y, Han L, Zhu Q, Xu J, Wang S, Wang D, Bai Y, Liang Y, Liu Y, Chen M, Xie C, Bo B, Li M, Zhang X, Ting W, Chen Z, Fang J, Li S, Jiang Y, Tan X, Zuo G, Xie Y, Li H, Tao Q, Li Y, Liu J, Liu Y, Hao M, Wang J, Wen H, Liu J, Yan Y, Zhang H, Sheng Y, Yu S, Liao X, Jiang X, Wang G, Liu H, Wang C, Feng N, Liu X, Ma K, Xu X, Han T, Cao H, Zheng H, Chen Y, Lu H, Yu Z, Zhang J, Wang B, Wang Z, Xie Q, Pan S, Liu C, Xu C, Cui L, Li Y, Liu S, Liao S, Chen A, Wu QF, Wang J, Liu Z, Sun Y, Mulder J, Yang H, Wang X, Li C, Yao J, Xu X, Liu L, Shen Z, Wei W, Sun YG. Single-cell spatial transcriptomic atlas of the whole mouse brain. Neuron 2025:S0896-6273(25)00133-3. [PMID: 40132589 DOI: 10.1016/j.neuron.2025.02.015] [Show More Authors] [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: 04/22/2024] [Revised: 10/24/2024] [Accepted: 02/14/2025] [Indexed: 03/27/2025]
Abstract
A comprehensive atlas of genes, cell types, and their spatial distribution across a whole mammalian brain is fundamental for understanding the function of the brain. Here, using single-nucleus RNA sequencing (snRNA-seq) and Stereo-seq techniques, we generated a mouse brain atlas with spatial information for 308 cell clusters at single-cell resolution, involving over 4 million cells, as well as for 29,655 genes. We have identified cell clusters exhibiting preference for cortical subregions and explored their associations with brain-related diseases. Additionally, we pinpointed 155 genes with distinct regional expression patterns within the brainstem and unveiled 513 long non-coding RNAs showing region-enriched expression in the adult brain. Parcellation of brain regions based on spatial transcriptomic information revealed fine structure for several brain areas. Furthermore, we have uncovered 411 transcription factor regulons showing distinct spatiotemporal dynamics during neurodevelopment. Thus, we have constructed a single-cell-resolution spatial transcriptomic atlas of the mouse brain with genome-wide coverage.
Collapse
Affiliation(s)
- Lei Han
- BGI Research, Hangzhou 310030, China
| | - Zhen Liu
- Lingang Laboratory, Shanghai 200031, China; CAS Key Laboratory of Computational Biology, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Zehua Jing
- BGI Research, Hangzhou 310030, China; College of Life Sciences, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Yuxuan Liu
- CAS Key Laboratory of Computational Biology, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | | | | | - Junjie Lei
- BGI Research, Hangzhou 310030, China; College of Life Sciences, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Kexin Wang
- Institute of Neuroscience, State Key Laboratory of Brain Cognition and Brain-inspired Intelligence Technology, CAS Center for Excellence in Brain Science and Intelligence Technology, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Yuanfang Xu
- Institute of Neuroscience, State Key Laboratory of Brain Cognition and Brain-inspired Intelligence Technology, CAS Center for Excellence in Brain Science and Intelligence Technology, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Wei Liu
- Lingang Laboratory, Shanghai 200031, China
| | - Zihan Wu
- Tencent AI Lab, Shenzhen 518057, China
| | - Qian Li
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing 100049, China; BGI Research, Shenzhen 518083, China
| | - Xiaoxue Shi
- Institute of Neuroscience, State Key Laboratory of Brain Cognition and Brain-inspired Intelligence Technology, CAS Center for Excellence in Brain Science and Intelligence Technology, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Mingyuan Zheng
- Institute of Neuroscience, State Key Laboratory of Brain Cognition and Brain-inspired Intelligence Technology, CAS Center for Excellence in Brain Science and Intelligence Technology, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - He Wang
- Institute of Neuroscience, State Key Laboratory of Brain Cognition and Brain-inspired Intelligence Technology, CAS Center for Excellence in Brain Science and Intelligence Technology, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Juan Deng
- Department of Anesthesiology, Huashan Hospital, State Key Laboratory of Medical Neurobiology, Institute for Translational Brain Research, MOE Frontiers Center for Brain Science, Fudan University, Shanghai 200032, China
