1
|
Khan SM, Wang AZ, Desai RR, McCornack CR, Sun R, Dahiya SM, Foltz JA, Sherpa ND, Leavitt L, West T, Wang AF, Krbanjevic A, Choi BD, Leuthardt EC, Patel B, Charest A, Kim AH, Dunn GP, Petti AA. Mapping the spatial architecture of glioblastoma from core to edge delineates niche-specific tumor cell states and intercellular interactions. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.04.04.647096. [PMID: 40235981 PMCID: PMC11996482 DOI: 10.1101/2025.04.04.647096] [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
Treatment resistance in glioblastoma (GBM) is largely driven by the extensive multi-level heterogeneity that typifies this disease. Despite significant progress toward elucidating GBM's genomic and transcriptional heterogeneity, a critical knowledge gap remains in defining this heterogeneity at the spatial level. To address this, we employed spatial transcriptomics to map the architecture of the GBM ecosystem. This revealed tumor cell states that are jointly defined by gene expression and spatial localization, and multicellular niches whose composition varies along the tumor core-edge axis. Ligand-receptor interaction analysis uncovered a complex network of intercellular communication, including niche- and region-specific interactions. Finally, we found that CD8 positive GZMK positive T cells colocalize with LYVE1 positive CD163 positive myeloid cells in vascular regions, suggesting a potential mechanism for immune evasion. These findings provide novel insights into the GBM tumor microenvironment, highlighting previously unrecognized patterns of spatial organization and intercellular interactions, and novel therapeutic avenues to disrupt tumor-promoting interactions and overcome immune resistance.
Collapse
|
2
|
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
|
3
|
Paniagua K, Jin YF, Chen Y, Gao SJ, Huang Y, Flores M. Dissection of tumoral niches using spatial transcriptomics and deep learning. iScience 2025; 28:112214. [PMID: 40230519 PMCID: PMC11994907 DOI: 10.1016/j.isci.2025.112214] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2024] [Revised: 10/05/2024] [Accepted: 03/10/2025] [Indexed: 04/16/2025] Open
Abstract
This study introduces TG-ME, an innovative computational framework that integrates transformer with graph variational autoencoder (GraphVAE) models for dissection of tumoral niches using spatial transcriptomics data and morphological images. TG-ME effectively identifies and characterizes niches in bench datasets and a high resolution NSCLC dataset. The pipeline consists in different stages that include normalization, spatial information integration, morphological feature extraction, gene expression quantification, single cell expression characterization, and tumor niche characterization. For this, TG-ME leverages advanced deep learning techniques that achieve robust clustering and profiling of niches across cancer stages. TG-ME can potentially provide insights into the spatial organization of tumor microenvironments (TME), highlighting specific niche compositions and their molecular changes along cancer progression. TG-ME is a promising tool for guiding personalized treatment strategies by uncovering microenvironmental signatures associated with disease prognosis and therapeutic outcomes.
Collapse
Affiliation(s)
- Karla Paniagua
- Department of Electrical and Computer Engineering, KLESSE School of Engineering and Integrated Design, University of Texas at San Antonio, San Antonio, TX 78249, USA
| | - Yu-Fang Jin
- Department of Electrical and Computer Engineering, KLESSE School of Engineering and Integrated Design, University of Texas at San Antonio, San Antonio, TX 78249, USA
| | - Yidong Chen
- Greehey Children Cancer Research Institute, The University of Texas Health Science Center at San Antonio, San Antonio, TX 78229, USA
- Department of Population Health Science, The University of Texas Health Science Center at San Antonio, San Antonio, TX 78229, USA
| | - Shou-Jiang Gao
- Cancer Virology Program, UPMC Hillman Cancer Center, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
- Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Yufei Huang
- Cancer Virology Program, UPMC Hillman Cancer Center, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
- Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
- Department of Electrical and Computer Engineering, Swanson School of Engineering, University of Pittsburgh, Pittsburgh, PA, USA
| | - Mario Flores
- Department of Electrical and Computer Engineering, KLESSE School of Engineering and Integrated Design, University of Texas at San Antonio, San Antonio, TX 78249, USA
| |
Collapse
|
4
|
Yates J, Van Allen EM. New horizons at the interface of artificial intelligence and translational cancer research. Cancer Cell 2025; 43:708-727. [PMID: 40233719 PMCID: PMC12007700 DOI: 10.1016/j.ccell.2025.03.018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/24/2025] [Revised: 03/04/2025] [Accepted: 03/12/2025] [Indexed: 04/17/2025]
Abstract
Artificial intelligence (AI) is increasingly being utilized in cancer research as a computational strategy for analyzing multiomics datasets. Advances in single-cell and spatial profiling technologies have contributed significantly to our understanding of tumor biology, and AI methodologies are now being applied to accelerate translational efforts, including target discovery, biomarker identification, patient stratification, and therapeutic response prediction. Despite these advancements, the integration of AI into clinical workflows remains limited, presenting both challenges and opportunities. This review discusses AI applications in multiomics analysis and translational oncology, emphasizing their role in advancing biological discoveries and informing clinical decision-making. Key areas of focus include cellular heterogeneity, tumor microenvironment interactions, and AI-aided diagnostics. Challenges such as reproducibility, interpretability of AI models, and clinical integration are explored, with attention to strategies for addressing these hurdles. Together, these developments underscore the potential of AI and multiomics to enhance precision oncology and contribute to advancements in cancer care.
Collapse
Affiliation(s)
- Josephine Yates
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA; Institute for Machine Learning, Department of Computer Science, ETH Zürich, Zurich, Switzerland; ETH AI Center, ETH Zurich, Zurich, Switzerland; Swiss Institute for Bioinformatics (SIB), Lausanne, Switzerland
| | - Eliezer M Van Allen
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA; Cancer Program, Broad Institute of MIT and Harvard, Cambridge, MA, USA; Division of Medical Sciences, Harvard University, Boston, MA, USA; Parker Institute for Cancer Immunotherapy, Dana-Farber Cancer Institute, Boston, MA, USA.
| |
Collapse
|
5
|
Zhang D, Li W, Sui X, Yu N, Wang S, Liu Z, Wang X, Yuan Z, Gao R, Zhang W. Space: reconciling multiple spatial domain identification algorithms via consensus clustering. BIOINFORMATICS ADVANCES 2025; 5:vbaf084. [PMID: 40297775 PMCID: PMC12037102 DOI: 10.1093/bioadv/vbaf084] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/30/2024] [Revised: 03/21/2025] [Accepted: 04/08/2025] [Indexed: 04/30/2025]
Abstract
Motivation The rapid development of spatially resolved transcriptomics (SRT) technologies has provided unprecedented opportunities for characterizing and understanding tissue architecture. As this field continues to advance, various methods have been developed to computationally identify spatial domains within tissues. However, the performance of different algorithms on the same dataset is not always consistent. This inconsistency makes it difficult for researchers to select the most reliable results for downstream analysis. Results To address this challenge, we propose a domain identification method named Space. Space measures consistency between different methods to select reliable algorithms. It then constructs a consensus matrix to integrate the outputs from multiple algorithms. We introduce similarity loss, spatial loss, and low-rank loss in Space to enhance the accuracy and optimize computational efficiency. This strategy not only resolves the inconsistent issue of clustering labels among different methods but also achieves highly reliable clustering output. Flexible interfaces are also provided for downstream analysis such as visualization, domain-specific gene analysis and trajectory inference. Testing results on multiple publicly available SRT datasets demonstrate that Space performs exceptionally well in deciphering key tissue structures and biological features. Availability and implementation The Space package can be easily installed through conda or mamba, and its source code is available at https://honchkrow.github.io/Space.
Collapse
Affiliation(s)
- Daoliang Zhang
- Center of Intelligent Medicine, School of Control Science and Engineering, Shandong University, Jinan, Shandong 250061, China
| | - Wenrui Li
- MOE Key Lab of Bioinformatics and Bioinformatics Division of BNRIST, Department of Automation, Tsinghua University, Beijing 100084, China
| | - Xinyi Sui
- Center of Intelligent Medicine, School of Control Science and Engineering, Shandong University, Jinan, Shandong 250061, China
| | - Na Yu
- Center of Intelligent Medicine, School of Control Science and Engineering, Shandong University, Jinan, Shandong 250061, China
| | - Shan Wang
- Center of Intelligent Medicine, School of Control Science and Engineering, Shandong University, Jinan, Shandong 250061, China
| | - Zhiping Liu
- Center of Intelligent Medicine, School of Control Science and Engineering, Shandong University, Jinan, Shandong 250061, China
| | - Xiaowo Wang
- MOE Key Lab of Bioinformatics and Bioinformatics Division of BNRIST, Department of Automation, Tsinghua University, Beijing 100084, China
| | - Zhiyuan Yuan
- Institute of Science and Technology for Brain-Inspired Intelligence, Center for Medical Research and Innovation, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Fudan University, Shanghai 200433, China
| | - Rui Gao
- Center of Intelligent Medicine, School of Control Science and Engineering, Shandong University, Jinan, Shandong 250061, China
| | - Wei Zhang
- Center of Intelligent Medicine, School of Control Science and Engineering, Shandong University, Jinan, Shandong 250061, China
| |
Collapse
|
6
|
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
|
7
|
Tu R, Zheng H, Zheng B, Zhong Q, Qian J, Wu F, Shiokawa T, Ochiai Y, Kobayashi H, Waterbury QT, Zamechek LB, Takahashi S, Mizuno S, Huang C, Li P, Hayakawa Y, Wang TC. Tff2 marks gastric corpus progenitors that give rise to pyloric metaplasia/SPEM following injury. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.04.09.647847. [PMID: 40291734 PMCID: PMC12027342 DOI: 10.1101/2025.04.09.647847] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/30/2025]
Abstract
In Brief Tu et al. show that Tff2 + corpus isthmus cells are TA progenitors, and they, not chief cells, are the primary source of SPEM following injury. Upon Kras mutation, these progenitors directly progress to dysplasia, bypassing metaplasia, highlighting them as a potential origin of gastric cancer. Highlights Tff2 + corpus cells are TA progenitors that give rise to secretory cells. Tff2 + progenitors, not chief cells, are the primary source of SPEM after injury. Kras-mutant Tff2 + progenitors progress directly to dysplasia, bypassing metaplasia. Multi-omics analysis reveals distinct trajectories for SPEM and gastric cancer. Abstract Figure Pyloric metaplasia, also known as spasmolytic polypeptide-expressing metaplasia (SPEM), arises in the corpus in response to oxyntic atrophy, but its origin and role in gastric cancer remain poorly understood. Using Tff2-CreERT knockin mice, we identified highly proliferative Tff2 + progenitors in the corpus isthmus that give rise to multiple secretory lineages, including chief cells. While lacking long-term self-renewal ability, Tff2 + corpus progenitors rapidly expand to form short-term SPEM following acute injury or loss of chief cells. Genetic ablation of Tff2 + progenitors abrogated SPEM formation, while genetic ablation of GIF + chief cells enhanced SPEM formation from Tff2 + progenitors. In response to H. pylori infection, Tff2 + progenitors progressed first to metaplasia and then later to dysplasia. Interestingly, induction of Kras G12D mutations in Tff2 + progenitors facilitated direct progression to dysplasia in part through the acquisition of stem cell-like properties. In contrast, Kras-mutated SPEM and chief cells were not able to progress to dysplasia. Tff2 mRNA was downregulated in isthmus cells during progression to dysplasia. Single-cell RNA sequencing and spatial transcriptomics of human tissues revealed distinct differentiation trajectories for SPEM and gastric cancer. These findings challenge the conventional interpretation of the stepwise progression through metaplasia and instead identify Tff2 + progenitor cells as potential cells of origin for SPEM and possibly for gastric cancer.
Collapse
|
8
|
Velu PP, Abhari RE, Henderson NC. Spatial genomics: Mapping the landscape of fibrosis. Sci Transl Med 2025; 17:eadm6783. [PMID: 40203082 DOI: 10.1126/scitranslmed.adm6783] [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: 07/31/2024] [Accepted: 03/19/2025] [Indexed: 04/11/2025]
Abstract
Organ fibrosis causes major morbidity and mortality worldwide. Treatments for fibrosis are limited, with organ transplantation being the only cure. Here, we review how various state-of-the-art spatial genomics approaches are being deployed to interrogate fibrosis across multiple organs, providing exciting insights into fibrotic disease pathogenesis. These include the detailed topographical annotation of pathogenic cell populations and states, detection of transcriptomic perturbations in morphologically normal tissue, characterization of fibrotic and homeostatic niches and their cellular constituents, and in situ interrogation of ligand-receptor interactions within these microenvironments. Together, these powerful readouts enable detailed analysis of fibrosis evolution across time and space.
Collapse
Affiliation(s)
- Prasad Palani Velu
- Centre for Inflammation Research, Institute for Regeneration and Repair, Edinburgh BioQuarter, University of Edinburgh, Edinburgh EH16 4UU, UK
| | - Roxanna E Abhari
- Centre for Inflammation Research, Institute for Regeneration and Repair, Edinburgh BioQuarter, University of Edinburgh, Edinburgh EH16 4UU, UK
| | - Neil C Henderson
- Centre for Inflammation Research, Institute for Regeneration and Repair, Edinburgh BioQuarter, University of Edinburgh, Edinburgh EH16 4UU, UK
- MRC Human Genetics Unit, Institute of Genetics and Cancer, University of Edinburgh, Edinburgh EH4 1QY, UK
| |
Collapse
|
9
|
Fumagalli L, Nazlie Mohebiany A, Premereur J, Polanco Miquel P, Bijnens B, Van de Walle P, Fattorelli N, Mancuso R. Microglia heterogeneity, modeling and cell-state annotation in development and neurodegeneration. Nat Neurosci 2025:10.1038/s41593-025-01931-4. [PMID: 40195564 DOI: 10.1038/s41593-025-01931-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2023] [Accepted: 02/20/2025] [Indexed: 04/09/2025]
Abstract
Within the CNS, microglia execute various functions associated with brain development, maintenance of homeostasis and elimination of pathogens and protein aggregates. This wide range of activities is closely associated with a plethora of cellular states, which may reciprocally influence or be influenced by their functional dynamics. Advancements in single-cell RNA sequencing have enabled a nuanced exploration of the intricate diversity of microglia, both in health and disease. Here, we review our current understanding of microglial transcriptional heterogeneity. We provide an overview of mouse and human microglial diversity encompassing aspects of development, neurodegeneration, sex and CNS regions. We offer an insight into state-of-the-art technologies and model systems that are poised to improve our understanding of microglial cell states and functions. We also provide suggestions and a tool to annotate microglial cell states on the basis of gene expression.
Collapse
Affiliation(s)
- Laura Fumagalli
- Microglia and Inflammation in Neurological Disorders (MIND) Lab, VIB Center for Molecular Neurology, VIB, Antwerp, Belgium
- Department of Biomedical Sciences, University of Antwerp, Antwerp, Belgium
| | - Alma Nazlie Mohebiany
- Microglia and Inflammation in Neurological Disorders (MIND) Lab, VIB Center for Molecular Neurology, VIB, Antwerp, Belgium
- Department of Biomedical Sciences, University of Antwerp, Antwerp, Belgium
| | - Jessie Premereur
- Microglia and Inflammation in Neurological Disorders (MIND) Lab, VIB Center for Molecular Neurology, VIB, Antwerp, Belgium
- Department of Biomedical Sciences, University of Antwerp, Antwerp, Belgium
| | - Paula Polanco Miquel
- Microglia and Inflammation in Neurological Disorders (MIND) Lab, VIB Center for Molecular Neurology, VIB, Antwerp, Belgium
- Department of Biomedical Sciences, University of Antwerp, Antwerp, Belgium
| | - Baukje Bijnens
- Microglia and Inflammation in Neurological Disorders (MIND) Lab, VIB Center for Molecular Neurology, VIB, Antwerp, Belgium
- Department of Biomedical Sciences, University of Antwerp, Antwerp, Belgium
| | | | - Nicola Fattorelli
- Microglia and Inflammation in Neurological Disorders (MIND) Lab, VIB Center for Molecular Neurology, VIB, Antwerp, Belgium
- Department of Biomedical Sciences, University of Antwerp, Antwerp, Belgium
| | - Renzo Mancuso
- Microglia and Inflammation in Neurological Disorders (MIND) Lab, VIB Center for Molecular Neurology, VIB, Antwerp, Belgium.
