1
|
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
|
2
|
Li L, Lu M, Guo L, Zhang X, Liu Q, Zhang M, Gao J, Xu M, Lu Y, Zhang F, Li Y, Zhang R, Liu X, Pan S, Zhang X, Li Z, Chen Y, Su X, Zhang N, Guo W, Yang T, Chen J, Qin Y, Zhang Z, Cui W, Yu L, Gu Y, Yang H, Xu X, Wang J, Burns CE, Burns CG, Han K, Zhao L, Fan G, Su Y. An organ-wide spatiotemporal transcriptomic and cellular atlas of the regenerating zebrafish heart. Nat Commun 2025; 16:3716. [PMID: 40253397 PMCID: PMC12009352 DOI: 10.1038/s41467-025-59070-0] [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: 06/12/2024] [Accepted: 04/10/2025] [Indexed: 04/21/2025] Open
Abstract
Adult zebrafish robustly regenerate injured hearts through a complex orchestration of molecular and cellular activities. However, this remarkable process, which is largely non-existent in humans, remains incompletely understood. Here, we utilize integrated spatial transcriptomics (Stereo-seq) and single-cell RNA-sequencing (scRNA-seq) to generate a spatially-resolved molecular and cellular atlas of regenerating zebrafish heart across eight stages. We characterize the cascade of cardiomyocyte cell states responsible for producing regenerated myocardium and explore a potential role for tpm4a in cardiomyocyte re-differentiation. Moreover, we uncover the activation of ifrd1 and atp6ap2 genes as a unique feature of regenerative hearts. Lastly, we reconstruct a 4D "virtual regenerating heart" comprising 569,896 cells/spots derived from 36 scRNA-seq libraries and 224 Stereo-seq slices. Our comprehensive atlas serves as a valuable resource to the cardiovascular and regeneration scientific communities and their ongoing efforts to understand the molecular and cellular mechanisms underlying vertebrate heart regeneration.
Collapse
Affiliation(s)
- Lei Li
- Qingdao Key Laboratory of Marine Genomics, BGI Research, Qingdao, 266555, China
- State Key Laboratory of Genome and Multi-omics Technologies, BGI Research, Shenzhen, 518083, China
| | - Meina Lu
- Key Laboratory of Evolution and Marine Biodiversity (Ministry of Education) and Institute of Evolution and Marine Biodiversity, Ocean University of China, Qingdao, 266003, China
- College of Fisheries, Ocean University of China, Qingdao, 266003, China
| | - Lidong Guo
- Qingdao Key Laboratory of Marine Genomics, BGI Research, Qingdao, 266555, China
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Xuejiao Zhang
- Key Laboratory of Evolution and Marine Biodiversity (Ministry of Education) and Institute of Evolution and Marine Biodiversity, Ocean University of China, Qingdao, 266003, China
- College of Fisheries, Ocean University of China, Qingdao, 266003, China
| | - Qun Liu
- Qingdao Key Laboratory of Marine Genomics, BGI Research, Qingdao, 266555, China
- Department of Biology, University of Copenhagen, Copenhagen, 2100, Denmark
| | - Meiling Zhang
- Key Laboratory of Evolution and Marine Biodiversity (Ministry of Education) and Institute of Evolution and Marine Biodiversity, Ocean University of China, Qingdao, 266003, China
- College of Fisheries, Ocean University of China, Qingdao, 266003, China
| | - Junying Gao
- Key Laboratory of Evolution and Marine Biodiversity (Ministry of Education) and Institute of Evolution and Marine Biodiversity, Ocean University of China, Qingdao, 266003, China
- College of Fisheries, Ocean University of China, Qingdao, 266003, China
| | - Mengyang Xu
- Qingdao Key Laboratory of Marine Genomics, BGI Research, Qingdao, 266555, China
- State Key Laboratory of Genome and Multi-omics Technologies, BGI Research, Shenzhen, 518083, China
| | - Yijian Lu
- Key Laboratory of Evolution and Marine Biodiversity (Ministry of Education) and Institute of Evolution and Marine Biodiversity, Ocean University of China, Qingdao, 266003, China
- College of Fisheries, Ocean University of China, Qingdao, 266003, China
| | - Fang Zhang
- Key Laboratory of Evolution and Marine Biodiversity (Ministry of Education) and Institute of Evolution and Marine Biodiversity, Ocean University of China, Qingdao, 266003, China
- College of Marine Life Sciences, Ocean University of China, Qingdao, 266003, China
| | - Yao Li
- Qingdao Key Laboratory of Marine Genomics, BGI Research, Qingdao, 266555, China
| | - Ruihua Zhang
- Qingdao Key Laboratory of Marine Genomics, BGI Research, Qingdao, 266555, China
| | - Xiawei Liu
- Qingdao Key Laboratory of Marine Genomics, BGI Research, Qingdao, 266555, China
| | - Shanshan Pan
- Qingdao Key Laboratory of Marine Genomics, BGI Research, Qingdao, 266555, China
| | - Xianghui Zhang
- Qingdao Key Laboratory of Marine Genomics, BGI Research, Qingdao, 266555, China
| | - Zhen Li
- Qingdao Key Laboratory of Marine Genomics, BGI Research, Qingdao, 266555, China
| | - Yadong Chen
- Qingdao Key Laboratory of Marine Genomics, BGI Research, Qingdao, 266555, China
| | - Xiaoshan Su
- Qingdao Key Laboratory of Marine Genomics, BGI Research, Qingdao, 266555, China
- Department of Biology, University of Copenhagen, Copenhagen, 2100, Denmark
| | - Nannan Zhang
- Qingdao Key Laboratory of Marine Genomics, BGI Research, Qingdao, 266555, China
| | - Wenjie Guo
- Qingdao Key Laboratory of Marine Genomics, BGI Research, Qingdao, 266555, China
| | - Tao Yang
- China National GeneBank, BGI Research, Shenzhen, 518120, China
| | - Jing Chen
- China National GeneBank, BGI Research, Shenzhen, 518120, China
| | - Yating Qin
- Qingdao Key Laboratory of Marine Genomics, BGI Research, Qingdao, 266555, China
- Department of Biology, University of Copenhagen, Copenhagen, 2100, Denmark
| | | | - Wei Cui
- Qingdao Key Laboratory of Marine Genomics, BGI Research, Qingdao, 266555, China
| | - Lindong Yu
- Key Laboratory of Evolution and Marine Biodiversity (Ministry of Education) and Institute of Evolution and Marine Biodiversity, Ocean University of China, Qingdao, 266003, China
- College of Fisheries, Ocean University of China, Qingdao, 266003, China
| | - Ying Gu
- State Key Laboratory of Genome and Multi-omics Technologies, BGI Research, Shenzhen, 518083, China
- BGI, Shenzhen, 518083, China
| | - Huanming Yang
- State Key Laboratory of Genome and Multi-omics Technologies, BGI Research, Shenzhen, 518083, China
- BGI, Shenzhen, 518083, China
| | - Xun Xu
- State Key Laboratory of Genome and Multi-omics Technologies, BGI Research, Shenzhen, 518083, China
- BGI, Shenzhen, 518083, China
| | - Jianxun Wang
- School of Basic Medicine, Qingdao University, Qingdao, 266071, China
| | - Caroline E Burns
- Division of Basic and Translational Cardiovascular Research, Department of Cardiology, Boston Children's Hospital, Boston, MA, 02115, USA
- Harvard Medical School, Boston, MA, 02115, USA
| | - C Geoffrey Burns
- Division of Basic and Translational Cardiovascular Research, Department of Cardiology, Boston Children's Hospital, Boston, MA, 02115, USA
- Harvard Medical School, Boston, MA, 02115, USA
| | - Kai Han
- Qingdao Key Laboratory of Marine Genomics, BGI Research, Qingdao, 266555, China.
- Department of Biology, University of Copenhagen, Copenhagen, 2100, Denmark.
| | - Long Zhao
- Key Laboratory of Evolution and Marine Biodiversity (Ministry of Education) and Institute of Evolution and Marine Biodiversity, Ocean University of China, Qingdao, 266003, China.
- College of Fisheries, Ocean University of China, Qingdao, 266003, China.
| | - Guangyi Fan
- Qingdao Key Laboratory of Marine Genomics, BGI Research, Qingdao, 266555, China.
- State Key Laboratory of Genome and Multi-omics Technologies, BGI Research, Shenzhen, 518083, China.
- BGI Research, Sanya, 572025, China.
- BGI Research, Hangzhou, 310030, China.
| | - Ying Su
- Key Laboratory of Evolution and Marine Biodiversity (Ministry of Education) and Institute of Evolution and Marine Biodiversity, Ocean University of China, Qingdao, 266003, China.