| | - Yanqing Zhong
- Institute of Neuroscience, State Key Laboratory of Brain Cognition and Brain-inspired Intelligence Technology, CAS Center for Excellence in Brain Science and Intelligence Technology, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | | | - Junkai Lin
- Institute of Neuroscience, State Key Laboratory of Brain Cognition and Brain-inspired Intelligence Technology, CAS Center for Excellence in Brain Science and Intelligence Technology, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Ruiyi Zhang
- Institute of Neuroscience, State Key Laboratory of Brain Cognition and Brain-inspired Intelligence Technology, CAS Center for Excellence in Brain Science and Intelligence Technology, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Yu Chen
- Institute of Neuroscience, State Key Laboratory of Brain Cognition and Brain-inspired Intelligence Technology, CAS Center for Excellence in Brain Science and Intelligence Technology, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Jinhua Wu
- Lingang Laboratory, Shanghai 200031, China
| | - Mingrui Xu
- State Key Laboratory of Molecular Development Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China
| | - Biyu Ren
- Institute of Neuroscience, State Key Laboratory of Brain Cognition and Brain-inspired Intelligence Technology, CAS Center for Excellence in Brain Science and Intelligence Technology, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | | | - Qian Yu
- Institute of Neuroscience, State Key Laboratory of Brain Cognition and Brain-inspired Intelligence Technology, CAS Center for Excellence in Brain Science and Intelligence Technology, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Xinxiang Song
- Institute of Neuroscience, State Key Laboratory of Brain Cognition and Brain-inspired Intelligence Technology, CAS Center for Excellence in Brain Science and Intelligence Technology, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Yanbing Lu
- Institute of Neuroscience, State Key Laboratory of Brain Cognition and Brain-inspired Intelligence Technology, CAS Center for Excellence in Brain Science and Intelligence Technology, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Yuanchun Tang
- BGI Research, Hangzhou 310030, China; BGI College & Henan Institute of Medical and Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450000, China
| | - Nini Yuan
- Institute of Neuroscience, State Key Laboratory of Brain Cognition and Brain-inspired Intelligence Technology, CAS Center for Excellence in Brain Science and Intelligence Technology, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Suhong Sun
- Institute of Neuroscience, State Key Laboratory of Brain Cognition and Brain-inspired Intelligence Technology, CAS Center for Excellence in Brain Science and Intelligence Technology, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Yingjie An
- Institute of Neuroscience, State Key Laboratory of Brain Cognition and Brain-inspired Intelligence Technology, CAS Center for Excellence in Brain Science and Intelligence Technology, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Wenqun Ding
- Institute of Neuroscience, State Key Laboratory of Brain Cognition and Brain-inspired Intelligence Technology, CAS Center for Excellence in Brain Science and Intelligence Technology, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Xing Sun
- Lingang Laboratory, Shanghai 200031, China; Institute of Neuroscience, State Key Laboratory of Brain Cognition and Brain-inspired Intelligence Technology, CAS Center for Excellence in Brain Science and Intelligence Technology, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Yanrong Wei
- BGI Research, Hangzhou 310030, China; College of Life Sciences, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Shuzhen Zhang
- Institute of Neuroscience, State Key Laboratory of Brain Cognition and Brain-inspired Intelligence Technology, CAS Center for Excellence in Brain Science and Intelligence Technology, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Yannong Dou
- Institute of Neuroscience, State Key Laboratory of Brain Cognition and Brain-inspired Intelligence Technology, CAS Center for Excellence in Brain Science and Intelligence Technology, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Yun Zhao
