1
|
Chen D, Zhuang Z, Huang M, Huang Y, Yan Y, Zhang Y, Lin Y, Jin X, Wang Y, Huang J, Xu W, Pan J, Wang H, Huang F, Liao K, Cheng M, Zhu Z, Bai Y, Niu Z, Zhang Z, Xiang Y, Wei X, Yang T, Zeng T, Dong Y, Lei Y, Sun Y, Wang J, Yang H, Sun Y, Cao G, Poo M, Liu L, Naumann RK, Xu C, Wang Z, Xu X, Liu S. Genomic evolution reshapes cell-type diversification in the amniote brain. Dev Cell 2025:S1534-5807(25)00252-7. [PMID: 40367951 DOI: 10.1016/j.devcel.2025.04.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2024] [Revised: 03/05/2025] [Accepted: 04/18/2025] [Indexed: 05/16/2025]
Abstract
Over 320 million years of evolution, amniotes have developed complex brains and cognition through largely unexplored genetic and gene expression mechanisms. We created a comprehensive single-cell atlas of over 1.3 million cells from the telencephalon and cerebellum of turtles, zebra finches, pigeons, mice, and macaques, employing single-cell resolution spatial transcriptomics to validate gene expression patterns across species. Our study identifies significant species-specific variations in cell types, highlighting their conservation and diversification in evolution. We found pronounced differences in telencephalon excitatory neurons (EXs) and cerebellar cell types between birds and mammals. Birds predominantly express SLC17A6 in EX, whereas mammals express SLC17A7 in the neocortex and SLC17A6 elsewhere, possibly due to loss of function of SLC17A7 in birds. Additionally, we identified a bird-specific Purkinje cell subtype (SVIL+), implicating the lysine-specific demethylase 11 (LSD1)/KDM1A pathway in learning and circadian rhythms and containing numerous positively selected genes, which suggests an evolutionary optimization of cerebellar functions for ecological and behavioral adaptation. Our findings elucidate the complex interplay between genetic evolution and environmental adaptation, underscoring the role of genetic diversification in the development of specialized cell types across amniotes.
Collapse
Affiliation(s)
- Duoyuan Chen
- BGI Research, Hangzhou 310030, China; BGI Research, Shenzhen 518083, China; Key Laboratory of Spatial Omics of Zhejiang Province, BGI Research, Hangzhou 310030, China; State Key Laboratory of Genome and Multi-omics Technologies, BGI Research, Hangzhou 310030, China
| | - Zhenkun Zhuang
- BGI Research, Hangzhou 310030, China; School of Biology and Biological Engineering, South China University of Technology, Guangzhou 510006, China; BGI Research, Shenzhen 518083, China; Key Laboratory of Spatial Omics of Zhejiang Province, BGI Research, Hangzhou 310030, China; State Key Laboratory of Genome and Multi-omics Technologies, BGI Research, Hangzhou 310030, China
| | - Maolin Huang
- School of Life Sciences, Zhengzhou University, Zhengzhou 450001, China
| | | | - Yuting Yan
- BGI Research, Hangzhou 310030, China; School of Biology and Biological Engineering, South China University of Technology, Guangzhou 510006, China
| | - Yanru Zhang
- BGI Research, Hangzhou 310030, China; School of Biology and Biological Engineering, South China University of Technology, Guangzhou 510006, China
| | - Youning Lin
- BGI Research, Hangzhou 310030, China; BGI Research, Shenzhen 518083, China; Key Laboratory of Spatial Omics of Zhejiang Province, BGI Research, Hangzhou 310030, China
| | - Xiaoying Jin
- BGI Research, Hangzhou 310030, China; College of Life Sciences, University of Chinese Academy of Sciences, Beijing 100049, China; Key Laboratory of Spatial Omics of Zhejiang Province, BGI Research, Hangzhou 310030, China
| | - Yuanmei Wang
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing 100049, China; BGI Research, Shenzhen 518083, China; HIM-BGI Omics Center, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou 310018, China
| | - Jinfeng Huang
- The Institute of Biomedical and Health Engineering, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen-Hong Kong Institute of Brain Science-Shenzhen Fundamental Research Institutions, Shenzhen, 1068 Xueyuan Avenue, Shenzhen University Town, Nanshan District, Shenzhen 518055, China
| | - Wenbo Xu
- School of Life Sciences, Zhengzhou University, Zhengzhou 450001, China
| | | | - Hong Wang
- The Institute of Biomedical and Health Engineering, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen-Hong Kong Institute of Brain Science-Shenzhen Fundamental Research Institutions, Shenzhen, 1068 Xueyuan Avenue, Shenzhen University Town, Nanshan District, Shenzhen 518055, China
| | - Fubaoqian Huang
- BGI Research, Hangzhou 310030, China; School of Biology and Biological Engineering, South China University of Technology, Guangzhou 510006, China; Key Laboratory of Spatial Omics of Zhejiang Province, BGI Research, Hangzhou 310030, China
| | - Kuo Liao
- BGI Research, Hangzhou 310030, China; School of Biology and Biological Engineering, South China University of Technology, Guangzhou 510006, China
| | - Mengnan Cheng
- BGI Research, Hangzhou 310030, China; Key Laboratory of Spatial Omics of Zhejiang Province, BGI Research, Hangzhou 310030, China
| | - Zhiyong Zhu
- BGI Research, Hangzhou 310030, China; College of Life Sciences, University of Chinese Academy of Sciences, Beijing 100049, China; Key Laboratory of Spatial Omics of Zhejiang Province, BGI Research, Hangzhou 310030, China
| | - Yinqi Bai
- BGI Research, Hangzhou 310030, China
| | - Zhiwei Niu
- BGI Research, Hangzhou 310030, China; College of Life Sciences, University of Chinese Academy of Sciences, Beijing 100049, China; Key Laboratory of Spatial Omics of Zhejiang Province, BGI Research, Hangzhou 310030, China
| | - Ze Zhang
- BGI Research, Hangzhou 310030, China; Key Laboratory of Systems Health Science of Zhejiang Province, School of Life Science, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou 310024, China; Key Laboratory of Spatial Omics of Zhejiang Province, BGI Research, Hangzhou 310030, China
| | - Ya Xiang
- BGI Research, Hangzhou 310030, China; Key Laboratory of Spatial Omics of Zhejiang Province, BGI Research, Hangzhou 310030, China; College of Life Sciences, Northwest University, Xi'an 710069, China
| | - Xiaofeng Wei
- China National GeneBank, BGI Research, Shenzhen 518120, China; Guangdong Genomics Data Center, BGl research, Shenzhen 518120, China
| | - Tao Yang
- China National GeneBank, BGI Research, Shenzhen 518120, China; Guangdong Genomics Data Center, BGl research, Shenzhen 518120, China
| | - Tao Zeng
- BGI Research, Hangzhou 310030, China; BGI Research, Shenzhen 518083, China
| | - Yuliang Dong
- BGI Research, Hangzhou 310030, China; BGI Research, Shenzhen 518083, China
| | - Ying Lei
- BGI Research, Hangzhou 310030, China; Key Laboratory of Spatial Omics of Zhejiang Province, BGI Research, Hangzhou 310030, China; Shanxi Medical University, BGI Collaborative Center for Future Medicine, Shanxi Medical University, Taiyuan 030001, China
| | - Yangang Sun
- Institute of Neuroscience, Key Laboratory of Brain Cognition and Brain-inspired Intelligence Technology, CAS Center for Excellence in Brain Science and Intelligence Technology, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Jian Wang
- BGI Research, Hangzhou 310030, China
| | - Huanming Yang
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing 100049, China; BGI Research, Shenzhen 518083, China; HIM-BGI Omics Center, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou 310018, China; James D. Watson Institute of Genome Sciences, Hangzhou 310029, China
| | - Yidi Sun
- Institute of Neuroscience, Key Laboratory of Brain Cognition and Brain-inspired Intelligence Technology, CAS Center for Excellence in Brain Science and Intelligence Technology, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Gang Cao
- Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
| | - Muming Poo
- Institute of Neuroscience, Key Laboratory of Brain Cognition and Brain-inspired Intelligence Technology, CAS Center for Excellence in Brain Science and Intelligence Technology, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Longqi Liu
- BGI Research, Hangzhou 310030, China; Key Laboratory of Spatial Omics of Zhejiang Province, BGI Research, Hangzhou 310030, China; Shanxi Medical University, BGI Collaborative Center for Future Medicine, Shanxi Medical University, Taiyuan 030001, China
| | - Robert K Naumann
- The Institute of Biomedical and Health Engineering, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen-Hong Kong Institute of Brain Science-Shenzhen Fundamental Research Institutions, Shenzhen, 1068 Xueyuan Avenue, Shenzhen University Town, Nanshan District, Shenzhen 518055, China.
| | - Chun Xu
- Institute of Neuroscience, Key Laboratory of Brain Cognition and Brain-inspired Intelligence Technology, CAS Center for Excellence in Brain Science and Intelligence Technology, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China.
| | - Zhenlong Wang
- School of Life Sciences, Zhengzhou University, Zhengzhou 450001, China.
| | - Xun Xu
- 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.
| | - Shiping Liu
- BGI Research, Hangzhou 310030, China; BGI Research, Shenzhen 518083, China; Key Laboratory of Spatial Omics of Zhejiang Province, BGI Research, Hangzhou 310030, China; State Key Laboratory of Genome and Multi-omics Technologies, BGI Research, Hangzhou 310030, China.
| |
Collapse
|
2
|
Mo F, Qian Q, Lu X, Zheng D, Cai W, Yao J, Chen H, Huang Y, Zhang X, Wu S, Shen Y, Bai Y, Wang Y, Jiang W, Fan L. mKmer: an unbiased K-mer embedding of microbiomic single-microbe RNA sequencing data. Brief Bioinform 2025; 26:bbaf227. [PMID: 40407385 PMCID: PMC12100620 DOI: 10.1093/bib/bbaf227] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2025] [Revised: 04/13/2025] [Accepted: 05/01/2025] [Indexed: 05/26/2025] Open
Abstract
The advanced single-microbe RNA sequencing (smRNA-seq) technique addresses the pressing need to understand the complexity and diversity of microbial communities, as well as the distinct microbial states defined by different gene expression profiles. Current analyses of smRNA-seq data heavily rely on the integrity of reference genomes within the queried microbiota. However, establishing a comprehensive collection of microbial reference genomes or gene sets remains a significant challenge for most real-world microbial ecosystems. Here, we developed an unbiased embedding algorithm utilizing K-mer signatures, named mKmer, which bypasses gene or genome alignment to enable species identification for individual microbes and downstream functional enrichment analysis. By substituting gene features in the canonical cell-by-gene matrix with highly conserved K-mers, we demonstrate that mKmer outperforms gene-based methods in clustering and motif inference tasks using benchmark datasets from crop soil and human gut microbiomes. Our method provides a reference genome-free analytical framework for advancing smRNA-seq studies.