- College of Marine Life Sciences, Ocean University of China, Qingdao, 266003, China.
| |
Collapse
|
3
|
Brooks ER, Moorman AR, Bhattacharya B, Prudhomme IS, Land M, Alcorn HL, Sharma R, Pe'er D, Zallen JA. A single-cell atlas of spatial and temporal gene expression in the mouse cranial neural plate. eLife 2025; 13:RP102819. [PMID: 40192104 PMCID: PMC11975377 DOI: 10.7554/elife.102819] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/09/2025] Open
Abstract
The formation of the mammalian brain requires regionalization and morphogenesis of the cranial neural plate, which transforms from an epithelial sheet into a closed tube that provides the structural foundation for neural patterning and circuit formation. Sonic hedgehog (SHH) signaling is important for cranial neural plate patterning and closure, but the transcriptional changes that give rise to the spatially regulated cell fates and behaviors that build the cranial neural tube have not been systematically analyzed. Here, we used single-cell RNA sequencing to generate an atlas of gene expression at six consecutive stages of cranial neural tube closure in the mouse embryo. Ordering transcriptional profiles relative to the major axes of gene expression predicted spatially regulated expression of 870 genes along the anterior-posterior and mediolateral axes of the cranial neural plate and reproduced known expression patterns with over 85% accuracy. Single-cell RNA sequencing of embryos with activated SHH signaling revealed distinct SHH-regulated transcriptional programs in the developing forebrain, midbrain, and hindbrain, suggesting a complex interplay between anterior-posterior and mediolateral patterning systems. These results define a spatiotemporally resolved map of gene expression during cranial neural tube closure and provide a resource for investigating the transcriptional events that drive early mammalian brain development.
Collapse
Affiliation(s)
- Eric R Brooks
- Department of Molecular Biomedical Sciences, College of Veterinary Medicine, North Carolina State UniversityRaleighUnited States
- Howard Hughes Medical Institute and Developmental Biology Program, Sloan Kettering InstituteNew YorkUnited States
| | - Andrew R Moorman
- Howard Hughes Medical Institute and Computational and Systems Biology Program, Sloan Kettering InstituteNew YorkUnited States
| | - Bhaswati Bhattacharya
- Howard Hughes Medical Institute and Developmental Biology Program, Sloan Kettering InstituteNew YorkUnited States
| | - Ian S Prudhomme
- Howard Hughes Medical Institute and Developmental Biology Program, Sloan Kettering InstituteNew YorkUnited States
| | - Max Land
- Howard Hughes Medical Institute and Computational and Systems Biology Program, Sloan Kettering InstituteNew YorkUnited States
| | - Heather L Alcorn
- Howard Hughes Medical Institute and Developmental Biology Program, Sloan Kettering InstituteNew YorkUnited States
| | - Roshan Sharma
- Howard Hughes Medical Institute and Computational and Systems Biology Program, Sloan Kettering InstituteNew YorkUnited States
| | - Dana Pe'er
- Howard Hughes Medical Institute and Computational and Systems Biology Program, Sloan Kettering InstituteNew YorkUnited States
| | - Jennifer A Zallen
- Howard Hughes Medical Institute and Developmental Biology Program, Sloan Kettering InstituteNew YorkUnited States
| |
Collapse
|
4
|
Jiang F, Wang H, Li Z, Cui G, Peng G. Leveraging spatial multiomics to unravel tissue architecture in embryo development. Trends Genet 2025; 41:271-274. [PMID: 39648060 DOI: 10.1016/j.tig.2024.11.007] [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: 04/30/2024] [Revised: 11/13/2024] [Accepted: 11/14/2024] [Indexed: 12/10/2024]
Abstract
Spatial multiomics technologies have revolutionized biomedical research by enabling the simultaneous measurement of multiple omics modalities within intact tissue sections. This approach facilitates the reconstruction of 3D molecular architectures, providing unprecedented insights into complex cellular interactions and the intricate organization of biological systems, such as those underlying embryonic development.
Collapse
Affiliation(s)
- Fuqing Jiang
- Center for Cell Lineage Technology and Bioengineering, Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, Guangdong-Hong Kong Joint Laboratory for Stem Cell and Regenerative Medicine, GIBH-CUHK Joint Research Laboratory on Stem Cell and Regenerative Medicine, GIBH-HKU Guangdong-Hong Kong Stem Cell and Regenerative Medicine Research Centre, China-New Zealand Belt and Road Joint Laboratory on Biomedicine and Health, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou 510530, China
| | - Haoxian Wang
- Center for Cell Lineage Technology and Bioengineering, Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, Guangdong-Hong Kong Joint Laboratory for Stem Cell and Regenerative Medicine, GIBH-CUHK Joint Research Laboratory on Stem Cell and Regenerative Medicine, GIBH-HKU Guangdong-Hong Kong Stem Cell and Regenerative Medicine Research Centre, China-New Zealand Belt and Road Joint Laboratory on Biomedicine and Health, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou 510530, China
| | - Zhuxia Li
- Center for Cell Lineage Technology and Bioengineering, Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, Guangdong-Hong Kong Joint Laboratory for Stem Cell and Regenerative Medicine, GIBH-CUHK Joint Research Laboratory on Stem Cell and Regenerative Medicine, GIBH-HKU Guangdong-Hong Kong Stem Cell and Regenerative Medicine Research Centre, China-New Zealand Belt and Road Joint Laboratory on Biomedicine and Health, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou 510530, China
| | | | - Guangdun Peng
- Center for Cell Lineage Technology and Bioengineering, Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, Guangdong-Hong Kong Joint Laboratory for Stem Cell and Regenerative Medicine, GIBH-CUHK Joint Research Laboratory on Stem Cell and Regenerative Medicine, GIBH-HKU Guangdong-Hong Kong Stem Cell and Regenerative Medicine Research Centre, China-New Zealand Belt and Road Joint Laboratory on Biomedicine and Health, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou 510530, China.
| |
Collapse
|
5
|
Hui T, Zhou J, Yao M, Xie Y, Zeng H. Advances in Spatial Omics Technologies. SMALL METHODS 2025:e2401171. [PMID: 40099571 DOI: 10.1002/smtd.202401171] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/29/2024] [Revised: 03/03/2025] [Indexed: 03/20/2025]
Abstract
Rapidly developing spatial omics technologies provide us with new approaches to deeply understanding the diversity and functions of cell types within organisms. Unlike traditional approaches, spatial omics technologies enable researchers to dissect the complex relationships between tissue structure and function at the cellular or even subcellular level. The application of spatial omics technologies provides new perspectives on key biological processes such as nervous system development, organ development, and tumor microenvironment. This review focuses on the advancements and strategies of spatial omics technologies, summarizes their applications in biomedical research, and highlights the power of spatial omics technologies in advancing the understanding of life sciences related to development and disease.
Collapse
Affiliation(s)
- Tianxiao Hui
- State Key Laboratory of Gene Function and Modulation Research, College of Future Technology, Peking-Tsinghua Center for Life Sciences, Peking University, Beijing, 100871, China
| | - Jian Zhou
- Peking-Tsinghua Center for Life Sciences, Tsinghua University, Beijing, 100084, China
| | - Muchen Yao
- College of Biological Sciences, China Agricultural University, Beijing, 100193, China
| | - Yige Xie
- School of Nursing, Peking University, Beijing, 100871, China
| | - Hu Zeng
- State Key Laboratory of Gene Function and Modulation Research, College of Future Technology, Peking-Tsinghua Center for Life Sciences, Peking University, Beijing, 100871, China
- Beijing Advanced Center of RNA Biology (BEACON), Peking University, Beijing, 100871, China
| |
Collapse
|
6
|
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
|
7
|
Braccioli L, van den Brand T, Alonso Saiz N, Fountas C, Celie PHN, Kazokaitė-Adomaitienė J, de Wit E. Identifying cross-lineage dependencies of cell-type-specific regulators in mouse gastruloids. Dev Cell 2025:S1534-5807(25)00118-2. [PMID: 40101716 DOI: 10.1016/j.devcel.2025.02.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2023] [Revised: 09/09/2024] [Accepted: 02/21/2025] [Indexed: 03/20/2025]
Abstract
Correct gene expression levels are crucial for normal development. Advances in genomics enable the inference of gene regulatory programs active during development but cannot capture the complex multicellular interactions occurring during mammalian embryogenesis in utero. In vitro models of mammalian development, like gastruloids, can overcome this limitation. Using time-resolved single-cell chromatin accessibility analysis, we delineated the regulatory profile during mouse gastruloid development, identifying critical drivers of developmental transitions. Gastruloids develop from bipotent progenitor cells driven by the transcription factors (TFs) OCT4, SOX2, and TBXT, differentiating into the mesoderm (characterized by the mesogenin 1 [MSGN1]) and spinal cord (characterized by CDX2). ΔCDX gastruloids fail to form spinal cord, while Msgn1 ablation inhibits paraxial mesoderm and spinal cord development. Chimeric gastruloids with ΔMSGN1 and wild-type cells formed both tissues, indicating that inter-tissue communication is necessary for spinal cord formation. Our work has important implications for studying inter-tissue communication and gene regulatory programs in development.