- Institute of Neuroscience, State Key Laboratory of Brain Cognition and Brain-inspired Intelligence Technology, CAS Center for Excellence in Brain Science and Intelligence Technology, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Luyao Han
- Institute of Neuroscience, State Key Laboratory of Brain Cognition and Brain-inspired Intelligence Technology, CAS Center for Excellence in Brain Science and Intelligence Technology, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | | | - Junfeng Xu
- Institute of Neuroscience, State Key Laboratory of Brain Cognition and Brain-inspired Intelligence Technology, CAS Center for Excellence in Brain Science and Intelligence Technology, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Shiwen Wang
- Institute of Neuroscience, State Key Laboratory of Brain Cognition and Brain-inspired Intelligence Technology, CAS Center for Excellence in Brain Science and Intelligence Technology, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Dan Wang
- Institute of Neuroscience, State Key Laboratory of Brain Cognition and Brain-inspired Intelligence Technology, CAS Center for Excellence in Brain Science and Intelligence Technology, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Yinqi Bai
- BGI Research, Hangzhou 310030, China
| | - Yikai Liang
- Institute of Neuroscience, State Key Laboratory of Brain Cognition and Brain-inspired Intelligence Technology, CAS Center for Excellence in Brain Science and Intelligence Technology, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Yuan Liu
- Institute of Neuroscience, State Key Laboratory of Brain Cognition and Brain-inspired Intelligence Technology, CAS Center for Excellence in Brain Science and Intelligence Technology, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Mengni Chen
- Institute of Neuroscience, State Key Laboratory of Brain Cognition and Brain-inspired Intelligence Technology, CAS Center for Excellence in Brain Science and Intelligence Technology, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Chun Xie
- Institute of Neuroscience, State Key Laboratory of Brain Cognition and Brain-inspired Intelligence Technology, CAS Center for Excellence in Brain Science and Intelligence Technology, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Binshi Bo
- Institute of Neuroscience, State Key Laboratory of Brain Cognition and Brain-inspired Intelligence Technology, CAS Center for Excellence in Brain Science and Intelligence Technology, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Mei Li
- BGI Research, Shenzhen 518083, China
| | - Xinyan Zhang
- Institute of Neuroscience, State Key Laboratory of Brain Cognition and Brain-inspired Intelligence Technology, CAS Center for Excellence in Brain Science and Intelligence Technology, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Wang Ting
- State Key Laboratory of Molecular Development Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China
| | - Zhenhua Chen
- State Key Laboratory of Molecular Development Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China
| | - Jiao Fang
- Institute of Neuroscience, State Key Laboratory of Brain Cognition and Brain-inspired Intelligence Technology, CAS Center for Excellence in Brain Science and Intelligence Technology, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Shuting Li
- Institute of Neuroscience, State Key Laboratory of Brain Cognition and Brain-inspired Intelligence Technology, CAS Center for Excellence in Brain Science and Intelligence Technology, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | | | - Xing Tan
- Institute of Neuroscience, State Key Laboratory of Brain Cognition and Brain-inspired Intelligence Technology, CAS Center for Excellence in Brain Science and Intelligence Technology, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Guolong Zuo
- Institute of Neuroscience, State Key Laboratory of Brain Cognition and Brain-inspired Intelligence Technology, CAS Center for Excellence in Brain Science and Intelligence Technology, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Yue Xie
- BGI Research, Shenzhen 518083, China
| | - Huanhuan Li
