1
|
Sadria M, Swaroop V. Discovering governing equations of biological systems through representation learning and sparse model discovery. NAR Genom Bioinform 2025; 7:lqaf048. [PMID: 40290314 PMCID: PMC12034105 DOI: 10.1093/nargab/lqaf048] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2024] [Revised: 03/19/2025] [Accepted: 04/11/2025] [Indexed: 04/30/2025] Open
Abstract
Understanding the governing rules of complex biological systems remains a significant challenge due to the nonlinear, high-dimensional nature of biological data. In this study, we present CLERA, a novel end-to-end computational framework designed to uncover parsimonious dynamical models and identify active gene programs from single-cell RNA sequencing data. By integrating a supervised autoencoder architecture with Sparse Identification of Nonlinear Dynamics, CLERA leverages prior knowledge to simultaneously extract related low-dimensional representation and uncover the underlying dynamical systems that drive the processes. Through the analysis of both synthetic and biological data, CLERA demonstrates robust performance in reconstructing gene expression dynamics, identifying key regulatory genes, and capturing temporal patterns across distinct cell types. CLERA's ability to generate dynamic interaction networks, combined with network rewiring using Personalized PageRank to highlight central genes and active gene programs, offers new insights into the complex regulatory mechanisms underlying cellular processes.
Collapse
Affiliation(s)
- Mehrshad Sadria
- Department of Applied Mathematics, University of Waterloo, Waterloo, Ontario N2L 3G1, Canada
| | - Vasu Swaroop
- Department of Computer Science Information Systems, BITS-Pilani, Pilani Campus, Pilani 333031, India
| |
Collapse
|
2
|
Zhang H, Zhang W, Zheng X, Li Y. scRDEN: single-cell dynamic gene rank differential expression network and robust trajectory inference. Sci Rep 2025; 15:16963. [PMID: 40374885 DOI: 10.1038/s41598-025-01969-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2024] [Accepted: 05/09/2025] [Indexed: 05/18/2025] Open
Abstract
The remarkable advancement of single-cell RNA sequencing (scRNA-seq) technology has empowered researchers to probe gene expression at the single-cell level with unprecedented precision. To gain a profound understanding of the heterogeneity inherent in cell fate determination, a central challenge lies in the comprehensive analysis of the dynamic regulatory alterations that underlie transcriptional differences and the accurate inference of the differentiation trajectory. Here, we propose the method scRDEN, a robust framework that infers important cell sub-populations and differential expression networks of multiple genes along the differentiation directions of each branch by converting the unstable gene expression values in cells into relatively stable gene-gene interactions (global features) and extracting the order of differential expression (network features), and further integrating the expression features of different dimension reduction methods. When applied to five published scRNA-seq datasets from human and mouse cell differentiation, scRDEN not only successfully captures the stable cell subpopulations with potential marker genes, measures the transcriptional differences of gene pairs to identify the rank differential expression network along the differentiation direction of each branch. In addition, in multiple gene rank differential expression networks, the rank expression directly related to transcription factors/marker genes shows a significant strengthening and weakening trend along with their expression changes, and the distribution of diversity and cluster coefficient show a non-monotonic change trend, including the cases of increasing first and then decreasing or decreasing first and then increasing. This may correspond to the mechanism of cells gradually differentiating into stable functions. It is particularly noteworthy that scRDEN method yielded exceptional results when applied to the large-scale, multi-branched, double-batch mouse dentate gyrus data. This outstanding performance provides novel and valuable insights into large-scale, multi-batch trajectory inference and the study of transcriptional mechanism regulation during the processes of differentiation and development.
Collapse
Affiliation(s)
- Han Zhang
- School of Mathematics and Physics, Wuhan Institute of Technology, Wuhan, 430073, China
| | - Wei Zhang
- School of Mathematics and Physics, Wuhan Institute of Technology, Wuhan, 430073, China
| | - Xiaoying Zheng
- School of Mathematics and Physics, Wuhan Institute of Technology, Wuhan, 430073, China.
| | - Yuanyuan Li
- School of Mathematics and Physics, Wuhan Institute of Technology, Wuhan, 430073, China.