Collapse
Affiliation(s)
- Luca Braccioli
- Division of Gene Regulation, Netherlands Cancer Institute, 1066 CX Amsterdam, the Netherlands.
| | - Teun van den Brand
- Division of Gene Regulation, Netherlands Cancer Institute, 1066 CX Amsterdam, the Netherlands
| | - Noemi Alonso Saiz
- Division of Gene Regulation, Netherlands Cancer Institute, 1066 CX Amsterdam, the Netherlands
| | - Charis Fountas
- Division of Gene Regulation, Netherlands Cancer Institute, 1066 CX Amsterdam, the Netherlands
| | - Patrick H N Celie
- Protein Facility, Division of Biochemistry, Netherlands Cancer Institute, 1066 CX Amsterdam, the Netherlands; Oncode Institute, 3521 AL Utrecht, the Netherlands
| | - Justina Kazokaitė-Adomaitienė
- Protein Facility, Division of Biochemistry, Netherlands Cancer Institute, 1066 CX Amsterdam, the Netherlands; Oncode Institute, 3521 AL Utrecht, the Netherlands
| | - Elzo de Wit
- Division of Gene Regulation, Netherlands Cancer Institute, 1066 CX Amsterdam, the Netherlands.
| |
Collapse
|
8
|
Brooks ER, Moorman AR, Bhattacharya B, Prudhomme IS, Land M, Alcorn HL, Sharma R, Pe’er D, Zallen JA. A single-cell atlas of spatial and temporal gene expression in the mouse cranial neural plate. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2024.08.25.609458. [PMID: 39229123 PMCID: PMC11370589 DOI: 10.1101/2024.08.25.609458] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/05/2024]
Abstract
The formation of the mammalian brain requires regionalization and morphogenesis of the cranial neural plate, which transforms from an epithelial sheet into a closed tube that provides the structural foundation for neural patterning and circuit formation. Sonic hedgehog (SHH) signaling is important for cranial neural plate patterning and closure, but the transcriptional changes that give rise to the spatially regulated cell fates and behaviors that build the cranial neural tube have not been systematically analyzed. Here we used single-cell RNA sequencing to generate an atlas of gene expression at six consecutive stages of cranial neural tube closure in the mouse embryo. Ordering transcriptional profiles relative to the major axes of gene expression predicted spatially regulated expression of 870 genes along the anterior-posterior and mediolateral axes of the cranial neural plate and reproduced known expression patterns with over 85% accuracy. Single-cell RNA sequencing of embryos with activated SHH signaling revealed distinct SHH-regulated transcriptional programs in the developing forebrain, midbrain, and hindbrain, suggesting a complex interplay between anterior-posterior and mediolateral patterning systems. These results define a spatiotemporally resolved map of gene expression during cranial neural tube closure and provide a resource for investigating the transcriptional events that drive early mammalian brain development.
Collapse
Affiliation(s)
- Eric R. Brooks
- HHMI and Developmental Biology Program, Sloan Kettering Institute
- Department of Molecular Biomedical Sciences, College of Veterinary Medicine, North Carolina State University
| | - Andrew R. Moorman
- HHMI and Computational and Systems Biology Program, Sloan Kettering Institute
| | | | - Ian S. Prudhomme
- HHMI and Developmental Biology Program, Sloan Kettering Institute
| | - Max Land
- HHMI and Computational and Systems Biology Program, Sloan Kettering Institute
| | | | - Roshan Sharma
- HHMI and Computational and Systems Biology Program, Sloan Kettering Institute
| | - Dana Pe’er
- HHMI and Computational and Systems Biology Program, Sloan Kettering Institute
| | | |
Collapse
|
9
|
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
|
10
|
Ito K, Hirakawa T, Shigenobu S, Fujiyoshi H, Yamashita T. Mouse-Geneformer: A deep learning model for mouse single-cell transcriptome and its cross-species utility. PLoS Genet 2025; 21:e1011420. [PMID: 40106407 PMCID: PMC11964219 DOI: 10.1371/journal.pgen.1011420] [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: 09/10/2024] [Revised: 04/02/2025] [Accepted: 02/17/2025] [Indexed: 03/22/2025] Open
Abstract
Deep learning techniques are increasingly utilized to analyze large-scale single-cell RNA sequencing (scRNA-seq) data, offering valuable insights from complex transcriptome datasets. Geneformer, a pre-trained model using a Transformer Encoder architecture and human scRNA-seq datasets, has demonstrated remarkable success in human transcriptome analysis. However, given the prominence of the mouse, Mus musculus, as a primary mammalian model in biological and medical research, there is an acute need for a mouse-specific version of Geneformer. In this study, we developed a mouse-specific Geneformer (mouse-Geneformer) by constructing a large transcriptome dataset consisting of 21 million mouse scRNA-seq profiles and pre-training Geneformer on this dataset. The mouse-Geneformer effectively models the mouse transcriptome and, upon fine-tuning for downstream tasks, enhances the accuracy of cell type classification. In silico perturbation experiments using mouse-Geneformer successfully identified disease-causing genes that have been validated in in vivo experiments. These results demonstrate the feasibility of analyzing mouse data with mouse-Geneformer and highlight the robustness of the Geneformer architecture, applicable to any species with large-scale transcriptome data available. Furthermore, we found that mouse-Geneformer can analyze human transcriptome data in a cross-species manner. After the ortholog-based gene name conversion, the analysis of human scRNA-seq data using mouse-Geneformer, followed by fine-tuning with human data, achieved cell type classification accuracy comparable to that obtained using the original human Geneformer. In in silico simulation experiments using human disease models, we obtained results similar to human-Geneformer for the myocardial infarction model but only partially consistent results for the COVID-19 model, a trait unique to humans (laboratory mice are not susceptible to SARS-CoV-2). These findings suggest the potential for cross-species application of the Geneformer model while emphasizing the importance of species-specific models for capturing the full complexity of disease mechanisms. Despite the existence of the original Geneformer tailored for humans, human research could benefit from mouse-Geneformer due to its inclusion of samples that are ethically or technically inaccessible for humans, such as embryonic tissues and certain disease models. Additionally, this cross-species approach indicates potential use for non-model organisms, where obtaining large-scale single-cell transcriptome data is challenging.
Collapse
Affiliation(s)
- Keita Ito
- Graduate School of Engineering, Chubu University, Kasugai, Aichi, Japan
| | - Tsubasa Hirakawa
- Department of Artificial Intelligence and Robotics, Center for Mathematical Science and Artificial Intelligence, Chubu University, Kasugai, Aichi, Japan
| | - Shuji Shigenobu
- Trans-Scale Biology Center, National Institute for Basic Biology, Okazaki, Aichi, Japan
- Life Science Center for Survival Dynamics, Tsukuba Advanced Research Alliance (TARA), University of Tsukuba, Tsukuba, Ibaraki, Japan
| | | | - Takayoshi Yamashita
- Department of Artificial Intelligence and Robotics, Chubu University, Kasugai, Aichi, Japan
| |
Collapse
|
11
|
Li KR, Yu PL, Zheng QQ, Wang X, Fang X, Li LC, Xu CR. Spatiotemporal and genetic cell lineage tracing of endodermal organogenesis at single-cell resolution. Cell 2025; 188:796-813.e24. [PMID: 39824184 DOI: 10.1016/j.cell.2024.12.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2024] [Revised: 09/30/2024] [Accepted: 12/09/2024] [Indexed: 01/20/2025]
Abstract
During early mammalian development, the endoderm germ layer forms the foundation of the respiratory and digestive systems through complex patterning. This intricate process, guided by a series of cell fate decisions, remains only partially understood. Our study introduces innovative genetic tracing codes for 14 distinct endodermal regions using novel mouse strains. By integrating high-throughput and high-precision single-cell RNA sequencing with sophisticated imaging, we detailed the spatiotemporal and genetic lineage differentiation of the endoderm at single-cell resolution. We discovered an unexpected multipotentiality within early endodermal regions, allowing differentiation into various organ primordia. This research illuminates the complex and underestimated phenomenon where endodermal organs develop from multiple origins, prompting a reevaluation of traditional differentiation models. Our findings advance understanding in developmental biology and have significant implications for regenerative medicine and the development of advanced organoid models, providing insights into the intricate mechanisms that guide organogenesis.
Collapse
Affiliation(s)
- Ke-Ran Li
- State Key Laboratory of Female Fertility Promotion, Department of Medical Genetics, School of Basic Medical Sciences, Peking University, Beijing 100191, China; Department of Human Anatomy, Histology, and Embryology, School of Basic Medical Sciences, Peking University, Beijing 100191, China; Peking-Tsinghua Center for Life Sciences, Peking University, Beijing 100871, China
| | - Pei-Long Yu
- State Key Laboratory of Female Fertility Promotion, Department of Medical Genetics, School of Basic Medical Sciences, Peking University, Beijing 100191, China; Peking-Tsinghua Center for Life Sciences, Peking University, Beijing 100871, China
| | - Qi-Qi Zheng
- PKU-Tsinghua-NIBS Graduate Program, Peking University, Beijing 100871, China; Peking-Tsinghua Center for Life Sciences, Peking University, Beijing 100871, China
| | - Xin Wang
- Peking-Tsinghua Center for Life Sciences, Peking University, Beijing 100871, China; School of Life Sciences, Peking University, Beijing 100871, China
| | - Xuan Fang
- Department of Human Anatomy, Histology, and Embryology, School of Basic Medical Sciences, Peking University, Beijing 100191, China
| | - Lin-Chen Li
- Department of Human Anatomy, Histology, and Embryology, School of Basic Medical Sciences, Peking University, Beijing 100191, China
| | - Cheng-Ran Xu
- State Key Laboratory of Female Fertility Promotion, Department of Medical Genetics, School of Basic Medical Sciences, Peking University, Beijing 100191, China; Peking-Tsinghua Center for Life Sciences, Peking University, Beijing 100871, China.