- Department of Biomedical Sciences, University of Antwerp, Antwerp, Belgium.
| |
Collapse
|
10
|
Leng J, Yu J, Wu LY, Chen H. Flexible integration of spatial and expression information for precise spot embedding via ZINB-based graph-enhanced autoencoder. Commun Biol 2025; 8:556. [PMID: 40186054 PMCID: PMC11971412 DOI: 10.1038/s42003-025-07965-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2024] [Accepted: 03/19/2025] [Indexed: 04/07/2025] Open
Abstract
Domain identification is a critical problem in spatially resolved transcriptomics data analysis, which aims to identify distinct spatial domains within a tissue that maintain both spatial continuity and expression consistency. The degree of coupling between expression data and spatial information in different datasets often varies significantly. Some regions have intact and clear boundaries, while others exhibit blurred boundaries with high intra-domain expression similarity. However, most domain identification methods do not adequately integrate expression and spatial information to flexibly identify different types of domains. To address these issues, we introduce Spot2vector, a computational framework that leverages a graph-enhanced autoencoder integrating zero-inflated negative binomial distribution modeling, combining both graph convolutional networks and graph attention networks to extract the latent embeddings of spots. Spot2vector encodes and integrates spatial and expression information, enabling effective identification of domains with diverse spatial patterns across spatially resolved transcriptomics data generated by different platforms. The decoders enable us to decipher the distribution and generation mechanisms of data while improving expression quality through denoising. Extensive validation and analyses demonstrate that Spot2vector excels in enhancing domain identification accuracy, effectively reducing data dimensionality, improving expression recovery and denoising, and precisely capturing spatial gene expression patterns.
Collapse
Affiliation(s)
| | - Jiating Yu
- School of Mathematics and Statistics, Nanjing University of Information Science & Technology, Nanjing, 210044, China
| | - Ling-Yun Wu
- IAM, MADIS, NCMIS, Academy of Mathematics and Systems Science, Chinese Academy of Sciences, Beijing, 100190, China
- School of Mathematical Sciences, University of Chinese Academy of Sciences, Beijing, 100049, China
| | | |
Collapse
|
11
|
Sun Y, Yu N, Zhang J, Yang B. Advances in Microfluidic Single-Cell RNA Sequencing and Spatial Transcriptomics. MICROMACHINES 2025; 16:426. [PMID: 40283301 PMCID: PMC12029715 DOI: 10.3390/mi16040426] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/25/2024] [Revised: 11/22/2024] [Accepted: 11/25/2024] [Indexed: 04/29/2025]
Abstract
The development of micro- and nano-fabrication technologies has greatly advanced single-cell and spatial omics technologies. With the advantages of integration and compartmentalization, microfluidic chips are capable of generating high-throughput parallel reaction systems for single-cell screening and analysis. As omics technologies improve, microfluidic chips can now integrate promising transcriptomics technologies, providing new insights from molecular characterization for tissue gene expression profiles and further revealing the static and even dynamic processes of tissues in homeostasis and disease. Here, we survey the current landscape of microfluidic methods in the field of single-cell and spatial multi-omics, as well as assessing their relative advantages and limitations. We highlight how microfluidics has been adapted and improved to provide new insights into multi-omics over the past decade. Last, we emphasize the contributions of microfluidic-based omics methods in development, neuroscience, and disease mechanisms, as well as further revealing some perspectives for technological advances in translational and clinical medicine.
Collapse
Affiliation(s)
- Yueqiu Sun
- State Key Laboratory of Supramolecular Structure and Materials, College of Chemistry, Jilin University, Changchun 130000, China
- Joint Laboratory of Opto-Functional Theranostics in Medicine and Chemistry, The First Hospital of Jilin University, Jilin University, Changchun 130000, China
| | - Nianzuo Yu
- State Key Laboratory of Supramolecular Structure and Materials, College of Chemistry, Jilin University, Changchun 130000, China
- Joint Laboratory of Opto-Functional Theranostics in Medicine and Chemistry, The First Hospital of Jilin University, Jilin University, Changchun 130000, China
| | - Junhu Zhang
- State Key Laboratory of Supramolecular Structure and Materials, College of Chemistry, Jilin University, Changchun 130000, China
- Joint Laboratory of Opto-Functional Theranostics in Medicine and Chemistry, The First Hospital of Jilin University, Jilin University, Changchun 130000, China
| | - Bai Yang
- State Key Laboratory of Supramolecular Structure and Materials, College of Chemistry, Jilin University, Changchun 130000, China
- Joint Laboratory of Opto-Functional Theranostics in Medicine and Chemistry, The First Hospital of Jilin University, Jilin University, Changchun 130000, China
| |
Collapse
|
12
|
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] [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
|
13
|
Wang H, Li J, Jing S, Lin P, Qiu Y, Yan X, Yuan J, Tang Z, Li Y, Zhang H, Chen Y, Wang Z, Li H. SOAPy: a Python package to dissect spatial architecture, dynamics, and communication. Genome Biol 2025; 26:80. [PMID: 40158115 PMCID: PMC11954224 DOI: 10.1186/s13059-025-03550-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2024] [Accepted: 03/18/2025] [Indexed: 04/01/2025] Open
Abstract
Advances in spatial omics enable deeper insights into tissue microenvironments while posing computational challenges. Therefore, we developed SOAPy, a comprehensive tool for analyzing spatial omics data, which offers methods for spatial domain identification, spatial expression tendency, spatiotemporal expression pattern, cellular co-localization, multi-cellular niches, cell-cell communication, and so on. SOAPy can be applied to diverse spatial omics technologies and multiple areas in physiological and pathological contexts, such as tumor biology and developmental biology. Its versatility and robust performance make it a universal platform for spatial omics analysis, providing diverse insights into the dynamics and architecture of tissue microenvironments.
Collapse
Affiliation(s)
- Heqi Wang
- CAS Key Laboratory of Computational Biology, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, 200031, China
| | - Jiarong Li
- CAS Key Laboratory of Computational Biology, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, 200031, China
| | - Siyu Jing
- CAS Key Laboratory of Computational Biology, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, 200031, China
| | - Ping Lin
- CAS Key Laboratory of Computational Biology, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, 200031, China
| | - Yiling Qiu
- CAS Key Laboratory of Computational Biology, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, 200031, China
| | - Xi Yan
- CAS Key Laboratory of Computational Biology, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, 200031, China
| | - Jiao Yuan
- CAS Key Laboratory of Computational Biology, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, 200031, China
| | - ZhiXuan Tang
- CAS Key Laboratory of Computational Biology, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, 200031, China
| | - Yu Li
- CAS Key Laboratory of Nutrition, Metabolism and Food Safety, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, 200031, China
| | - Haibing Zhang
- CAS Key Laboratory of Nutrition, Metabolism and Food Safety, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, 200031, China
| | - Yujie Chen
- CAS Key Laboratory of Computational Biology, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, 200031, China
| | - Zhen Wang
- CAS Key Laboratory of Computational Biology, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, 200031, China
| | - Hong Li
- CAS Key Laboratory of Computational Biology, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, 200031, China.
| |
Collapse
|
14
|
Troulé K, Petryszak R, Cakir B, Cranley J, Harasty A, Prete M, Tuong ZK, Teichmann SA, Garcia-Alonso L, Vento-Tormo R. CellPhoneDB v5: inferring cell-cell communication from single-cell multiomics data. Nat Protoc 2025:10.1038/s41596-024-01137-1. [PMID: 40133495 DOI: 10.1038/s41596-024-01137-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2023] [Accepted: 12/20/2024] [Indexed: 03/27/2025]
Abstract
Cell-cell communication is essential for tissue development, function and regeneration. The revolution of single-cell genomics technologies offers an unprecedented opportunity to uncover how cells communicate in vivo within their tissue niches and how disruption of these niches can lead to diseases and developmental abnormalities. CellPhoneDB is a bioinformatics toolkit designed to infer cell-cell communication by combining a curated repository of bona fide ligand-receptor interactions with methods to integrate these interactions with single-cell genomics data. Here we present a protocol for the latest version of CellPhoneDB (v5), offering several new features. First, the repository has been expanded by one-third with the addition of new interactions, including ~1,000 interactions mediated by nonpeptidic ligands such as steroidogenic hormones, neurotransmitters and small G-protein-coupled receptor (GPCR)-binding ligands. Second, we outline a new way of using the database that allows users to tailor queries to their experimental designs. Third, the update incorporates novel strategies to prioritize specific cell-cell interactions, leveraging information from other modalities such as tissue microenvironments derived from spatial transcriptomics technologies or transcription factor activities derived from a single-cell assay for transposase accessible chromatin assays. Finally, we describe the new CellPhoneDBViz module to interactively visualize and share results. Altogether, CellPhoneDB v5 enhances the precision of cell-cell communication inference, offering new insights into tissue biology in physiological microenvironments. This protocol typically takes ~15 min and requires basic knowledge of python.
Collapse
Affiliation(s)
| | | | | | | | - Alicia Harasty
- Ian Frazer Centre for Children's Immunotherapy Research, Child Health Research Centre, Faculty of Medicine, The University of Queensland, Brisbane, Queensland, Australia
| | | | - Zewen Kelvin Tuong
- Wellcome Sanger Institute, Cambridge, UK
- Ian Frazer Centre for Children's Immunotherapy Research, Child Health Research Centre, Faculty of Medicine, The University of Queensland, Brisbane, Queensland, Australia
| | - Sarah A Teichmann
- Wellcome Sanger Institute, Cambridge, UK
- Department of Physics, Cavendish Laboratory, University of Cambridge, Cambridge, UK
- Department of Medicine and Cambridge Stem Cell Institute Clinical School, University of Cambridge, Cambridge, UK
| | | | | |
Collapse
|
15
|
Bryan JP, Farhi SL, Cleary B. Accurate trajectory inference in time-series spatial transcriptomics with structurally-constrained optimal transport. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.03.19.644194. [PMID: 40166168 PMCID: PMC11957147 DOI: 10.1101/2025.03.19.644194] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/02/2025]
Abstract
New experimental and computational methods use genetic or gene expression observations with single cell resolution to study the relationship between single-cell molecular profiles and developmental trajectories. Most tissues contain spatially contiguous regions that develop as a unit, such as follicles in the ovary, or tubules and glomeruli in the kidney. We find that existing approaches designed to use time series spatial transcriptomics (ST) data produce biologically incoherent trajectories that fail to maintain these structural units over time. We present Spatiotemporal Optimal transport with Contiguous Structures (SOCS), an Optimal Transport-based trajectory inference method for time-series ST that produces trajectory inferences preserving the structural integrity of contiguous biologically meaningful units, along with gene expression similarity and global geometric structure. We show that SOCS produces more plausible trajectory estimates, maintaining the spatial coherence of biological structures across time, enabling more accurate trajectory inference and biological insight than other approaches.
Collapse
Affiliation(s)
- John P Bryan
- Department of Electrical Engineering and Computer Science, Massachusetts Institute of Technology, Cambridge, MA
- Spatial Technology Platform, Broad Institute of MIT and Harvard, Cambridge, MA
| | - Samouil L Farhi
- Spatial Technology Platform, Broad Institute of MIT and Harvard, Cambridge, MA
| | - Brian Cleary
- Faculty of Computing and Data Sciences, Boston University, Boston, MA; Department of Biology, Boston University, Boston, MA; Department of Biomedical Engineering, Boston University, Boston, MA; Program in Bioinformatics, Boston University, Boston, MA; Biological Design Center, Boston University, Boston, MA
| |
Collapse
|
16
|
Liang X, Torkel M, Cao Y, Yang JYH. Multi-task benchmarking of spatially resolved gene expression simulation models. Genome Biol 2025; 26:57. [PMID: 40098171 PMCID: PMC11912772 DOI: 10.1186/s13059-025-03505-w] [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: 05/15/2024] [Accepted: 02/12/2025] [Indexed: 03/19/2025] Open
Abstract
BACKGROUND Computational methods for spatially resolved transcriptomics (SRT) are often developed and assessed using simulated data. The effectiveness of these evaluations relies on the ability of simulation methods to accurately reflect experimental data. However, a systematic evaluation framework for spatial simulators is currently lacking. RESULTS Here, we present SpatialSimBench, a comprehensive evaluation framework that assesses 13 simulation methods using ten distinct STR datasets. We introduce simAdaptor, a tool that extends single-cell simulators by incorporating spatial variables, enabling them to simulate spatial data. SimAdaptor ensures SpatialSimBench is backwards compatible, facilitating direct comparisons between spatially aware simulators and existing non-spatial single-cell simulators through the adaption. Using SpatialSimBench, we demonstrate the feasibility of leveraging existing single-cell simulators for SRT data and highlight performance differences among methods. Additionally, we evaluate the simulation methods based on a total of 35 metrics across data property estimation, various downstream analyses, and scalability. In total, we generated 4550 results from 13 simulation methods, ten spatial datasets, and 35 metrics. CONCLUSIONS Our findings reveal that model estimation can be influenced by distribution assumptions and dataset characteristics. In summary, our evaluation framework provides guidelines for selecting appropriate methods for specific scenarios and informs future method development.
Collapse
Affiliation(s)
- Xiaoqi Liang
- School of Mathematics and Statistics, The University of Sydney, Sydney, NSW, 2006, Australia
- Sydney Precision Data Science Centre, The University of Sydney, Sydney, NSW, 2006, Australia
- Charles Perkins Centre, The University of Sydney, Sydney, NSW, 2006, Australia
| | - Marni Torkel
- School of Mathematics and Statistics, The University of Sydney, Sydney, NSW, 2006, Australia
- Sydney Precision Data Science Centre, The University of Sydney, Sydney, NSW, 2006, Australia
- Charles Perkins Centre, The University of Sydney, Sydney, NSW, 2006, Australia
| | - Yue Cao
- School of Mathematics and Statistics, The University of Sydney, Sydney, NSW, 2006, Australia.
- Sydney Precision Data Science Centre, The University of Sydney, Sydney, NSW, 2006, Australia.
- Charles Perkins Centre, The University of Sydney, Sydney, NSW, 2006, Australia.
- Laboratory of Data Discovery for Health Limited (D24H), Science Park, Hong Kong SAR, China.
| | - Jean Yee Hwa Yang
- School of Mathematics and Statistics, The University of Sydney, Sydney, NSW, 2006, Australia.
- Sydney Precision Data Science Centre, The University of Sydney, Sydney, NSW, 2006, Australia.
- Charles Perkins Centre, The University of Sydney, Sydney, NSW, 2006, Australia.
- Laboratory of Data Discovery for Health Limited (D24H), Science Park, Hong Kong SAR, China.
| |
Collapse
|
17
|
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] [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
|
18
|
Fang Z, Krusen K, Priest H, Wang M, Kim S, Sriram A, Yellanki A, Singh A, Horwitz E, Coskun AF. Graph-Based 3-Dimensional Spatial Gene Neighborhood Networks of Single Cells in Gels and Tissues. BME FRONTIERS 2025; 6:0110. [PMID: 40084126 PMCID: PMC11906096 DOI: 10.34133/bmef.0110] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2024] [Revised: 02/12/2025] [Accepted: 02/13/2025] [Indexed: 03/16/2025] Open
Abstract
Objective: We developed 3-dimensional spatially resolved gene neighborhood network embedding (3D-spaGNN-E) to find subcellular gene proximity relationships and identify key subcellular motifs in cell-cell communication (CCC). Impact Statement: The pipeline combines 3D imaging-based spatial transcriptomics and graph-based deep learning to identify subcellular motifs. Introduction: Advancements in imaging and experimental technology allow the study of 3D spatially resolved transcriptomics and capture better spatial context than approximating the samples as 2D. However, the third spatial dimension increases the data complexity and requires new analyses. Methods: 3D-spaGNN-E detects single transcripts in 3D cell culture samples and identifies subcellular gene proximity relationships. Then, a graph autoencoder projects the gene proximity relationships into a latent space. We then applied explainability analysis to identify subcellular CCC motifs. Results: We first applied the pipeline to mesenchymal stem cells (MSCs) cultured in hydrogel. After clustering the cells based on the RNA count, we identified cells belonging to the same cluster as homotypic and those belonging to different clusters as heterotypic. We identified changes in local gene proximity near the border between homotypic and heterotypic cells. When applying the pipeline to the MSC-peripheral blood mononuclear cell (PBMC) coculture system, we identified CD4+ and CD8+ T cells. Local gene proximity and autoencoder embedding changes can distinguish strong and weak suppression of different immune cells. Lastly, we compared astrocyte-neuron CCC in mouse hypothalamus and cortex by analyzing 3D multiplexed-error-robust fluorescence in situ hybridization (MERFISH) data and identified regional gene proximity differences. Conclusion: 3D-spaGNN-E distinguished distinct CCCs in cell culture and tissue by examining subcellular motifs.