Collapse
Affiliation(s)
- Fangyu Mo
- Hainan Institute, Zhejiang University, Zhenzhou Road, Yazhou Bay Science and Technology City, Yazhou District, Sanya 572025, Hainan Province, China
- Institute of Crop Science, Zhejiang University, 866 Yuhangtang Road, Xihu District, Hangzhou 310058, Zhejiang Province, China
| | - Qinghong Qian
- Institute of Crop Science, Zhejiang University, 866 Yuhangtang Road, Xihu District, Hangzhou 310058, Zhejiang Province, China
| | - Xiaolin Lu
- Institute of Bioinformatics and James D. Watson Institute of Genome Sciences, Zhejiang University, 866 Yuhangtang Road, Xihu District, Hangzhou 310058, Zhejiang Province, China
| | - Dihuai Zheng
- Institute of Crop Science, Zhejiang University, 866 Yuhangtang Road, Xihu District, Hangzhou 310058, Zhejiang Province, China
| | - Wenjie Cai
- Liangzhu Laboratory (Zhejiang Provincial Laboratory for Systems Medicine and Precision Diagnosis), Zhejiang University, 1369 Wenyi West Road, Yuhang District, Hangzhou 311121, Zhejiang Province, China
| | - Jie Yao
- Institute of Bioinformatics and James D. Watson Institute of Genome Sciences, Zhejiang University, 866 Yuhangtang Road, Xihu District, Hangzhou 310058, Zhejiang Province, China
| | - Hongyu Chen
- Institute of Crop Science, Zhejiang University, 866 Yuhangtang Road, Xihu District, Hangzhou 310058, Zhejiang Province, China
| | - Yujie Huang
- Institute of Crop Science, Zhejiang University, 866 Yuhangtang Road, Xihu District, Hangzhou 310058, Zhejiang Province, China
| | - Xiang Zhang
- Department of Colorectal Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, 79 Qingchun Road, Shangcheng District, Hangzhou 310003, Zhejiang Province, China
| | - Sanling Wu
- Analysis Center of Agrobiology and Environmental Sciences, Zhejiang University, 866 Yuhangtang Road, Xihu District, Hangzhou 310058, Zhejiang Province, China
| | - Yifei Shen
- Department of Laboratory Medicine, The First Affiliated Hospital, Zhejiang University School of Medicine, 79 Qingchun Road, Shangcheng District, Hangzhou 310003, Zhejiang Province, China
| | - Yinqi Bai
- BGI-Sanya, Zhenzhou Road, Yazhou Bay Science and Technology City, Yazhou District, Sanya 572025, Hainan Province, China
| | - Yongcheng Wang
- Liangzhu Laboratory, Zhejiang University, 1369 Wenyi West Road, Yuhang District, Hangzhou 311113, Zhejiang Province, China
| | - Weiqin Jiang
- Department of Colorectal Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, 79 Qingchun Road, Shangcheng District, Hangzhou 310003, Zhejiang Province, China
| | - Longjiang Fan
- Hainan Institute, Zhejiang University, Zhenzhou Road, Yazhou Bay Science and Technology City, Yazhou District, Sanya 572025, Hainan Province, China
- Institute of Crop Science, Zhejiang University, 866 Yuhangtang Road, Xihu District, Hangzhou 310058, Zhejiang Province, China
- Institute of Bioinformatics and James D. Watson Institute of Genome Sciences, Zhejiang University, 866 Yuhangtang Road, Xihu District, Hangzhou 310058, Zhejiang Province, China
| |
Collapse
|
3
|
Guo B, Ling W, Kwon SH, Panwar P, Ghazanfar S, Martinowich K, Hicks SC. Integrating Spatially-Resolved Transcriptomics Data Across Tissues and Individuals: Challenges and Opportunities. SMALL METHODS 2025; 9:e2401194. [PMID: 39935130 PMCID: PMC12103234 DOI: 10.1002/smtd.202401194] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/01/2024] [Revised: 12/13/2024] [Indexed: 02/13/2025]
Abstract
Advances in spatially-resolved transcriptomics (SRT) technologies have propelled the development of new computational analysis methods to unlock biological insights. The lowering cost of SRT data generation presents an unprecedented opportunity to create large-scale spatial atlases and enable population-level investigation, integrating SRT data across multiple tissues, individuals, species, or phenotypes. Here, unique challenges are described in the SRT data integration, where the analytic impact of varying spatial and biological resolutions is characterized and explored. A succinct review of spatially-aware integration methods and computational strategies is provided. Exciting opportunities to advance computational algorithms amenable to atlas-scale datasets along with standardized preprocessing methods, leading to improved sensitivity and reproducibility in the future are further highlighted.
Collapse
Affiliation(s)
- Boyi Guo
- Department of BiostatisticsJohns Hopkins Bloomberg School of Public HealthBaltimoreMD21205USA
| | - Wodan Ling
- Division of BiostatisticsDepartment of Population Health SciencesWeill Cornell MedicineNew YorkNY10065USA
| | - Sang Ho Kwon
- Lieber Institute for Brain DevelopmentJohns Hopkins Medical CampusBaltimoreMD21205USA
- Solomon H. Snyder Department of NeuroscienceJohns Hopkins School of MedicineBaltimoreMD21205USA
- Biochemistry, Cellular, and Molecular Biology Graduate ProgramJohns Hopkins School of MedicineBaltimoreMD21205USA
| | - Pratibha Panwar
- School of Mathematics and StatisticsThe University of SydneyCamperdownNSW2006Australia
- Sydney Precision Data Science CentreUniversity of SydneyCamperdownNSW2006Australia
- Charles Perkins CentreThe University of SydneyCamperdownNSW2006Australia
| | - Shila Ghazanfar
- School of Mathematics and StatisticsThe University of SydneyCamperdownNSW2006Australia
- Sydney Precision Data Science CentreUniversity of SydneyCamperdownNSW2006Australia
- Charles Perkins CentreThe University of SydneyCamperdownNSW2006Australia
| | - Keri Martinowich
- Lieber Institute for Brain DevelopmentJohns Hopkins Medical CampusBaltimoreMD21205USA
- Solomon H. Snyder Department of NeuroscienceJohns Hopkins School of MedicineBaltimoreMD21205USA
- Department of Psychiatry and Behavioral SciencesJohns Hopkins School of MedicineBaltimoreMDUSA
- Johns Hopkins Kavli Neuroscience Discovery InstituteJohns Hopkins UniversityBaltimoreMD21218USA
- Department of Biomedical EngineeringJohns Hopkins UniversityBaltimoreMD21218USA
| | - Stephanie C. Hicks
- Department of BiostatisticsJohns Hopkins Bloomberg School of Public HealthBaltimoreMD21205USA
- Center for Computational BiologyJohns Hopkins UniversityBaltimoreMD21218USA
- Malone Center for Engineering in HealthcareJohns Hopkins UniversityBaltimoreMD21218USA
| |
Collapse
|
4
|
Wang J, Ye F, Chai H, Jiang Y, Wang T, Ran X, Xia Q, Xu Z, Fu Y, Zhang G, Wu H, Guo G, Guo H, Ruan Y, Wang Y, Xing D, Xu X, Zhang Z. Advances and applications in single-cell and spatial genomics. SCIENCE CHINA. LIFE SCIENCES 2025; 68:1226-1282. [PMID: 39792333 DOI: 10.1007/s11427-024-2770-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/20/2024] [Accepted: 10/10/2024] [Indexed: 01/12/2025]
Abstract
The applications of single-cell and spatial technologies in recent times have revolutionized the present understanding of cellular states and the cellular heterogeneity inherent in complex biological systems. These advancements offer unprecedented resolution in the examination of the functional genomics of individual cells and their spatial context within tissues. In this review, we have comprehensively discussed the historical development and recent progress in the field of single-cell and spatial genomics. We have reviewed the breakthroughs in single-cell multi-omics technologies, spatial genomics methods, and the computational strategies employed toward the analyses of single-cell atlas data. Furthermore, we have highlighted the advances made in constructing cellular atlases and their clinical applications, particularly in the context of disease. Finally, we have discussed the emerging trends, challenges, and opportunities in this rapidly evolving field.
Collapse
Affiliation(s)
- Jingjing Wang
- Bone Marrow Transplantation Center of the First Affiliated Hospital & Liangzhu Laboratory, Zhejiang University School of Medicine, Hangzhou, 310058, China
| | - Fang Ye
- Bone Marrow Transplantation Center of the First Affiliated Hospital & Liangzhu Laboratory, Zhejiang University School of Medicine, Hangzhou, 310058, China
| | - Haoxi Chai
- Life Sciences Institute and The Second Affiliated Hospital, Zhejiang University, Hangzhou, 310058, China
| | - Yujia Jiang
- BGI Research, Shenzhen, 518083, China
- BGI Research, Hangzhou, 310030, China
| | - Teng Wang
- Biomedical Pioneering Innovation Center (BIOPIC) and School of Life Sciences, Peking University, Beijing, 100871, China
- Academy for Advanced Interdisciplinary Studies, Peking University, Beijing, 100871, China
| | - Xia Ran
- Bone Marrow Transplantation Center of the First Affiliated Hospital & Liangzhu Laboratory, Zhejiang University School of Medicine, Hangzhou, 310058, China
- Institute of Hematology, Zhejiang University, Hangzhou, 310000, China
| | - Qimin Xia
- Biomedical Pioneering Innovation Center (BIOPIC) and School of Life Sciences, Peking University, Beijing, 100871, China
| | - Ziye Xu
- Department of Laboratory Medicine of The First Affiliated Hospital & Liangzhu Laboratory, Zhejiang University School of Medicine, Hangzhou, 310058, China
| | - Yuting Fu
- Bone Marrow Transplantation Center of the First Affiliated Hospital & Liangzhu Laboratory, Zhejiang University School of Medicine, Hangzhou, 310058, China
- Center for Stem Cell and Regenerative Medicine, Zhejiang University School of Medicine, Hangzhou, 310058, China
| | - Guodong Zhang
- Bone Marrow Transplantation Center of the First Affiliated Hospital & Liangzhu Laboratory, Zhejiang University School of Medicine, Hangzhou, 310058, China
- Center for Stem Cell and Regenerative Medicine, Zhejiang University School of Medicine, Hangzhou, 310058, China
| | - Hanyu Wu
- Bone Marrow Transplantation Center of the First Affiliated Hospital & Liangzhu Laboratory, Zhejiang University School of Medicine, Hangzhou, 310058, China
- Center for Stem Cell and Regenerative Medicine, Zhejiang University School of Medicine, Hangzhou, 310058, China
| | - Guoji Guo
- Bone Marrow Transplantation Center of the First Affiliated Hospital & Liangzhu Laboratory, Zhejiang University School of Medicine, Hangzhou, 310058, China.
- Center for Stem Cell and Regenerative Medicine, Zhejiang University School of Medicine, Hangzhou, 310058, China.
- Zhejiang Provincial Key Lab for Tissue Engineering and Regenerative Medicine, Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cell and Regenerative Medicine, Hangzhou, 310058, China.
- Institute of Hematology, Zhejiang University, Hangzhou, 310000, China.
| | - Hongshan Guo
- Bone Marrow Transplantation Center of the First Affiliated Hospital & Liangzhu Laboratory, Zhejiang University School of Medicine, Hangzhou, 310058, China.