- Institute of Neuroscience, State Key Laboratory of Brain Cognition and Brain-inspired Intelligence Technology, CAS Center for Excellence in Brain Science and Intelligence Technology, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Quyuan Tao
- BGI Research, Hangzhou 310030, China; College of Life Sciences, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Yan Li
- Institute of Neuroscience, State Key Laboratory of Brain Cognition and Brain-inspired Intelligence Technology, CAS Center for Excellence in Brain Science and Intelligence Technology, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Jianfeng Liu
- Institute of Neuroscience, State Key Laboratory of Brain Cognition and Brain-inspired Intelligence Technology, CAS Center for Excellence in Brain Science and Intelligence Technology, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Yuyang Liu
- BGI Research, Hangzhou 310030, China; College of Life Sciences, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Mingkun Hao
- Lingang Laboratory, Shanghai 200031, China; Institute of Neuroscience, State Key Laboratory of Brain Cognition and Brain-inspired Intelligence Technology, CAS Center for Excellence in Brain Science and Intelligence Technology, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Jingjing Wang
- State Key Laboratory of Molecular Development Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China
| | - Huiying Wen
- BGI Research, Hangzhou 310030, China; School of Biology and Biological Engineering, South China University of Technology, Guangzhou 510006, China
| | - Jiabing Liu
- Institute of Neuroscience, State Key Laboratory of Brain Cognition and Brain-inspired Intelligence Technology, CAS Center for Excellence in Brain Science and Intelligence Technology, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | | | - Hui Zhang
- Institute of Neuroscience, State Key Laboratory of Brain Cognition and Brain-inspired Intelligence Technology, CAS Center for Excellence in Brain Science and Intelligence Technology, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Yifan Sheng
- Lingang Laboratory, Shanghai 200031, China; Institute of Neuroscience, State Key Laboratory of Brain Cognition and Brain-inspired Intelligence Technology, CAS Center for Excellence in Brain Science and Intelligence Technology, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Shui Yu
- Institute of Neuroscience, State Key Laboratory of Brain Cognition and Brain-inspired Intelligence Technology, CAS Center for Excellence in Brain Science and Intelligence Technology, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | | | - Xuyin Jiang
- Institute of Neuroscience, State Key Laboratory of Brain Cognition and Brain-inspired Intelligence Technology, CAS Center for Excellence in Brain Science and Intelligence Technology, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Guangling Wang
- Institute of Neuroscience, State Key Laboratory of Brain Cognition and Brain-inspired Intelligence Technology, CAS Center for Excellence in Brain Science and Intelligence Technology, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | | | - Congcong Wang
- Lingang Laboratory, Shanghai 200031, China; Institute of Neuroscience, State Key Laboratory of Brain Cognition and Brain-inspired Intelligence Technology, CAS Center for Excellence in Brain Science and Intelligence Technology, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Ning Feng
- BGI Research, Shenzhen 518083, China
| | - Xin Liu
- Institute of Neuroscience, State Key Laboratory of Brain Cognition and Brain-inspired Intelligence Technology, CAS Center for Excellence in Brain Science and Intelligence Technology, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | | | - Xiangjie Xu
- Institute of Neuroscience, State Key Laboratory of Brain Cognition and Brain-inspired Intelligence Technology, CAS Center for Excellence in Brain Science and Intelligence Technology, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | | | - Huateng Cao
- Institute of Neuroscience, State Key Laboratory of Brain Cognition and Brain-inspired Intelligence Technology, CAS Center for Excellence in Brain Science and Intelligence Technology, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Huiwen Zheng
- BGI Research, Hangzhou 310030, China; College of Life Sciences, University of Chinese Academy of Sciences, Beijing 100049, China
| | | | - Haorong Lu