| |
Collapse
|
12
|
Sarkar H, Lee E, Lopez-Darwin SL, Kang Y. Deciphering normal and cancer stem cell niches by spatial transcriptomics: opportunities and challenges. Genes Dev 2025; 39:64-85. [PMID: 39496456 PMCID: PMC11789490 DOI: 10.1101/gad.351956.124] [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] [Indexed: 11/06/2024]
Abstract
Cancer stem cells (CSCs) often exhibit stem-like attributes that depend on an intricate stemness-promoting cellular ecosystem within their niche. The interplay between CSCs and their niche has been implicated in tumor heterogeneity and therapeutic resistance. Normal stem cells (NSCs) and CSCs share stemness features and common microenvironmental components, displaying significant phenotypic and functional plasticity. Investigating these properties across diverse organs during normal development and tumorigenesis is of paramount research interest and translational potential. Advancements in next-generation sequencing (NGS), single-cell transcriptomics, and spatial transcriptomics have ushered in a new era in cancer research, providing high-resolution and comprehensive molecular maps of diseased tissues. Various spatial technologies, with their unique ability to measure the location and molecular profile of a cell within tissue, have enabled studies on intratumoral architecture and cellular cross-talk within the specific niches. Moreover, delineation of spatial patterns for niche-specific properties such as hypoxia, glucose deprivation, and other microenvironmental remodeling are revealed through multilevel spatial sequencing. This tremendous progress in technology has also been paired with the advent of computational tools to mitigate technology-specific bottlenecks. Here we discuss how different spatial technologies are used to identify NSCs and CSCs, as well as their associated niches. Additionally, by exploring related public data sets, we review the current challenges in characterizing such niches, which are often hindered by technological limitations, and the computational solutions used to address them.
Collapse
Affiliation(s)
- Hirak Sarkar
- Department of Molecular Biology, Princeton University, Princeton, New Jersey 08544, USA
- Ludwig Institute for Cancer Research Princeton Branch, Princeton, New Jersey 08544, USA
- Department of Computer Science, Princeton, New Jersey 08544, USA
| | - Eunmi Lee
- Department of Molecular Biology, Princeton University, Princeton, New Jersey 08544, USA
| | - Sereno L Lopez-Darwin
- Lewis-Sigler Institute for Integrative Genomics, Princeton University, Princeton, New Jersey 08544, USA
| | - Yibin Kang
- Department of Molecular Biology, Princeton University, Princeton, New Jersey 08544, USA;
- Ludwig Institute for Cancer Research Princeton Branch, Princeton, New Jersey 08544, USA
- Cancer Metabolism and Growth Program, Rutgers Cancer Institute of New Jersey, New Brunswick, New Jersey 08903, USA
| |
Collapse
|
13
|
Samadi Z, Hao K, Askary A. SMORE: spatial motifs reveal patterns in cellular architecture of complex tissues. Genome Biol 2025; 26:3. [PMID: 39754206 PMCID: PMC11697875 DOI: 10.1186/s13059-024-03467-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2024] [Accepted: 12/20/2024] [Indexed: 01/07/2025] Open
Abstract
Deciphering the link between tissue architecture and function requires methods to identify and interpret patterns in spatial arrangement of cells. We present SMORE, an approach to detect patterns in sequential arrangements of cells and examine their associated gene expression specializations. Applied to retina, brain, and embryonic tissue maps, SMORE identifies novel spatial motifs, including one that offers a new mechanism of action for type 1b bipolar cells. Structural signatures detected by SMORE also form a basis for classifying tissues. Together, our method provides a new framework for uncovering spatial complexity in tissue organization and offers novel insights into tissue function.
Collapse
Affiliation(s)
- Zainalabedin Samadi
- Department of Molecular, Cell and Developmental Biology, University of California, Los Angeles, 90095, CA, USA
| | - Kai Hao
- Department of Molecular, Cell and Developmental Biology, University of California, Los Angeles, 90095, CA, USA
| | - Amjad Askary
- Department of Molecular, Cell and Developmental Biology, University of California, Los Angeles, 90095, CA, USA.
| |
Collapse
|
14
|
Kulasinghe A, Berrell N, Donovan ML, Nilges BS. Spatial-Omics Methods and Applications. Methods Mol Biol 2025; 2880:101-146. [PMID: 39900756 DOI: 10.1007/978-1-0716-4276-4_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: 02/05/2025]
Abstract
Traditional tissue profiling approaches have evolved from bulk studies to single-cell analysis over the last decade; however, the spatial context in tissues and microenvironments has always been lost. Over the last 5 years, spatial technologies have emerged that enabled researchers to investigate tissues in situ for proteins and transcripts without losing anatomy and histology. The field of spatial-omics enables highly multiplexed analysis of biomolecules like RNAs and proteins in their native spatial context-and has matured from initial proof-of-concept studies to a thriving field with widespread applications from basic research to translational and clinical studies. While there has been wide adoption of spatial technologies, there remain challenges with the standardization of methodologies, sample compatibility, throughput, resolution, and ease of use. In this chapter, we discuss the current state of the field and highlight technological advances and limitations.
Collapse
Affiliation(s)
- Arutha Kulasinghe
- Frazer Institute, Faculty of Medicine, University of Queensland, Brisbane, QLD, Australia
- Queensland Spatial Biology Centre, Wesley Research Institute, The Wesley Hospital, Auchenflower, QLD, Australia
| | - Naomi Berrell
- Queensland Spatial Biology Centre, Wesley Research Institute, The Wesley Hospital, Auchenflower, QLD, Australia
| | - Meg L Donovan
- Queensland Spatial Biology Centre, Wesley Research Institute, The Wesley Hospital, Auchenflower, QLD, Australia
| | | |
Collapse
|
15
|
Qiu X, Zhu DY, Lu Y, Yao J, Jing Z, Min KH, Cheng M, Pan H, Zuo L, King S, Fang Q, Zheng H, Wang M, Wang S, Zhang Q, Yu S, Liao S, Liu C, Wu X, Lai Y, Hao S, Zhang Z, Wu L, Zhang Y, Li M, Tu Z, Lin J, Yang Z, Li Y, Gu Y, Ellison D, Chen A, Liu L, Weissman JS, Ma J, Xu X, Liu S, Bai Y. Spatiotemporal modeling of molecular holograms. Cell 2024; 187:7351-7373.e61. [PMID: 39532097 DOI: 10.1016/j.cell.2024.10.011] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2022] [Revised: 05/29/2024] [Accepted: 10/08/2024] [Indexed: 11/16/2024]
Abstract
Quantifying spatiotemporal dynamics during embryogenesis is crucial for understanding congenital diseases. We developed Spateo (https://github.com/aristoteleo/spateo-release), a 3D spatiotemporal modeling framework, and applied it to a 3D mouse embryogenesis atlas at E9.5 and E11.5, capturing eight million cells. Spateo enables scalable, partial, non-rigid alignment, multi-slice refinement, and mesh correction to create molecular holograms of whole embryos. It introduces digitization methods to uncover multi-level biology from subcellular to whole organ, identifying expression gradients along orthogonal axes of emergent 3D structures, e.g., secondary organizers such as midbrain-hindbrain boundary (MHB). Spateo further jointly models intercellular and intracellular interaction to dissect signaling landscapes in 3D structures, including the zona limitans intrathalamica (ZLI). Lastly, Spateo introduces "morphometric vector fields" of cell migration and integrates spatial differential geometry to unveil molecular programs underlying asymmetrical murine heart organogenesis and others, bridging macroscopic changes with molecular dynamics. Thus, Spateo enables the study of organ ecology at a molecular level in 3D space over time.
Collapse
Affiliation(s)
- Xiaojie Qiu
- Department of Genetics, Stanford University School of Medicine, Stanford, CA, USA; Basic Sciences and Engineering Initiative, Betty Irene Moore Children's Heart Center, Lucile Packard Children's Hospital, Stanford, CA, USA; Department of Computer Science, Stanford University, Stanford, CA 94305, USA; Stanford Cardiovascular Institute, Stanford University, Stanford, CA, USA.