| |
Collapse
|
3
|
Zhao W, Larschan E, Sandstede B, Singh R. Optimal transport reveals dynamic gene regulatory networks via gene velocity estimation. PLoS Comput Biol 2025; 21:e1012476. [PMID: 40341271 DOI: 10.1371/journal.pcbi.1012476] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2024] [Accepted: 04/10/2025] [Indexed: 05/10/2025] Open
Abstract
Inferring gene regulatory networks from gene expression data is an important and challenging problem in the biology community. We propose OTVelo, a methodology that takes time-stamped single-cell gene expression data as input and predicts gene regulation across two time points. It is known that the rate of change of gene expression, which we will refer to as gene velocity, provides crucial information that enhances such inference; however, this information is not always available due to the limitations in sequencing depth. Our algorithm overcomes this limitation by estimating gene velocities using optimal transport. We then infer gene regulation using time-lagged correlation and Granger causality via regularized linear regression. Instead of providing an aggregated network across all time points, our method uncovers the underlying dynamical mechanism across time points. We validate our algorithm on 13 simulated datasets with both synthetic and curated networks and demonstrate its efficacy on 9 experimental data sets.
Collapse
Affiliation(s)
- Wenjun Zhao
- Division of Applied Mathematics, Brown University, Providence, Rhode Island, United States of America
- Department of Mathematics, University of British Columbia, Vancouver, Canada
| | - Erica Larschan
- Department of Molecular Biology, Cell Biology and Biochemistry, Center for Computational Molecular Biology, Brown University, Providence, Rhode Island, United States of America
| | - Björn Sandstede
- Division of Applied Mathematics, Brown University, Providence, Rhode Island, United States of America
| | - Ritambhara Singh
- Department of Computer Science, Center for Computational Molecular Biology, Brown University, Providence, Rhode Island, United States of America
| |
Collapse
|
4
|
Pan Q, Ding L, Hladyshau S, Yao X, Zhou J, Yan L, Dhungana Y, Shi H, Qian C, Dong X, Burdyshaw C, Veloso JP, Khatamian A, Xie Z, Risch I, Yang X, Yang J, Huang X, Fang J, Jain A, Jain A, Rusch M, Brewer M, Peng J, Yan KK, Chi H, Yu J. scMINER: a mutual information-based framework for clustering and hidden driver inference from single-cell transcriptomics data. Nat Commun 2025; 16:4305. [PMID: 40341143 PMCID: PMC12062461 DOI: 10.1038/s41467-025-59620-6] [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: 01/17/2025] [Accepted: 04/28/2025] [Indexed: 05/10/2025] Open
Abstract
Single-cell transcriptomics data present challenges due to their inherent stochasticity and sparsity, complicating both cell clustering and cell type-specific network inference. To address these challenges, we introduce scMINER (single-cell Mutual Information-based Network Engineering Ranger), an integrative framework for unsupervised cell clustering, transcription factor and signaling protein network inference, and identification of hidden drivers from single-cell transcriptomic data. scMINER demonstrates superior accuracy in cell clustering, outperforming five state-of-the-art algorithms and excelling in distinguishing closely related cell populations. For network inference, scMINER outperforms three established methods, as validated by ATAC-seq and CROP-seq. In particular, it surpasses SCENIC in revealing key transcription factor drivers involved in T cell exhaustion and Treg tissue specification. Moreover, scMINER enables the inference of signaling protein networks and drivers with high accuracy, which presents an advantage in multimodal single cell data analysis. In addition, we establish scMINER Portal, an interactive visualization tool to facilitate exploration of scMINER results.