- China National GeneBank, BGI Research, Shenzhen 518120, China
| | - Zixian Yu
- Institute of Neuroscience, State Key Laboratory of Brain Cognition and Brain-inspired Intelligence Technology, CAS Center for Excellence in Brain Science and Intelligence Technology, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | | | - Bo Wang
- China National GeneBank, BGI Research, Shenzhen 518120, China
| | | | - Qing Xie
- BGI Research, Shenzhen 518083, China
| | | | - Chuanyu Liu
- BGI Research, Shenzhen 518083, China; Shenzhen Proof-of-Concept Center of Digital Cytopathology, BGI Research, Shenzhen 518083, China; Shanxi Medical University-BGI Collaborative Center for Future Medicine, Shanxi Medical University, Taiyuan 030001, China
| | - Chan Xu
- BGI Research, Qingdao 266555, China
| | - Luman Cui
- BGI Research, Shenzhen 518083, China
| | - Yuxiang Li
- BGI Research, Shenzhen 518083, China; BGI Research, Wuhan 430074, China
| | - Shiping Liu
- BGI Research, Hangzhou 310030, China; BGI Research, Shenzhen 518083, China
| | - Sha Liao
- BGI Research, Shenzhen 518083, China; BGI Research, Chongqing 401329, China; JFL-BGI STOmics Center, Jinfeng Laboratory, Chongqing 401329, China
| | - Ao Chen
- BGI Research, Shenzhen 518083, China; BGI Research, Chongqing 401329, China; JFL-BGI STOmics Center, Jinfeng Laboratory, Chongqing 401329, China; Department of Biology, University of Copenhagen, Copenhagen 2200, Denmark
| | - Qing-Feng Wu
- State Key Laboratory of Molecular Development Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China
| | - Jian Wang
- BGI Research, Shenzhen 518083, China; China National GeneBank, BGI Research, Shenzhen 518120, China
| | - Zhiyong Liu
- Institute of Neuroscience, State Key Laboratory of Brain Cognition and Brain-inspired Intelligence Technology, CAS Center for Excellence in Brain Science and Intelligence Technology, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China; Shanghai Center for Brain Science and Brain-Inspired Technology, Shanghai 201602, China
| | - Yidi Sun
- Institute of Neuroscience, State Key Laboratory of Brain Cognition and Brain-inspired Intelligence Technology, CAS Center for Excellence in Brain Science and Intelligence Technology, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Jan Mulder
- Department of Protein Science, Science for Life Laboratory, KTH-Royal Institute of Technology, Stockholm 17121, Sweden; Department of Neuroscience, Karolinska Institute, Stockholm 17177, Sweden
| | | | - Xiaofei Wang
- Institute of Neuroscience, State Key Laboratory of Brain Cognition and Brain-inspired Intelligence Technology, CAS Center for Excellence in Brain Science and Intelligence Technology, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China.
| | - Chao Li
- Institute of Neuroscience, State Key Laboratory of Brain Cognition and Brain-inspired Intelligence Technology, CAS Center for Excellence in Brain Science and Intelligence Technology, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China.
| | | | - Xun Xu
- BGI Research, Shenzhen 518083, China; Shanxi Medical University-BGI Collaborative Center for Future Medicine, Shanxi Medical University, Taiyuan 030001, China; Guangdong Provincial Key Laboratory of Genome Read and Write, BGI Research, Shenzhen 518083, China.
| | - Longqi Liu
- BGI Research, Hangzhou 310030, China; BGI Research, Shenzhen 518083, China; Shanxi Medical University-BGI Collaborative Center for Future Medicine, Shanxi Medical University, Taiyuan 030001, China.
| | - Zhiming Shen
- Institute of Neuroscience, State Key Laboratory of Brain Cognition and Brain-inspired Intelligence Technology, CAS Center for Excellence in Brain Science and Intelligence Technology, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China; Shanghai Center for Brain Science and Brain-Inspired Technology, Shanghai 201602, China.
| | - Wu Wei
- Lingang Laboratory, Shanghai 200031, China; CAS Key Laboratory of Computational Biology, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China.
| | - Yan-Gang Sun
- Institute of Neuroscience, State Key Laboratory of Brain Cognition and Brain-inspired Intelligence Technology, CAS Center for Excellence in Brain Science and Intelligence Technology, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China.