- Institute of Hematology, Zhejiang University, Hangzhou, 310000, China.
| | - Yijun Ruan
- Life Sciences Institute and The Second Affiliated Hospital, Zhejiang University, Hangzhou, 310058, China.
| | - Yongcheng Wang
- Department of Laboratory Medicine of The First Affiliated Hospital & Liangzhu Laboratory, Zhejiang University School of Medicine, Hangzhou, 310058, China.
| | - Dong Xing
- Biomedical Pioneering Innovation Center (BIOPIC) and School of Life Sciences, Peking University, Beijing, 100871, China.
- Beijing Advanced Innovation Center for Genomics (ICG), Peking University, Beijing, 100871, China.
| | - Xun Xu
- BGI Research, Shenzhen, 518083, China.
- BGI Research, Hangzhou, 310030, China.
- Guangdong Provincial Key Laboratory of Genome Read and Write, BGI Research, Shenzhen, 518083, China.
| | - Zemin Zhang
- Biomedical Pioneering Innovation Center (BIOPIC) and School of Life Sciences, Peking University, Beijing, 100871, China.
| |
Collapse
|
5
|
Binder N, Khavaran A, Sankowski R. Primer on machine learning applications in brain immunology. FRONTIERS IN BIOINFORMATICS 2025; 5:1554010. [PMID: 40313869 PMCID: PMC12043695 DOI: 10.3389/fbinf.2025.1554010] [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: 01/07/2025] [Accepted: 03/24/2025] [Indexed: 05/03/2025] Open
Abstract
Single-cell and spatial technologies have transformed our understanding of brain immunology, providing unprecedented insights into immune cell heterogeneity and spatial organisation within the central nervous system. These methods have uncovered complex cellular interactions, rare cell populations, and the dynamic immune landscape in neurological disorders. This review highlights recent advances in single-cell "omics" data analysis and discusses their applicability for brain immunology. Traditional statistical techniques, adapted for single-cell omics, have been crucial in categorizing cell types and identifying gene signatures, overcoming challenges posed by increasingly complex datasets. We explore how machine learning, particularly deep learning methods like autoencoders and graph neural networks, is addressing these challenges by enhancing dimensionality reduction, data integration, and feature extraction. Newly developed foundation models present exciting opportunities for uncovering gene expression programs and predicting genetic perturbations. Focusing on brain development, we demonstrate how single-cell analyses have resolved immune cell heterogeneity, identified temporal maturation trajectories, and uncovered potential therapeutic links to various pathologies, including brain malignancies and neurodegeneration. The integration of single-cell and spatial omics has elucidated the intricate cellular interplay within the developing brain. This mini-review is intended for wet lab biologists at all career stages, offering a concise overview of the evolving landscape of single-cell omics in the age of widely available artificial intelligence.
Collapse
Affiliation(s)
| | | | - Roman Sankowski
- Institute of Neuropathology, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| |
Collapse
|
6
|
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
|
7
|
He C, Filippidis P, Kleinstein SH, Guan L. Partially characterized topology guides reliable anchor-free scRNA-integration. Commun Biol 2025; 8:561. [PMID: 40185996 PMCID: PMC11971424 DOI: 10.1038/s42003-025-07988-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2024] [Accepted: 03/21/2025] [Indexed: 04/07/2025] Open
Abstract
Single-cell RNA sequencing (scRNA-seq) is an important technique for obtaining biological insights at cellular resolution, with scRNA-seq batch integration a key step before downstream statistical analysis. Despite the plethora of methods proposed, achieving reliable batch correction while preserving the heterogeneity of biological signals that define cell type continues to pose a challenge. To address this, we propose scCRAFT, an autoencoder model that separates cell-type-related signals from batch effects for reliable multi-batch integration. scCRAFT integrates three key loss components: a reconstruction loss for observation reconstruction, a multi-domain adaptation loss to eliminate batch effects, and an innovative dual-resolution triplet loss to preserve intra-batch, introduced as an effective mechanism to counteract the over-correction effect of domain adaptation loss amid heterogeneous cell distributions across batches. We show that scCRAFT effectively manages unbalanced batches, rare cell types, and batch-specific cell phenotypes in simulations, and surpasses state-of-the-art methods in a diverse set of real datasets.
Collapse
Affiliation(s)
- Chuan He
- Department of Biostatistics, Yale School of Public Health, New Haven, CT, US
| | | | - Steven H Kleinstein
- Department of Pathology, Yale School of Medicine, New Haven, CT, US
- Department of Immunobiology, Yale School of Medicine, New Haven, CT, US
- Program in Computational Biology and Biomedical Informatics, Yale University, New Haven, CT, US
| | - Leying Guan
- Department of Biostatistics, Yale School of Public Health, New Haven, CT, US.
- Program in Computational Biology and Biomedical Informatics, Yale University, New Haven, CT, US.
| |
Collapse
|
8
|
Wei S, Lu Y, Wang P, Li Q, Shuai J, Zhao Q, Lin H, Peng Y. Investigation of cell development and tissue structure network based on natural Language processing of scRNA-seq data. J Transl Med 2025; 23:264. [PMID: 40038714 PMCID: PMC11877821 DOI: 10.1186/s12967-025-06263-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2024] [Accepted: 02/14/2025] [Indexed: 03/06/2025] Open
Abstract
BACKGROUND Single-cell multi-omics technologies, particularly single-cell RNA sequencing (scRNA-seq), have revolutionized our understanding of cellular heterogeneity and development by providing insights into gene expression at the single-cell level. Investigating the influence of genes on cellular behavior is crucial for elucidating cell fate determination and differentiation, cell development processes, and disease mechanisms. METHODS Inspired by NLP, we present a novel scRNA-seq analysis method that treats genes as analogous to words. Using word2vec to embed gene sequences derived from gene networks, we generate vector representations of genes, which are then used to represent cells by summing gene vectors and subsequently tissues by aggregating cell vectors. RESULTS Our NLP-based approach analyzes scRNA-seq data by generating vector representations of genes, cells, and tissues. This multi-scale analysis includes mapping cell states in vector space to reveal developmental trajectories, quantifying cell similarity using Euclidean distance, and constructing inter-tissue relationship networks from aggregated cell vectors. CONCLUSIONS This method offers a computationally efficient approach for analyzing scRNA-seq data by constructing embedding representations similar to those used in large language model pre-training, but without requiring high-performance computing clusters. By generating gene embeddings that capture functional relationships, this method facilitates the study of cell development trajectories, the impact of gene perturbations, cell clustering, and the construction and analysis of tissue networks. This provides a valuable tool for single-cell data analysis.
Collapse
Affiliation(s)
- Suwen Wei
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, 325001, Zhejiang, P. R. China
| | - Yuer Lu
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, 325001, Zhejiang, P. R. China
| | - Peng Wang
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, 325001, Zhejiang, P. R. China
- Postgraduate Training Base Alliance of Wenzhou Medical University, Wenzhou, 325001, Zhejiang, P. R. China
| | - Qichao Li
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, 325001, Zhejiang, P. R. China
- Postgraduate Training Base Alliance of Wenzhou Medical University, Wenzhou, 325001, Zhejiang, P. R. China
| | - Jianwei Shuai
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, 325001, Zhejiang, P. R. China
| | - Qi Zhao
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, 325001, Zhejiang, P. R. China.
- School of Computer Science and Software Engineering, University of Science and Technology Liaoning, Anshan, 114051, P.R. China.
| | - Hai Lin
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, 325001, Zhejiang, P. R. China.
| | - Yuming Peng
- Department of General Practice, Central Hospital of Karamay, Xinjiang, 834000, P. R. China.
| |
Collapse
|
9
|
Guo Y, Ma F, Li P, Guo L, Liu Z, Huo C, Shi C, Zhu L, Gu M, Na R, Zhang W. Comprehensive SHAP Values and Single-Cell Sequencing Technology Reveal Key Cell Clusters in Bovine Skeletal Muscle. Int J Mol Sci 2025; 26:2054. [PMID: 40076676 PMCID: PMC11900076 DOI: 10.3390/ijms26052054] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2025] [Revised: 02/24/2025] [Accepted: 02/25/2025] [Indexed: 03/14/2025] Open
Abstract
The skeletal muscle of cattle is the main component of their muscular system, responsible for supporting and movement functions. However, there are still many unknown areas regarding the ranking of the importance of different types of cell populations within it. This study conducted in-depth research and made a series of significant findings. First, we trained 15 bovine skeletal muscle models and selected the best-performing model as the initial model. Based on the SHAP (Shapley Additive exPlanations) analysis of this initial model, we obtained the SHAP values of 476 important genes. Using the contributions of these 476 genes, we reconstructed a 476-gene SHAP value matrix, and relying solely on the interactions among these 476 genes, successfully mapped the single-cell atlas of bovine skeletal muscle. After retraining the model and further interpretation, we found that Myofiber cells are the most representative cell type in bovine skeletal muscle, followed by neutrophils. By determining the key genes of each cell type through SHAP values, we conducted analyses on the correlations among key genes and between cells for Myofiber cells, revealing the critical role these genes play in muscle growth and development. Further, by using protein language models, we performed cross-species comparisons between cattle and pigs, deepening our understanding of Myofiber cells as key cells in skeletal muscle, and exploring the common regulatory mechanisms of muscle development across species.
Collapse
Affiliation(s)
- Yaqiang Guo
- College of Animal Science, Inner Mongolia Agricultural University, Hohhot 010010, China; (Y.G.); (F.M.); (L.G.); (C.H.); (C.S.); (L.Z.); (M.G.)
- Inner Mongolia Engineering Research Center of Genomic Big Data for Agriculture, Hohhot 010018, China
| | - Fengying Ma
- College of Animal Science, Inner Mongolia Agricultural University, Hohhot 010010, China; (Y.G.); (F.M.); (L.G.); (C.H.); (C.S.); (L.Z.); (M.G.)