Collapse
Affiliation(s)
- Qingfei Pan
- Department of Computational Biology, St. Jude Children's Research Hospital, Memphis, TN, 38105, USA
| | - Liang Ding
- Department of Computational Biology, St. Jude Children's Research Hospital, Memphis, TN, 38105, USA
| | - Siarhei Hladyshau
- Department of Computational Biology, St. Jude Children's Research Hospital, Memphis, TN, 38105, USA
| | - Xiangyu Yao
- Department of Computational Biology, St. Jude Children's Research Hospital, Memphis, TN, 38105, USA
| | - Jiayu Zhou
- Department of Computational Biology, St. Jude Children's Research Hospital, Memphis, TN, 38105, USA
| | - Lei Yan
- Department of Computational Biology, St. Jude Children's Research Hospital, Memphis, TN, 38105, USA
| | - Yogesh Dhungana
- Department of Computational Biology, St. Jude Children's Research Hospital, Memphis, TN, 38105, USA
- Graduate School of Biomedical Sciences, St. Jude Children's Research Hospital, Memphis, TN, 38105, USA
| | - Hao Shi
- Department of Immunology, St. Jude Children's Research Hospital, Memphis, TN, 38105, USA
| | - Chenxi Qian
- Department of Computational Biology, St. Jude Children's Research Hospital, Memphis, TN, 38105, USA
| | - Xinran Dong
- Center for Molecular Medicine, Children's Hospital of Fudan University, Shanghai, 201102, P.R. China
| | - Chad Burdyshaw
- Department of Information Services, St. Jude Children's Research Hospital, Memphis, TN, 38105, USA
| | - Joao Pedro Veloso
- Department of Computational Biology, St. Jude Children's Research Hospital, Memphis, TN, 38105, USA
| | - Alireza Khatamian
- Department of Computational Biology, St. Jude Children's Research Hospital, Memphis, TN, 38105, USA
| | - Zhen Xie
- Department of Computational Biology, St. Jude Children's Research Hospital, Memphis, TN, 38105, USA
- Department of Physiology, University of Tennessee Health Science Center, Memphis, TN, 38163, USA
| | - Isabel Risch
- Department of Computational Biology, St. Jude Children's Research Hospital, Memphis, TN, 38105, USA
- Department of Immunology, St. Jude Children's Research Hospital, Memphis, TN, 38105, USA
| | - Xu Yang
- Department of Computational Biology, St. Jude Children's Research Hospital, Memphis, TN, 38105, USA
| | - Jiyuan Yang
- Department of Computational Biology, St. Jude Children's Research Hospital, Memphis, TN, 38105, USA
| | - Xin Huang
- Department of Computational Biology, St. Jude Children's Research Hospital, Memphis, TN, 38105, USA
- Precision Research Center for Refractory Diseases, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 201620, China
| | - Jason Fang
- Department of Computational Biology, St. Jude Children's Research Hospital, Memphis, TN, 38105, USA
| | - Anuj Jain
- Department of Computational Biology, St. Jude Children's Research Hospital, Memphis, TN, 38105, USA
| | - Arihant Jain
- Department of Computational Biology, St. Jude Children's Research Hospital, Memphis, TN, 38105, USA
| | - Michael Rusch
- Department of Computational Biology, St. Jude Children's Research Hospital, Memphis, TN, 38105, USA
| | - Michael Brewer
- Department of Information Services, St. Jude Children's Research Hospital, Memphis, TN, 38105, USA
| | - Junmin Peng
- Department of Structural Biology and Developmental Neurobiology, St. Jude Children's Research Hospital, Memphis, TN, 38105, USA
| | - Koon-Kiu Yan
- Department of Computational Biology, St. Jude Children's Research Hospital, Memphis, TN, 38105, USA
| | - Hongbo Chi
- Graduate School of Biomedical Sciences, St. Jude Children's Research Hospital, Memphis, TN, 38105, USA
- Department of Immunology, St. Jude Children's Research Hospital, Memphis, TN, 38105, USA
| | - Jiyang Yu
- Department of Computational Biology, St. Jude Children's Research Hospital, Memphis, TN, 38105, USA.
- Graduate School of Biomedical Sciences, St. Jude Children's Research Hospital, Memphis, TN, 38105, USA.
| |
Collapse
|
5
|
Wang JC, Chen YJ, Zou Q. GRACE: Unveiling Gene Regulatory Networks With Causal Mechanistic Graph Neural Networks in Single-Cell RNA-Sequencing Data. IEEE TRANSACTIONS ON NEURAL NETWORKS AND LEARNING SYSTEMS 2025; 36:9005-9017. [PMID: 38896510 DOI: 10.1109/tnnls.2024.3412753] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/21/2024]
Abstract
Reconstructing gene regulatory networks (GRNs) using single-cell RNA sequencing (scRNA-seq) data holds great promise for unraveling cellular fate development and heterogeneity. While numerous machine-learning methods have been proposed to infer GRNs from scRNA-seq gene expression data, many of them operate solely in a statistical or black box manner, limiting their capacity for making causal inferences between genes. In this study, we introduce GRN inference with Accuracy and Causal Explanation (GRACE), a novel graph-based causal autoencoder framework that combines a structural causal model (SCM) with graph neural networks (GNNs) to enable GRN inference and gene causal reasoning from scRNA-seq data. By explicitly modeling causal relationships between genes, GRACE facilitates the learning of regulatory context and gene embeddings. With the learned gene signals, our model successfully decoding the causal structures and alleviates the accurate determination of multiple attributes of gene regulation that is important to determine the regulatory levels. Through extensive evaluations on seven benchmarks, we demonstrate that GRACE outperforms 14 state-of-the-art GRN inference methods, with the incorporation of causal mechanisms significantly enhancing the accuracy of GRN and gene causality inference. Furthermore, the application to human peripheral blood mononuclear cell (PBMC) samples reveals cell type-specific regulators in monocyte phagocytosis and immune regulation, validated through network analysis and functional enrichment analysis.