Collapse
Affiliation(s)
- Zhou Fang
- Wallace H. Coulter Department of Biomedical Engineering,
Georgia Institute of Technology and Emory University, Atlanta, GA, USA
- Machine Learning Graduate Program,
Georgia Institute of Technology, Atlanta, GA, USA
| | - Kelsey Krusen
- Wallace H. Coulter Department of Biomedical Engineering,
Georgia Institute of Technology and Emory University, Atlanta, GA, USA
| | - Hannah Priest
- Wallace H. Coulter Department of Biomedical Engineering,
Georgia Institute of Technology and Emory University, Atlanta, GA, USA
| | - Mingshuang Wang
- Wallace H. Coulter Department of Biomedical Engineering,
Georgia Institute of Technology and Emory University, Atlanta, GA, USA
| | - Sungwoong Kim
- Wallace H. Coulter Department of Biomedical Engineering,
Georgia Institute of Technology and Emory University, Atlanta, GA, USA
| | - Anirudh Sriram
- Wallace H. Coulter Department of Biomedical Engineering,
Georgia Institute of Technology and Emory University, Atlanta, GA, USA
| | - Ashritha Yellanki
- Wallace H. Coulter Department of Biomedical Engineering,
Georgia Institute of Technology and Emory University, Atlanta, GA, USA
| | - Ankur Singh
- Wallace H. Coulter Department of Biomedical Engineering,
Georgia Institute of Technology and Emory University, Atlanta, GA, USA
- Woodruff School of Mechanical Engineering,
Georgia Institute of Technology, Atlanta, GA, USA
- Interdisciplinary Bioengineering Graduate Program,
Georgia Institute of Technology, Atlanta, GA, USA
- Parker H. Petit Institute for Bioengineering and Bioscience, GeorgiaInstitute of Technology, Atlanta, GA 30332, USA
| | - Edwin Horwitz
- Department of Pediatrics,
Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Ahmet F. Coskun
- Wallace H. Coulter Department of Biomedical Engineering,
Georgia Institute of Technology and Emory University, Atlanta, GA, USA
- Interdisciplinary Bioengineering Graduate Program,
Georgia Institute of Technology, Atlanta, GA, USA
- Parker H. Petit Institute for Bioengineering and Bioscience, GeorgiaInstitute of Technology, Atlanta, GA 30332, USA
| |
Collapse
|
19
|
Jing SY, Wang HQ, Lin P, Yuan J, Tang ZX, Li H. Quantifying and interpreting biologically meaningful spatial signatures within tumor microenvironments. NPJ Precis Oncol 2025; 9:68. [PMID: 40069556 PMCID: PMC11897387 DOI: 10.1038/s41698-025-00857-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2024] [Accepted: 02/25/2025] [Indexed: 03/15/2025] Open
Abstract
The tumor microenvironment (TME) plays a crucial role in orchestrating tumor cell behavior and cancer progression. Recent advances in spatial profiling technologies have uncovered novel spatial signatures, including univariate distribution patterns, bivariate spatial relationships, and higher-order structures. These signatures have the potential to revolutionize tumor mechanism and treatment. In this review, we summarize the current state of spatial signature research, highlighting computational methods to uncover spatially relevant biological significance. We discuss the impact of these advances on fundamental cancer biology and translational research, address current challenges and future research directions.
Collapse
Affiliation(s)
- Si-Yu Jing
- CAS Key Laboratory of Computational Biology, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, People's Republic of China
| | - He-Qi Wang
- CAS Key Laboratory of Computational Biology, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, People's Republic of China
| | - Ping Lin
- CAS Key Laboratory of Computational Biology, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, People's Republic of China
| | - Jiao Yuan
- CAS Key Laboratory of Computational Biology, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, People's Republic of China
| | - Zhi-Xuan Tang
- CAS Key Laboratory of Computational Biology, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, People's Republic of China
| | - Hong Li
- CAS Key Laboratory of Computational Biology, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, People's Republic of China.
| |
Collapse
|
20
|
Zhang P, Chen W, Tran TN, Zhou M, Carter KN, Kandel I, Li S, Hoi XP, Youker K, Lai L, Song Q, Yang Y, Nikolos F, Chan KS, Wang G. Thor: a platform for cell-level investigation of spatial transcriptomics and histology. RESEARCH SQUARE 2025:rs.3.rs-4909620. [PMID: 40162205 PMCID: PMC11952649 DOI: 10.21203/rs.3.rs-4909620/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/02/2025]
Abstract
Spatial transcriptomics integrates transcriptomics data with histological tissue images, offering deeper insights into cellular organization and molecular functions. However, existing computational platforms mainly focus on genomic analysis, leaving a gap in the seamless integration of genomic and image analysis. To address this, we introduce Thor, a comprehensive computational platform for multi-modal analysis of spatial transcriptomics and histological images. Thor utilizes an anti-shrinking Markov diffusion method to infer single-cell spatial transcriptomes from spot-level data, effectively integrating cell morphology with spatial transcriptomics. The platform features 10 modules designed for cell-level genomic and image analysis. Additionally, we present Mjolnir, a web-based tool for interactive tissue analysis using vivid gigapixel images that display information on histology, gene expression, pathway enrichment, and immune response. Thor's accuracy was validated through simulations and ISH, MERFISH, Xenium, and Stereo-seq datasets. To demonstrate its versatility, we applied Thor for joint genomic-histology analysis across various datasets. In in-house heart failure patient samples, Thor identified a regenerative signature in heart failure, with protein presence confirmed in blood vessels through immunofluorescence staining. Thor also revealed the layered structure of the mouse olfactory bulb, performed unbiased screening of breast cancer hallmarks, elucidated the heterogeneity of immune responses, and annotated fibrotic regions in multiple heart failure zones using a semi-supervised approach. Furthermore, Thor imputed high-resolution spatial transcriptomics data in an in-house bladder cancer sample sequenced using Visium HD, uncovering stronger spatial patterns that align more closely with histology. Bridging the gap between genomic and image analysis in spatial biology, Thor offers a powerful tool for comprehensive cellular and molecular analysis.
Collapse
Affiliation(s)
- Pengzhi Zhang
- Center for Bioinformatics and Computational Biology, Houston Methodist Research Institute, Houston, TX, 77030, USA
- Center for Cardiovascular Regeneration, Houston Methodist Research Institute, Houston, TX, 77030, USA
- Center for RNA Therapeutics, Houston Methodist Research Institute, Houston, TX, 77030, USA
- Department of Cardiothoracic Surgery, Weill Cornell Medicine, Cornell University, New York, NY, 10065, USA
| | - Weiqing Chen
- Department of Physiology, Biophysics & Systems Biology, Weill Cornell Graduate School of Medical Science, Weill Cornell Medicine, Cornell University, New York, NY, 10065, USA
| | - Tu Nhi Tran
- Center for Bioinformatics and Computational Biology, Houston Methodist Research Institute, Houston, TX, 77030, USA
- Center for Cardiovascular Regeneration, Houston Methodist Research Institute, Houston, TX, 77030, USA
- Center for RNA Therapeutics, Houston Methodist Research Institute, Houston, TX, 77030, USA
- Department of Cardiothoracic Surgery, Weill Cornell Medicine, Cornell University, New York, NY, 10065, USA
| | - Minghao Zhou
- Department of Health Outcomes and Biomedical Informatics, University of Florida, Gainesville, FL, 32610, USA
| | - Kaylee N Carter
- Center for Cardiovascular Regeneration, Houston Methodist Research Institute, Houston, TX, 77030, USA
| | - Ibrahem Kandel
- Center for Bioinformatics and Computational Biology, Houston Methodist Research Institute, Houston, TX, 77030, USA
- Center for Cardiovascular Regeneration, Houston Methodist Research Institute, Houston, TX, 77030, USA
- Center for RNA Therapeutics, Houston Methodist Research Institute, Houston, TX, 77030, USA
- Department of Cardiothoracic Surgery, Weill Cornell Medicine, Cornell University, New York, NY, 10065, USA
| | - Shengyu Li
- Center for Bioinformatics and Computational Biology, Houston Methodist Research Institute, Houston, TX, 77030, USA
- Center for Cardiovascular Regeneration, Houston Methodist Research Institute, Houston, TX, 77030, USA
- Center for RNA Therapeutics, Houston Methodist Research Institute, Houston, TX, 77030, USA
- Department of Cardiothoracic Surgery, Weill Cornell Medicine, Cornell University, New York, NY, 10065, USA
| | - Xen Ping Hoi
- Department of Urology, Houston Methodist Research Institute, Houston, TX, 77030, USA
- Spatial Omics Core, Neal Cancer Center, Houston Methodist Research Institute, Houston, TX, 77030, USA
- Graduate Program in Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA 90069, USA
| | - Keith Youker
- Department of Cardiothoracic Surgery, Weill Cornell Medicine, Cornell University, New York, NY, 10065, USA
- Department of Cardiovascular Sciences, Houston Methodist Research Institute, Houston, TX, 77030, USA
| | - Li Lai
- Center for Cardiovascular Regeneration, Houston Methodist Research Institute, Houston, TX, 77030, USA
| | - Qianqian Song
- Department of Health Outcomes and Biomedical Informatics, University of Florida, Gainesville, FL, 32610, USA
| | - Yu Yang
- Department of Pathology, Immunology and Laboratory Medicine, College of Medicine, University of Florida, Gainesville, FL, 32608, USA
| | - Fotis Nikolos
- Department of Urology, Houston Methodist Research Institute, Houston, TX, 77030, USA
- Spatial Omics Core, Neal Cancer Center, Houston Methodist Research Institute, Houston, TX, 77030, USA
| | - Keith Syson Chan
- Department of Urology, Houston Methodist Research Institute, Houston, TX, 77030, USA
- Spatial Omics Core, Neal Cancer Center, Houston Methodist Research Institute, Houston, TX, 77030, USA
| | - Guangyu Wang
- Center for Bioinformatics and Computational Biology, Houston Methodist Research Institute, Houston, TX, 77030, USA
- Center for Cardiovascular Regeneration, Houston Methodist Research Institute, Houston, TX, 77030, USA
- Center for RNA Therapeutics, Houston Methodist Research Institute, Houston, TX, 77030, USA
- Department of Cardiothoracic Surgery, Weill Cornell Medicine, Cornell University, New York, NY, 10065, USA
| |
Collapse
|
21
|
Sakai SA, Saeki K, Chi S, Hamaya Y, Du J, Nakamura M, Hojo H, Kojima T, Nakamura Y, Bando H, Kojima M, Suzuki A, Suzuki Y, Akimoto T, Tsuchihara K, Haeno H, Yamashita R, Kageyama SI. Mathematical Modeling Predicts Optimal Immune Checkpoint Inhibitor and Radiotherapy Combinations and Timing of Administration. Cancer Immunol Res 2025; 13:353-364. [PMID: 39666379 PMCID: PMC11876959 DOI: 10.1158/2326-6066.cir-24-0610] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2024] [Revised: 09/04/2024] [Accepted: 12/10/2024] [Indexed: 12/13/2024]
Abstract
Radiotherapy (RT) combined with immune checkpoint inhibitor (ICI) therapy has attracted substantial attention due to its potential to improve outcomes for patients with several types of cancer. However, the optimal administration timepoints and drug combinations remain unclear because the mechanisms underlying RT-induced changes in immune checkpoint molecule expression and interaction with their ligand(s) remain unclear. In this study, we demonstrated the dynamics of lymphocyte-mediated molecular interactions in tissue samples from patients with esophageal cancer throughout RT schedules. Single-cell RNA sequencing and spatial transcriptomic analyses were performed to investigate the dynamics of these interactions. The biological signal in lymphocytes transitioned from innate to adaptive immune reaction, with increases in ligand-receptor interactions, such as PD-1-PD-L1, CTLA4-CD80/86, and TIGIT-PVR interactions. A mathematical model was constructed to predict the efficacy of five types of ICIs when administered at four different timepoints. The model suggested that concurrent anti-PD-1/PD-L1 therapy or concurrent/adjuvant anti-CTLA4/TIGIT therapy would exert a maximal effect with RT. This study provides rationale for clinical trials of RT combined with defined ICI therapy, and these findings will support future studies to search for more effective targets and timing of therapy administration.
Collapse
Affiliation(s)
- Shunsuke A. Sakai
- Division of Translational Informatics, Exploratory Oncology Research and Clinical Trial Center, National Cancer Center, Kashiwa, Japan
- Department of Integrated Biosciences, Graduate School of Frontier Sciences, The University of Tokyo, Chiba, Japan
- Department of Radiation Oncology, National Cancer Center Hospital East, Kashiwa, Japan
| | - Koichi Saeki
- Department of Computational Biology and Medical Sciences, Graduate School of Frontier Sciences, The University of Tokyo, Chiba, Japan
| | - SungGi Chi
- Division of Translational Informatics, Exploratory Oncology Research and Clinical Trial Center, National Cancer Center, Kashiwa, Japan
| | - Yamato Hamaya
- Division of Translational Informatics, Exploratory Oncology Research and Clinical Trial Center, National Cancer Center, Kashiwa, Japan
- Department of Integrated Biosciences, Graduate School of Frontier Sciences, The University of Tokyo, Chiba, Japan
| | - Junyan Du
- Division of Translational Informatics, Exploratory Oncology Research and Clinical Trial Center, National Cancer Center, Kashiwa, Japan
- Department of Integrated Biosciences, Graduate School of Frontier Sciences, The University of Tokyo, Chiba, Japan
| | - Masaki Nakamura
- Department of Radiation Oncology, National Cancer Center Hospital East, Kashiwa, Japan
- Division of Radiation Oncology and Particle Therapy, Exploratory Oncology Research and Clinical Trial Center, National Cancer Center, Kashiwa, Japan
| | - Hidehiro Hojo
- Department of Radiation Oncology, National Cancer Center Hospital East, Kashiwa, Japan
- Division of Radiation Oncology and Particle Therapy, Exploratory Oncology Research and Clinical Trial Center, National Cancer Center, Kashiwa, Japan
| | - Takashi Kojima
- Department of Gastroenterology and Gastrointestinal Oncology, National Cancer Center Hospital East, Kashiwa, Japan
| | - Yoshiaki Nakamura
- Department of Gastroenterology and Gastrointestinal Oncology, National Cancer Center Hospital East, Kashiwa, Japan
- Translational Research Support Office, National Cancer Center Hospital East, Kashiwa, Japan
| | - Hideaki Bando
- Department of Gastroenterology and Gastrointestinal Oncology, National Cancer Center Hospital East, Kashiwa, Japan
- Translational Research Support Office, National Cancer Center Hospital East, Kashiwa, Japan
| | - Motohiro Kojima
- Department of Pathology and Clinical Laboratories, National Cancer Center Hospital East, Kashiwa, Japan
- Pathology Division, Exploratory Oncology Research and Clinical Trial Center, National Cancer Center, Kashiwa, Japan
| | - Ayako Suzuki
- Department of Computational Biology and Medical Sciences, Graduate School of Frontier Sciences, The University of Tokyo, Chiba, Japan
| | - Yutaka Suzuki
- Department of Computational Biology and Medical Sciences, Graduate School of Frontier Sciences, The University of Tokyo, Chiba, Japan
| | - Tetsuo Akimoto
- Department of Radiation Oncology, National Cancer Center Hospital East, Kashiwa, Japan
| | - Katsuya Tsuchihara
- Division of Translational Informatics, Exploratory Oncology Research and Clinical Trial Center, National Cancer Center, Kashiwa, Japan
- Department of Integrated Biosciences, Graduate School of Frontier Sciences, The University of Tokyo, Chiba, Japan
| | - Hiroshi Haeno
- Division of Integrated Research, Research Institute for Biomedical Sciences, Tokyo University of Science, Chiba, Japan
| | - Riu Yamashita
- Division of Translational Informatics, Exploratory Oncology Research and Clinical Trial Center, National Cancer Center, Kashiwa, Japan
- Department of Computational Biology and Medical Sciences, Graduate School of Frontier Sciences, The University of Tokyo, Chiba, Japan
| | - Shun-Ichiro Kageyama
- Department of Radiation Oncology, National Cancer Center Hospital East, Kashiwa, Japan
- Division of Radiation Oncology and Particle Therapy, Exploratory Oncology Research and Clinical Trial Center, National Cancer Center, Kashiwa, Japan
| |
Collapse
|
22
|
Li Y, Hu Q, Han S, Wang-Sattler R, Du W. Multi-Manifolds fusing hyperbolic graph network balanced by pareto optimization for identifying spatial domains of spatial transcriptomics. Brief Bioinform 2025; 26:bbaf162. [PMID: 40220278 PMCID: PMC11992958 DOI: 10.1093/bib/bbaf162] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2024] [Revised: 02/28/2025] [Accepted: 03/19/2025] [Indexed: 04/14/2025] Open
Abstract
Identifying spatial domains for spatial transcriptomics is crucial for achieving comprehensive insights into the pathogenesis of gene expression. Increasingly, computational methods based on graph neural networks are being developed for spatial transcriptomics. However, previous methods have solely focused on the Euclidean manifold. To effectively exploit and explore the informative and deeper topological structures of inherent manifolds, we presented a Multi-Manifolds fusing hyperbolic graph network, balanced by Pareto optimization, for identifying spatial domains in Spatial Transcriptomics (MManiST). First, we developed multi-manifolds encoders for distinct manifolds using the hyperbolic neural network. Features from different manifolds were then combined using an attention mechanism, with multiple reconstruction losses balanced by Pareto optimization. Extensive experiments on commonly used benchmark datasets show that our method consistently outperforms seven state-of-the-art methods. Additionally, we investigated the validity of each component and the impact of fusion methods in ablation experiments.