- Inner Mongolia Engineering Research Center of Genomic Big Data for Agriculture, Hohhot 010018, China
| | - Peipei Li
- College of Animal Science, Inner Mongolia Agricultural University, Hohhot 010010, China; (Y.G.); (F.M.); (L.G.); (C.H.); (C.S.); (L.Z.); (M.G.)
| | - Lili Guo
- College of Animal Science, Inner Mongolia Agricultural University, Hohhot 010010, China; (Y.G.); (F.M.); (L.G.); (C.H.); (C.S.); (L.Z.); (M.G.)
| | - Zaixia Liu
- College of Life Sciences, Inner Mongolia University, Hohhot 010020, China;
| | - Chenxi Huo
- College of Animal Science, Inner Mongolia Agricultural University, Hohhot 010010, China; (Y.G.); (F.M.); (L.G.); (C.H.); (C.S.); (L.Z.); (M.G.)
| | - Caixia Shi
- College of Animal Science, Inner Mongolia Agricultural University, Hohhot 010010, China; (Y.G.); (F.M.); (L.G.); (C.H.); (C.S.); (L.Z.); (M.G.)
| | - Lin Zhu
- College of Animal Science, Inner Mongolia Agricultural University, Hohhot 010010, China; (Y.G.); (F.M.); (L.G.); (C.H.); (C.S.); (L.Z.); (M.G.)
| | - Mingjuan Gu
- College of Animal Science, Inner Mongolia Agricultural University, Hohhot 010010, China; (Y.G.); (F.M.); (L.G.); (C.H.); (C.S.); (L.Z.); (M.G.)
| | - Risu Na
- College of Animal Science, Inner Mongolia Agricultural University, Hohhot 010010, China; (Y.G.); (F.M.); (L.G.); (C.H.); (C.S.); (L.Z.); (M.G.)
| | - Wenguang Zhang
- College of Animal Science, Inner Mongolia Agricultural University, Hohhot 010010, China; (Y.G.); (F.M.); (L.G.); (C.H.); (C.S.); (L.Z.); (M.G.)
| |
Collapse
|
10
|
Zhang R, Yang M, Schreiber J, O'Day DR, Turner JMA, Shendure J, Noble WS, Disteche CM, Deng X. Cross-species imputation and comparison of single-cell transcriptomic profiles. Genome Biol 2025; 26:40. [PMID: 40012008 PMCID: PMC11863430 DOI: 10.1186/s13059-025-03493-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2023] [Accepted: 01/31/2025] [Indexed: 02/28/2025] Open
Abstract
Cross-species comparison and prediction of gene expression profiles are important to understand regulatory changes during evolution and to transfer knowledge learned from model organisms to humans. Single-cell RNA-seq (scRNA-seq) profiles enable us to capture gene expression profiles with respect to variations among individual cells; however, cross-species comparison of scRNA-seq profiles is challenging because of data sparsity, batch effects, and the lack of one-to-one cell matching across species. Moreover, single-cell profiles are challenging to obtain in certain biological contexts, limiting the scope of hypothesis generation. Here we developed Icebear, a neural network framework that decomposes single-cell measurements into factors representing cell identity, species, and batch factors. Icebear enables accurate prediction of single-cell gene expression profiles across species, thereby providing high-resolution cell type and disease profiles in under-characterized contexts. Icebear also facilitates direct cross-species comparison of single-cell expression profiles for conserved genes that are located on the X chromosome in eutherian mammals but on autosomes in chicken. This comparison, for the first time, revealed evolutionary and diverse adaptations of X-chromosome upregulation in mammals.
Collapse
Affiliation(s)
- Ran Zhang
- Department of Genome Sciences, University of Washington, Seattle, USA
- eScience Institute, University of Washington, Seattle, USA
| | - Mu Yang
- Department of Biomedical Informatics and Medical Education, University of Washington, Seattle, USA
| | | | - Diana R O'Day
- Brotman Baty Institute for Precision Medicine, University of Washington, Seattle, USA
| | - James M A Turner
- Sex Chromosome Biology Laboratory, The Francis Crick Institute, London, UK
| | - Jay Shendure
- Department of Genome Sciences, University of Washington, Seattle, USA
- Brotman Baty Institute for Precision Medicine, University of Washington, Seattle, USA
- Howard Hughes Medical Institute, Chevy Chase, USA
- Allen Center for Cell Lineage Tracing, Seattle, USA
| | - William Stafford Noble
- Department of Genome Sciences, University of Washington, Seattle, USA.
- Paul G. Allen School of Computer Science and Engineering, University of Washington, Seattle, USA.
| | - Christine M Disteche
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, USA.
- Department of Medicine, University of Washington, Seattle, USA.
| | - Xinxian Deng
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, USA.
| |
Collapse
|
11
|
Zhong H, Han W, Gomez-Cabrero D, Tegner J, Gao X, Cui G, Aranda M. Benchmarking cross-species single-cell RNA-seq data integration methods: towards a cell type tree of life. Nucleic Acids Res 2025; 53:gkae1316. [PMID: 39778870 PMCID: PMC11707536 DOI: 10.1093/nar/gkae1316] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2024] [Revised: 11/23/2024] [Accepted: 12/27/2024] [Indexed: 01/11/2025] Open
Abstract
Cross-species single-cell RNA-seq data hold immense potential for unraveling cell type evolution and transferring knowledge between well-explored and less-studied species. However, challenges arise from interspecific genetic variation, batch effects stemming from experimental discrepancies and inherent individual biological differences. Here, we benchmarked nine data-integration methods across 20 species, encompassing 4.7 million cells, spanning eight phyla and the entire animal taxonomic hierarchy. Our evaluation reveals notable differences between the methods in removing batch effects and preserving biological variance across taxonomic distances. Methods that effectively leverage gene sequence information capture underlying biological variances, while generative model-based approaches excel in batch effect removal. SATURN demonstrates robust performance across diverse taxonomic levels, from cross-genus to cross-phylum, emphasizing its versatility. SAMap excels in integrating species beyond the cross-family level, especially for atlas-level cross-species integration, while scGen shines within or below the cross-class hierarchy. As a result, our analysis offers recommendations and guidelines for selecting suitable integration methods, enhancing cross-species single-cell RNA-seq analyses and advancing algorithm development.
Collapse
Affiliation(s)
- Huawen Zhong
- BioEngineering Program, Biological and Environmental Science and Engineering Division, King Abdullah University of Science and Technology (KAUST), Thuwal 23955-6900, Kingdom of Saudi Arabia
| | - Wenkai Han
- Computer Science Program, Computer, Electrical and Mathematical Sciences and Engineering Division, King Abdullah University of Science and Technology (KAUST), Thuwal 23955-6900, Kingdom of Saudi Arabia
- Klarman Cell Observatory, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - David Gomez-Cabrero
- BioEngineering Program, Biological and Environmental Science and Engineering Division, King Abdullah University of Science and Technology (KAUST), Thuwal 23955-6900, Kingdom of Saudi Arabia
- Unit of Translational Bioinformatics, Navarrabiomed—Fundación Miguel Servet, Universidad Pública de Navarra (UPNA), IdiSNA, Pamplona, Spain
| | - Jesper Tegner
- BioEngineering Program, Biological and Environmental Science and Engineering Division, King Abdullah University of Science and Technology (KAUST), Thuwal 23955-6900, Kingdom of Saudi Arabia
- Computer Science Program, Computer, Electrical and Mathematical Sciences and Engineering Division, King Abdullah University of Science and Technology (KAUST), Thuwal 23955-6900, Kingdom of Saudi Arabia
- Unit of Computational Medicine, Department of Medicine, Center for Molecular Medicine, Karolinska Institutet, Karolinska University Hospital, L8:05, SE-171 76 Stockholm, Sweden
- Science for Life Laboratory, Tomtebodavagen 23A, SE-17165 Solna, Sweden
| | - Xin Gao
- Computer Science Program, Computer, Electrical and Mathematical Sciences and Engineering Division, King Abdullah University of Science and Technology (KAUST), Thuwal 23955-6900, Kingdom of Saudi Arabia
- Center of Excellence on Smart Health, King Abdullah University of Science and Technology (KAUST), Thuwal 23955-6900, Kingdom of Saudi Arabia
- Center of Excellence for Generative AI, King Abdullah University of Science and Technology (KAUST), Thuwal 23955-6900, Kingdom of Saudi Arabia
| | - Guoxin Cui
- BioEngineering Program, Biological and Environmental Science and Engineering Division, King Abdullah University of Science and Technology (KAUST), Thuwal 23955-6900, Kingdom of Saudi Arabia
- Marine Science Program, Biological and Environmental Science and Engineering Division, King Abdullah University of Science and Technology (KAUST), Thuwal 23955-6900, Kingdom of Saudi Arabia
| | - Manuel Aranda
- BioEngineering Program, Biological and Environmental Science and Engineering Division, King Abdullah University of Science and Technology (KAUST), Thuwal 23955-6900, Kingdom of Saudi Arabia
- Marine Science Program, Biological and Environmental Science and Engineering Division, King Abdullah University of Science and Technology (KAUST), Thuwal 23955-6900, Kingdom of Saudi Arabia
| |
Collapse
|
12
|
Hecker N, Kempynck N, Mauduit D, Abaffyová D, Vandepoel R, Dieltiens S, Borm L, Sarropoulos I, González-Blas CB, De Man J, Davie K, Leysen E, Vandensteen J, Moors R, Hulselmans G, Lim L, De Wit J, Christiaens V, Poovathingal S, Aerts S. Enhancer-driven cell type comparison reveals similarities between the mammalian and bird pallium. Science 2025; 387:eadp3957. [PMID: 39946451 DOI: 10.1126/science.adp3957] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2024] [Accepted: 11/26/2024] [Indexed: 04/23/2025]
Abstract
Combinations of transcription factors govern the identity of cell types, which is reflected by genomic enhancer codes. We used deep learning to characterize these enhancer codes and devised three metrics to compare cell types in the telencephalon across amniotes. To this end, we generated single-cell multiome and spatially resolved transcriptomics data of the chicken telencephalon. Enhancer codes of orthologous nonneuronal and γ-aminobutyric acid-mediated (GABAergic) cell types show a high degree of similarity across amniotes, whereas excitatory neurons of the mammalian neocortex and avian pallium exhibit varying degrees of similarity. Enhancer codes of avian mesopallial neurons are most similar to those of mammalian deep-layer neurons. With this study, we present generally applicable deep learning approaches to characterize and compare cell types on the basis of genomic regulatory sequences.