Collapse
|
6
|
Su G, Wang H, Zhang Y, Wilkins MR, Canete PF, Yu D, Yang Y, Zhang W. Inferring gene regulatory networks by hypergraph generative model. CELL REPORTS METHODS 2025; 5:101026. [PMID: 40220759 DOI: 10.1016/j.crmeth.2025.101026] [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: 06/23/2024] [Revised: 01/16/2025] [Accepted: 03/20/2025] [Indexed: 04/14/2025]
Abstract
We present hypergraph variational autoencoder (HyperG-VAE), a Bayesian deep generative model that leverages hypergraph representation to model single-cell RNA sequencing (scRNA-seq) data. The model features a cell encoder with a structural equation model to account for cellular heterogeneity and construct gene regulatory networks (GRNs) alongside a gene encoder using hypergraph self-attention to identify gene modules. The synergistic optimization of encoders via a decoder improves GRN inference, single-cell clustering, and data visualization, as validated by benchmarks. HyperG-VAE effectively uncovers gene regulation patterns and demonstrates robustness in downstream analyses, as shown in B cell development data from bone marrow. Gene set enrichment analysis of overlapping genes in predicted GRNs confirms the gene encoder's role in refining GRN inference. Offering an efficient solution for scRNA-seq analysis and GRN construction, HyperG-VAE also holds the potential for extending GRN modeling to temporal and multimodal single-cell omics.
Collapse
Affiliation(s)
- Guangxin Su
- School of Computer Science and Engineering, The University of New South Wales, Sydney, NSW, Australia; ARC Centre of Excellence for the Mathematical Analysis of Cellular Systems (MACSYS), Melbourne, VIC, Australia
| | - Hanchen Wang
- ARC Centre of Excellence for the Mathematical Analysis of Cellular Systems (MACSYS), Melbourne, VIC, Australia; Australian Artificial Intelligence Institute, The University of Technology Sydney, Sydney, NSW, Australia
| | - Ying Zhang
- School of Computer Science and Technology, Zhejiang Gongshang University, Zhejiang, China
| | - Marc R Wilkins
- ARC Centre of Excellence for the Mathematical Analysis of Cellular Systems (MACSYS), Melbourne, VIC, Australia; Systems Biology Initiative, School of Biotechnology and Biomolecular Sciences, The University of New South Wales, Sydney, NSW, Australia
| | - Pablo F Canete
- Frazer Institute, Faculty of Health, Medicine and Behaviour Sciences, The University of Queensland, Brisbane, QLD, Australia
| | - Di Yu
- Frazer Institute, Faculty of Health, Medicine and Behaviour Sciences, The University of Queensland, Brisbane, QLD, Australia; Ian Frazer Centre for Children's Immunotherapy Research, Child Health Research Centre, Faculty of Health, Medicine and Behaviour Sciences, The University of Queensland, Brisbane, QLD, Australia
| | - Yang Yang
- Frazer Institute, Faculty of Health, Medicine and Behaviour Sciences, The University of Queensland, Brisbane, QLD, Australia.
| | - Wenjie Zhang
- School of Computer Science and Engineering, The University of New South Wales, Sydney, NSW, Australia; ARC Centre of Excellence for the Mathematical Analysis of Cellular Systems (MACSYS), Melbourne, VIC, Australia.