| | - Daniel Y Zhu
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Yifan Lu
- Department of Genetics, Stanford University School of Medicine, Stanford, CA, USA; Basic Sciences and Engineering Initiative, Betty Irene Moore Children's Heart Center, Lucile Packard Children's Hospital, Stanford, CA, USA; Department of Computer Science, Stanford University, Stanford, CA 94305, USA; Electronic Information School, Wuhan University, Wuhan 430072, China
| | - Jiajun Yao
- BGI Research, Hangzhou 310030, China; BGI Research, Sanya 572025, China; College of Life Sciences, Northwest University, Xi'an 710069, China
| | - Zehua Jing
- BGI Research, Hangzhou 310030, China; BGI Research, Sanya 572025, China; College of Life Sciences, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Kyung Hoi Min
- Ginkgo Bioworks, The Innovation and Design Building, Boston, MA 02210, USA
| | - Mengnan Cheng
- BGI Research, Hangzhou 310030, China; BGI Research, Shenzhen 518083, China
| | | | - Lulu Zuo
- BGI Research, Shenzhen 518083, China
| | - Samuel King
- Department of Bioengineering, Stanford University School of Medicine, Stanford, CA, USA
| | - Qi Fang
- BGI Research, Hangzhou 310030, China; BGI Research, Shenzhen 518083, China
| | - Huiwen Zheng
- BGI Research, Hangzhou 310030, China; College of Life Sciences, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Mingyue Wang
- BGI Research, Hangzhou 310030, China; Shenzhen Key Laboratory of Gene Regulation and Systems Biology, School of Life Sciences, Southern University of Science and Technology, Shenzhen 518055, China
| | - Shuai Wang
- BGI Research, Hangzhou 310030, China; College of Life Sciences, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Qingquan Zhang
- Department of Medicine, Division of Cardiology, University of California, San Diego, La Jolla, CA, USA
| | - Sichao Yu
- Whitehead Institute for Biomedical Research, Cambridge, MA, USA
| | - Sha Liao
- BGI Research, Shenzhen 518083, China; STOmics Tech Co., Ltd, Shenzhen 518083, China; BGI Research, Chongqing 401329, China
| | - Chao Liu
- BGI Research, Wuhan 430074, China
| | - Xinchao Wu
- BGI Research, Hangzhou 310030, China; BGI Research, Sanya 572025, China; School of Life Sciences, Southern University of Science and Technology, Shenzhen 518055, China
| | - Yiwei Lai
- BGI Research, Shenzhen 518083, China
| | | | - Zhewei Zhang
- BGI Research, Hangzhou 310030, China; BGI Research, Sanya 572025, China; School of Life Sciences, Southern University of Science and Technology, Shenzhen 518055, China
| | - Liang Wu
- BGI Research, Chongqing 401329, China
| | | | - Mei Li
- STOmics Tech Co., Ltd, Shenzhen 518083, China
| | - Zhencheng Tu
- BGI Research, Hangzhou 310030, China; College of Life Sciences, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Jinpei Lin
- BGI Research, Hangzhou 310030, China; BGI Research, Sanya 572025, China
| | - Zhuoxuan Yang
- BGI Research, Hangzhou 310030, China; School of Life Sciences, Southern University of Science and Technology, Shenzhen 518055, China
| | | | - Ying Gu
- BGI Research, Hangzhou 310030, China; BGI Research, Shenzhen 518083, China; College of Life Sciences, University of Chinese Academy of Sciences, Beijing 100049, China
| | | | - Ao Chen
- BGI Research, Shenzhen 518083, China; STOmics Tech Co., Ltd, Shenzhen 518083, China; BGI Research, Chongqing 401329, China
| | - Longqi Liu
- BGI Research, Hangzhou 310030, China; Shenzhen Bay Laboratory, Shenzhen 518132, China; Shenzhen Key Laboratory of Single-Cell Omics, BGI-Shenzhen, Shenzhen 518120, China
| | - Jonathan S Weissman
- Whitehead Institute for Biomedical Research, Cambridge, MA, USA; Department of Biology and Howard Hughes Medical Institute, Massachusetts Institute of Technology, Cambridge, MA, USA; Koch Institute for Integrative Cancer Research at MIT, MIT, Cambridge, MA, USA
| | - Jiayi Ma
- Electronic Information School, Wuhan University, Wuhan 430072, China.
| | - Xun Xu
- BGI Research, Hangzhou 310030, China; College of Life Sciences, University of Chinese Academy of Sciences, Beijing 100049, China; Guangdong Provincial Key Laboratory of Genome Read and Write, BGI-Shenzhen, Shenzhen 518120, China.
| | - Shiping Liu
- BGI Research, Hangzhou 310030, China; Shenzhen Bay Laboratory, Shenzhen 518132, China; Shenzhen Key Laboratory of Single-Cell Omics, BGI-Shenzhen, Shenzhen 518120, China; The Guangdong-Hong Kong Joint Laboratory on Immunological and Genetic Kidney Diseases, Guangzhou, Guangdong, China.
| | - Yinqi Bai
- BGI Research, Sanya 572025, China; Hainan Technology Innovation Center for Marine Biological Resources Utilization (Preparatory Period), BGI Research, Sanya 572025, China.
| |
Collapse
|
16
|
Wang J, Ye F, Chai H, Jiang Y, Wang T, Ran X, Xia Q, Xu Z, Fu Y, Zhang G, Wu H, Guo G, Guo H, Ruan Y, Wang Y, Xing D, Xu X, Zhang Z. Advances and applications in single-cell and spatial genomics. SCIENCE CHINA. LIFE SCIENCES 2024:10.1007/s11427-024-2770-x. [PMID: 39792333 DOI: 10.1007/s11427-024-2770-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/20/2024] [Accepted: 10/10/2024] [Indexed: 01/12/2025]
Abstract
The applications of single-cell and spatial technologies in recent times have revolutionized the present understanding of cellular states and the cellular heterogeneity inherent in complex biological systems. These advancements offer unprecedented resolution in the examination of the functional genomics of individual cells and their spatial context within tissues. In this review, we have comprehensively discussed the historical development and recent progress in the field of single-cell and spatial genomics. We have reviewed the breakthroughs in single-cell multi-omics technologies, spatial genomics methods, and the computational strategies employed toward the analyses of single-cell atlas data. Furthermore, we have highlighted the advances made in constructing cellular atlases and their clinical applications, particularly in the context of disease. Finally, we have discussed the emerging trends, challenges, and opportunities in this rapidly evolving field.
Collapse
Affiliation(s)
- Jingjing Wang
- Bone Marrow Transplantation Center of the First Affiliated Hospital & Liangzhu Laboratory, Zhejiang University School of Medicine, Hangzhou, 310058, China
| | - Fang Ye
- Bone Marrow Transplantation Center of the First Affiliated Hospital & Liangzhu Laboratory, Zhejiang University School of Medicine, Hangzhou, 310058, China
| | - Haoxi Chai
- Life Sciences Institute and The Second Affiliated Hospital, Zhejiang University, Hangzhou, 310058, China
| | - Yujia Jiang
- BGI Research, Shenzhen, 518083, China
- BGI Research, Hangzhou, 310030, China
| | - Teng Wang
- Biomedical Pioneering Innovation Center (BIOPIC) and School of Life Sciences, Peking University, Beijing, 100871, China
- Academy for Advanced Interdisciplinary Studies, Peking University, Beijing, 100871, China
| | - Xia Ran
- Bone Marrow Transplantation Center of the First Affiliated Hospital & Liangzhu Laboratory, Zhejiang University School of Medicine, Hangzhou, 310058, China
- Institute of Hematology, Zhejiang University, Hangzhou, 310000, China
| | - Qimin Xia
- Biomedical Pioneering Innovation Center (BIOPIC) and School of Life Sciences, Peking University, Beijing, 100871, China
| | - Ziye Xu
- Department of Laboratory Medicine of The First Affiliated Hospital & Liangzhu Laboratory, Zhejiang University School of Medicine, Hangzhou, 310058, China
| | - Yuting Fu
- Bone Marrow Transplantation Center of the First Affiliated Hospital & Liangzhu Laboratory, Zhejiang University School of Medicine, Hangzhou, 310058, China
- Center for Stem Cell and Regenerative Medicine, Zhejiang University School of Medicine, Hangzhou, 310058, China
| | - Guodong Zhang
- Bone Marrow Transplantation Center of the First Affiliated Hospital & Liangzhu Laboratory, Zhejiang University School of Medicine, Hangzhou, 310058, China
- Center for Stem Cell and Regenerative Medicine, Zhejiang University School of Medicine, Hangzhou, 310058, China
| | - Hanyu Wu
- Bone Marrow Transplantation Center of the First Affiliated Hospital & Liangzhu Laboratory, Zhejiang University School of Medicine, Hangzhou, 310058, China
- Center for Stem Cell and Regenerative Medicine, Zhejiang University School of Medicine, Hangzhou, 310058, China
| | - Guoji Guo
- Bone Marrow Transplantation Center of the First Affiliated Hospital & Liangzhu Laboratory, Zhejiang University School of Medicine, Hangzhou, 310058, China.
- Center for Stem Cell and Regenerative Medicine, Zhejiang University School of Medicine, Hangzhou, 310058, China.
- Zhejiang Provincial Key Lab for Tissue Engineering and Regenerative Medicine, Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cell and Regenerative Medicine, Hangzhou, 310058, China.
- Institute of Hematology, Zhejiang University, Hangzhou, 310000, China.
| | - Hongshan Guo
- Bone Marrow Transplantation Center of the First Affiliated Hospital & Liangzhu Laboratory, Zhejiang University School of Medicine, Hangzhou, 310058, China.
- Institute of Hematology, Zhejiang University, Hangzhou, 310000, China.
| | - Yijun Ruan
- Life Sciences Institute and The Second Affiliated Hospital, Zhejiang University, Hangzhou, 310058, China.
| | - Yongcheng Wang
- Department of Laboratory Medicine of The First Affiliated Hospital & Liangzhu Laboratory, Zhejiang University School of Medicine, Hangzhou, 310058, China.
| | - Dong Xing
- Biomedical Pioneering Innovation Center (BIOPIC) and School of Life Sciences, Peking University, Beijing, 100871, China.
- Beijing Advanced Innovation Center for Genomics (ICG), Peking University, Beijing, 100871, China.
| | - Xun Xu
- BGI Research, Shenzhen, 518083, China.
- BGI Research, Hangzhou, 310030, China.