Collapse
Affiliation(s)
- Ying Li
- Key Laboratory of Symbol Computation and Knowledge Engineering of Ministry of Education, College of Computer Science and Technology, Jilin University, Qianjin Street 2699, Changchun 130012, Jilin, China
| | - Qifeng Hu
- Key Laboratory of Symbol Computation and Knowledge Engineering of Ministry of Education, College of Computer Science and Technology, Jilin University, Qianjin Street 2699, Changchun 130012, Jilin, China
| | - Siyu Han
- TUM School of Medicine, Technical University of Munich, Ismaninger Straße 22, D-81675 Munich, Bavaria, Germany
| | - Rui Wang-Sattler
- Institute of Translational Genomics, Helmholtz Zentrum Munchen, Ingolstadter Landstraße 1, D-85764 Neuherberg, Bavaria, Germany
| | - Wei Du
- Key Laboratory of Symbol Computation and Knowledge Engineering of Ministry of Education, College of Computer Science and Technology, Jilin University, Qianjin Street 2699, Changchun 130012, Jilin, China
| |
Collapse
|
23
|
Liu C, Li X, Hu Q, Jia Z, Ye Q, Wang X, Zhao K, Liu L, Wang M. Decoding the blueprints of embryo development with single-cell and spatial omics. Semin Cell Dev Biol 2025; 167:22-39. [PMID: 39889540 DOI: 10.1016/j.semcdb.2025.01.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2023] [Revised: 01/18/2025] [Accepted: 01/18/2025] [Indexed: 02/03/2025]
Abstract
Embryonic development is a complex and intricately regulated process that encompasses precise control over cell differentiation, morphogenesis, and the underlying gene expression changes. Recent years have witnessed a remarkable acceleration in the development of single-cell and spatial omic technologies, enabling high-throughput profiling of transcriptomic and other multi-omic information at the individual cell level. These innovations offer fresh and multifaceted perspectives for investigating the intricate cellular and molecular mechanisms that govern embryonic development. In this review, we provide an in-depth exploration of the latest technical advancements in single-cell and spatial multi-omic methodologies and compile a systematic catalog of their applications in the field of embryonic development. We deconstruct the research strategies employed by recent studies that leverage single-cell sequencing techniques and underscore the unique advantages of spatial transcriptomics. Furthermore, we delve into both the current applications, data analysis algorithms and the untapped potential of these technologies in advancing our understanding of embryonic development. With the continuous evolution of multi-omic technologies, we anticipate their widespread adoption and profound contributions to unraveling the intricate molecular foundations underpinning embryo development in the foreseeable future.
Collapse
Affiliation(s)
- Chang 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; Shenzhen Proof-of-Concept Center of Digital Cytopathology, BGI Research, Shenzhen 518083, China
| | | | - Qinan Hu
- Shenzhen Key Laboratory of Gene Regulation and Systems Biology, School of Life Sciences, Southern University of Science and Technology, Shenzhen 518005, China; Department of Pharmacology, School of Medicine, Southern University of Science and Technology, Shenzhen 518005, China
| | - Zihan Jia
- BGI Research, Hangzhou 310030, China; College of Life Sciences, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Qing Ye
- BGI Research, Hangzhou 310030, China; China Jiliang University, Hangzhou 310018, China
| | | | - Kaichen Zhao
- College of Biomedicine and Health, College of Life Science and Technology, Huazhong Agricultural University, Wuhan, Hubei 430070, China
| | - Longqi Liu
- BGI Research, Hangzhou 310030, China; Shanxi Medical University-BGI Collaborative Center for Future Medicine, Shanxi Medical University, Taiyuan 030001, China.
| | - Mingyue Wang
- BGI Research, Hangzhou 310030, China; Key Laboratory of Spatial Omics of Zhejiang Province, BGI Research, Hangzhou 310030, China.
| |
Collapse
|
24
|
Lee CYC, McCaffrey J, McGovern D, Clatworthy MR. Profiling immune cell tissue niches in the spatial -omics era. J Allergy Clin Immunol 2025; 155:663-677. [PMID: 39522655 DOI: 10.1016/j.jaci.2024.11.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2024] [Revised: 10/29/2024] [Accepted: 11/04/2024] [Indexed: 11/16/2024]
Abstract
Immune responses require complex, spatially coordinated interactions between immune cells and their tissue environment. For decades, we have imaged tissue sections to visualize a limited number of immune-related macromolecules in situ, functioning as surrogates for cell types or processes of interest. However, this inevitably provides a limited snapshot of the tissue's immune landscape. Recent developments in high-throughput spatial -omics technologies, particularly spatial transcriptomics, and its application to human samples has facilitated a more comprehensive understanding of tissue immunity by mapping fine-grained immune cell states to their precise tissue location while providing contextual information about their immediate cellular and tissue environment. These data provide opportunities to investigate mechanisms underlying the spatial distribution of immune cells and its functional implications, including the identification of immune niches, although the criteria used to define this term have been inconsistent. Here, we review recent technological and analytic advances in multiparameter spatial profiling, focusing on how these methods have generated new insights in translational immunology. We propose a 3-step framework for the definition and characterization of immune niches, which is powerfully facilitated by new spatial profiling methodologies. Finally, we summarize current approaches to analyze adaptive immune repertoires and lymphocyte clonal expansion in a spatially resolved manner.
Collapse
Affiliation(s)
- Colin Y C Lee
- Cambridge Institute of Therapeutic Immunology and Infectious Disease, University of Cambridge, Cambridge, United Kingdom; Cellular Genetics, the Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridge, United Kingdom
| | - James McCaffrey
- Cambridge Institute of Therapeutic Immunology and Infectious Disease, University of Cambridge, Cambridge, United Kingdom; Cellular Genetics, the Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridge, United Kingdom; Cambridge University Hospitals NHS Foundation Trust, Cambridge, United Kingdom
| | - Dominic McGovern
- Cambridge Institute of Therapeutic Immunology and Infectious Disease, University of Cambridge, Cambridge, United Kingdom; Cellular Genetics, the Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridge, United Kingdom; Cambridge University Hospitals NHS Foundation Trust, Cambridge, United Kingdom
| | - Menna R Clatworthy
- Cambridge Institute of Therapeutic Immunology and Infectious Disease, University of Cambridge, Cambridge, United Kingdom; Cellular Genetics, the Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridge, United Kingdom; Cambridge University Hospitals NHS Foundation Trust, Cambridge, United Kingdom.
| |
Collapse
|
25
|
Whittle JR, Kriel J, Fatunla OE, Lu T, Moffet JJD, Spiteri M, Best SA, Freytag S. Spatial omics shed light on the tumour organisation of glioblastoma. Semin Cell Dev Biol 2025; 167:1-9. [PMID: 39787997 DOI: 10.1016/j.semcdb.2024.12.006] [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: 06/27/2024] [Revised: 10/23/2024] [Accepted: 12/30/2024] [Indexed: 01/12/2025]
Abstract
The glioblastoma tumour microenvironment is characterised by immense heterogeneity, with malignant and non-malignant cells that interact in a complex ecosystem. Emerging evidence suggests that the tumour microenvironment is key in facilitating rapid proliferation, invasion, migration and cancer cell survival, crucial for treatment resistance. Spatial omics technologies have enabled the molecular characterisation of regions or individual cells within their spatial context, providing previously unattainable insights into the complex organisation of the glioblastoma tumour microenvironment. Understanding this organisation is crucial for the development of new therapeutics and novel diagnostic tools that guide patient care. This review explores spatial omics technologies and how they have contributed to the development of a model outlining the architecture of the glioblastoma tumour microenvironment.
Collapse
Affiliation(s)
- James R Whittle
- Personalised Oncology Division, The Walter and Eliza Hall Institute of Medical Research, Melbourne, Australia; Department of Medical Biology, University of Melbourne, Melbourne, Australia; Department of Medical Oncology, Peter MacCallum Cancer Centre, Melbourne, Australia
| | - Jurgen Kriel
- Personalised Oncology Division, The Walter and Eliza Hall Institute of Medical Research, Melbourne, Australia; Department of Medical Biology, University of Melbourne, Melbourne, Australia
| | - Oluwaseun E Fatunla
- Personalised Oncology Division, The Walter and Eliza Hall Institute of Medical Research, Melbourne, Australia; Department of Medical Biology, University of Melbourne, Melbourne, Australia
| | - Tianyao Lu
- Personalised Oncology Division, The Walter and Eliza Hall Institute of Medical Research, Melbourne, Australia
| | - Joel J D Moffet
- Personalised Oncology Division, The Walter and Eliza Hall Institute of Medical Research, Melbourne, Australia; Department of Medical Biology, University of Melbourne, Melbourne, Australia
| | - Montana Spiteri
- Personalised Oncology Division, The Walter and Eliza Hall Institute of Medical Research, Melbourne, Australia
| | - Sarah A Best
- Personalised Oncology Division, The Walter and Eliza Hall Institute of Medical Research, Melbourne, Australia; Department of Medical Biology, University of Melbourne, Melbourne, Australia.
| | - Saskia Freytag
- Personalised Oncology Division, The Walter and Eliza Hall Institute of Medical Research, Melbourne, Australia; Department of Medical Biology, University of Melbourne, Melbourne, Australia.
| |
Collapse
|
26
|
Vo T, Prakrithi P, Jones K, Yoon S, Lam PY, Kao Y, Ma N, Tan SX, Jin X, Zhou C, Crawford J, Walters S, Gupta I, Soyer PH, Khosrotehrani K, Stark MS, Nguyen Q. Assessing spatial sequencing and imaging approaches to capture the molecular and pathological heterogeneity of archived cancer tissues. J Pathol 2025; 265:274-288. [PMID: 39846232 PMCID: PMC11794982 DOI: 10.1002/path.6383] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2024] [Revised: 10/10/2024] [Accepted: 11/22/2024] [Indexed: 01/24/2025]
Abstract
Spatial transcriptomics (ST) offers enormous potential to decipher the biological and pathological heterogeneity in precious archival cancer tissues. Traditionally, these tissues have rarely been used and only examined at a low throughput, most commonly by histopathological staining. ST adds thousands of times as many molecular features to histopathological images, but critical technical issues and limitations require more assessment of how ST performs on fixed archival tissues. In this work, we addressed this in a cancer-heterogeneity pipeline, starting with an exploration of the whole transcriptome by two sequencing-based ST protocols capable of measuring coding and non-coding RNAs. We optimised the two protocols to work with challenging formalin-fixed paraffin-embedded (FFPE) tissues, derived from skin. We then assessed alternative imaging methods, including multiplex RNAScope single-molecule imaging and multiplex protein imaging (CODEX). We evaluated the methods' performance for tissues stored from 4 to 14 years ago, covering a range of RNA qualities, allowing us to assess variation. In addition to technical performance metrics, we determined the ability of these methods to quantify tumour heterogeneity. We integrated gene expression profiles with pathological information, charting a new molecular landscape on the pathologically defined tissue regions. Together, this work provides important and comprehensive experimental technical perspectives to consider the applications of ST in deciphering the cancer heterogeneity in archived tissues. © 2025 The Author(s). The Journal of Pathology published by John Wiley & Sons Ltd on behalf of The Pathological Society of Great Britain and Ireland.
Collapse
Affiliation(s)
- Tuan Vo
- The Institute for Molecular BioscienceThe University of QueenslandSt LuciaQueenslandAustralia
- Queensland Institute of Medical ResearchHerstonQueenslandAustralia
- School of Biomedical Sciences and PharmacyUniversity of NewcastleCallaghanNew South WalesAustralia
- Precision Medicine Research ProgramHunter Medical Research InstituteNew Lambton HeightsNew South WalesAustralia
| | - P Prakrithi
- The Institute for Molecular BioscienceThe University of QueenslandSt LuciaQueenslandAustralia
- Queensland Institute of Medical ResearchHerstonQueenslandAustralia
- University of Queensland – IIT Delhi Research Academy (UQIDRA)New DelhiIndia
- Department of Biochemical Engineering and BiotechnologyIndian Institute of Technology DelhiNew DelhiIndia
| | - Kahli Jones
- The Institute for Molecular BioscienceThe University of QueenslandSt LuciaQueenslandAustralia
| | - Sohye Yoon
- Genome Innovation HubThe University of QueenslandSt LuciaQueenslandAustralia
| | - Pui Yeng Lam
- The Institute for Molecular BioscienceThe University of QueenslandSt LuciaQueenslandAustralia
| | - Yung‐Ching Kao
- Dermatology Research CentreThe Frazer Institute, The University of QueenslandWoolloongabbaQueenslandAustralia
| | - Ning Ma
- Akoya Biosciences IncMarlboroughMassachusettsUSA
| | - Samuel X Tan
- Dermatology Research CentreThe Frazer Institute, The University of QueenslandWoolloongabbaQueenslandAustralia
| | - Xinnan Jin
- The Institute for Molecular BioscienceThe University of QueenslandSt LuciaQueenslandAustralia
- Queensland Institute of Medical ResearchHerstonQueenslandAustralia
| | - Chenhao Zhou
- Dermatology Research CentreThe Frazer Institute, The University of QueenslandWoolloongabbaQueenslandAustralia
| | - Joanna Crawford
- The Institute for Molecular BioscienceThe University of QueenslandSt LuciaQueenslandAustralia
| | - Shaun Walters
- School of Biomedical SciencesThe University of QueenslandSt LuciaQueenslandAustralia
| | - Ishaan Gupta
- University of Queensland – IIT Delhi Research Academy (UQIDRA)New DelhiIndia
- Department of Biochemical Engineering and BiotechnologyIndian Institute of Technology DelhiNew DelhiIndia
| | - Peter H Soyer
- Dermatology Research CentreThe Frazer Institute, The University of QueenslandWoolloongabbaQueenslandAustralia
| | - Kiarash Khosrotehrani
- Dermatology Research CentreThe Frazer Institute, The University of QueenslandWoolloongabbaQueenslandAustralia
| | - Mitchell S Stark
- Dermatology Research CentreThe Frazer Institute, The University of QueenslandWoolloongabbaQueenslandAustralia
| | - Quan Nguyen
- The Institute for Molecular BioscienceThe University of QueenslandSt LuciaQueenslandAustralia
- Queensland Institute of Medical ResearchHerstonQueenslandAustralia
| |
Collapse
|
27
|
Zuo Y, Jin Y, Li G, Ming Y, Fan T, Pan Y, Yao X, Peng Y. Spatial transcriptomic analysis of tumor microenvironment in esophageal squamous cell carcinoma with HIV infection. Mol Cancer 2025; 24:54. [PMID: 39994631 PMCID: PMC11853777 DOI: 10.1186/s12943-025-02248-3] [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: 12/02/2024] [Accepted: 01/27/2025] [Indexed: 02/26/2025] Open
Abstract
BACKGROUND Human Immunodeficiency Virus (HIV) is one of the most prevalent viruses, causing significant immune depletion in affected individuals. Current treatments can control HIV and prolong patients' lives, but new challenges have emerged. Increasing incidence of cancers occur in HIV patients. Esophageal squamous cell carcinoma (ESCC) is one of the most common cancers observed in HIV patients. However, the spatial cellular characteristics of HIV-related ESCC have not been explored, and the differences between HIV-ESCC and typical ESCC remain unclear. METHODS We performed spatial transcriptome sequencing on HIV-ESCC samples to depict the microenvironment and employed cell communication analysis and multiplex immunofluorescence to investigate the molecular mechanism in HIV-ESCC. RESULTS We found that HIV-ESCC exhibited a unique cellular composition, with fibroblasts and epithelial cells intermixed throughout the tumor tissue, lacking obvious spatial separation, while other cell types were sparse. Besides, HIV-ESCC exhibited an immune desert phenotype, characterized by a low degree of immune cell infiltration, with only a few SPP1+ macrophages showing immune resistance functions. Cell communication analysis and multiplex immunofluorescence staining revealed that tumor fibroblasts in HIV-ESCC interact with CD44+ epithelial cells via COL1A2, promoting the expression of PIK3R1 in epithelial cells. This interaction activates the PI3K-AKT signaling pathway, which contributes to the progression of HIV-ESCC. CONCLUSIONS Our findings depict the spatial microenvironment of HIV-ESCC and elucidate a molecular mechanism in the progression of HIV-ESCC. This will provide us insights into the molecular basis of HIV-ESCC and potential treatment strategies.