Collapse
Affiliation(s)
- Nikolai Hecker
- Laboratory of Computational Biology, VIB Center for AI & Computational Biology, Leuven, Belgium
- VIB-KU Leuven Center for Brain & Disease Research, Leuven, Belgium
- Department of Human Genetics, KU Leuven, Leuven, Belgium
| | - Niklas Kempynck
- Laboratory of Computational Biology, VIB Center for AI & Computational Biology, Leuven, Belgium
- VIB-KU Leuven Center for Brain & Disease Research, Leuven, Belgium
- Department of Human Genetics, KU Leuven, Leuven, Belgium
| | - David Mauduit
- Laboratory of Computational Biology, VIB Center for AI & Computational Biology, Leuven, Belgium
- VIB-KU Leuven Center for Brain & Disease Research, Leuven, Belgium
- Department of Human Genetics, KU Leuven, Leuven, Belgium
| | - Darina Abaffyová
- Laboratory of Computational Biology, VIB Center for AI & Computational Biology, Leuven, Belgium
- VIB-KU Leuven Center for Brain & Disease Research, Leuven, Belgium
- Department of Human Genetics, KU Leuven, Leuven, Belgium
| | - Roel Vandepoel
- Laboratory of Computational Biology, VIB Center for AI & Computational Biology, Leuven, Belgium
- VIB-KU Leuven Center for Brain & Disease Research, Leuven, Belgium
- Department of Human Genetics, KU Leuven, Leuven, Belgium
| | - Sam Dieltiens
- Laboratory of Computational Biology, VIB Center for AI & Computational Biology, Leuven, Belgium
- VIB-KU Leuven Center for Brain & Disease Research, Leuven, Belgium
- Department of Human Genetics, KU Leuven, Leuven, Belgium
| | - Lars Borm
- Laboratory of Computational Biology, VIB Center for AI & Computational Biology, Leuven, Belgium
- VIB-KU Leuven Center for Brain & Disease Research, Leuven, Belgium
- Department of Human Genetics, KU Leuven, Leuven, Belgium
| | - Ioannis Sarropoulos
- Center for Molecular Biology of Heidelberg University, Heidelberg University, Heidelberg, Germany
| | - Carmen Bravo González-Blas
- VIB-KU Leuven Center for Brain & Disease Research, Leuven, Belgium
- Department of Human Genetics, KU Leuven, Leuven, Belgium
| | - Julie De Man
- Laboratory of Computational Biology, VIB Center for AI & Computational Biology, Leuven, Belgium
- VIB-KU Leuven Center for Brain & Disease Research, Leuven, Belgium
- Department of Human Genetics, KU Leuven, Leuven, Belgium
| | - Kristofer Davie
- VIB-KU Leuven Center for Brain & Disease Research, Leuven, Belgium
- Department of Neurosciences, KU Leuven, Leuven, Belgium
| | - Elke Leysen
- VIB-KU Leuven Center for Brain & Disease Research, Leuven, Belgium
- Department of Neurosciences, KU Leuven, Leuven, Belgium
| | - Jeroen Vandensteen
- VIB-KU Leuven Center for Brain & Disease Research, Leuven, Belgium
- Department of Neurosciences, KU Leuven, Leuven, Belgium
| | - Rani Moors
- VIB-KU Leuven Center for Brain & Disease Research, Leuven, Belgium
- Department of Neurosciences, KU Leuven, Leuven, Belgium
| | - Gert Hulselmans
- Laboratory of Computational Biology, VIB Center for AI & Computational Biology, Leuven, Belgium
- VIB-KU Leuven Center for Brain & Disease Research, Leuven, Belgium
- Department of Human Genetics, KU Leuven, Leuven, Belgium
| | - Lynette Lim
- VIB-KU Leuven Center for Brain & Disease Research, Leuven, Belgium
- Department of Neurosciences, KU Leuven, Leuven, Belgium
| | - Joris De Wit
- VIB-KU Leuven Center for Brain & Disease Research, Leuven, Belgium
- Department of Neurosciences, KU Leuven, Leuven, Belgium
| | - Valerie Christiaens
- Laboratory of Computational Biology, VIB Center for AI & Computational Biology, Leuven, Belgium
- VIB-KU Leuven Center for Brain & Disease Research, Leuven, Belgium
- Department of Human Genetics, KU Leuven, Leuven, Belgium
| | | | - Stein Aerts
- Laboratory of Computational Biology, VIB Center for AI & Computational Biology, Leuven, Belgium
- VIB-KU Leuven Center for Brain & Disease Research, Leuven, Belgium
- Department of Human Genetics, KU Leuven, Leuven, Belgium
| |
Collapse
|
13
|
Coorens THH, Guillaumet-Adkins A, Kovner R, Linn RL, Roberts VHJ, Sule A, Van Hoose PM. The human and non-human primate developmental GTEx projects. Nature 2025; 637:557-564. [PMID: 39815096 PMCID: PMC12013525 DOI: 10.1038/s41586-024-08244-9] [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: 06/14/2024] [Accepted: 10/17/2024] [Indexed: 01/18/2025]
Abstract
Many human diseases are the result of early developmental defects. As most paediatric diseases and disorders are rare, children are critically underrepresented in research. Functional genomics studies primarily rely on adult tissues and lack critical cell states in specific developmental windows. In parallel, little is known about the conservation of developmental programmes across non-human primate (NHP) species, with implications for human evolution. Here we introduce the developmental Genotype-Tissue Expression (dGTEx) projects, which span humans and NHPs and aim to integrate gene expression, regulation and genetics data across development and species. The dGTEx cohort will consist of 74 tissue sites across 120 human donors from birth to adulthood, and developmentally matched NHP age groups, with additional prenatal and adult animals, with 126 rhesus macaques (Macaca mulatta) and 72 common marmosets (Callithrix jacchus). The data will comprise whole-genome sequencing, extensive bulk, single-cell and spatial gene expression profiles, and chromatin accessibility data across tissues and development. Through community engagement and donor diversity, the human dGTEx study seeks to address disparities in genomic research. Thus, dGTEx will provide a reference human and NHP dataset and tissue bank, enabling research into developmental changes in expression and gene regulation, childhood disorders and the effect of genetic variation on development.
Collapse
Affiliation(s)
| | | | | | - Rebecca L Linn
- Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Victoria H J Roberts
- Division of Reproductive and Developmental Sciences, Oregon National Primate Research Center, Oregon Health and Sciences University, Portland, OR, USA
| | - Amrita Sule
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | | |
Collapse
|
14
|
Bunne C, Roohani Y, Rosen Y, Gupta A, Zhang X, Roed M, Alexandrov T, AlQuraishi M, Brennan P, Burkhardt DB, Califano A, Cool J, Dernburg AF, Ewing K, Fox EB, Haury M, Herr AE, Horvitz E, Hsu PD, Jain V, Johnson GR, Kalil T, Kelley DR, Kelley SO, Kreshuk A, Mitchison T, Otte S, Shendure J, Sofroniew NJ, Theis F, Theodoris CV, Upadhyayula S, Valer M, Wang B, Xing E, Yeung-Levy S, Zitnik M, Karaletsos T, Regev A, Lundberg E, Leskovec J, Quake SR. How to build the virtual cell with artificial intelligence: Priorities and opportunities. Cell 2024; 187:7045-7063. [PMID: 39672099 PMCID: PMC12148494 DOI: 10.1016/j.cell.2024.11.015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2024] [Revised: 11/02/2024] [Accepted: 11/12/2024] [Indexed: 12/15/2024]
Abstract
Cells are essential to understanding health and disease, yet traditional models fall short of modeling and simulating their function and behavior. Advances in AI and omics offer groundbreaking opportunities to create an AI virtual cell (AIVC), a multi-scale, multi-modal large-neural-network-based model that can represent and simulate the behavior of molecules, cells, and tissues across diverse states. This Perspective provides a vision on their design and how collaborative efforts to build AIVCs will transform biological research by allowing high-fidelity simulations, accelerating discoveries, and guiding experimental studies, offering new opportunities for understanding cellular functions and fostering interdisciplinary collaborations in open science.
Collapse
Affiliation(s)
- Charlotte Bunne
- Department of Computer Science, Stanford University, Stanford, CA, USA; Genentech, South San Francisco, CA, USA; Chan Zuckerberg Initiative, Redwood City, CA, USA; School of Computer and Communication Sciences and School of Life Sciences, EPFL, Lausanne, Switzerland
| | - Yusuf Roohani
- Department of Computer Science, Stanford University, Stanford, CA, USA; Chan Zuckerberg Initiative, Redwood City, CA, USA; Arc Institute, Palo Alto, CA, USA
| | - Yanay Rosen
- Department of Computer Science, Stanford University, Stanford, CA, USA; Chan Zuckerberg Initiative, Redwood City, CA, USA
| | - Ankit Gupta
- Chan Zuckerberg Initiative, Redwood City, CA, USA; Department of Protein Science, Science for Life Laboratory, KTH Royal Institute of Technology, Stockholm, Sweden
| | - Xikun Zhang
- Department of Computer Science, Stanford University, Stanford, CA, USA; Chan Zuckerberg Initiative, Redwood City, CA, USA; Department of Bioengineering, Stanford University, Stanford, CA, USA
| | - Marcel Roed
- Department of Computer Science, Stanford University, Stanford, CA, USA; Chan Zuckerberg Initiative, Redwood City, CA, USA
| | - Theo Alexandrov
- Department of Pharmacology, University of California, San Diego, San Diego, CA, USA; Department of Bioengineering, University of California, San Diego, San Diego, CA, USA
| | - Mohammed AlQuraishi
- Department of Bioengineering, University of California, San Diego, San Diego, CA, USA
| | | | | | - Andrea Califano
- Department of Systems Biology, Columbia University, New York, NY, USA; Vagelos College of Physicians and Surgeons, Columbia University Irving Medical Center, New York, NY, USA; Chan Zuckerberg Biohub, New York, NY, USA
| | - Jonah Cool
- Chan Zuckerberg Initiative, Redwood City, CA, USA
| | - Abby F Dernburg
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA, USA
| | - Kirsty Ewing
- Chan Zuckerberg Initiative, Redwood City, CA, USA
| | - Emily B Fox
- Department of Computer Science, Stanford University, Stanford, CA, USA; Department of Statistics, Stanford University, Stanford, CA, USA; Chan Zuckerberg Biohub, San Francisco, CA, USA
| | - Matthias Haury
- Chan Zuckerberg Institute for Advanced Biological Imaging, Redwood City, CA, USA
| | - Amy E Herr
- Chan Zuckerberg Biohub, San Francisco, CA, USA; Department of Bioengineering, University of California, Berkeley, Berkeley, CA, USA
| | | | - Patrick D Hsu
- Arc Institute, Palo Alto, CA, USA; Department of Bioengineering, University of California, Berkeley, Berkeley, CA, USA; Center for Computational Biology, University of California, Berkeley, Berkeley, CA, USA
| | | | | | | | | | - Shana O Kelley
- Chan Zuckerberg Biohub, Chicago, IL, USA; Northwestern University, Evanston, IL, USA
| | - Anna Kreshuk
- Cell Biology and Biophysics Unit, European Molecular Biology Laboratory, Heidelberg, Germany
| | - Tim Mitchison
- Department of Systems Biology, Harvard Medical School, Boston, MA, USA
| | - Stephani Otte
- Chan Zuckerberg Institute for Advanced Biological Imaging, Redwood City, CA, USA
| | - Jay Shendure
- Department of Genome Sciences, University of Washington, Seattle, WA, USA; Brotman Baty Institute for Precision Medicine, Seattle, WA, USA; Seattle Hub for Synthetic Biology, Seattle, WA, USA; Howard Hughes Medical Institute, Seattle, WA, USA
| | | | - Fabian Theis
- Institute of Computational Biology, Helmholtz Center Munich, Munich, Germany; School of Computing, Information and Technology, Technical University of Munich, Munich, Germany; TUM School of Life Sciences Weihenstephan, Technical University of Munich, Munich, Germany
| | - Christina V Theodoris
- Gladstone Institute of Cardiovascular Disease, Gladstone Institute of Data Science and Biotechnology, San Francisco, CA, USA; Department of Pediatrics, University of California, San Francisco, San Francisco, CA, USA
| | - Srigokul Upadhyayula
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA, USA; Chan Zuckerberg Biohub, San Francisco, CA, USA; Molecular Biophysics and Integrated Bioimaging Division, Lawrence Berkeley National Laboratory, Berkeley, CA, USA
| | - Marc Valer
- Chan Zuckerberg Initiative, Redwood City, CA, USA
| | - Bo Wang
- Department of Computer Science, University of Toronto, Toronto, ON, Canada; Vector Institute, Toronto, ON, Canada
| | - Eric Xing
- Carnegie Mellon University, School of Computer Science, Pittsburgh, PA, USA; Mohamed Bin Zayed University of Artificial Intelligence, Abu Dhabi, United Arab Emirates
| | - Serena Yeung-Levy
- Department of Computer Science, Stanford University, Stanford, CA, USA; Department of Biomedical Data Science, Stanford University, Stanford, CA, USA
| | - Marinka Zitnik
- Department of Biomedical Informatics, Harvard Medical School, Boston, MA, USA; Kempner Institute for the Study of Natural and Artificial Intelligence, Harvard University, Cambridge, MA, USA; Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | | | - Aviv Regev
- Genentech, South San Francisco, CA, USA.