| |
Collapse
|
7
|
Dong Z, Liu X, Guo X, Liu X, Wang B, Shao W, Tian C, Zheng Y, Yu Q, Zhong L, Sun J, Li S, Xin T, Zhang B, Yang T, Lu H, Rose JKC, Lucas WJ, Xu X, Huang S, Liu H, Yang X. Developmental innovation of inferior ovaries and flower sex orchestrated by KNOX1 in cucurbits. NATURE PLANTS 2025; 11:861-877. [PMID: 40169874 DOI: 10.1038/s41477-025-01950-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/01/2024] [Accepted: 01/17/2025] [Indexed: 04/03/2025]
Abstract
In flowering plants, inferior ovaries are key morphological innovations that evolved multiple times from superior ovaries to protect female parts of the flower. However, the developmental mechanisms underlying inferior ovary formation remain largely unknown. Comparative spatial transcriptome mapping and cell lineage reconstructions in developing floral buds of cucumber and tomato, which have inferior and superior ovaries, respectively, revealed that inferior ovaries develop from accelerated receptacle growth resulting from the continuous activity of meristematic stems cells at the base of the cucumber floral organs. Genetic knockout of a receptacle-specific KNOX1 transcription factor in cucumber caused arrest in receptacle growth and yielded bisexual flowers with superior ovaries similar to those of tomato. Here we provide developmental and mechanistic insights into inferior ovary formation and sex determination in cucurbits.
Collapse
Affiliation(s)
- Zhaonian Dong
- State Key Laboratory of Vegetable Biobreeding, Institute of Vegetables and Flowers, Chinese Academy of Agricultural Sciences, Beijing, China
- Shenzhen Branch, Guangdong Laboratory of Lingnan Modern Agriculture, Key Laboratory of Synthetic Biology, Ministry of Agriculture and Rural Affairs, Agricultural Genomics Institute at Shenzhen, Chinese Academy of Agricultural Sciences, Shenzhen, China
| | - Xiaolin Liu
- State Key Laboratory of Vegetable Biobreeding, Institute of Vegetables and Flowers, Chinese Academy of Agricultural Sciences, Beijing, China
| | | | - Xun Liu
- State Key Laboratory of Vegetable Biobreeding, Institute of Vegetables and Flowers, Chinese Academy of Agricultural Sciences, Beijing, China
| | - Bowen Wang
- State Key Laboratory of Vegetable Biobreeding, Institute of Vegetables and Flowers, Chinese Academy of Agricultural Sciences, Beijing, China
- Shenzhen Branch, Guangdong Laboratory of Lingnan Modern Agriculture, Key Laboratory of Synthetic Biology, Ministry of Agriculture and Rural Affairs, Agricultural Genomics Institute at Shenzhen, Chinese Academy of Agricultural Sciences, Shenzhen, China
| | - Wenwen Shao
- BGI Research, Wuhan, China
- State Key Laboratory of Agricultural Genomics, BGI Research, Shenzhen, China
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, China
| | - Caihuan Tian
- State Key Laboratory of Vegetable Biobreeding, Institute of Vegetables and Flowers, Chinese Academy of Agricultural Sciences, Beijing, China
- State Key Laboratory of Plant Genomics, Institute of Genetics and Developmental Biology, The Innovative Academy of Seed Design, Chinese Academy of Sciences, Beijing, China
| | - Yingying Zheng
- State Key Laboratory of Vegetable Biobreeding, Institute of Vegetables and Flowers, Chinese Academy of Agricultural Sciences, Beijing, China
| | - Qiong Yu
- State Key Laboratory of Vegetable Biobreeding, Institute of Vegetables and Flowers, Chinese Academy of Agricultural Sciences, Beijing, China
| | - Liyuan Zhong
- BGI Research, Wuhan, China
- State Key Laboratory of Agricultural Genomics, BGI Research, Shenzhen, China
| | - Jinjing Sun
- State Key Laboratory of Vegetable Biobreeding, Institute of Vegetables and Flowers, Chinese Academy of Agricultural Sciences, Beijing, China
| | - Shengkang Li
- State Key Laboratory of Agricultural Genomics, BGI Research, Shenzhen, China
| | - Tongxu Xin
- State Key Laboratory of Vegetable Biobreeding, Institute of Vegetables and Flowers, Chinese Academy of Agricultural Sciences, Beijing, China
| | - Bohan Zhang
- State Key Laboratory of Agricultural Genomics, BGI Research, Shenzhen, China
| | - Tao Yang
- China National GeneBank, Shenzhen, China
| | - Haorong Lu
- China National GeneBank, Shenzhen, China
| | - Jocelyn K C Rose
- Plant Biology Section, School of Integrative Plant Science, Cornell University, Ithaca, NY, USA
| | - William J Lucas
- Department of Plant Biology, College of Biological Sciences, University of California, Davis, CA, USA
| | - Xun Xu
- State Key Laboratory of Agricultural Genomics, BGI Research, Shenzhen, China
| | - Sanwen Huang
- Shenzhen Branch, Guangdong Laboratory of Lingnan Modern Agriculture, Key Laboratory of Synthetic Biology, Ministry of Agriculture and Rural Affairs, Agricultural Genomics Institute at Shenzhen, Chinese Academy of Agricultural Sciences, Shenzhen, China
- National Key Laboratory of Tropical Crop Breeding, Chinese Academy of Tropical Agricultural Sciences, Haikou, China
| | - Huan Liu
- State Key Laboratory of Agricultural Genomics, BGI Research, Shenzhen, China.