- Guangdong Provincial Key Laboratory of Genome Read and Write, BGI Research, Shenzhen, 518083, China.
| | - Zemin Zhang
- Biomedical Pioneering Innovation Center (BIOPIC) and School of Life Sciences, Peking University, Beijing, 100871, China.
| |
Collapse
|
17
|
Zhu J, Pang K, Hu B, He R, Wang N, Jiang Z, Ji P, Zhao F. Custom microfluidic chip design enables cost-effective three-dimensional spatiotemporal transcriptomics with a wide field of view. Nat Genet 2024; 56:2259-2270. [PMID: 39256584 PMCID: PMC11525186 DOI: 10.1038/s41588-024-01906-4] [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: 01/02/2024] [Accepted: 08/13/2024] [Indexed: 09/12/2024]
Abstract
Spatial transcriptomic techniques offer unprecedented insights into the molecular organization of complex tissues. However, integrating cost-effectiveness, high throughput, a wide field of view and compatibility with three-dimensional (3D) volumes has been challenging. Here we introduce microfluidics-assisted grid chips for spatial transcriptome sequencing (MAGIC-seq), a new method that combines carbodiimide chemistry, spatial combinatorial indexing and innovative microfluidics design. This technique allows sensitive and reproducible profiling of diverse tissue types, achieving an eightfold increase in throughput, minimal cost and reduced batch effects. MAGIC-seq breaks conventional microfluidics limits by enhancing barcoding efficiency and enables analysis of whole postnatal mouse sections, providing comprehensive cellular structure elucidation at near single-cell resolution, uncovering transcriptional variations and dynamic trajectories of mouse organogenesis. Our 3D transcriptomic atlas of the developing mouse brain, consisting of 93 sections, reveals the molecular and cellular landscape, serving as a valuable resource for neuroscience and developmental biology. Overall, MAGIC-seq is a high-throughput, cost-effective, large field of view and versatile method for spatial transcriptomic studies.
Collapse
Affiliation(s)
- Junjie Zhu
- Institute of Zoology, Chinese Academy of Sciences, Beijing, China
- Key Laboratory of Systems Biology, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou, China
| | - Kun Pang
- Institute of Zoology, Chinese Academy of Sciences, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Beiyu Hu
- Institute of Zoology, Chinese Academy of Sciences, Beijing, China
- Key Laboratory of Systems Biology, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou, China
| | - Ruiqiao He
- Institute of Zoology, Chinese Academy of Sciences, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Ning Wang
- Institute of Zoology, Chinese Academy of Sciences, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Zewen Jiang
- Institute of Zoology, Chinese Academy of Sciences, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Peifeng Ji
- Institute of Zoology, Chinese Academy of Sciences, Beijing, China
| | - Fangqing Zhao
- Institute of Zoology, Chinese Academy of Sciences, Beijing, China.
- Key Laboratory of Systems Biology, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou, China.
- University of Chinese Academy of Sciences, Beijing, China.
| |
Collapse
|
18
|
Asami S, Yin C, Garza LA, Kalhor R. Deconvolving organogenesis in space and time via spatial transcriptomics in thick tissues. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.09.24.614640. [PMID: 39386671 PMCID: PMC11463617 DOI: 10.1101/2024.09.24.614640] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 10/12/2024]
Abstract
Organ development is guided by a space-time landscape that constraints cell behavior. This landscape is challenging to characterize for the hair follicle - the most abundant mini organ - due to its complex microscopic structure and asynchronous development. We developed 3DEEP, a tissue clearing and spatial transcriptomic strategy for characterizing tissue blocks up to 400 µm in thickness. We captured 371 hair follicles at different stages of organogenesis in 1 mm3 of skin of a 12-hour-old mouse with 6 million transcripts from 81 genes. From this single time point, we deconvoluted follicles by age based on whole-organ molecular pseudotimes to animate a stop-motion 3D atlas of follicle development along its trajectory. We defined molecular stages for hair follicle organogenesis and characterized the order of emergence for its structures, differential signaling dynamics at its top and bottom, morphogen shifts preceding and accompanying structural changes, and series of structural changes leading to the formation of its canal and opening. We further found that hair follicle stem cells and their niche are established and stratified early in organogenesis, before the formation of the hair bulb. Overall, this work demonstrates the power of increased depth of spatial transcriptomics to provide a four-dimensional analysis of organogenesis.
Collapse
Affiliation(s)
- Soichiro Asami
- Department of Biomedical Engineering, Center for Epigenetics, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Chenshuo Yin
- Department of Biomedical Engineering, Center for Epigenetics, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Luis A. Garza
- Department of Dermatology, Department of Cell Biology, Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Reza Kalhor
- Department of Biomedical Engineering, Center for Epigenetics, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Molecular Biology and Genetics, Department of Medicine, Department of Neuroscience, Department of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| |
Collapse
|
19
|
Mao G, Yang Y, Luo Z, Lin C, Xie P. SpatialQC: automated quality control for spatial transcriptome data. Bioinformatics 2024; 40:btae458. [PMID: 39051702 PMCID: PMC11333854 DOI: 10.1093/bioinformatics/btae458] [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: 02/14/2024] [Revised: 07/04/2024] [Accepted: 07/24/2024] [Indexed: 07/27/2024] Open
Abstract
SUMMARY The advent of spatial transcriptomics has revolutionized our understanding of the spatial heterogeneity in tissues, providing unprecedented insights into the cellular and molecular mechanisms underlying biological processes. Although quality control (QC) critical for downstream data analyses, there is currently a lack of specialized tools for one-stop spatial transcriptome QC. Here, we introduce SpatialQC, a one-stop QC pipeline, which generates comprehensive QC reports and produces clean data in an interactive fashion. SpatialQC is widely applicable to spatial transcriptomic techniques. AVAILABILITY AND IMPLEMENTATION source code and user manuals are available via https://github.com/mgy520/spatialQC, and deposited on Zenodo (https://doi.org/10.5281/zenodo.12634669).
Collapse
Affiliation(s)
- Guangyao Mao
- Key Laboratory of Developmental Genes and Human Disease, School of Life Science and Technology, Southeast University, Nanjing 210000, China
- Co-innovation Center of Neuroregeneration, Nantong University, Nantong 226000, China
| | - Yi Yang
- Key Laboratory of Developmental Genes and Human Disease, School of Life Science and Technology, Southeast University, Nanjing 210000, China
| | - Zhuojuan Luo
- Key Laboratory of Developmental Genes and Human Disease, School of Life Science and Technology, Southeast University, Nanjing 210000, China
- Co-innovation Center of Neuroregeneration, Nantong University, Nantong 226000, China
- Center of Reproductive Medicine, Fujian Maternity and Child Health Hospital, Fuzhou 350000, China
- Shenzhen Research Institute, Southeast University, Shenzhen, China
| | - Chengqi Lin
- Key Laboratory of Developmental Genes and Human Disease, School of Life Science and Technology, Southeast University, Nanjing 210000, China
- Co-innovation Center of Neuroregeneration, Nantong University, Nantong 226000, China
- Center of Reproductive Medicine, Fujian Maternity and Child Health Hospital, Fuzhou 350000, China
- Shenzhen Research Institute, Southeast University, Shenzhen, China
| | - Peng Xie
- School of Biological Science & Medical Engineering, Southeast University, Nanjing 518000, China
| |
Collapse
|
20
|
Bolondi A, Law BK, Kretzmer H, Gassaloglu SI, Buschow R, Riemenschneider C, Yang D, Walther M, Veenvliet JV, Meissner A, Smith ZD, Chan MM. Reconstructing axial progenitor field dynamics in mouse stem cell-derived embryoids. Dev Cell 2024; 59:1489-1505.e14. [PMID: 38579718 PMCID: PMC11187653 DOI: 10.1016/j.devcel.2024.03.024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2023] [Revised: 12/13/2023] [Accepted: 03/12/2024] [Indexed: 04/07/2024]
Abstract
Embryogenesis requires substantial coordination to translate genetic programs to the collective behavior of differentiating cells, but understanding how cellular decisions control tissue morphology remains conceptually and technically challenging. Here, we combine continuous Cas9-based molecular recording with a mouse embryonic stem cell-based model of the embryonic trunk to build single-cell phylogenies that describe the behavior of transient, multipotent neuro-mesodermal progenitors (NMPs) as they commit into neural and somitic cell types. We find that NMPs show subtle transcriptional signatures related to their recent differentiation and contribute to downstream lineages through a surprisingly broad distribution of individual fate outcomes. Although decision-making can be heavily influenced by environmental cues to induce morphological phenotypes, axial progenitors intrinsically mature over developmental time to favor the neural lineage. Using these data, we present an experimental and analytical framework for exploring the non-homeostatic dynamics of transient progenitor populations as they shape complex tissues during critical developmental windows.