Collapse
Affiliation(s)
- Yuanli Zuo
- Center for Molecular Oncology, Frontiers Science Center for Disease-Related Molecular Network, State Key Laboratory of Respiratory Health and Multimorbidity, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Yang Jin
- Center for Molecular Oncology, Frontiers Science Center for Disease-Related Molecular Network, State Key Laboratory of Respiratory Health and Multimorbidity, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Gang Li
- Department of Thoracic Surgery, The Public Health Clinical Center of Chengdu, Chengdu, 610061, China
| | - Yue Ming
- Center for Molecular Oncology, Frontiers Science Center for Disease-Related Molecular Network, State Key Laboratory of Respiratory Health and Multimorbidity, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Ting Fan
- Center for Molecular Oncology, Frontiers Science Center for Disease-Related Molecular Network, State Key Laboratory of Respiratory Health and Multimorbidity, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Yitong Pan
- Center for Molecular Oncology, Frontiers Science Center for Disease-Related Molecular Network, State Key Laboratory of Respiratory Health and Multimorbidity, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Xiaojun Yao
- Department of Thoracic Surgery, The Public Health Clinical Center of Chengdu, Chengdu, 610061, China.
| | - Yong Peng
- Center for Molecular Oncology, Frontiers Science Center for Disease-Related Molecular Network, State Key Laboratory of Respiratory Health and Multimorbidity, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, 610041, China.
| |
Collapse
|
28
|
Shen X, Zuo L, Ye Z, Yuan Z, Huang K, Li Z, Yu Q, Zou X, Wei X, Xu P, Deng Y, Jin X, Xu X, Wu L, Zhu H, Qin P. Inferring cell trajectories of spatial transcriptomics via optimal transport analysis. Cell Syst 2025; 16:101194. [PMID: 39904341 DOI: 10.1016/j.cels.2025.101194] [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: 12/10/2023] [Revised: 09/30/2024] [Accepted: 01/10/2025] [Indexed: 02/06/2025]
Abstract
The integration of cell transcriptomics and spatial position to organize differentiation trajectories remains a challenge. Here, we introduce SpaTrack, which leverages optimal transport to reconcile both gene expression and spatial position from spatial transcriptomics into the transition costs, thereby reconstructing cell differentiation. SpaTrack can construct detailed spatial trajectories that reflect the differentiation topology and trace cell dynamics across multiple samples over temporal intervals. To capture the dynamic drivers of differentiation, SpaTrack models cell fate as a function of expression profiles influenced by transcription factors over time. By applying SpaTrack, we successfully disentangle spatiotemporal trajectories of axolotl telencephalon regeneration and mouse midbrain development. Diverse malignant lineages expanding within a primary tumor are uncovered. One lineage, characterized by upregulated epithelial mesenchymal transition, implants at the metastatic site and subsequently colonizes to form a secondary tumor. Overall, SpaTrack efficiently advances trajectory inference from spatial transcriptomics, providing valuable insights into differentiation processes.
Collapse
Affiliation(s)
- Xunan Shen
- BGI Research, Chongqing 401329, China; BGI Research, Beijing 102601, China
| | | | | | - Zhongyang Yuan
- BGI Research, Chongqing 401329, China; State Key Laboratory of Medicinal Chemical Biology, College of Life Sciences, Nankai University, Tianjin 300071, China
| | - Ke Huang
- BGI Research, Chongqing 401329, China
| | - Zeyu Li
- BGI Research, Chongqing 401329, China
| | - Qichao Yu
- BGI Research, Chongqing 401329, China
| | - Xuanxuan Zou
- BGI Research, Chongqing 401329, China; Department of Neurology, Hubei Provincial Clinical Research Center for Parkinson's Disease, Xiangyang No. 1 People's Hospital, Hubei University of Medicine, Xiangyang 441000, China
| | | | - Ping Xu
- BGI Research, Chongqing 401329, China; BGI College & Henan Institute of Medical and Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450000, China
| | - Yaqi Deng
- Key Laboratory of Major Brain Disease and Aging Research (Ministry of Education), Institute for Brain Science and Disease, Chongqing Medical University, Chongqing, China
| | - Xin Jin
- BGI Research, Shenzhen 518083, China
| | - Xun Xu
- BGI Research, Shenzhen 518083, China.
| | - Liang Wu
- BGI Research, Chongqing 401329, China; BGI Research, Shenzhen 518083, China.
| | | | - Pengfei Qin
- BGI Research, Chongqing 401329, China; BGI Research, Shenzhen 518083, China.
| |
Collapse
|
29
|
Kordowski A, Mulay O, Tan X, Vo T, Baumgartner U, Maybury MK, Hassall T, Harris L, Nguyen Q, Day BW. Spatial analysis of a complete DIPG-infiltrated brainstem reveals novel ligand-receptor mediators of tumour-to-TME crosstalk. Acta Neuropathol Commun 2025; 13:35. [PMID: 39972389 PMCID: PMC11837654 DOI: 10.1186/s40478-025-01952-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2024] [Accepted: 02/09/2025] [Indexed: 02/21/2025] Open
Abstract
Previous studies have highlighted the capacity of brain cancer cells to functionally interact with the tumour microenvironment (TME). This TME-cancer crosstalk crucially contributes to tumour cell invasion and disease progression. In this study, we performed spatial transcriptomic sequencing analysis of a complete annotated tumour-infiltrated brainstem from a single diffuse intrinsic pontine glioma (DIPG) patient. Gene signatures from ten sequential tumour regions were analysed to assess mechanisms of disease progression and oncogenic interactions with the TME. We identified four distinct tumour subpopulations and assessed respective ligand-receptor pairs that actively promote DIPG tumour progression via crosstalk with endothelial, neuronal and immune cell communities. Our analysis found potential targetable mediators of tumour-to-TME communication, including members of the complement component system and the neuropeptide/GPCR ligand-receptor pair ADCYAP1-ADCYAP1R1. These interactions could influence DIPG tumour progression and represent novel therapeutic targets.
Collapse
Affiliation(s)
- Anja Kordowski
- Queensland Institute for Medical Research, Brisbane, QLD, 4006, Australia.
| | - Onkar Mulay
- Queensland Institute for Medical Research, Brisbane, QLD, 4006, Australia
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, QLD, 4067, Australia
| | - Xiao Tan
- Queensland Institute for Medical Research, Brisbane, QLD, 4006, Australia
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, QLD, 4067, Australia
| | - Tuan Vo
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, QLD, 4067, Australia
- Hunter Medical Research Institute, Precision Medicine Research Program, Newcastle, NSW, 2305, Australia
- School of Biomedical Sciences and Pharmacy, The University of Newcastle, Newcastle, NSW, 2308, Australia
| | - Ulrich Baumgartner
- School of Biomedical Sciences, The University of Queensland, Brisbane, QLD, 4067, Australia
| | - Mellissa K Maybury
- Child Health Research Centre, The University of Queensland, Brisbane, QLD, 4101, Australia
| | - Timothy Hassall
- Children's Brain Cancer Centre, UQ Frazer Institute, Brisbane, QLD, 4102, Australia
- Queensland Children's Hospital, Brisbane, QLD, 4101, Australia
| | - Lachlan Harris
- Queensland Institute for Medical Research, Brisbane, QLD, 4006, Australia
- School of Biomedical Sciences, The University of Queensland, Brisbane, QLD, 4067, Australia
- School of Biomedical Sciences, Queensland University of Technology, Brisbane, QLD, 4059, Australia
| | - Quan Nguyen
- Queensland Institute for Medical Research, Brisbane, QLD, 4006, Australia
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, QLD, 4067, Australia
| | - Bryan W Day
- Queensland Institute for Medical Research, Brisbane, QLD, 4006, Australia.
- School of Biomedical Sciences, The University of Queensland, Brisbane, QLD, 4067, Australia.
- Children's Brain Cancer Centre, UQ Frazer Institute, Brisbane, QLD, 4102, Australia.
| |
Collapse
|
30
|
Zeng Y, Jiang Z. Analysis of fatty acid metabolism in prostate cancer and discovery of a new programmed cell death-associated luminal cell subpopulation. Discov Oncol 2025; 16:194. [PMID: 39961915 PMCID: PMC11832849 DOI: 10.1007/s12672-025-01982-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/15/2024] [Accepted: 02/12/2025] [Indexed: 02/20/2025] Open
Abstract
INTRODUCTION This study focuses on the role of fatty acid metabolism in prostate cancer, particularly in oncogenic luminal cells associated with programmed cell death under the influence of metabolic reprogramming. MATERIALS AND METHODS Prostate cancer was analyzed using single-cell transcriptomics and spatial transcriptomics data. Fatty acid metabolism levels in the tumor microenvironment were quantified by multiple gene set scoring methods, and data were processed using NMF and deconvolution methods to identify different cell populations and their interactions in the tumor microenvironment. RESULTS Luminal cells have significantly increased activity in fatty acid metabolism, which is associated with the aggressiveness and metastatic capability of tumors. Luminal cell subpopulations have been found to play a key role in the development of prostate cancer, especially their close association with programmed cell death. CONCLUSION This study deepens the understanding of the role of fatty acid metabolism in prostate cancer, identifies fatty acid metabolism-related luminal cell subtypes, and proposes new therapeutic targets, providing new insights into prostate cancer treatment.
Collapse
Affiliation(s)
- Yan Zeng
- Department of Laboratory Medicine, Traditional Chinese Medicine Hospital of Emeishan City, Emeishan, 614000, China.
| | - Zhaolin Jiang
- Clinical Medical College, North Sichuan Medical College, NanChong, 637000, China
| |
Collapse
|
31
|
Zhang H, Zhang Y, Ting KM, Zhang J, Zhao Q. Kernel-bounded clustering for spatial transcriptomics enables scalable discovery of complex spatial domains. Genome Res 2025; 35:355-367. [PMID: 39909714 PMCID: PMC11874963 DOI: 10.1101/gr.278983.124] [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: 01/17/2024] [Accepted: 12/19/2024] [Indexed: 02/07/2025]
Abstract
Spatial transcriptomics are a collection of technologies that have enabled characterization of gene expression profiles and spatial information in tissue samples. Existing methods for clustering spatial transcriptomics data have primarily focused on data transformation techniques to represent the data suitably for subsequent clustering analysis, often using an existing clustering algorithm. These methods have limitations in handling complex data characteristics with varying densities, sizes, and shapes (in the transformed space on which clustering is performed), and they have high computational complexity, resulting in unsatisfactory clustering outcomes and slow execution time even with GPUs. Rather than focusing on data transformation techniques, we propose a new clustering algorithm called kernel-bounded clustering (KBC). It has two unique features: (1) It is the first clustering algorithm that employs a distributional kernel to recruit members of a cluster, enabling clusters of varying densities, sizes, and shapes to be discovered, and (2) it is a linear-time clustering algorithm that significantly enhances the speed of clustering analysis, enabling researchers to effectively handle large-scale spatial transcriptomics data sets. We show that (1) KBC works well with a simple data transformation technique called the Weisfeiler-Lehman scheme, and (2) a combination of KBC and the Weisfeiler-Lehman scheme produces good clustering outcomes, and it is faster and easier-to-use than many methods that employ existing clustering algorithms and data transformation techniques.
Collapse
Affiliation(s)
- Hang Zhang
- National Key Laboratory for Novel Software Technology, Nanjing University, Nanjing 210023, China
- School of Artificial Intelligence, Nanjing University, Nanjing 210023, China
| | - Yi Zhang
- National Key Laboratory for Novel Software Technology, Nanjing University, Nanjing 210023, China
- School of Artificial Intelligence, Nanjing University, Nanjing 210023, China
| | - Kai Ming Ting
- National Key Laboratory for Novel Software Technology, Nanjing University, Nanjing 210023, China;
- School of Artificial Intelligence, Nanjing University, Nanjing 210023, China
| | - Jie Zhang
- National Key Laboratory for Novel Software Technology, Nanjing University, Nanjing 210023, China;
- School of Artificial Intelligence, Nanjing University, Nanjing 210023, China
| | - Qiuran Zhao
- National Key Laboratory for Novel Software Technology, Nanjing University, Nanjing 210023, China
- School of Artificial Intelligence, Nanjing University, Nanjing 210023, China
| |
Collapse
|
32
|
Sun P, Bush SJ, Wang S, Jia P, Li M, Xu T, Zhang P, Yang X, Wang C, Xu L, Wang T, Ye K. STMiner: Gene-centric spatial transcriptomics for deciphering tumor tissues. CELL GENOMICS 2025; 5:100771. [PMID: 39947134 PMCID: PMC11872602 DOI: 10.1016/j.xgen.2025.100771] [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: 09/14/2024] [Revised: 12/09/2024] [Accepted: 01/17/2025] [Indexed: 03/05/2025]
Abstract
Analyzing spatial transcriptomics data from tumor tissues poses several challenges beyond those of healthy samples, including unclear boundaries between different regions, uneven cell densities, and relatively higher cellular heterogeneity. Collectively, these bias the background against which spatially variable genes are identified, which can result in misidentification of spatial structures and hinder potential insight into complex pathologies. To overcome this problem, STMiner leverages 2D Gaussian mixture models and optimal transport theory to directly characterize the spatial distribution of genes rather than the capture locations of the cells expressing them (spots). By effectively mitigating the impacts of both background bias and data sparsity, STMiner reveals key gene sets and spatial structures overlooked by spot-based analytic tools, facilitating novel biological discoveries. The core concept of directly analyzing overall gene expression patterns also allows for a broader application beyond spatial transcriptomics, positioning STMiner for continuous expansion as spatial omics technologies evolve.
Collapse
Affiliation(s)
- Peisen Sun
- School of Automation Science and Engineering, Faculty of Electronic and Information Engineering, Xi'an Jiaotong University, Xi'an, China; MOE Key Lab for Intelligent Networks & Networks Security, Faculty of Electronic and Information Engineering, Xi'an Jiaotong University, Xi'an, China
| | - Stephen J Bush
- School of Automation Science and Engineering, Faculty of Electronic and Information Engineering, Xi'an Jiaotong University, Xi'an, China; MOE Key Lab for Intelligent Networks & Networks Security, Faculty of Electronic and Information Engineering, Xi'an Jiaotong University, Xi'an, China
| | - Songbo Wang
- School of Automation Science and Engineering, Faculty of Electronic and Information Engineering, Xi'an Jiaotong University, Xi'an, China; MOE Key Lab for Intelligent Networks & Networks Security, Faculty of Electronic and Information Engineering, Xi'an Jiaotong University, Xi'an, China
| | - Peng Jia
- School of Automation Science and Engineering, Faculty of Electronic and Information Engineering, Xi'an Jiaotong University, Xi'an, China; MOE Key Lab for Intelligent Networks & Networks Security, Faculty of Electronic and Information Engineering, Xi'an Jiaotong University, Xi'an, China; Department of Gynecology and Obstetrics, Center for Mathematical Medical, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Mingxuan Li
- School of Automation Science and Engineering, Faculty of Electronic and Information Engineering, Xi'an Jiaotong University, Xi'an, China; MOE Key Lab for Intelligent Networks & Networks Security, Faculty of Electronic and Information Engineering, Xi'an Jiaotong University, Xi'an, China
| | - Tun Xu
- School of Automation Science and Engineering, Faculty of Electronic and Information Engineering, Xi'an Jiaotong University, Xi'an, China; MOE Key Lab for Intelligent Networks & Networks Security, Faculty of Electronic and Information Engineering, Xi'an Jiaotong University, Xi'an, China
| | - Pengyu Zhang
- School of Automation Science and Engineering, Faculty of Electronic and Information Engineering, Xi'an Jiaotong University, Xi'an, China; MOE Key Lab for Intelligent Networks & Networks Security, Faculty of Electronic and Information Engineering, Xi'an Jiaotong University, Xi'an, China
| | - Xiaofei Yang
- MOE Key Lab for Intelligent Networks & Networks Security, Faculty of Electronic and Information Engineering, Xi'an Jiaotong University, Xi'an, China; School of Computer Science and Technology, Faculty of Electronic and Information Engineering, Xi'an Jiaotong University, Xi'an, China
| | - Chengyao Wang
- Department of Endocrinology, Genome Institute, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Linfeng Xu
- School of Automation Science and Engineering, Faculty of Electronic and Information Engineering, Xi'an Jiaotong University, Xi'an, China; MOE Key Lab for Intelligent Networks & Networks Security, Faculty of Electronic and Information Engineering, Xi'an Jiaotong University, Xi'an, China
| | - Tingjie Wang
- The Affiliated Cancer Hospital of Zhengzhou University, Zhengzhou, China
| | - Kai Ye
- School of Automation Science and Engineering, Faculty of Electronic and Information Engineering, Xi'an Jiaotong University, Xi'an, China; MOE Key Lab for Intelligent Networks & Networks Security, Faculty of Electronic and Information Engineering, Xi'an Jiaotong University, Xi'an, China; Department of Gynecology and Obstetrics, Center for Mathematical Medical, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China; Genome Institute, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China; Faculty of Science, Leiden University, Leiden, the Netherlands.
| |
Collapse
|
33
|
Ji L, Wang D, Zhuo G, Chen Z, Wang L, Zhang Q, Wan Y, Liu G, Pan Y. Spatial Metabolomics and Transcriptomics Reveal Metabolic Reprogramming and Cellular Interactions in Nasopharyngeal Carcinoma with High PD-1 Expression and Therapeutic Response. Theranostics 2025; 15:3035-3054. [PMID: 40083932 PMCID: PMC11898293 DOI: 10.7150/thno.102822] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2024] [Accepted: 01/29/2025] [Indexed: 03/16/2025] Open
Abstract
Nasopharyngeal carcinoma (NPC) is a heterogeneous cancer with variable therapeutic responses, highlighting the need to better understand the molecular factors influencing treatment outcomes. This study aims to explore spatially metabolic and gene expression alterations in NPC patients with different therapeutic responses and PD-1 expression levels. Methods: This study employs spatial metabolomics (SM) and spatial transcriptomics (ST) to investigate significant alterations in metabolic pathways and metabolites in NPC patients exhibiting therapeutic sensitivity or elevated programmed death 1 (PD-1) expression. The spatial distribution of various cell types within the TME and their complex interactions were also investigated. Identified prognostic targets were validated using public datasets from TCGA, and further substantiated by in vitro functional analyses. Results: SM analysis revealed substantial reprogramming in lipid metabolism, branched-chain amino acid (BCAA) metabolism, and glutamine metabolism, which were closely associated with therapeutic response and PD-1 expression. ST analysis highlighted the critical role of interactions between precursor T cells and malignant epithelial cells in modulating therapeutic response in NPC. Notably, six key genes involved in BCAA metabolism (IL4I1, OXCT1, BCAT2, DLD, ALDH1B1, HADH) were identified in distinguishing patients with therapeutic sensitivity from those with therapeutic resistance. Functional validation of DLD and IL4I1 revealed that gene silencing significantly inhibited NPC cell proliferation, colony formation, wound healing, and invasion. Silencing DLD or IL4I1 induced cell cycle arrest. Reduction in α-Ketomethylvaleric acid (KMV) levels was demonstrated upon IL4I1 silencing. Immunohistochemical analysis further confirmed that high expression of these six genes was significantly associated with poor prognosis in NPC patients, a trend corroborated by data from the TCGA head and neck cancer cohort. Conclusions: This study highlights the pivotal roles of key molecular players in therapeutic response in NPC, providing compelling evidence for their potential application as prognostic biomarkers and therapeutic targets, thereby contributing to precision oncology strategies aimed at improving patient outcomes.