| | - Xueyong Yang
- State Key Laboratory of Vegetable Biobreeding, Institute of Vegetables and Flowers, Chinese Academy of Agricultural Sciences, Beijing, China.
| |
Collapse
|
8
|
Hu Q, Lu X, Xue Z, Wang R. Gene regulatory network inference during cell fate decisions by perturbation strategies. NPJ Syst Biol Appl 2025; 11:23. [PMID: 40032872 PMCID: PMC11876352 DOI: 10.1038/s41540-025-00504-2] [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: 09/23/2024] [Accepted: 02/21/2025] [Indexed: 03/05/2025] Open
Abstract
With rapid advances in biological technology and computational approaches, inferring specific gene regulatory networks from data alone during cell fate decisions, including determining direct regulations and their intensities between biomolecules, remains one of the most significant challenges. In this study, we propose a general computational approach based on systematic perturbation, statistical, and differential analyses to infer network topologies and identify network differences during cell fate decisions. For each cell fate state, we first theoretically show how to calculate local response matrices based on perturbation data under systematic perturbation analysis, and we also derive the wild-type (WT) local response matrix for specific ordinary differential equations. To make the inferred network more accurate and eliminate the impact of perturbation degrees, the confidence interval (CI) of local response matrices under multiple perturbations is applied, and the redefined local response matrix is proposed in statistical analysis to determine network topologies across all cell fates. Then in differential analysis, we introduce the concept of relative local response matrix, which enables us to identify critical regulations governing each cell state and dominant cell states associated with specific regulations. The epithelial to mesenchymal transition (EMT) network is chosen as an illustrative example to verify the feasibility of the approach. Largely consistent with experimental observations, the differences of inferred networks at the three cell states can be quantitatively identified. The approach presented here can be also applied to infer other regulatory networks related to cell fate decisions.
Collapse
Affiliation(s)
- Qing Hu
- Department of Mathematics, Shanghai University, Shanghai, China
| | - Xiaoqi Lu
- Department of Mathematics, Shanghai University, Shanghai, China
| | - Zhuozhen Xue
- Department of Mathematics, Shanghai University, Shanghai, China
| | - Ruiqi Wang
- Department of Mathematics, Shanghai University, Shanghai, China.
- Newtouch Center for Mathematics of Shanghai University, Shanghai, China.
| |
Collapse
|
9
|
Han M, Chen X, Li X, Ma J, Chen T, Yang C, Wang J, Li Y, Guo W, Zhu Y. MulNet: a scalable framework for reconstructing intra- and intercellular signaling networks from bulk and single-cell RNA-seq data. Brief Bioinform 2025; 26:bbaf081. [PMID: 40095604 PMCID: PMC11912874 DOI: 10.1093/bib/bbaf081] [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: 05/24/2024] [Revised: 02/02/2025] [Accepted: 02/13/2025] [Indexed: 03/19/2025] Open
Abstract
Gene expression involves complex interactions between DNA, RNA, proteins, and small molecules. However, most existing molecular networks are built on limited interaction types, resulting in a fragmented understanding of gene regulation. Here, we present MulNet, a framework that organizes diverse molecular interactions underlying gene expression data into a scalable multilayer network. Additionally, MulNet can accurately identify gene modules and key regulators within this network. When applied across diverse cancer datasets, MulNet outperformed state-of-the-art methods in identifying biologically relevant modules. MulNet analysis of RNA-seq data from colon cancer revealed numerous well-established cancer regulators and a promising new therapeutic target, miR-8485, along with several downstream pathways it governs to inhibit tumor growth. MulNet analysis of single-cell RNA-seq data from head and neck cancer revealed intricate communication networks between fibroblasts and malignant cells mediated by transcription factors and cytokines. Overall, MulNet enables high-resolution reconstruction of intra- and intercellular communication from both bulk and single-cell data. The MulNet code and application are available at https://github.com/free1234hm/MulNet.