| |
Collapse
|
45
|
Chowdhury S, Kamatkar NG, Wang WX, Akerele CA, Huang J, Wu J, Nwankpa A, Kane CM, Bhave VM, Huang H, Wang X, Nectow AR. Brainstem neuropeptidergic neurons link a neurohumoral axis to satiation. Cell 2025; 188:1563-1579.e18. [PMID: 39914383 DOI: 10.1016/j.cell.2025.01.018] [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: 02/16/2024] [Revised: 12/03/2024] [Accepted: 01/10/2025] [Indexed: 03/23/2025]
Abstract
Hunger is evolutionarily hardwired to ensure that an animal has sufficient energy to survive and reproduce. Just as important as knowing when to start eating is knowing when to stop eating. Here, using spatially resolved single-cell phenotyping, we characterize a population of neuropeptidergic neurons in the brainstem's dorsal raphe nucleus (DRN) and describe how they regulate satiation. These neurons track food from sensory presentation through ingestion, integrate these signals with slower-acting humoral cues, and express cholecystokinin (CCK). These CCK neurons bidirectionally regulate meal size, driving a sustained meal termination signal with a built-in delay. They are also well positioned to sense and respond to ingestion: they express a host of metabolic signaling factors and are integrated into an extended network known to regulate feeding. Together, this work demonstrates how DRN CCK neurons regulate satiation and identifies a likely conserved cellular mechanism that transforms diverse neurohumoral signals into a key behavioral output.
Collapse
Affiliation(s)
| | | | - Wendy Xueyi Wang
- Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA; Department of Chemistry, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; John A. Paulson School of Engineering and Applied Sciences, Harvard University, Boston, MA 02134, USA
| | - Christa A Akerele
- Department of Medicine, Columbia University, New York, NY 10032, USA
| | - Jiahao Huang
- Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA; Department of Chemistry, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Junlin Wu
- Department of Medicine, Columbia University, New York, NY 10032, USA
| | - Amajindi Nwankpa
- Department of Medicine, Columbia University, New York, NY 10032, USA
| | - Charlotte M Kane
- Department of Medicine, Columbia University, New York, NY 10032, USA
| | - Varun M Bhave
- Department of Medicine, Columbia University, New York, NY 10032, USA
| | - Hao Huang
- Princeton Neuroscience Institute, Princeton University, Princeton, NJ 08540, USA
| | - Xiao Wang
- Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA; Department of Chemistry, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Alexander R Nectow
- Department of Medicine, Columbia University, New York, NY 10032, USA; Division of Cardiology, Columbia University, New York, NY 10032, USA.
| |
Collapse
|
46
|
Zhao PA, Li R, Adewunmi T, Garber J, Gustafson C, Kim J, Malone J, Savage A, Skene P, Li XJ. SPARROW reveals microenvironment-zone-specific cell states in healthy and diseased tissues. Cell Syst 2025; 16:101235. [PMID: 40112778 DOI: 10.1016/j.cels.2025.101235] [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: 04/05/2024] [Revised: 10/23/2024] [Accepted: 02/19/2025] [Indexed: 03/22/2025]
Abstract
Spatially resolved transcriptomics technologies have advanced our understanding of cellular characteristics within tissue contexts. However, current analytical tools often treat cell-type inference and cellular neighborhood identification as separate and hard clustering processes, limiting comparability across scales and samples. SPARROW addresses these challenges by jointly learning latent embeddings and soft clusterings of cell types and cellular organization. It outperformed state-of-the-art methods in cell-type inference and microenvironment zone delineation and uncovered zone-specific cell states in human and mouse tissues that competing methods missed. By integrating spatially resolved transcriptomics and single-cell RNA sequencing (scRNA-seq) data in a shared latent space, SPARROW achieves single-cell spatial resolution and whole-transcriptome coverage, enabling the discovery of both established and unknown microenvironment zone-specific ligand-receptor interactions in the human tonsil. Overall, SPARROW is a computational framework that provides a comprehensive characterization of tissue features across scales, samples, and conditions.