Collapse
Affiliation(s)
- Lili Ji
- Department of Laboratory Medicine, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan 430071, China
- Hubei Provincial Clinical Research Center for Molecular Diagnostics, Wuhan 430071, China
| | - Dujuan Wang
- Department of Clinical Pathology, Houjie Hospital of Dongguan, The Affiliated Houjie Hospital of Guangdong Medical University, Dongguan 523960, China
| | - Guangzheng Zhuo
- Department of Laboratory Medicine, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan 430071, China
| | - Zhe Chen
- Department of Otolaryngology Head and Neck Surgery, Zhongnan Hospital of Wuhan University, Wuhan 430071, China
| | - Liping Wang
- Department of Laboratory Medicine, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan 430071, China
- Hubei Provincial Clinical Research Center for Molecular Diagnostics, Wuhan 430071, China
| | - Qian Zhang
- Department of Laboratory Medicine, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan 430071, China
| | - Yuhang Wan
- Department of Laboratory Medicine, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan 430071, China
| | - Guohong Liu
- Department of Radiology, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan 430071, China
| | - Yunbao Pan
- Department of Laboratory Medicine, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan 430071, China
- Hubei Provincial Clinical Research Center for Molecular Diagnostics, Wuhan 430071, China
- Wuhan Research Center for Infectious Diseases and Tumors of the Chinese Academy of Medical Sciences, Wuhan 430071, China
| |
Collapse
|
34
|
Chitra U, Arnold BJ, Sarkar H, Sanno K, Ma C, Lopez-Darwin S, Raphael BJ. Mapping the topography of spatial gene expression with interpretable deep learning. Nat Methods 2025; 22:298-309. [PMID: 39849132 DOI: 10.1038/s41592-024-02503-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2023] [Accepted: 10/14/2024] [Indexed: 01/25/2025]
Abstract
Spatially resolved transcriptomics technologies provide high-throughput measurements of gene expression in a tissue slice, but the sparsity of these data complicates analysis of spatial gene expression patterns. We address this issue by deriving a topographic map of a tissue slice-analogous to a map of elevation in a landscape-using a quantity called the isodepth. Contours of constant isodepths enclose domains with distinct cell type composition, while gradients of the isodepth indicate spatial directions of maximum change in expression. We develop GASTON (gradient analysis of spatial transcriptomics organization with neural networks), an unsupervised and interpretable deep learning algorithm that simultaneously learns the isodepth, spatial gradients and piecewise linear expression functions that model both continuous gradients and discontinuous variation in gene expression. We show that GASTON accurately identifies spatial domains and marker genes across several tissues, gradients of neuronal differentiation and firing in the brain, and gradients of metabolism and immune activity in the tumor microenvironment.
Collapse
Affiliation(s)
- Uthsav Chitra
- Department of Computer Science, Princeton University, Princeton, NJ, USA
| | - Brian J Arnold
- Department of Computer Science, Princeton University, Princeton, NJ, USA
- Center for Statistics and Machine Learning, Princeton University, Princeton, NJ, USA
| | - Hirak Sarkar
- Department of Computer Science, Princeton University, Princeton, NJ, USA
- Ludwig Cancer Institute, Princeton Branch, Princeton University, Princeton, NJ, USA
| | - Kohei Sanno
- Department of Computer Science, Princeton University, Princeton, NJ, USA
| | - Cong Ma
- Department of Computer Science, Princeton University, Princeton, NJ, USA
| | - Sereno Lopez-Darwin
- Lewis-Sigler Institute for Integrative Genomics, Princeton University, Princeton, NJ, USA
| | - Benjamin J Raphael
- Department of Computer Science, Princeton University, Princeton, NJ, USA.
| |
Collapse
|
35
|
Sakai SA, Nomura R, Nagasawa S, Chi S, Suzuki A, Suzuki Y, Imai M, Nakamura Y, Yoshino T, Ishikawa S, Tsuchihara K, Kageyama SI, Yamashita R. SpatialKNifeY (SKNY): Extending from spatial domain to surrounding area to identify microenvironment features with single-cell spatial omics data. PLoS Comput Biol 2025; 21:e1012854. [PMID: 39965034 PMCID: PMC11849985 DOI: 10.1371/journal.pcbi.1012854] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2024] [Revised: 02/24/2025] [Accepted: 02/03/2025] [Indexed: 02/20/2025] Open
Abstract
Single-cell spatial omics analysis requires consideration of biological functions and mechanisms in a microenvironment. However, microenvironment analysis using bioinformatic methods is limited by the need to detect histological morphology and extend it to the surrounding area. In this study, we developed SpatialKNifeY (SKNY), an image-processing-based toolkit that detects spatial domains that potentially reflect histology and extends these domains to the microenvironment. Using spatial transcriptomic data from breast cancer, we applied the SKNY algorithm to identify tumor spatial domains, followed by clustering of the domains, trajectory estimation, and spatial extension to the tumor microenvironment (TME). The results of the trajectory estimation were consistent with the known mechanisms of cancer progression. We observed tumor vascularization and immunodeficiency at mid- and late-stage progression in TME. Furthermore, we applied the SKNY to integrate and cluster the spatial domains of 14 patients with metastatic colorectal cancer, and the clusters were divided based on the TME characteristics. In conclusion, the SKNY facilitates the determination of the functions and mechanisms in the microenvironment and cataloguing of the features.
Collapse
Affiliation(s)
- Shunsuke A. Sakai
- Division of Translational Informatics, Exploratory Oncology Research & Clinical Trial Center, National Cancer Center, Kashiwa, Chiba, Japan
- Department of Integrated Biosciences, Graduate School of Frontier Sciences, The University of Tokyo, Kashiwa, Chiba, Japan
- Department of Radiation Oncology, National Cancer Center Hospital East, Kashiwa, Chiba, Japan
| | - Ryosuke Nomura
- Division of Translational Informatics, Exploratory Oncology Research & Clinical Trial Center, National Cancer Center, Kashiwa, Chiba, Japan
- Department of Computational Biology and Medical Sciences, Graduate School of Frontier Sciences, The University of Tokyo, Kashiwa, Chiba, Japan
| | - Satoi Nagasawa
- Department of Computational Biology and Medical Sciences, Graduate School of Frontier Sciences, The University of Tokyo, Kashiwa, Chiba, Japan
- Department of Breast Surgery, National Cancer Center Hospital East, Kashiwa, Chiba, Japan
| | - SungGi Chi
- Division of Translational Informatics, Exploratory Oncology Research & Clinical Trial Center, National Cancer Center, Kashiwa, Chiba, Japan
| | - Ayako Suzuki
- Department of Computational Biology and Medical Sciences, Graduate School of Frontier Sciences, The University of Tokyo, Kashiwa, Chiba, Japan
| | - Yutaka Suzuki
- Department of Computational Biology and Medical Sciences, Graduate School of Frontier Sciences, The University of Tokyo, Kashiwa, Chiba, Japan
| | - Mitsuho Imai
- Translational Research Support Office, National Cancer Center Hospital East, Chiba, Japan
- Department of Genetic Medicine and Services, National Cancer Center Hospital East, Chiba, Japan
| | - Yoshiaki Nakamura
- Translational Research Support Office, National Cancer Center Hospital East, Chiba, Japan
- Department of Gastroenterology and Gastrointestinal Oncology, National Cancer Center Hospital East, Chiba, Japan
| | - Takayuki Yoshino
- Translational Research Support Office, National Cancer Center Hospital East, Chiba, Japan
- Department of Gastroenterology and Gastrointestinal Oncology, National Cancer Center Hospital East, Chiba, Japan
| | - Shumpei Ishikawa
- Department of Preventive Medicine, Graduate School of Medicine, The University of Tokyo, Bunkyo-ku, Tokyo, Japan
- Division of Pathology, National Cancer Center Exploratory Oncology Research & Clinical Trial Center, Kashiwa, Chiba, Japan
| | - Katsuya Tsuchihara
- Division of Translational Informatics, Exploratory Oncology Research & Clinical Trial Center, National Cancer Center, Kashiwa, Chiba, Japan
- Department of Integrated Biosciences, Graduate School of Frontier Sciences, The University of Tokyo, Kashiwa, Chiba, Japan
| | - Shun-Ichiro Kageyama
- Department of Radiation Oncology, National Cancer Center Hospital East, Kashiwa, Chiba, Japan
- Division of Radiation Oncology and Particle Therapy, Exploratory Oncology Research & Clinical Trial Center, National Cancer Center, Kashiwa, Chiba, Japan
| | - Riu Yamashita
- Division of Translational Informatics, Exploratory Oncology Research & Clinical Trial Center, National Cancer Center, Kashiwa, Chiba, Japan
- Department of Computational Biology and Medical Sciences, Graduate School of Frontier Sciences, The University of Tokyo, Kashiwa, Chiba, Japan
| |
Collapse
|
36
|
Yan G, Hua SH, Li JJ. Categorization of 34 computational methods to detect spatially variable genes from spatially resolved transcriptomics data. Nat Commun 2025; 16:1141. [PMID: 39880807 PMCID: PMC11779979 DOI: 10.1038/s41467-025-56080-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2024] [Accepted: 01/06/2025] [Indexed: 01/31/2025] Open
Abstract
In the analysis of spatially resolved transcriptomics data, detecting spatially variable genes (SVGs) is crucial. Numerous computational methods exist, but varying SVG definitions and methodologies lead to incomparable results. We review 34 state-of-the-art methods, classifying SVGs into three categories: overall, cell-type-specific, and spatial-domain-marker SVGs. Our review explains the intuitions underlying these methods, summarizes their applications, and categorizes the hypothesis tests they use in the trade-off between generality and specificity for SVG detection. We discuss challenges in SVG detection and propose future directions for improvement. Our review offers insights for method developers and users, advocating for category-specific benchmarking.
Collapse
Affiliation(s)
- Guanao Yan
- Department of Statistics and Data Science, University of California, Los Angeles, CA, 90095-1554, USA
| | - Shuo Harper Hua
- Department of Biomedical Data Science, Stanford University, Stanford, CA, 94305, USA
| | - Jingyi Jessica Li
- Department of Statistics and Data Science, University of California, Los Angeles, CA, 90095-1554, USA.
- Department of Human Genetics, University of California, Los Angeles, CA, 90095-7088, USA.
- Department of Computational Medicine, University of California, Los Angeles, CA, 90095-1766, USA.
- Department of Biostatistics, University of California, Los Angeles, CA, 90095-1772, USA.
- Radcliffe Institute for Advanced Study, Harvard University, Cambridge, MA, 02138, USA.
| |
Collapse
|
37
|
Yang Y, Cui Y, Zeng X, Zhang Y, Loza M, Park SJ, Nakai K. STAIG: Spatial transcriptomics analysis via image-aided graph contrastive learning for domain exploration and alignment-free integration. Nat Commun 2025; 16:1067. [PMID: 39870633 PMCID: PMC11772580 DOI: 10.1038/s41467-025-56276-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2024] [Accepted: 01/06/2025] [Indexed: 01/29/2025] Open
Abstract
Spatial transcriptomics is an essential application for investigating cellular structures and interactions and requires multimodal information to precisely study spatial domains. Here, we propose STAIG, a deep-learning model that integrates gene expression, spatial coordinates, and histological images using graph-contrastive learning coupled with high-performance feature extraction. STAIG can integrate tissue slices without prealignment and remove batch effects. Moreover, it is designed to accept data acquired from various platforms, with or without histological images. By performing extensive benchmarks, we demonstrate the capability of STAIG to recognize spatial regions with high precision and uncover new insights into tumor microenvironments, highlighting its promising potential in deciphering spatial biological intricates.
Collapse
Affiliation(s)
- Yitao Yang
- Department of Computational Biology and Medical Science, Graduate School of Frontier Sciences, the University of Tokyo, Tokyo, Japan
| | - Yang Cui
- Department of Computational Biology and Medical Science, Graduate School of Frontier Sciences, the University of Tokyo, Tokyo, Japan
| | - Xin Zeng
- Department of Computational Biology and Medical Science, Graduate School of Frontier Sciences, the University of Tokyo, Tokyo, Japan
| | - Yubo Zhang
- Department of Computational Biology and Medical Science, Graduate School of Frontier Sciences, the University of Tokyo, Tokyo, Japan
| | - Martin Loza
- Human Genome Center, the Institute of Medical Science, the University of Tokyo, Tokyo, Japan
| | - Sung-Joon Park
- Human Genome Center, the Institute of Medical Science, the University of Tokyo, Tokyo, Japan
| | - Kenta Nakai
- Department of Computational Biology and Medical Science, Graduate School of Frontier Sciences, the University of Tokyo, Tokyo, Japan.
- Human Genome Center, the Institute of Medical Science, the University of Tokyo, Tokyo, Japan.
| |
Collapse
|
38
|
Kim K, Han M, Lee D. InTiCAR: Network-based identification of significant inter-tissue communicators for autoimmune diseases. Comput Struct Biotechnol J 2025; 27:333-345. [PMID: 39897058 PMCID: PMC11782887 DOI: 10.1016/j.csbj.2025.01.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2024] [Revised: 01/03/2025] [Accepted: 01/04/2025] [Indexed: 02/04/2025] Open
Abstract
Inter-tissue communicators (ITCs) are intricate and essential aspects of our body, as they are the keepers of homeostatic equilibrium. It is no surprise that the dysregulation of the exchange between tissues are at the core of various disorders. Among such conditions, autoimmune diseases (AIDs) refer to a collection of pathological conditions where the miscommunication drives the immune system to mistakenly attack one's own body. Due to their myriad and diverse pathophysiologies, AIDs cannot be easily diagnosed or treated, and continuous efforts are required to seek for potential diagnostic markers or therapeutic targets. The identification of ITCs with significant involvement in the disease states is therefore crucial. Here, we present InTiCAR, Inter-Tissue Communicators for Autoimmune diseases by Random walk with restart, which is a network exploration-based analysis method that suggests disease-specific ITCs based on prior knowledge of disease genes, without the need for the external expression data. We first show that distinct ITC profile s can be acquired for various diseases by InTiCAR. We further illustrate that, for autoimmune diseases (AIDs) specifically, the disease-specific ITCs outperform disease genes in diagnosing patients using the UK Biobank plasma proteome dataset. Also, through CMap LINCS dataset, we find that high perturbation on the AIDs genes can be observed by the disease-specific ITCs. Our results provide and highlight unique perspectives on biological network analysis by focusing on the entities of extracellular communications.
Collapse
Affiliation(s)
- Kwansoo Kim
- Department of Bio and Brain Engineering, KAIST, Daejeon 34141, Republic of Korea
| | - Manyoung Han
- Department of Bio and Brain Engineering, KAIST, Daejeon 34141, Republic of Korea
| | - Doheon Lee
- Department of Bio and Brain Engineering, KAIST, Daejeon 34141, Republic of Korea
| |
Collapse
|
39
|
Song X, Chavez-Fuentes JC, Ma W, Fu W, Wang P, Yuan GC. sCCIgen: A high-fidelity spatially resolved transcriptomics data simulator for cell-cell interaction studies. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.01.07.631830. [PMID: 39829773 PMCID: PMC11741276 DOI: 10.1101/2025.01.07.631830] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/22/2025]
Abstract
Spatially resolved transcriptomics (SRT) provides an invaluable avenue for examining cell-cell interactions within native tissue environments. The development and evaluation of analytical tools for SRT data necessitate tools for generating synthetic datasets with known ground truth of cell-cell interaction induced features. To address this gap, we introduce sCCIgen, a novel real-data-based simulator tailored to generate high-fidelity SRT data with a focus on cell-cell interactions. sCCIgen preserves transcriptomic and spatial characteristics in SRT data, while comprehensively models various cell-cell interaction features, including cell colocalization, spatial dependence among gene expressions, and gene-gene interactions between nearby cells. We implemented sCCIgen as an interactive, easy-to-use, realistic, reproducible, and well-documented tool for studying cellular interactions and spatial biology.