Collapse
Affiliation(s)
- Adriano Bolondi
- Department of Genome Regulation, Max Planck Institute for Molecular Genetics, 14195 Berlin, Germany
| | - Benjamin K Law
- Lewis-Sigler Institute for Integrative Genomics, Princeton University, Princeton, NJ 08544, USA; Department of Molecular Biology, Princeton University, Princeton, NJ 08544, USA
| | - Helene Kretzmer
- Department of Genome Regulation, Max Planck Institute for Molecular Genetics, 14195 Berlin, Germany
| | - Seher Ipek Gassaloglu
- Department of Genome Regulation, Max Planck Institute for Molecular Genetics, 14195 Berlin, Germany; Max Planck Institute of Molecular Cell Biology and Genetics, 01307 Dresden, Germany
| | - René Buschow
- Microscopy Core Facility, Max Planck Institute for Molecular Genetics, 14195 Berlin, Germany
| | | | - Dian Yang
- Department of Molecular Pharmacology and Therapeutics & Systems Biology, Columbia University, New York, NY 10032, USA
| | - Maria Walther
- Department of Genome Regulation, Max Planck Institute for Molecular Genetics, 14195 Berlin, Germany
| | - Jesse V Veenvliet
- Max Planck Institute of Molecular Cell Biology and Genetics, 01307 Dresden, Germany; Cluster of Excellence Physics of Life, Technische Universität Dresden, 01307 Dresden, Germany; Center for Systems Biology Dresden, 01307 Dresden, Germany
| | - Alexander Meissner
- Department of Genome Regulation, Max Planck Institute for Molecular Genetics, 14195 Berlin, Germany; Institute of Chemistry and Biochemistry, Freie Universität Berlin, 14195 Berlin, Germany.
| | - Zachary D Smith
- Department of Genetics, Yale Stem Cell Center, Yale School of Medicine, New Haven, CT 06519, USA.
| | - Michelle M Chan
- Lewis-Sigler Institute for Integrative Genomics, Princeton University, Princeton, NJ 08544, USA; Department of Molecular Biology, Princeton University, Princeton, NJ 08544, USA.
| |
Collapse
|
21
|
Dupont C. A comprehensive review: synergizing stem cell and embryonic development knowledge in mouse and human integrated stem cell-based embryo models. Front Cell Dev Biol 2024; 12:1386739. [PMID: 38715920 PMCID: PMC11074781 DOI: 10.3389/fcell.2024.1386739] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2024] [Accepted: 04/05/2024] [Indexed: 01/06/2025] Open
Abstract
Mammalian stem cell-based embryo models have emerged as innovative tools for investigating early embryogenesis in both mice and primates. They not only reduce the need for sacrificing mice but also overcome ethical limitations associated with human embryo research. Furthermore, they provide a platform to address scientific questions that are otherwise challenging to explore in vivo. The usefulness of a stem cell-based embryo model depends on its fidelity in replicating development, efficiency and reproducibility; all essential for addressing biological queries in a quantitative manner, enabling statistical analysis. Achieving such fidelity and efficiency requires robust systems that demand extensive optimization efforts. A profound understanding of pre- and post-implantation development, cellular plasticity, lineage specification, and existing models is imperative for making informed decisions in constructing these models. This review aims to highlight essential differences in embryo development and stem cell biology between mice and humans, assess how these variances influence the formation of partially and fully integrated stem cell models, and identify critical challenges in the field.
Collapse
Affiliation(s)
- Cathérine Dupont
- Department of Developmental Biology, Erasmus University Medical Center, Rotterdam, Netherlands
| |
Collapse
|
22
|
Herlin MK. Genetics of Mayer-Rokitansky-Küster-Hauser (MRKH) syndrome: advancements and implications. Front Endocrinol (Lausanne) 2024; 15:1368990. [PMID: 38699388 PMCID: PMC11063329 DOI: 10.3389/fendo.2024.1368990] [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: 01/11/2024] [Accepted: 04/04/2024] [Indexed: 05/05/2024] Open
Abstract
Mayer-Rokitansky-Küster-Hauser (MRKH) syndrome is a congenital anomaly characterized by agenesis/aplasia of the uterus and upper part of the vagina in females with normal external genitalia and a normal female karyotype (46,XX). Patients typically present during adolescence with complaints of primary amenorrhea where the diagnosis is established with significant implications including absolute infertility. Most often cases appear isolated with no family history of MRKH syndrome or related anomalies. However, cumulative reports of familial recurrence suggest genetic factors to be involved. Early candidate gene studies had limited success in their search for genetic causes of MRKH syndrome. More recently, genomic investigations using chromosomal microarray and genome-wide sequencing have been successful in detecting promising genetic variants associated with MRKH syndrome, including 17q12 (LHX1, HNF1B) and 16p11.2 (TBX6) deletions and sequence variations in GREB1L and PAX8, pointing towards a heterogeneous etiology with various genes involved. With uterus transplantation as an emerging fertility treatment in MRKH syndrome and increasing evidence for genetic etiologies, the need for genetic counseling concerning the recurrence risk in offspring will likely increase. This review presents the advancements in MRKH syndrome genetics from early familial occurrences and candidate gene searches to current genomic studies. Moreover, the review provides suggestions for future genetic investigations and discusses potential implications for clinical practice.
Collapse
Affiliation(s)
- Morten Krogh Herlin
- Department of Clinical Genetics, Aarhus University Hospital, Aarhus N, Denmark
| |
Collapse
|
23
|
Zhuang H, Ji Z. PreTSA: computationally efficient modeling of temporal and spatial gene expression patterns. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.03.20.585926. [PMID: 38585819 PMCID: PMC10996487 DOI: 10.1101/2024.03.20.585926] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/09/2024]
Abstract
Modeling temporal and spatial gene expression patterns in large-scale single-cell and spatial transcriptomics data is a computationally intensive task. We present PreTSA, a method that offers computational efficiency in modeling these patterns and is applicable to single-cell and spatial transcriptomics data comprising millions of cells. PreTSA consistently matches the results of state-of-the-art methods while significantly reducing computational time. PreTSA provides a unique solution for studying gene expression patterns in extremely large datasets.
Collapse
Affiliation(s)
- Haotian Zhuang
- Department of Biostatistics and Bioinformatics, Duke University School of Medicine, Durham, NC, USA
| | - Zhicheng Ji
- Department of Biostatistics and Bioinformatics, Duke University School of Medicine, Durham, NC, USA
| |
Collapse
|
24
|
Helms HR, Oyama KA, Ware JP, Ibsen SD, Bertassoni LE. Multiplex Single-Cell Bioprinting for Engineering of Heterogeneous Tissue Constructs with Subcellular Spatial Resolution. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.02.01.578499. [PMID: 38352428 PMCID: PMC10862823 DOI: 10.1101/2024.02.01.578499] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 02/24/2024]
Abstract
Tissue development, function, and disease are largely driven by the spatial organization of individual cells and their cell-cell interactions. Precision engineered tissues with single-cell spatial resolution, therefore, have tremendous potential for next generation disease models, drug discovery, and regenerative therapeutics. Despite significant advancements in biofabrication approaches to improve feature resolution, strategies to fabricate tissues with the exact same organization of individual cells in their native cellular microenvironment have remained virtually non-existent to date. Here we report a method to spatially pattern single cells with up to eight cell phenotypes and subcellular spatial precision. As proof-of-concept we first demonstrate the ability to systematically assess the influence of cellular microenvironments on cell behavior by controllably altering the spatial arrangement of cell types in bioprinted precision cell-cell interaction arrays. We then demonstrate, for the first time, the ability to produce high-fidelity replicas of a patient's annotated cancer biopsy with subcellular resolution. The ability to replicate native cellular microenvironments marks a significant advancement for precision biofabricated in-vitro models, where heterogenous tissues can be engineered with single-cell spatial precision to advance our understanding of complex biological systems in a controlled and systematic manner.
Collapse
Affiliation(s)
- Haylie R Helms
- Knight Cancer Precision Biofabrication Hub, Knight Cancer Institute, Oregon Health and Science University, Portland, OR 97201, USA
- Department of Biomedical Engineering, School of Medicine, Oregon Health and Science University, Portland, OR 97201, USA
- Cancer Early Detection Advanced Research Center (CEDAR), Knight Cancer Institute, Oregon Health and Science University, Portland, OR 97201, USA
| | - Kody A Oyama
- Knight Cancer Precision Biofabrication Hub, Knight Cancer Institute, Oregon Health and Science University, Portland, OR 97201, USA
| | - Jason P Ware
- Department of Biomedical Engineering, School of Medicine, Oregon Health and Science University, Portland, OR 97201, USA
- Cancer Early Detection Advanced Research Center (CEDAR), Knight Cancer Institute, Oregon Health and Science University, Portland, OR 97201, USA
| | - Stuart D Ibsen
- Department of Biomedical Engineering, School of Medicine, Oregon Health and Science University, Portland, OR 97201, USA
- Cancer Early Detection Advanced Research Center (CEDAR), Knight Cancer Institute, Oregon Health and Science University, Portland, OR 97201, USA
| | - Luiz E Bertassoni
- Knight Cancer Precision Biofabrication Hub, Knight Cancer Institute, Oregon Health and Science University, Portland, OR 97201, USA
- Department of Biomedical Engineering, School of Medicine, Oregon Health and Science University, Portland, OR 97201, USA
- Cancer Early Detection Advanced Research Center (CEDAR), Knight Cancer Institute, Oregon Health and Science University, Portland, OR 97201, USA
- Division of Oncological Sciences, Knight Cancer Institute, Oregon Health and Science University, Portland, OR 97201, USA
- Division of Biomaterials and Biomechanics, Department of Oral Rehabilitation and Biosciences, School of Dentistry, Oregon Health and Science University, Portland, OR 97201, USA
- Center for Regenerative Medicine, School of Medicine, Oregon Health and Science University, Portland, OR 97201, USA
| |
Collapse
|
25
|
Ye F, Wang J, Li J, Mei Y, Guo G. Mapping Cell Atlases at the Single-Cell Level. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2305449. [PMID: 38145338 PMCID: PMC10885669 DOI: 10.1002/advs.202305449] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/07/2023] [Revised: 12/01/2023] [Indexed: 12/26/2023]
Abstract
Recent advancements in single-cell technologies have led to rapid developments in the construction of cell atlases. These atlases have the potential to provide detailed information about every cell type in different organisms, enabling the characterization of cellular diversity at the single-cell level. Global efforts in developing comprehensive cell atlases have profound implications for both basic research and clinical applications. This review provides a broad overview of the cellular diversity and dynamics across various biological systems. In addition, the incorporation of machine learning techniques into cell atlas analyses opens up exciting prospects for the field of integrative biology.