| | - Emma Lundberg
- Chan Zuckerberg Initiative, Redwood City, CA, USA; Department of Protein Science, Science for Life Laboratory, KTH Royal Institute of Technology, Stockholm, Sweden; Department of Bioengineering, Stanford University, Stanford, CA, USA; Department of Pathology, Stanford University, Stanford, CA, USA.
| | - Jure Leskovec
- Department of Computer Science, Stanford University, Stanford, CA, USA; Chan Zuckerberg Initiative, Redwood City, CA, USA.
| | - Stephen R Quake
- Chan Zuckerberg Initiative, Redwood City, CA, USA; Department of Bioengineering, Stanford University, Stanford, CA, USA; Department of Applied Physics, Stanford University, Stanford, CA, USA.
| |
Collapse
|
15
|
Karimzadeh M, Momen-Roknabadi A, Cavazos TB, Fang Y, Chen NC, Multhaup M, Yen J, Ku J, Wang J, Zhao X, Murzynowski P, Wang K, Hanna R, Huang A, Corti D, Nguyen D, Lam T, Kilinc S, Arensdorf P, Chau KH, Hartwig A, Fish L, Li H, Behsaz B, Elemento O, Zou J, Hormozdiari F, Alipanahi B, Goodarzi H. Deep generative AI models analyzing circulating orphan non-coding RNAs enable detection of early-stage lung cancer. Nat Commun 2024; 15:10090. [PMID: 39572521 PMCID: PMC11582319 DOI: 10.1038/s41467-024-53851-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2024] [Accepted: 10/22/2024] [Indexed: 11/24/2024] Open
Abstract
Liquid biopsies have the potential to revolutionize cancer care through non-invasive early detection of tumors. Developing a robust liquid biopsy test requires collecting high-dimensional data from a large number of blood samples across heterogeneous groups of patients. We propose that the generative capability of variational auto-encoders enables learning a robust and generalizable signature of blood-based biomarkers. In this study, we analyze orphan non-coding RNAs (oncRNAs) from serum samples of 1050 individuals diagnosed with non-small cell lung cancer (NSCLC) at various stages, as well as sex-, age-, and BMI-matched controls. We demonstrate that our multi-task generative AI model, Orion, surpasses commonly used methods in both overall performance and generalizability to held-out datasets. Orion achieves an overall sensitivity of 94% (95% CI: 87%-98%) at 87% (95% CI: 81%-93%) specificity for cancer detection across all stages, outperforming the sensitivity of other methods on held-out validation datasets by more than ~ 30%.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | - Ti Lam
- Exai Bio Inc., Palo Alto, CA, US
| | | | | | | | | | | | - Helen Li
- Exai Bio Inc., Palo Alto, CA, US
| | | | | | - James Zou
- Stanford University, Stanford, CA, US
| | | | | | - Hani Goodarzi
- University of California, San Francisco, CA, US.
- Arc Institute, Palo Alto, CA, US.
| |
Collapse
|
16
|
Nourreddine S, Doctor Y, Dailamy A, Forget A, Lee YH, Chinn B, Khaliq H, Polacco B, Muralidharan M, Pan E, Zhang Y, Sigaeva A, Hansen JN, Gao J, Parker JA, Obernier K, Clark T, Chen JY, Metallo C, Lundberg E, Ideker T, Krogan N, Mali P. A PERTURBATION CELL ATLAS OF HUMAN INDUCED PLURIPOTENT STEM CELLS. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.11.03.621734. [PMID: 39574586 PMCID: PMC11580897 DOI: 10.1101/2024.11.03.621734] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/30/2024]
Abstract
Towards comprehensively investigating the genotype-phenotype relationships governing the human pluripotent stem cell state, we generated an expressed genome-scale CRISPRi Perturbation Cell Atlas in KOLF2.1J human induced pluripotent stem cells (hiPSCs) mapping transcriptional and fitness phenotypes associated with 11,739 targeted genes. Using the transcriptional phenotypes, we created a minimum distortion embedding map of the pluripotent state, demonstrating rich recapitulation of protein complexes, such as strong co-clustering of MRPL, BAF, SAGA, and Ragulator family members. Additionally, we uncovered transcriptional regulators that are uncoupled from cell fitness, discovering potential novel pluripotency (JOSD1, RNF7) and metabolic factors (ZBTB41). We validated these findings via phenotypic, protein-interaction, and metabolic tracing assays. Finally, we propose a contrastive human-cell engineering framework (CHEF), a machine learning architecture that learns from perturbation cell atlases to predict perturbation recipes that achieve desired transcriptional states. Taken together, our study presents a comprehensive resource for interrogating the regulatory networks governing pluripotency.
Collapse
Affiliation(s)
- Sami Nourreddine
- Department of Bioengineering, University of California San Diego, CA, USA
| | - Yesh Doctor
- Department of Bioengineering, University of California San Diego, CA, USA
| | - Amir Dailamy
- Department of Bioengineering, University of California San Diego, CA, USA
| | - Antoine Forget
- Quantitative Biosciences Institute (QBI), University of California San Francisco, CA, USA
- Gladstone Institute of Data Science and Biotechnology, J. David Gladstone Institutes, San Francisco, CA, USA
| | - Yi-Hung Lee
- Department of Bioengineering, University of California San Diego, CA, USA
| | - Becky Chinn
- Department of Bioengineering, University of California San Diego, CA, USA
- School of Medicine, University of California San Diego, CA, USA
| | - Hammza Khaliq
- Department of Bioengineering, University of California San Diego, CA, USA
| | - Benjamin Polacco
- Quantitative Biosciences Institute (QBI), University of California San Francisco, CA, USA
- Gladstone Institute of Data Science and Biotechnology, J. David Gladstone Institutes, San Francisco, CA, USA
- Department of Cellular and Molecular Pharmacology, University of California San Francisco, CA, USA
| | - Monita Muralidharan
- Quantitative Biosciences Institute (QBI), University of California San Francisco, CA, USA
- Gladstone Institute of Data Science and Biotechnology, J. David Gladstone Institutes, San Francisco, CA, USA
| | - Emily Pan
- Department of Bioengineering, University of California San Diego, CA, USA
| | - Yifan Zhang
- Department of Bioengineering, University of California San Diego, CA, USA
| | - Alina Sigaeva
- Division of Cellular and Clinical Proteomics, Department of Protein Science, SciLifeLab, KTH Royal Institute of Technology, Stockholm, Sweden
| | | | - Jiahao Gao
- School of Medicine, University of California San Diego, CA, USA
| | | | - Kirsten Obernier
- Gladstone Institute of Data Science and Biotechnology, J. David Gladstone Institutes, San Francisco, CA, USA
- Department of Cellular and Molecular Pharmacology, University of California San Francisco, CA, USA
| | - Timothy Clark
- Department of Medicine, University of Virginia, VA, USA
| | - Jake Y. Chen
- Department of Computer Science, The University of Alabama at Birmingham, VA, USA
| | - Christian Metallo
- Department of Bioengineering, University of California San Diego, CA, USA
- Molecular and Cell Biology Laboratory, The Salk Institute for Biological Studies, CA, USA
| | - Emma Lundberg
- Department of Bioengineering, Stanford University, CA, USA
- Department of Pathology, Stanford University, CA, USA
| | - Trey Ideker
- Department of Bioengineering, University of California San Diego, CA, USA
- School of Medicine, University of California San Diego, CA, USA
| | - Nevan Krogan
- Quantitative Biosciences Institute (QBI), University of California San Francisco, CA, USA
- Gladstone Institute of Data Science and Biotechnology, J. David Gladstone Institutes, San Francisco, CA, USA
- Department of Cellular and Molecular Pharmacology, University of California San Francisco, CA, USA
- Department of Bioengineering and Therapeutics Sciences, University of California San Francisco, CA, USA
| | - Prashant Mali
- Department of Bioengineering, University of California San Diego, CA, USA
| |
Collapse
|
17
|
Ahn K, Park HS, Choi S, Lee H, Choi H, Hong SB, Han J, Han JW, Ahn J, Song J, Park K, Cha B, Kim M, Liu HW, Song H, Kim SJ, Chung S, Kim JI, Mook-Jung I. Differentiating visceral sensory ganglion organoids from induced pluripotent stem cells. Nat Methods 2024; 21:2135-2146. [PMID: 39438735 DOI: 10.1038/s41592-024-02455-8] [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: 08/29/2023] [Accepted: 09/06/2024] [Indexed: 10/25/2024]
Abstract
The ability to generate visceral sensory neurons (VSN) from induced pluripotent stem (iPS) cells may help to gain insights into how the gut-nerve-brain axis is involved in neurological disorders. We established a protocol to differentiate human iPS-cell-derived visceral sensory ganglion organoids (VSGOs). VSGOs exhibit canonical VSN markers, and single-cell RNA sequencing revealed heterogenous molecular signatures and developmental trajectories of VSGOs aligned with native VSN. We integrated VSGOs with human colon organoids on a microfluidic device and applied this axis-on-a-chip model to Alzheimer's disease. Our results suggest that VSN could be a potential mediator for propagating gut-derived amyloid and tau to the brain in an APOE4- and LRP1-dependent manner. Furthermore, our approach was extended to include patient-derived iPS cells, which demonstrated a strong correlation with clinical data.
Collapse
Affiliation(s)
- Kyusik Ahn
- Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul, Republic of Korea
- Convergence Dementia Research Center, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Hwee-Seon Park
- Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul, Republic of Korea
- Genomic Medicine Institute, Medical Research Center, Seoul National University, Seoul, Republic of Korea
| | - Sieun Choi
- KU-KIST Graduate School of Converging Science and Technology, Korea University, Seoul, Republic of Korea
| | - Hojeong Lee
- Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul, Republic of Korea
- Department of Physiology, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Hyunjung Choi
- Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul, Republic of Korea
- Convergence Dementia Research Center, Seoul National University College of Medicine, Seoul, Republic of Korea
- Genomic Medicine Institute, Medical Research Center, Seoul National University, Seoul, Republic of Korea
| | - Seok Beom Hong
- Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul, Republic of Korea
- Convergence Dementia Research Center, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Jihui Han
- Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul, Republic of Korea
- Convergence Dementia Research Center, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Jong Won Han
- Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul, Republic of Korea
- Convergence Dementia Research Center, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Jinchul Ahn
- School of Mechanical Engineering, Korea University, Seoul, Republic of Korea
| | - Jaehoon Song
- Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul, Republic of Korea
- Convergence Dementia Research Center, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Kyunghyuk Park
- Genomic Medicine Institute, Medical Research Center, Seoul National University, Seoul, Republic of Korea
| | - Bukyung Cha
- Genomic Medicine Institute, Medical Research Center, Seoul National University, Seoul, Republic of Korea
| | - Minseop Kim
- KU-KIST Graduate School of Converging Science and Technology, Korea University, Seoul, Republic of Korea
| | - Hui-Wen Liu
- School of Mechanical Engineering, Korea University, Seoul, Republic of Korea
| | - Hyeonggyu Song
- KU-KIST Graduate School of Converging Science and Technology, Korea University, Seoul, Republic of Korea
| | - Sang Jeong Kim
- Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul, Republic of Korea
- Department of Physiology, Seoul National University College of Medicine, Seoul, Republic of Korea
- Memory Network Medical Research Center, Neuroscience Research Institute, Wide River Institute of Immunology, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Seok Chung
- KU-KIST Graduate School of Converging Science and Technology, Korea University, Seoul, Republic of Korea.