Collapse
Affiliation(s)
- Peiyao A Zhao
- Allen Institute for Immunology, Seattle, WA 98109, USA.
| | - Ruoxin Li
- Allen Institute for Immunology, Seattle, WA 98109, USA
| | - Temi Adewunmi
- Allen Institute for Immunology, Seattle, WA 98109, USA
| | | | | | - June Kim
- Allen Institute for Immunology, Seattle, WA 98109, USA
| | | | - Adam Savage
- Allen Institute for Immunology, Seattle, WA 98109, USA
| | - Peter Skene
- Allen Institute for Immunology, Seattle, WA 98109, USA
| | - Xiao-Jun Li
- Allen Institute for Immunology, Seattle, WA 98109, USA.
| |
Collapse
|
47
|
Nguyen DH, Tian J, Shanahan SL, Wang CK, Jacks T, Wang X, Li P. A tissue-scale strategy for sensing threats in barrier organs. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.03.19.644134. [PMID: 40166266 PMCID: PMC11957033 DOI: 10.1101/2025.03.19.644134] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/02/2025]
Abstract
Barrier organs rely on a limited set of pattern recognition receptors (PRRs) to detect diverse immunogenic challenges. How organs assess threats and adjust immune responses to balance host protection with collateral tissue damage remains unclear. Here, by analyzing influenza infection in the lung using single-molecule imaging and spatial transcriptomics, we discovered a tiered threat-sensing strategy at the tissue scale, where the probability of detecting and responding to infection is lowest in the outermost epithelia and highest in the inner stroma. This strategy emerges from spatially graded PRR expression that results in cell-type-specific probabilities of threat-sensing across the tissue, a design broadly adopted by barrier organs. Selectively increasing PRR expression in lung epithelia in vivo exacerbated tissue damage upon inflammatory challenge. These results reveal a spatially tiered strategy to tolerate threats restricted within the epithelia, and yet enable progressively potent immune responses as threats invade deeper into the tissue.
Collapse
|
48
|
Liu T, Lin Y, Luo X, Sun Y, Zhao H. VISTA Uncovers Missing Gene Expression and Spatial-induced Information for Spatial Transcriptomic Data Analysis. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2024.08.26.609718. [PMID: 40166134 PMCID: PMC11957009 DOI: 10.1101/2024.08.26.609718] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 04/02/2025]
Abstract
Characterizing cell activities within a spatially resolved context is essential to enhance our understanding of spatially-induced cellular states and features. While single-cell RNA-seq (scRNA-seq) offers comprehensive profiling of cells within a tissue, it fails to capture spatial context. Conversely, subcellular spatial transcriptomics (SST) technologies provide high-resolution spatial profiles of gene expression, yet their utility is constrained by the limited number of genes they can simultaneously profile. To address this limitation, we introduce VISTA, a novel approach designed to predict the expression levels of unobserved genes specifically tailored for SST data. VISTA jointly models scRNA-seq data and SST data based on variational inference and geometric deep learning, and incorporates uncertainty quantification. Using four SST datasets, we demonstrate VISTA's superior performance in imputation and in analyzing large-scale SST datasets with satisfactory time efficiency and memory consumption. The imputation of VISTA enables a multitude of downstream applications, including the detection of new spatially variable genes, the discovery of novel ligand-receptor interactions, the inference of spatial RNA velocity, the generation for spatial transcriptomics with in-silico perturbation, and an improved decomposition of spatial and intrinsic variations.