Collapse
|
40
|
Cui T, Li YY, Li BL, Zhang H, Yu TT, Zhang JN, Qian FC, Yin MX, Fang QL, Hu ZH, Yan YX, Wang QY, Li CQ, Shang DS. SpatialRef: a reference of spatial omics with known spot annotation. Nucleic Acids Res 2025; 53:D1215-D1223. [PMID: 39417483 PMCID: PMC11701618 DOI: 10.1093/nar/gkae892] [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: 08/15/2024] [Revised: 09/19/2024] [Accepted: 09/26/2024] [Indexed: 10/19/2024] Open
Abstract
Spatial omics technologies have enabled the creation of intricate spatial maps that capture molecular features and tissue morphology, providing valuable insights into the spatial associations and functional organization of tissues. Accurate annotation of spot or domain types is essential for downstream spatial omics analyses, but this remains challenging. Therefore, this study aimed to develop a manually curated spatial omics database (SpatialRef, https://bio.liclab.net/spatialref/), to provide comprehensive and high-quality spatial omics data with known spot labels across multiple species. The current version of SpatialRef aggregates >9 million manually annotated spots across 17 Human, Mouse and Drosophila tissue types through extensive review and strict quality control, covering multiple spatial sequencing technologies and >400 spot/domain types from original studies. Furthermore, SpatialRef supports various spatial omics analyses about known spot types, including differentially expressed genes, spatially variable genes, Gene Ontology (GO)/KEGG annotation, spatial communication and spatial trajectories. With a user-friendly interface, SpatialRef facilitates querying, browsing and visualizing, thereby aiding in elucidating the functional relevance of spatial domains within the tissue and uncovering potential biological effects.
Collapse
Affiliation(s)
- Ting Cui
- The First Affiliated Hospital & National Health Commission Key Laboratory of Birth Defect Research and Prevention, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, China
- Insititute of Biochemistry and Molecular Biology, Hengyang Medical College, University of South China, Hengyang, Hunan 421001, China
- The First Affiliated Hospital, Cardiovascular Lab of Big Data and Imaging Artificial Intelligence, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, China
| | - Yan-Yu Li
- The First Affiliated Hospital & National Health Commission Key Laboratory of Birth Defect Research and Prevention, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, China
- Insititute of Biochemistry and Molecular Biology, Hengyang Medical College, University of South China, Hengyang, Hunan 421001, China
- Hunan Provincial Key Laboratory of Multi-omics And Artificial Intelligence of Cardiovascular Diseases, University of South China, Hengyang, Hunan 421001, China
- School of Computer, University of South China, Hengyang, Hunan 421001, China
- The First Affiliated Hospital, Cardiovascular Lab of Big Data and Imaging Artificial Intelligence, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, China
- The First Affiliated Hospital, Institute of Cardiovascular Disease, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, China
| | - Bing-Long Li
- The First Affiliated Hospital & National Health Commission Key Laboratory of Birth Defect Research and Prevention, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, China
- Insititute of Biochemistry and Molecular Biology, Hengyang Medical College, University of South China, Hengyang, Hunan 421001, China
- The First Affiliated Hospital, Cardiovascular Lab of Big Data and Imaging Artificial Intelligence, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, China
| | - Han Zhang
- School of Computer, University of South China, Hengyang, Hunan 421001, China
- The First Affiliated Hospital, Cardiovascular Lab of Big Data and Imaging Artificial Intelligence, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, China
| | - Ting-Ting Yu
- School of Computer, University of South China, Hengyang, Hunan 421001, China
- The First Affiliated Hospital, Cardiovascular Lab of Big Data and Imaging Artificial Intelligence, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, China
| | - Jia-Ning Zhang
- The First Affiliated Hospital & National Health Commission Key Laboratory of Birth Defect Research and Prevention, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, China
- Insititute of Biochemistry and Molecular Biology, Hengyang Medical College, University of South China, Hengyang, Hunan 421001, China
- Hunan Provincial Key Laboratory of Multi-omics And Artificial Intelligence of Cardiovascular Diseases, University of South China, Hengyang, Hunan 421001, China
- The First Affiliated Hospital, Cardiovascular Lab of Big Data and Imaging Artificial Intelligence, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, China
| | - Feng-Cui Qian
- The First Affiliated Hospital & National Health Commission Key Laboratory of Birth Defect Research and Prevention, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, China
- Insititute of Biochemistry and Molecular Biology, Hengyang Medical College, University of South China, Hengyang, Hunan 421001, China
- Hunan Provincial Key Laboratory of Multi-omics And Artificial Intelligence of Cardiovascular Diseases, University of South China, Hengyang, Hunan 421001, China
- School of Computer, University of South China, Hengyang, Hunan 421001, China
- The First Affiliated Hospital, Cardiovascular Lab of Big Data and Imaging Artificial Intelligence, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, China
- The First Affiliated Hospital, Institute of Cardiovascular Disease, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, China
| | - Ming-Xue Yin
- The First Affiliated Hospital & National Health Commission Key Laboratory of Birth Defect Research and Prevention, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, China
- Insititute of Biochemistry and Molecular Biology, Hengyang Medical College, University of South China, Hengyang, Hunan 421001, China
- The First Affiliated Hospital, Cardiovascular Lab of Big Data and Imaging Artificial Intelligence, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, China
| | - Qiao-Li Fang
- School of Computer, University of South China, Hengyang, Hunan 421001, China
- The First Affiliated Hospital, Cardiovascular Lab of Big Data and Imaging Artificial Intelligence, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, China
| | - Zi-Hao Hu
- School of Computer, University of South China, Hengyang, Hunan 421001, China
- The First Affiliated Hospital, Cardiovascular Lab of Big Data and Imaging Artificial Intelligence, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, China
| | - Yu-Xiang Yan
- School of Computer, University of South China, Hengyang, Hunan 421001, China
- The First Affiliated Hospital, Cardiovascular Lab of Big Data and Imaging Artificial Intelligence, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, China
| | - Qiu-Yu Wang
- The First Affiliated Hospital & National Health Commission Key Laboratory of Birth Defect Research and Prevention, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, China
- Insititute of Biochemistry and Molecular Biology, Hengyang Medical College, University of South China, Hengyang, Hunan 421001, China
- Hunan Provincial Key Laboratory of Multi-omics And Artificial Intelligence of Cardiovascular Diseases, University of South China, Hengyang, Hunan 421001, China
- School of Computer, University of South China, Hengyang, Hunan 421001, China
- The First Affiliated Hospital, Cardiovascular Lab of Big Data and Imaging Artificial Intelligence, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, China
- The First Affiliated Hospital, Institute of Cardiovascular Disease, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, China
| | - Chun-Quan Li
- The First Affiliated Hospital & National Health Commission Key Laboratory of Birth Defect Research and Prevention, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, China
- Insititute of Biochemistry and Molecular Biology, Hengyang Medical College, University of South China, Hengyang, Hunan 421001, China
- Hunan Provincial Key Laboratory of Multi-omics And Artificial Intelligence of Cardiovascular Diseases, University of South China, Hengyang, Hunan 421001, China
- School of Computer, University of South China, Hengyang, Hunan 421001, China
- The First Affiliated Hospital, Cardiovascular Lab of Big Data and Imaging Artificial Intelligence, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, China
- Key Laboratory of Rare Pediatric Diseases, Ministry of Education, University of South China, Hengyang, Hunan 421001, China
- The First Affiliated Hospital, Institute of Cardiovascular Disease, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, China
| | - De-Si Shang
- The First Affiliated Hospital & National Health Commission Key Laboratory of Birth Defect Research and Prevention, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, China
- Insititute of Biochemistry and Molecular Biology, Hengyang Medical College, University of South China, Hengyang, Hunan 421001, China
- Hunan Provincial Key Laboratory of Multi-omics And Artificial Intelligence of Cardiovascular Diseases, University of South China, Hengyang, Hunan 421001, China
- School of Computer, University of South China, Hengyang, Hunan 421001, China
- The First Affiliated Hospital, Cardiovascular Lab of Big Data and Imaging Artificial Intelligence, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, China
- The First Affiliated Hospital, Institute of Cardiovascular Disease, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, China
| |
Collapse
|
41
|
Wu S, Zhang J, Wang Y, Qin X, Zhang Z, Lu Z, Kim P, Zhou X, Huang L. metsDB: a knowledgebase of cancer metastasis at bulk, single-cell and spatial levels. Nucleic Acids Res 2025; 53:D1427-D1434. [PMID: 39436035 PMCID: PMC11701579 DOI: 10.1093/nar/gkae916] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2024] [Revised: 09/18/2024] [Accepted: 10/18/2024] [Indexed: 10/23/2024] Open
Abstract
Cancer metastasis, the process by which tumour cells migrate and colonize distant organs from a primary site, is responsible for the majority of cancer-related deaths. Understanding the cellular and molecular mechanisms underlying this complex process is essential for developing effective metastasis prevention and therapy strategies. To this end, we systematically analysed 1786 bulk tissue samples from 13 cancer types, 988 463 single cells from 17 cancer types, and 40 252 spots from 45 spatial slides across 10 cancer types. The results of these analyses are compiled in the metsDB database, accessible at https://relab.xidian.edu.cn/metsDB/. This database provides insights into alterations in cell constitutions, cell relationships, biological pathways, molecular biomarkers, and drug responses during cancer metastasis at bulk, single-cell, and spatial levels. Users can perform cell or gene searches to obtain multi-view and multi-scale metastasis-related data. This comprehensive resource is invaluable for understanding the metastasis process and for designing molecular therapies.
Collapse
Affiliation(s)
- Sijia Wu
- School of Life Science and Technology, Xidian University, Xi’an, Shaanxi 710126, PR China
| | - Jiajin Zhang
- School of Life Science and Technology, Xidian University, Xi’an, Shaanxi 710126, PR China
| | - Yanfei Wang
- School of Biomedical Informatics, University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| | - Xinyu Qin
- School of Life Science and Technology, Xidian University, Xi’an, Shaanxi 710126, PR China
| | - Zhaocan Zhang
- School of Life Science and Technology, Xidian University, Xi’an, Shaanxi 710126, PR China
| | - Zhennan Lu
- School of Life Science and Technology, Xidian University, Xi’an, Shaanxi 710126, PR China
| | - Pora Kim
- School of Biomedical Informatics, University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| | - Xiaobo Zhou
- School of Biomedical Informatics, University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| | - Liyu Huang
- School of Life Science and Technology, Xidian University, Xi’an, Shaanxi 710126, PR China
| |
Collapse
|
42
|
Zhou L, Peng X, Chen M, He X, Tian G, Yang J, Peng L. Unveiling patterns in spatial transcriptomics data: a novel approach utilizing graph attention autoencoder and multiscale deep subspace clustering network. Gigascience 2025; 14:giae103. [PMID: 39804726 PMCID: PMC11727722 DOI: 10.1093/gigascience/giae103] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2024] [Revised: 07/06/2024] [Accepted: 11/21/2024] [Indexed: 01/16/2025] Open
Abstract
BACKGROUND The accurate deciphering of spatial domains, along with the identification of differentially expressed genes and the inference of cellular trajectory based on spatial transcriptomic (ST) data, holds significant potential for enhancing our understanding of tissue organization and biological functions. However, most of spatial clustering methods can neither decipher complex structures in ST data nor entirely employ features embedded in different layers. RESULTS This article introduces STMSGAL, a novel framework for analyzing ST data by incorporating graph attention autoencoder and multiscale deep subspace clustering. First, STMSGAL constructs ctaSNN, a cell type-aware shared nearest neighbor graph, using Louvian clustering exclusively based on gene expression profiles. Subsequently, it integrates expression profiles and ctaSNN to generate spot latent representations using a graph attention autoencoder and multiscale deep subspace clustering. Lastly, STMSGAL implements spatial clustering, differential expression analysis, and trajectory inference, providing comprehensive capabilities for thorough data exploration and interpretation. STMSGAL was evaluated against 7 methods, including SCANPY, SEDR, CCST, DeepST, GraphST, STAGATE, and SiGra, using four 10x Genomics Visium datasets, 1 mouse visual cortex STARmap dataset, and 2 Stereo-seq mouse embryo datasets. The comparison showcased STMSGAL's remarkable performance across Davies-Bouldin, Calinski-Harabasz, S_Dbw, and ARI values. STMSGAL significantly enhanced the identification of layer structures across ST data with different spatial resolutions and accurately delineated spatial domains in 2 breast cancer tissues, adult mouse brain (FFPE), and mouse embryos. CONCLUSIONS STMSGAL can serve as an essential tool for bridging the analysis of cellular spatial organization and disease pathology, offering valuable insights for researchers in the field.
Collapse
Affiliation(s)
- Liqian Zhou
- School of Computer Science, Hunan University of Technology, Zhuzhou 412007, Hunan, China
| | - Xinhuai Peng
- School of Computer Science, Hunan University of Technology, Zhuzhou 412007, Hunan, China
| | - Min Chen
- School of Computer Science, Hunan Institute of Technology, Hengyang 421002, Hunan, China
| | - Xianzhi He
- School of Computer Science, Hunan University of Technology, Zhuzhou 412007, Hunan, China
| | - Geng Tian
- Geneis (Beijing) Co. Ltd., Beijing 100102, China
| | | | - Lihong Peng
- School of Computer Science, Hunan University of Technology, Zhuzhou 412007, Hunan, China
- College of Life Science and Chemistry, Hunan University of Technology, Zhuzhou 412007, Hunan, China
| |
Collapse
|
43
|
Zheng BW, Guo W. Multi-omics analysis unveils the role of inflammatory cancer-associated fibroblasts in chordoma progression. J Pathol 2025; 265:69-83. [PMID: 39611243 DOI: 10.1002/path.6369] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2024] [Revised: 09/25/2024] [Accepted: 10/13/2024] [Indexed: 11/30/2024]
Abstract
Cancer-associated fibroblasts (CAFs) constitute the primary cellular component of the stroma in chordomas, characterized by an abundance of mucinous stromal elements, potentially facilitating their initiation and progression; however, this inference has yet to be fully confirmed. In this study, single-cell RNA sequencing (scRNA-seq), spatial transcriptomics (ST), bulk RNA-seq, multiplexed quantitative immunofluorescence (QIF), and in vivo and in vitro experiments were performed to determine the heterogeneity, spatial distribution, and clinical significance of CAFs in chordoma. ScRNA-seq was performed on 87,693 single cells derived from seven tumor samples and four control nucleus pulposus samples. A distinct CAF cluster distinguished by the upregulated expression of inflammatory genes and enriched functionality in activating inflammation-associated cells was identified. Pseudotime trajectory and cell communication analyses suggested that this inflammatory CAF (iCAF) subset originated from normal fibroblasts and interacted extensively with tumors and various other cell types. By integrating the scRNA-seq results with ST, the presence of iCAF in chordoma tissue was further confirmed, indicating their positioning at a distance from the tumor cells. Bulk RNA-seq data analysis from 126 patients revealed a correlation between iCAF signature scores, chordoma invasiveness, and poor prognosis. QIF validation involving an additional 116 patients found that although iCAFs were not in close proximity to tumor cells compared with other CAF subsets, their density correlated with malignant tumor phenotypes and adverse outcomes. In vivo and in vitro experiments further confirmed that iCAFs accelerate the malignant progression of chordomas. These findings could provide insights into the development of novel therapeutic strategies. © 2024 The Pathological Society of Great Britain and Ireland.
Collapse
Affiliation(s)
- Bo-Wen Zheng
- Department of Musculoskeletal Tumor, Peking University People's Hospital, Peking University, Beijing, PR China
- Beijing Key Laboratory of Musculoskeletal Tumor, Peking University People's Hospital, Beijing, PR China
| | - Wei Guo
- Department of Musculoskeletal Tumor, Peking University People's Hospital, Peking University, Beijing, PR China
- Beijing Key Laboratory of Musculoskeletal Tumor, Peking University People's Hospital, Beijing, PR China
| |
Collapse
|
44
|
Defard T, Desrentes A, Fouillade C, Mueller F. Homebuilt Imaging-Based Spatial Transcriptomics: Tertiary Lymphoid Structures as a Case Example. Methods Mol Biol 2025; 2864:77-105. [PMID: 39527218 DOI: 10.1007/978-1-0716-4184-2_5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2024]
Abstract
Spatial transcriptomics methods provide insight into the cellular heterogeneity and spatial architecture of complex, multicellular systems. Combining molecular and spatial information provides important clues to study tissue architecture in development and disease. Here, we present a comprehensive do-it-yourself (DIY) guide to perform such experiments at reduced costs leveraging open-source approaches. This guide spans the entire life cycle of a project, from its initial definition to experimental choices, wet lab approaches, instrumentation, and analysis. As a concrete example, we focus on tertiary lymphoid structures (TLS), which we use to develop typical questions that can be addressed by these approaches.