- School of Mechanical Engineering, Korea University, Seoul, Republic of Korea.
- Center for Brain Technology, Brain Science Institute, Korea Institute of Science and Technology (KIST), Seoul, Republic of Korea.
| | - Jong-Il Kim
- Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul, Republic of Korea.
- Genomic Medicine Institute, Medical Research Center, Seoul National University, Seoul, Republic of Korea.
| | - Inhee Mook-Jung
- Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul, Republic of Korea.
- Convergence Dementia Research Center, Seoul National University College of Medicine, Seoul, Republic of Korea.
- Neuroscience Research Institute, Seoul National University College of Medicine, Seoul, Republic of Korea.
| |
Collapse
|
18
|
Schroeder ME, McCormack DM, Metzner L, Kang J, Li KX, Yu E, Levandowski KM, Zaniewski H, Zhang Q, Boyden ES, Krienen FM, Feng G. Astrocyte regional specialization is shaped by postnatal development. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.10.11.617802. [PMID: 39416060 PMCID: PMC11482951 DOI: 10.1101/2024.10.11.617802] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/19/2024]
Abstract
Astrocytes are an abundant class of glial cells with critical roles in neural circuit assembly and function. Though many studies have uncovered significant molecular distinctions between astrocytes from different brain regions, how this regionalization unfolds over development is not fully understood. We used single-nucleus RNA sequencing to characterize the molecular diversity of brain cells across six developmental stages and four brain regions in the mouse and marmoset brain. Our analysis of over 170,000 single astrocyte nuclei revealed striking regional heterogeneity among astrocytes, particularly between telencephalic and diencephalic regions, at all developmental time points surveyed in both species. At the stages sampled, most of the region patterning was private to astrocytes and not shared with neurons or other glial types. Though astrocytes were already regionally patterned in late embryonic stages, this region-specific astrocyte gene expression signature changed dramatically over postnatal development, and its composition suggests that regional astrocytes further specialize postnatally to support their local neuronal circuits. Comparing across species, we found divergence in the expression of astrocytic region- and age-differentially expressed genes and the timing of astrocyte maturation relative to birth between mouse and marmoset, as well as hundreds of species differentially expressed genes. Finally, we used expansion microscopy to show that astrocyte morphology is largely conserved across gray matter regions of prefrontal cortex, striatum, and thalamus in the mouse, despite substantial molecular divergence.
Collapse
Affiliation(s)
- Margaret E Schroeder
- McGovern Institute for Brain Research, MIT, Cambridge, MA, USA
- Department of Brain and Cognitive Sciences, MIT, Cambridge, MA, USA
| | | | - Lukas Metzner
- McGovern Institute for Brain Research, MIT, Cambridge, MA, USA
| | - Jinyoung Kang
- McGovern Institute for Brain Research, MIT, Cambridge, MA, USA
- Department of Brain and Cognitive Sciences, MIT, Cambridge, MA, USA
| | - Katelyn X Li
- McGovern Institute for Brain Research, MIT, Cambridge, MA, USA
| | - Eunah Yu
- McGovern Institute for Brain Research, MIT, Cambridge, MA, USA
| | - Kirsten M Levandowski
- McGovern Institute for Brain Research, MIT, Cambridge, MA, USA
- The Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | | | - Qiangge Zhang
- McGovern Institute for Brain Research, MIT, Cambridge, MA, USA
- The Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Edward S Boyden
- McGovern Institute for Brain Research, MIT, Cambridge, MA, USA
- Department of Brain and Cognitive Sciences, MIT, Cambridge, MA, USA
- Yang Tan Collective, MIT, Cambridge, MA, USA
- Center for Neurobiological Engineering and K. Lisa Yang Center for Bionics, MIT, Cambridge, MA, USA
- Department of Biological Engineering, MIT, Cambridge, MA, USA
- Koch Institute, MIT, Cambridge, MA, USA
- Howard Hughes Medical Institute, Cambridge, MA, USA
- Media Arts and Sciences, MIT, Cambridge, MA, USA
| | - Fenna M Krienen
- Princeton Neuroscience Institute, Princeton University, Princeton, NJ, USA
| | - Guoping Feng
- McGovern Institute for Brain Research, MIT, Cambridge, MA, USA
- Department of Brain and Cognitive Sciences, MIT, Cambridge, MA, USA
- Yang Tan Collective, MIT, Cambridge, MA, USA
- The Broad Institute of MIT and Harvard, Cambridge, MA, USA
| |
Collapse
|
19
|
Bunne C, Roohani Y, Rosen Y, Gupta A, Zhang X, Roed M, Alexandrov T, AlQuraishi M, Brennan P, Burkhardt DB, Califano A, Cool J, Dernburg AF, Ewing K, Fox EB, Haury M, Herr AE, Horvitz E, Hsu PD, Jain V, Johnson GR, Kalil T, Kelley DR, Kelley SO, Kreshuk A, Mitchison T, Otte S, Shendure J, Sofroniew NJ, Theis F, Theodoris CV, Upadhyayula S, Valer M, Wang B, Xing E, Yeung-Levy S, Zitnik M, Karaletsos T, Regev A, Lundberg E, Leskovec J, Quake SR. How to Build the Virtual Cell with Artificial Intelligence: Priorities and Opportunities. ARXIV 2024:arXiv:2409.11654v2. [PMID: 39398201 PMCID: PMC11468656] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Subscribe] [Scholar Register] [Indexed: 10/15/2024]
Abstract
The cell is arguably the most fundamental unit of life and is central to understanding biology. Accurate modeling of cells is important for this understanding as well as for determining the root causes of disease. Recent advances in artificial intelligence (AI), combined with the ability to generate large-scale experimental data, present novel opportunities to model cells. Here we propose a vision of leveraging advances in AI to construct virtual cells, high-fidelity simulations of cells and cellular systems under different conditions that are directly learned from biological data across measurements and scales. We discuss desired capabilities of such AI Virtual Cells, including generating universal representations of biological entities across scales, and facilitating interpretable in silico experiments to predict and understand their behavior using Virtual Instruments. We further address the challenges, opportunities and requirements to realize this vision including data needs, evaluation strategies, and community standards and engagement to ensure biological accuracy and broad utility. We envision a future where AI Virtual Cells help identify new drug targets, predict cellular responses to perturbations, as well as scale hypothesis exploration. With open science collaborations across the biomedical ecosystem that includes academia, philanthropy, and the biopharma and AI industries, a comprehensive predictive understanding of cell mechanisms and interactions has come into reach.
Collapse
Affiliation(s)
- Charlotte Bunne
- Department of Computer Science, Stanford University, Stanford, CA, USA
- Genentech, South San Francisco, CA, USA
- Chan Zuckerberg Initiative, Redwood City, CA, USA
- School of Computer and Communication Sciences and School of Life Sciences, EPFL, Lausanne, Switzerland
| | - Yusuf Roohani
- Department of Computer Science, Stanford University, Stanford, CA, USA
- Chan Zuckerberg Initiative, Redwood City, CA, USA
- Arc Institute, Palo Alto, CA, USA
| | - Yanay Rosen
- Department of Computer Science, Stanford University, Stanford, CA, USA
- Chan Zuckerberg Initiative, Redwood City, CA, USA
| | - Ankit Gupta
- Chan Zuckerberg Initiative, Redwood City, CA, USA
- KTH Royal Institute of Technology, Science for Life Laboratory, Department of Protein Science, Stockholm, Sweden
| | - Xikun Zhang
- Department of Computer Science, Stanford University, Stanford, CA, USA
- Chan Zuckerberg Initiative, Redwood City, CA, USA
- Department of Bioengineering, Stanford University, Stanford, CA, USA
| | - Marcel Roed
- Department of Computer Science, Stanford University, Stanford, CA, USA
- Chan Zuckerberg Initiative, Redwood City, CA, USA
| | - Theo Alexandrov
- Department of Pharmacology, University of California, San Diego, CA, USA
- Department of Bioengineering, University of California, San Diego, CA, USA
| | | | | | | | - Andrea Califano
- Department of Systems Biology, Columbia University, New York, NY, USA
- Vagelos College of Physicians and Surgeons, Columbia University Irving Medical Center, New York, NY, USA
- Chan Zuckerberg Biohub New York, NY, USA
| | - Jonah Cool
- Chan Zuckerberg Initiative, Redwood City, CA, USA
| | - Abby F Dernburg
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA, USA
| | - Kirsty Ewing
- Chan Zuckerberg Initiative, Redwood City, CA, USA
| | - Emily B Fox
- Department of Computer Science, Stanford University, Stanford, CA, USA
- Department of Statistics, Stanford University, Stanford, CA, USA
- Chan Zuckerberg Biohub San Francisco, CA, USA
| | - Matthias Haury
- Chan Zuckerberg Institute for Advanced Biological Imaging, Redwood City, CA, USA
| | - Amy E Herr
- Chan Zuckerberg Biohub San Francisco, CA, USA
- Department of Bioengineering, University of California, Berkeley, CA, USA
| | | | - Patrick D Hsu
- Arc Institute, Palo Alto, CA, USA
- Department of Bioengineering, University of California, Berkeley, CA, USA
- Center for Computational Biology, University of California, Berkeley, Berkeley, CA, USA
| | | | | | | | | | - Shana O Kelley
- Chan Zuckerberg Biohub Chicago, IL, USA
- Northwestern University, Evanston, IL, USA
| | - Anna Kreshuk
- Cell Biology and Biophysics Unit, European Molecular Biology Laboratory, Heidelberg, Germany
| | - Tim Mitchison
- Department of Systems Biology, Harvard Medical School, Boston, MA, USA
| | - Stephani Otte
- Chan Zuckerberg Institute for Advanced Biological Imaging, Redwood City, CA, USA
| | - Jay Shendure
- Department of Genome Sciences, University of Washington, Seattle, WA, USA
- Brotman Baty Institute for Precision Medicine, Seattle, WA, USA
- Seattle Hub for Synthetic Biology, Seattle, WA, USA
- Howard Hughes Medical Institute, Seattle, WA, USA
| | | | - Fabian Theis
- Institute of Computational Biology, Helmholtz Center Munich, Munich, Germany
- School of Computing, Information and Technology, Technical University of Munich, Munich, Germany
- TUM School of Life Sciences Weihenstephan, Technical University of Munich, Munich, Germany
| | - Christina V Theodoris
- Gladstone Institute of Cardiovascular Disease, Gladstone Institute of Data Science and Biotechnology, San Francisco, CA, USA
- Department of Pediatrics, University of California, San Francisco, CA, USA
| | - Srigokul Upadhyayula
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA, USA
- Chan Zuckerberg Biohub San Francisco, CA, USA
- Molecular Biophysics and Integrated Bioimaging Division, Lawrence Berkeley National Laboratory, Berkeley, CA, USA
| | - Marc Valer
- Chan Zuckerberg Initiative, Redwood City, CA, USA
| | - Bo Wang
- Department of Computer Science, University of Toronto, Toronto, Ontario, Canada
- Vector Institute, Toronto, Ontario, Canada
| | - Eric Xing
- Carnegie Mellon University, School of Computer Science, Pittsburgh, PA, USA
- Mohamed Bin Zayed University of Artificial Intelligence, Abu Dhabi, United Arab Emirates
| | - Serena Yeung-Levy
- Department of Computer Science, Stanford University, Stanford, CA, USA
- Department of Biomedical Data Science, Stanford University, Stanford, CA, USA
| | - Marinka Zitnik
- Department of Biomedical Informatics, Harvard Medical School, Boston, MA, USA
- Kempner Institute for the Study of Natural and Artificial Intelligence, Harvard University, Cambridge, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | | | | | - Emma Lundberg
- Chan Zuckerberg Initiative, Redwood City, CA, USA
- KTH Royal Institute of Technology, Science for Life Laboratory, Department of Protein Science, Stockholm, Sweden
- Department of Bioengineering, Stanford University, Stanford, CA, USA
- Department of Pathology, Stanford University, Stanford, CA, USA
| | - Jure Leskovec
- Department of Computer Science, Stanford University, Stanford, CA, USA
- Chan Zuckerberg Initiative, Redwood City, CA, USA
| | - Stephen R Quake
- Chan Zuckerberg Initiative, Redwood City, CA, USA
- Department of Bioengineering, Stanford University, Stanford, CA, USA
- Department of Applied Physics, Stanford University, Stanford, CA, USA
| |
Collapse
|
20
|
Todhunter ME, Jubair S, Verma R, Saqe R, Shen K, Duffy B. Artificial intelligence and machine learning applications for cultured meat. Front Artif Intell 2024; 7:1424012. [PMID: 39381621 PMCID: PMC11460582 DOI: 10.3389/frai.2024.1424012] [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: 04/26/2024] [Accepted: 08/21/2024] [Indexed: 10/10/2024] Open
Abstract
Cultured meat has the potential to provide a complementary meat industry with reduced environmental, ethical, and health impacts. However, major technological challenges remain which require time-and resource-intensive research and development efforts. Machine learning has the potential to accelerate cultured meat technology by streamlining experiments, predicting optimal results, and reducing experimentation time and resources. However, the use of machine learning in cultured meat is in its infancy. This review covers the work available to date on the use of machine learning in cultured meat and explores future possibilities. We address four major areas of cultured meat research and development: establishing cell lines, cell culture media design, microscopy and image analysis, and bioprocessing and food processing optimization. In addition, we have included a survey of datasets relevant to CM research. This review aims to provide the foundation necessary for both cultured meat and machine learning scientists to identify research opportunities at the intersection between cultured meat and machine learning.