Collapse
Affiliation(s)
- Tianyu Liu
- Interdepartmental Program in Computational Biology & Bioinformatics, Yale University, New Haven, 06511, CT, USA
| | - Yingxin Lin
- Department of Biostatistics, Yale University, New Haven, 06511, CT, USA
| | - Xiao Luo
- Department of Computer Science, University of California, Los Angeles, Los Angeles, 90095, CA, USA
| | - Yizhou Sun
- Department of Computer Science, University of California, Los Angeles, Los Angeles, 90095, CA, USA
| | - Hongyu Zhao
- Interdepartmental Program in Computational Biology & Bioinformatics, Yale University, New Haven, 06511, CT, USA
- Department of Biostatistics, Yale University, New Haven, 06511, CT, USA
| |
Collapse
|
49
|
Wang Y, Liu Z, Ma X. MuCST: restoring and integrating heterogeneous morphology images and spatial transcriptomics data with contrastive learning. Genome Med 2025; 17:21. [PMID: 40082941 PMCID: PMC11907906 DOI: 10.1186/s13073-025-01449-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2024] [Accepted: 03/07/2025] [Indexed: 03/16/2025] Open
Abstract
Spatially resolved transcriptomics (SRT) simultaneously measure spatial location, histology images, and transcriptional profiles of cells or regions in undissociated tissues. Integrative analysis of multi-modal SRT data holds immense potential for understanding biological mechanisms. Here, we present a flexible multi-modal contrastive learning for the integration of SRT data (MuCST), which joins denoising, heterogeneity elimination, and compatible feature learning. MuCST accurately identifies spatial domains and is applicable to diverse datasets platforms. Overall, MuCST provides an alternative for integrative analysis of multi-modal SRT data ( https://github.com/xkmaxidian/MuCST ).
Collapse
Affiliation(s)
- Yu Wang
- School of Computer Science and Technology, Xidian University, No.2 South Taibai Road, Xi'an, 710071, Shaanxi, China
- Key Laboratory of Smart Human-Computer Interaction and Wearable Technology of Shaanxi Province, Xidian University, No.2 South Taibai Road, Xi'an, 710071, Shaanxi, China
| | - Zaiyi Liu
- Department of Radiology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, 106 Zhongshan Er Road, Guangzhou, 510080, Guangdong, China
- Guangdong Provincial Key Laboratory of Artificial Intelligence in Medical Image Analysis and Application, 106 Zhongshan Er Road, Guangzhou, 510080, Guangdong, China
| | - Xiaoke Ma
- School of Computer Science and Technology, Xidian University, No.2 South Taibai Road, Xi'an, 710071, Shaanxi, China.
- Key Laboratory of Smart Human-Computer Interaction and Wearable Technology of Shaanxi Province, Xidian University, No.2 South Taibai Road, Xi'an, 710071, Shaanxi, China.
| |
Collapse
|
50
|
Shyer AE, Rodrigues AR. Transcending the hegemony of the molecular machine through an organic renewal of biology and biomedicine. Cells Dev 2025:204018. [PMID: 40074200 DOI: 10.1016/j.cdev.2025.204018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2025] [Revised: 03/05/2025] [Accepted: 03/06/2025] [Indexed: 03/14/2025]
Abstract
The dominant approach to the study of living systems in the 20th century into today has been that of a reductionist approach focused on genetics and biochemistry. The hunt for genes and the elucidation of their biochemical outputs has organized funding in research, educational curricula, academic promotion, and the distribution of prestige through awards. Such reductionism has gone hand in hand with an ontology of the machine. We will discuss how viewing life as if it emanated from a set of molecular machines is the main bottleneck in addressing key questions in biology. We will discuss how moving beyond it is not contingent on new technologies but rather a refreshed perspective of life that can be termed "organic". Furthermore, we suggest that the study of how form arises, morphogenesis, is the key to an organic renewal of biology and biomedicine. Although morphogenesis is currently seen as a subsidiary branch of developmental biology as well as the consequence of molecular patterning processes at the subcellular scale, we will argue that morphology and its self-organizing capacity at the supracellular scale is the fundamental nexus in embryonic development as well as disease. We see the inability to appreciate form through an organic supracellular perspective as the principal bottleneck for making inroads into health issues such as cancer and the chronic disease epidemic.
Collapse
Affiliation(s)
- Amy E Shyer
- Laboratory of Morphogenesis, The Rockefeller University, New York, NY 10065, United States of America.
| | - Alan R Rodrigues
- Laboratory of Morphogenesis, The Rockefeller University, New York, NY 10065, United States of America.
| |
Collapse
|