Collapse
Affiliation(s)
- Thomas Defard
- Institut Pasteur, Université Paris Cité, Photonic Bio-Imaging, Centre de Ressources et Recherches Technologiques (UTechS-PBI, C2RT), Paris, France
- Institut Pasteur, Université Paris Cité, Imaging and Modeling Unit, Paris, France
- Centre for Computational Biology (CBIO), Mines Paris, PSL University, Paris, France
- Institut Curie, PSL University, Paris, France
- INSERM, U900, Paris, France
| | - Auxence Desrentes
- UMRS1135 Sorbonne University, Paris, France
- INSERM U1135, Paris, France
- Team "Immune Microenvironment and Immunotherapy", Centre for Immunology and Microbial Infections (CIMI), Paris, France
| | - Charles Fouillade
- Institut Curie, Inserm U1021-CNRS UMR 3347, University Paris-Saclay, PSL Research University, Centre Universitaire, Orsay, France
| | - Florian Mueller
- Institut Pasteur, Université Paris Cité, Photonic Bio-Imaging, Centre de Ressources et Recherches Technologiques (UTechS-PBI, C2RT), Paris, France.
- Institut Pasteur, Université Paris Cité, Imaging and Modeling Unit, Paris, France.
| |
Collapse
|
45
|
Gulati GS, D'Silva JP, Liu Y, Wang L, Newman AM. Profiling cell identity and tissue architecture with single-cell and spatial transcriptomics. Nat Rev Mol Cell Biol 2025; 26:11-31. [PMID: 39169166 DOI: 10.1038/s41580-024-00768-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/16/2024] [Indexed: 08/23/2024]
Abstract
Single-cell transcriptomics has broadened our understanding of cellular diversity and gene expression dynamics in healthy and diseased tissues. Recently, spatial transcriptomics has emerged as a tool to contextualize single cells in multicellular neighbourhoods and to identify spatially recurrent phenotypes, or ecotypes. These technologies have generated vast datasets with targeted-transcriptome and whole-transcriptome profiles of hundreds to millions of cells. Such data have provided new insights into developmental hierarchies, cellular plasticity and diverse tissue microenvironments, and spurred a burst of innovation in computational methods for single-cell analysis. In this Review, we discuss recent advancements, ongoing challenges and prospects in identifying and characterizing cell states and multicellular neighbourhoods. We discuss recent progress in sample processing, data integration, identification of subtle cell states, trajectory modelling, deconvolution and spatial analysis. Furthermore, we discuss the increasing application of deep learning, including foundation models, in analysing single-cell and spatial transcriptomics data. Finally, we discuss recent applications of these tools in the fields of stem cell biology, immunology, and tumour biology, and the future of single-cell and spatial transcriptomics in biological research and its translation to the clinic.
Collapse
Affiliation(s)
- Gunsagar S Gulati
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | | | - Yunhe Liu
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Linghua Wang
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
- The University of Texas MD Anderson Cancer Center UTHealth Houston Graduate School of Biomedical Sciences, Houston, TX, USA
| | - Aaron M Newman
- Department of Biomedical Data Science, Stanford University, Stanford, CA, USA.
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University, Stanford, CA, USA.
- Stanford Cancer Institute, Stanford University, Stanford, CA, USA.
- Chan Zuckerberg Biohub - San Francisco, San Francisco, CA, USA.
| |
Collapse
|
46
|
Pei Y, Mou Z, Jiang L, Yang J, Gu Y, Min J, Sunzhang L, Xiong N, Xu X, Chi H, Xu K, Liu S, Luo H. Aging and head and neck cancer insights from single cell and spatial transcriptomic analyses. Discov Oncol 2024; 15:801. [PMID: 39692961 PMCID: PMC11655923 DOI: 10.1007/s12672-024-01672-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/03/2024] [Accepted: 12/04/2024] [Indexed: 12/19/2024] Open
Abstract
BACKGROUND Head and neck squamous cell carcinoma(HNSCC) is the sixth most common malignancy worldwide, with more than 890,000 new cases and 450,000 deaths annually. Its major risk factors include smoking, alcohol abuse, aging, and poor oral hygiene. Due to the lack of early and effective detection and screening methods, many patients are diagnosed at advanced stages with a five-year survival rate of less than 50%. In this study, we deeply explored the expression of Aging-related genes(ARGs) in HNSCC and analyzed their prognostic significance using single-cell sequencing and spatial transcriptomics analysis. This research aims to provide new theoretical support and directions for personalized treatment. Annually, more than 890,000 new cases of head and neck squamous cell carcinoma (HNSCC) are diagnosed globally, leading to 450,000 deaths, making it the sixth most common malignancy worldwide. The primary risk factors for HNSCC include smoking, alcohol abuse, aging, and poor oral hygiene. Many patients are diagnosed at advanced stages due to the absence of early and effective detection and screening methods, resulting in a five-year survival rate of less than 50%. In this research, single cell sequencing and spatial transcriptome analysis were used to investigate the expression of Aging-related genes (ARGs) in HNSCC and to analyse their prognostic significance. This research aims to provide new theoretical support and directions for personalized treatment. METHODS In this study, we investigated the association between HNSCC and AGRs by utilizing the GSE139324 series in the GEO database alongside the TCGA database, combined with single-cell sequencing and spatial transcriptomics analysis. The data were analyzed using Seurat and tSNE tools to reveal intercellular communication networks. For the spatial transcriptome data, SCTransform and RunPCA were applied to examine the metabolic activities of the cells. Gene expression differences were determined through spacerxr and RCTD tools, while the limma package was employed to identify differentially expressed genes and to predict recurrence rates using Cox regression analysis and column line plots. These findings underscore the potential importance of molecular classification, prognostic assessment, and personalized treatment of HNSCC. RESULTS This study utilized HNSCC single-cell sequencing data to highlight the significance of ARGs in the onset and prognosis of HNSCC. It revealed that the proportion of monocytes and macrophages increased, while the proportion of B cells decreased. Notably, high expression of the APOE gene in monocytes was closely associated with patient prognosis. Additionally, a Cox regression model was developed based on GSTP1 and age to provide personalized prediction tools for clinical use in predicting patient survival. CONCLUSIONS We utilized single-cell sequencing and spatial transcriptomics to explore the cellular characteristics of HNSCC and its interaction with the tumor microenvironment. Our findings reveal that HNSCC tissues show increased mononuclear cells and demonstrate enhanced activity in ARGs, thereby advancing our understanding of HNSCC development mechanisms.
Collapse
Affiliation(s)
- Yi Pei
- School of Stomatology, Southwest Medical University, Luzhou, 646000, China
| | - Zhuying Mou
- School of Stomatology, Southwest Medical University, Luzhou, 646000, China
| | - Lai Jiang
- Clinical Medical College, Southwest Medical University, Luzhou, 646000, China
| | - Jinyan Yang
- School of Stomatology, Southwest Medical University, Luzhou, 646000, China
| | - Yuheng Gu
- Clinical Medical College, Southwest Medical University, Luzhou, 646000, China
| | - Jie Min
- Clinical Medical College, Southwest Medical University, Luzhou, 646000, China
| | - Lingyi Sunzhang
- Clinical Medical College, Southwest Medical University, Luzhou, 646000, China
| | - Nan Xiong
- Clinical Medical College, Southwest Medical University, Luzhou, 646000, China
| | - Xiang Xu
- School of Stomatology, Southwest Medical University, Luzhou, 646000, China
| | - Hao Chi
- Clinical Medical College, Southwest Medical University, Luzhou, 646000, China
| | - Ke Xu
- Department of Oncology, Chongqing General Hospital, Chongqing University, Chongqing, 401147, China.
| | - Sinian Liu
- Department of Pathology, Xichong People's Hospital, Nanchong, 637200, China.
| | - Huiyan Luo
- Department of Oncology, Chongqing General Hospital, Chongqing University, Chongqing, 401147, China.
| |
Collapse
|
47
|
Niu J, Zhu F, Fang D, Min W. SpatialCVGAE: Consensus Clustering Improves Spatial Domain Identification of Spatial Transcriptomics Using VGAE. Interdiscip Sci 2024:10.1007/s12539-024-00676-1. [PMID: 39680300 DOI: 10.1007/s12539-024-00676-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2024] [Revised: 11/04/2024] [Accepted: 11/14/2024] [Indexed: 12/17/2024]
Abstract
The advent of spatially resolved transcriptomics (SRT) has provided critical insights into the spatial context of tissue microenvironments. Spatial clustering is a fundamental aspect of analyzing spatial transcriptomics data. However, spatial clustering methods often suffer from instability caused by the sparsity and high noise in the SRT data. To address this challenge, we propose SpatialCVGAE, a consensus clustering framework designed for SRT data analysis. SpatialCVGAE adopts the expression of high-variable genes from different dimensions along with multiple spatial graphs as inputs to variational graph autoencoders (VGAEs), learning multiple latent representations for clustering. These clustering results are then integrated using a consensus clustering approach, which enhances the model's stability and robustness by combining multiple clustering outcomes. Experiments demonstrate that SpatialCVGAE effectively mitigates the instability typically associated with non-ensemble deep learning methods, significantly improving both the stability and accuracy of the results. Compared to previous non-ensemble methods in representation learning and post-processing, our method fully leverages the diversity of multiple representations to accurately identify spatial domains, showing superior robustness and adaptability. All code and public datasets used in this paper are available at https://github.com/wenwenmin/SpatialCVGAE .
Collapse
Affiliation(s)
- Jinyun Niu
- School of Information Science and Engineering, Yunnan University, Kunming, 650500, China
| | - Fangfang Zhu
- School of Health and Nursing, Yunnan Open University, Kunming, 650599, China
| | - Donghai Fang
- School of Information Science and Engineering, Yunnan University, Kunming, 650500, China
| | - Wenwen Min
- School of Information Science and Engineering, Yunnan University, Kunming, 650500, China.
| |
Collapse
|
48
|
Sun Y, Kong L, Huang J, Deng H, Bian X, Li X, Cui F, Dou L, Cao C, Zou Q, Zhang Z. A comprehensive survey of dimensionality reduction and clustering methods for single-cell and spatial transcriptomics data. Brief Funct Genomics 2024; 23:733-744. [PMID: 38860675 DOI: 10.1093/bfgp/elae023] [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: 12/26/2023] [Revised: 02/29/2024] [Accepted: 05/27/2024] [Indexed: 06/12/2024] Open
Abstract
In recent years, the application of single-cell transcriptomics and spatial transcriptomics analysis techniques has become increasingly widespread. Whether dealing with single-cell transcriptomic or spatial transcriptomic data, dimensionality reduction and clustering are indispensable. Both single-cell and spatial transcriptomic data are often high-dimensional, making the analysis and visualization of such data challenging. Through dimensionality reduction, it becomes possible to visualize the data in a lower-dimensional space, allowing for the observation of relationships and differences between cell subpopulations. Clustering enables the grouping of similar cells into the same cluster, aiding in the identification of distinct cell subpopulations and revealing cellular diversity, providing guidance for downstream analyses. In this review, we systematically summarized the most widely recognized algorithms employed for the dimensionality reduction and clustering analysis of single-cell transcriptomic and spatial transcriptomic data. This endeavor provides valuable insights and ideas that can contribute to the development of novel tools in this rapidly evolving field.
Collapse
Affiliation(s)
- Yidi Sun
- School of Computer Science and Technology, Hainan University, Haikou 570228, China
| | - Lingling Kong
- School of Computer Science and Technology, Hainan University, Haikou 570228, China
| | - Jiayi Huang
- School of Computer Science and Technology, Hainan University, Haikou 570228, China
| | - Hongyan Deng
- School of Computer Science and Technology, Hainan University, Haikou 570228, China
| | - Xinling Bian
- School of Computer Science and Technology, Hainan University, Haikou 570228, China
| | - Xingfeng Li
- School of Computer Science and Technology, Hainan University, Haikou 570228, China
| | - Feifei Cui
- School of Computer Science and Technology, Hainan University, Haikou 570228, China
| | - Lijun Dou
- Genomic Medicine Institute, Lerner Research Institute, Cleveland, OH 44106, United States
| | - Chen Cao
- School of Biomedical Engineering and Informatics, Nanjing Medical University, Nanjing 210029, China
| | - Quan Zou
- Institute of Fundamental and Frontier Sciences, University of Electronic Science and Technology of China, Chengdu 610054, China
- Yangtze Delta Region Institute (Quzhou), University of Electronic Science and Technology of China, Quzhou 324000, China
| | - Zilong Zhang
- School of Computer Science and Technology, Hainan University, Haikou 570228, China
| |
Collapse
|
49
|
Wang Z, Geng A, Duan H, Cui F, Zou Q, Zhang Z. A comprehensive review of approaches for spatial domain recognition of spatial transcriptomes. Brief Funct Genomics 2024; 23:702-712. [PMID: 39426802 DOI: 10.1093/bfgp/elae040] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2024] [Revised: 10/02/2024] [Accepted: 10/07/2024] [Indexed: 10/21/2024] Open
Abstract
In current bioinformatics research, spatial transcriptomics (ST) as a rapidly evolving technology is gradually receiving widespread attention from researchers. Spatial domains are regions where gene expression and histology are consistent in space, and detecting spatial domains can better understand the organization and functional distribution of tissues. Spatial domain recognition is a fundamental step in the process of ST data interpretation, which is also a major challenge in ST analysis. Therefore, developing more accurate, efficient, and general spatial domain recognition methods has become an important and urgent research direction. This article aims to review the current status and progress of spatial domain recognition research, explore the advantages and limitations of existing methods, and provide suggestions and directions for future tool development.
Collapse
Affiliation(s)
- Ziyi Wang
- School of Computer Science and Technology, Hainan University, Haikou 570228, China
| | - Aoyun Geng
- School of Computer Science and Technology, Hainan University, Haikou 570228, China
| | - Hao Duan
- School of Computer Science and Technology, Hainan University, Haikou 570228, China
| | - Feifei Cui
- School of Computer Science and Technology, Hainan University, Haikou 570228, China
| | - Quan Zou
- Institute of Fundamental and Frontier Sciences, University of Electronic Science and Technology of China, Chengdu 610054, China
- Yangtze Delta Region Institute (Quzhou), University of Electronic Science and Technology of China, Quzhou 324000, China
| | - Zilong Zhang
- School of Computer Science and Technology, Hainan University, Haikou 570228, China
| |
Collapse
|
50
|
Agrawal A, Thomann S, Basu S, Grün D. NiCo identifies extrinsic drivers of cell state modulation by niche covariation analysis. Nat Commun 2024; 15:10628. [PMID: 39639035 PMCID: PMC11621405 DOI: 10.1038/s41467-024-54973-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2024] [Accepted: 11/22/2024] [Indexed: 12/07/2024] Open
Abstract
Cell states are modulated by intrinsic driving forces such as gene expression noise and extrinsic signals from the tissue microenvironment. The distinction between intrinsic and extrinsic cell state determinants is essential for understanding the regulation of cell fate in tissues during development, homeostasis and disease. The rapidly growing availability of single-cell resolution spatial transcriptomics makes it possible to meet this challenge. However, available computational methods to infer topological tissue domains, spatially variable genes, or ligand-receptor interactions are limited in their capacity to capture cell state changes driven by crosstalk between individual cell types within the same niche. We present NiCo, a computational framework for integrating single-cell resolution spatial transcriptomics with matched single-cell RNA-sequencing reference data to infer the influence of the spatial niche on the cell state. By applying NiCo to mouse embryogenesis, adult small intestine and liver data, we demonstrate the ability to predict novel niche interactions that govern cell state variation underlying tissue development and homeostasis. In particular, NiCo predicts a feedback mechanism between Kupffer cells and neighboring stellate cells dampening stellate cell activation in the normal liver. NiCo provides a powerful tool to elucidate tissue architecture and to identify drivers of cellular states in local niches.
Collapse
Affiliation(s)
- Ankit Agrawal
- Würzburg Institute of Systems Immunology, Julius-Maximilians-Universität Würzburg, Würzburg, Germany
| | - Stefan Thomann
- Würzburg Institute of Systems Immunology, Julius-Maximilians-Universität Würzburg, Würzburg, Germany
| | - Sukanya Basu
- Würzburg Institute of Systems Immunology, Julius-Maximilians-Universität Würzburg, Würzburg, Germany
| | - Dominic Grün
- Würzburg Institute of Systems Immunology, Julius-Maximilians-Universität Würzburg, Würzburg, Germany.
- CAIDAS - Center for Artificial Intelligence and Data Science, Würzburg, Germany.
| |
Collapse
|