Collapse
Affiliation(s)
- Fang Ye
- Bone Marrow Transplantation Center of the First Affiliated Hospital, and Center for Stem Cell and Regenerative MedicineZhejiang University School of MedicineHangzhouZhejiang310000China
- Liangzhu LaboratoryZhejiang UniversityHangzhouZhejiang311121China
| | - Jingjing Wang
- Bone Marrow Transplantation Center of the First Affiliated Hospital, and Center for Stem Cell and Regenerative MedicineZhejiang University School of MedicineHangzhouZhejiang310000China
- Liangzhu LaboratoryZhejiang UniversityHangzhouZhejiang311121China
| | - Jiaqi Li
- Bone Marrow Transplantation Center of the First Affiliated Hospital, and Center for Stem Cell and Regenerative MedicineZhejiang University School of MedicineHangzhouZhejiang310000China
| | - Yuqing Mei
- Bone Marrow Transplantation Center of the First Affiliated Hospital, and Center for Stem Cell and Regenerative MedicineZhejiang University School of MedicineHangzhouZhejiang310000China
| | - Guoji Guo
- Bone Marrow Transplantation Center of the First Affiliated Hospital, and Center for Stem Cell and Regenerative MedicineZhejiang University School of MedicineHangzhouZhejiang310000China
- Liangzhu LaboratoryZhejiang UniversityHangzhouZhejiang311121China
- Zhejiang Provincial Key Lab for Tissue Engineering and Regenerative MedicineDr. Li Dak Sum & Yip Yio Chin Center for Stem Cell and Regenerative MedicineHangzhouZhejiang310058China
- Institute of HematologyZhejiang UniversityHangzhouZhejiang310000China
| |
Collapse
|
26
|
Deng Y, Lu Y, Li M, Shen J, Qin S, Zhang W, Zhang Q, Shen Z, Li C, Jia T, Chen P, Peng L, Chen Y, Zhang W, Liu H, Zhang L, Rong L, Wang X, Chen D. SCAN: Spatiotemporal Cloud Atlas for Neural cells. Nucleic Acids Res 2024; 52:D998-D1009. [PMID: 37930842 PMCID: PMC10767991 DOI: 10.1093/nar/gkad895] [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: 08/14/2023] [Revised: 09/20/2023] [Accepted: 10/05/2023] [Indexed: 11/08/2023] Open
Abstract
The nervous system is one of the most complicated and enigmatic systems within the animal kingdom. Recently, the emergence and development of spatial transcriptomics (ST) and single-cell RNA sequencing (scRNA-seq) technologies have provided an unprecedented ability to systematically decipher the cellular heterogeneity and spatial locations of the nervous system from multiple unbiased aspects. However, efficiently integrating, presenting and analyzing massive multiomic data remains a huge challenge. Here, we manually collected and comprehensively analyzed high-quality scRNA-seq and ST data from the nervous system, covering 10 679 684 cells. In addition, multi-omic datasets from more than 900 species were included for extensive data mining from an evolutionary perspective. Furthermore, over 100 neurological diseases (e.g. Alzheimer's disease, Parkinson's disease, Down syndrome) were systematically analyzed for high-throughput screening of putative biomarkers. Differential expression patterns across developmental time points, cell types and ST spots were discerned and subsequently subjected to extensive interpretation. To provide researchers with efficient data exploration, we created a new database with interactive interfaces and integrated functions called the Spatiotemporal Cloud Atlas for Neural cells (SCAN), freely accessible at http://47.98.139.124:8799 or http://scanatlas.net. SCAN will benefit the neuroscience research community to better exploit the spatiotemporal atlas of the neural system and promote the development of diagnostic strategies for various neurological disorders.
Collapse
Affiliation(s)
- Yushan Deng
- State Key Laboratory of Common Mechanism Research for Major Diseases, Suzhou Institute of Systems Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Suzhou 215123, China
| | - Yubao Lu
- Department of Spine Surgery, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou 510630, China
| | - Mengrou Li
- State Key Laboratory of Common Mechanism Research for Major Diseases, Suzhou Institute of Systems Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Suzhou 215123, China
- Institutes of Biology and Medical Sciences (IBMS), Soochow University, Suzhou 215123, China
| | - Jiayi Shen
- State Key Laboratory of Common Mechanism Research for Major Diseases, Suzhou Institute of Systems Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Suzhou 215123, China
- Peninsula Cancer Research Center, School of Basic Medical Sciences, Binzhou Medical University, Yantai 264003, China
| | - Siying Qin
- State Key Laboratory of Common Mechanism Research for Major Diseases, Suzhou Institute of Systems Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Suzhou 215123, China
| | - Wei Zhang
- Department of Spine Surgery, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou 510630, China
| | - Qiang Zhang
- State Key Laboratory of Common Mechanism Research for Major Diseases, Suzhou Institute of Systems Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Suzhou 215123, China
| | - Zhaoyang Shen
- Life Sciences and Technology College, China Pharmaceutical University, Nanjing 211198, China
| | - Changxiao Li
- State Key Laboratory of Common Mechanism Research for Major Diseases, Suzhou Institute of Systems Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Suzhou 215123, China
| | - Tengfei Jia
- State Key Laboratory of Common Mechanism Research for Major Diseases, Suzhou Institute of Systems Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Suzhou 215123, China
- Institutes of Biology and Medical Sciences (IBMS), Soochow University, Suzhou 215123, China
| | - Peixin Chen
- State Key Laboratory of Common Mechanism Research for Major Diseases, Suzhou Institute of Systems Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Suzhou 215123, China
- Cam-Su Genomic Resource Center, Medical College of Soochow University, Suzhou 215123, China
| | - Lingmin Peng
- State Key Laboratory of Common Mechanism Research for Major Diseases, Suzhou Institute of Systems Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Suzhou 215123, China
| | - Yangfeng Chen
- State Key Laboratory of Common Mechanism Research for Major Diseases, Suzhou Institute of Systems Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Suzhou 215123, China
| | - Wensheng Zhang
- Peninsula Cancer Research Center, School of Basic Medical Sciences, Binzhou Medical University, Yantai 264003, China
- Cam-Su Genomic Resource Center, Medical College of Soochow University, Suzhou 215123, China
| | - Hebin Liu
- Institutes of Biology and Medical Sciences (IBMS), Soochow University, Suzhou 215123, China
| | - Liangming Zhang
- Department of Spine Surgery, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou 510630, China
| | - Limin Rong
- Department of Spine Surgery, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou 510630, China
| | - Xiangdong Wang
- Zhongshan Hospital, Department of Pulmonary and Critical Care Medicine, Institute for Clinical Science, Shanghai Institute of Clinical Bioinformatics, Shanghai 200000, China
| | - Dongsheng Chen
- State Key Laboratory of Common Mechanism Research for Major Diseases, Suzhou Institute of Systems Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Suzhou 215123, China
| |
Collapse
|
27
|
Richard S, Ren J, Flamant F. Thyroid hormone action during GABAergic neuron maturation: The quest for mechanisms. Front Endocrinol (Lausanne) 2023; 14:1256877. [PMID: 37854197 PMCID: PMC10579935 DOI: 10.3389/fendo.2023.1256877] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/11/2023] [Accepted: 09/18/2023] [Indexed: 10/20/2023] Open
Abstract
Thyroid hormone (TH) signaling plays a major role in mammalian brain development. Data obtained in the past years in animal models have pinpointed GABAergic neurons as a major target of TH signaling during development, which opens up new perspectives to further investigate the mechanisms by which TH affects brain development. The aim of the present review is to gather the available information about the involvement of TH in the maturation of GABAergic neurons. After giving an overview of the kinds of neurological disorders that may arise from disruption of TH signaling during brain development in humans, we will take a historical perspective to show how rodent models of hypothyroidism have gradually pointed to GABAergic neurons as a main target of TH signaling during brain development. The third part of this review underscores the challenges that are encountered when conducting gene expression studies to investigate the molecular mechanisms that are at play downstream of TH receptors during brain development. Unravelling the mechanisms of action of TH in the developing brain should help make progress in the prevention and treatment of several neurological disorders, including autism and epilepsy.
Collapse
Affiliation(s)
| | | | - Frédéric Flamant
- Institut de Génomique Fonctionnelle de Lyon, UMR5242, Ecole Normale Supérieure de Lyon, Centre National de la Recherche Scientifique, Université Claude Bernard-Lyon 1, USC1370 Institut National de Recherche pour l’Agriculture, l’Alimentation et l’Environnement, Lyon, France
| |
Collapse
|