Collapse
Affiliation(s)
| | - Sheikh Jubair
- Alberta Machine Intelligence Institute, Edmonton, AB, Canada
| | - Ruchika Verma
- Alberta Machine Intelligence Institute, Edmonton, AB, Canada
| | - Rikard Saqe
- Department of Biology, University of Waterloo, Waterloo, ON, Canada
| | - Kevin Shen
- Department of Mathematics, University of Waterloo, Waterloo, ON, Canada
| | | |
Collapse
|
21
|
Yuan H, Mancuso CA, Johnson K, Braasch I, Krishnan A. Computational strategies for cross-species knowledge transfer and translational biomedicine. ARXIV 2024:arXiv:2408.08503v1. [PMID: 39184546 PMCID: PMC11343225] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Subscribe] [Scholar Register] [Indexed: 08/27/2024]
Abstract
Research organisms provide invaluable insights into human biology and diseases, serving as essential tools for functional experiments, disease modeling, and drug testing. However, evolutionary divergence between humans and research organisms hinders effective knowledge transfer across species. Here, we review state-of-the-art methods for computationally transferring knowledge across species, primarily focusing on methods that utilize transcriptome data and/or molecular networks. We introduce the term "agnology" to describe the functional equivalence of molecular components regardless of evolutionary origin, as this concept is becoming pervasive in integrative data-driven models where the role of evolutionary origin can become unclear. Our review addresses four key areas of information and knowledge transfer across species: (1) transferring disease and gene annotation knowledge, (2) identifying agnologous molecular components, (3) inferring equivalent perturbed genes or gene sets, and (4) identifying agnologous cell types. We conclude with an outlook on future directions and several key challenges that remain in cross-species knowledge transfer.
Collapse
Affiliation(s)
- Hao Yuan
- Genetics and Genome Science Program; Ecology, Evolution, and Behavior Program, Michigan State University
| | - Christopher A. Mancuso
- Department of Biostatistics & Informatics, University of Colorado Anschutz Medical Campus
| | - Kayla Johnson
- Department of Biomedical Informatics, University of Colorado Anschutz Medical Campus
| | - Ingo Braasch
- Department of Integrative Biology; Genetics and Genome Science Program; Ecology, Evolution, and Behavior Program, Michigan State University
| | - Arjun Krishnan
- Department of Biomedical Informatics, University of Colorado Anschutz Medical Campus
| |
Collapse
|
22
|
Ma Q, Jiang Y, Cheng H, Xu D. Harnessing the deep learning power of foundation models in single-cell omics. Nat Rev Mol Cell Biol 2024; 25:593-594. [PMID: 38926531 DOI: 10.1038/s41580-024-00756-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/28/2024]
Affiliation(s)
- Qin Ma
- Department of Biomedical Informatics, College of Medicine, The Ohio State University, Columbus, OH, USA.
- Pelotonia Institute for Immuno-Oncology, The James Comprehensive Cancer Center, The Ohio State University, Columbus, OH, USA.
| | - Yi Jiang
- Department of Biomedical Informatics, College of Medicine, The Ohio State University, Columbus, OH, USA
| | - Hao Cheng
- Department of Biomedical Informatics, College of Medicine, The Ohio State University, Columbus, OH, USA
| | - Dong Xu
- Department of Electrical Engineering and Computer Science, Bond Life Sciences Center, University of Missouri, Columbia, MO, USA
| |
Collapse
|
23
|
Embedding AI in biology. Nat Methods 2024; 21:1365-1366. [PMID: 39122959 DOI: 10.1038/s41592-024-02391-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/12/2024]
|
24
|
Guo B, Ling W, Kwon SH, Panwar P, Ghazanfar S, Martinowich K, Hicks SC. Integrating spatially-resolved transcriptomics data across tissues and individuals: challenges and opportunities. ARXIV 2024:arXiv:2408.00367v1. [PMID: 39130195 PMCID: PMC11312629] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 08/13/2024]
Abstract
Advances in spatially-resolved transcriptomics (SRT) technologies have propelled the development of new computational analysis methods to unlock biological insights. As the cost of generating these data decreases, these technologies provide an exciting opportunity to create large-scale atlases that integrate SRT data across multiple tissues, individuals, species, or phenotypes to perform population-level analyses. Here, we describe unique challenges of varying spatial resolutions in SRT data, as well as highlight the opportunities for standardized preprocessing methods along with computational algorithms amenable to atlas-scale datasets leading to improved sensitivity and reproducibility in the future.
Collapse
Affiliation(s)
- Boyi Guo
- Department of Biostatistics, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Wodan Ling
- Division of Biostatistics, Department of Population Health Sciences, Weill Cornell Medicine, NY, USA
| | - Sang Ho Kwon
- Lieber Institute for Brain Development, Johns Hopkins Medical Campus, Baltimore, MD, USA
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins School of Medicine, Baltimore, MD, USA
- Biochemistry, Cellular, and Molecular Biology Graduate Program, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Pratibha Panwar
- School of Mathematics and Statistics, The University of Sydney, NSW 2006, Australia
- Sydney Precision Data Science Centre, University of Sydney, NSW 2006, Australia
- Charles Perkins Centre, The University of Sydney, NSW 2006, Australia
| | - Shila Ghazanfar
- School of Mathematics and Statistics, The University of Sydney, NSW 2006, Australia
- Sydney Precision Data Science Centre, University of Sydney, NSW 2006, Australia
- Charles Perkins Centre, The University of Sydney, NSW 2006, Australia
| | - Keri Martinowich
- Lieber Institute for Brain Development, Johns Hopkins Medical Campus, Baltimore, MD, USA
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins School of Medicine, Baltimore, MD, USA
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins School of Medicine, Baltimore, MD, USA
- Johns Hopkins Kavli Neuroscience Discovery Institute, Baltimore, MD, USA
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD, USA
| | - Stephanie C. Hicks
- Department of Biostatistics, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
- Center for Computational Biology, Johns Hopkins University, Baltimore, MD, USA
- Malone Center for Engineering in Healthcare, Johns Hopkins University, Baltimore, MD, USA
| |
Collapse
|
25
|
Palmer JA, Rosenthal N, Teichmann SA, Litvinukova M. Revisiting Cardiac Biology in the Era of Single Cell and Spatial Omics. Circ Res 2024; 134:1681-1702. [PMID: 38843288 PMCID: PMC11149945 DOI: 10.1161/circresaha.124.323672] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/05/2024] [Revised: 04/16/2024] [Accepted: 04/24/2024] [Indexed: 06/09/2024]
Abstract
Throughout our lifetime, each beat of the heart requires the coordinated action of multiple cardiac cell types. Understanding cardiac cell biology, its intricate microenvironments, and the mechanisms that govern their function in health and disease are crucial to designing novel therapeutical and behavioral interventions. Recent advances in single-cell and spatial omics technologies have significantly propelled this understanding, offering novel insights into the cellular diversity and function and the complex interactions of cardiac tissue. This review provides a comprehensive overview of the cellular landscape of the heart, bridging the gap between suspension-based and emerging in situ approaches, focusing on the experimental and computational challenges, comparative analyses of mouse and human cardiac systems, and the rising contextualization of cardiac cells within their niches. As we explore the heart at this unprecedented resolution, integrating insights from both mouse and human studies will pave the way for novel diagnostic tools and therapeutic interventions, ultimately improving outcomes for patients with cardiovascular diseases.
Collapse
Affiliation(s)
- Jack A. Palmer
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridge, United Kingdom (J.A.P., S.A.T.)
- Wellcome-MRC Cambridge Stem Cell Institute, Jeffrey Cheah Biomedical Centre, Cambridge Biomedical Campus (J.A.P., S.A.T.), University of Cambridge, United Kingdom
| | - Nadia Rosenthal
- The Jackson Laboratory for Mammalian Genetics, Bar Harbor, ME (N.R.)
- National Heart and Lung Institute, Imperial College London, United Kingdom (N.R.)
| | - Sarah A. Teichmann
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridge, United Kingdom (J.A.P., S.A.T.)
- Wellcome-MRC Cambridge Stem Cell Institute, Jeffrey Cheah Biomedical Centre, Cambridge Biomedical Campus (J.A.P., S.A.T.), University of Cambridge, United Kingdom
- Theory of Condensed Matter Group, Department of Physics, Cavendish Laboratory (S.A.T.), University of Cambridge, United Kingdom
| | - Monika Litvinukova
- University Hospital Würzburg, Germany (M.L.)
- Würzburg Institute of Systems Immunology, Max Planck Research Group at the Julius-Maximilians-Universität Würzburg, Germany (M.L.)
- Helmholtz Pioneer Campus, Helmholtz Munich, Germany (M.L.)
| |
Collapse
|