Collapse
Affiliation(s)
- Mingfei Han
- State Key Laboratory of Medical Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences (Beijing), Beijing Institute of Lifeomics, No. 38, Life Science Park Road, Changping District, Beijing 102206, China
| | - Xiaoqing Chen
- State Key Laboratory of Medical Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences (Beijing), Beijing Institute of Lifeomics, No. 38, Life Science Park Road, Changping District, Beijing 102206, China
| | - Xiao Li
- State Key Laboratory of Medical Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences (Beijing), Beijing Institute of Lifeomics, No. 38, Life Science Park Road, Changping District, Beijing 102206, China
| | - Jie Ma
- State Key Laboratory of Medical Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences (Beijing), Beijing Institute of Lifeomics, No. 38, Life Science Park Road, Changping District, Beijing 102206, China
| | - Tao Chen
- State Key Laboratory of Medical Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences (Beijing), Beijing Institute of Lifeomics, No. 38, Life Science Park Road, Changping District, Beijing 102206, China
| | - Chunyuan Yang
- State Key Laboratory of Medical Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences (Beijing), Beijing Institute of Lifeomics, No. 38, Life Science Park Road, Changping District, Beijing 102206, China
| | - Juan Wang
- State Key Laboratory of Medical Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences (Beijing), Beijing Institute of Lifeomics, No. 38, Life Science Park Road, Changping District, Beijing 102206, China
| | - Yingxing Li
- Central Research Laboratory, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, No. 1 Shuaifuyuan Wangfujing Dongcheng District, Beijing 100730, China
| | - Wenting Guo
- Central Research Laboratory, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, No. 1 Shuaifuyuan Wangfujing Dongcheng District, Beijing 100730, China
| | - Yunping Zhu
- State Key Laboratory of Medical Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences (Beijing), Beijing Institute of Lifeomics, No. 38, Life Science Park Road, Changping District, Beijing 102206, China
| |
Collapse
|
10
|
Chen L, Dautle M, Gao R, Zhang S, Chen Y. Inferring gene regulatory networks from time-series scRNA-seq data via GRANGER causal recurrent autoencoders. Brief Bioinform 2025; 26:bbaf089. [PMID: 40062616 PMCID: PMC11891664 DOI: 10.1093/bib/bbaf089] [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: 10/31/2024] [Revised: 01/26/2025] [Accepted: 02/18/2025] [Indexed: 05/13/2025] Open
Abstract
The development of single-cell RNA sequencing (scRNA-seq) technology provides valuable data resources for inferring gene regulatory networks (GRNs), enabling deeper insights into cellular mechanisms and diseases. While many methods exist for inferring GRNs from static scRNA-seq data, current approaches face challenges in accurately handling time-series scRNA-seq data due to high noise levels and data sparsity. The temporal dimension introduces additional complexity by requiring models to capture dynamic changes, increasing sensitivity to noise, and exacerbating data sparsity across time points. In this study, we introduce GRANGER, an unsupervised deep learning-based method that integrates multiple advanced techniques, including a recurrent variational autoencoder, GRANGER causality, sparsity-inducing penalties, and negative binomial (NB)-based loss functions, to infer GRNs. GRANGER was evaluated using multiple popular benchmarking datasets, where it demonstrated superior performance compared to eight well-known GRN inference methods. The integration of a NB-based loss function and sparsity-inducing penalties in GRANGER significantly enhanced its capacity to address dropout noise and sparsity in scRNA-seq data. Additionally, GRANGER exhibited robustness against high levels of dropout noise. We applied GRANGER to scRNA-seq data from the whole mouse brain obtained through the BRAIN Initiative project and identified GRNs for five transcription regulators: E2f7, Gbx1, Sox10, Prox1, and Onecut2, which play crucial roles in diverse brain cell types. The inferred GRNs not only recalled many known regulatory relationships but also revealed sets of novel regulatory interactions with functional potential. These findings demonstrate that GRANGER is a highly effective tool for real-world applications in discovering novel gene regulatory relationships.
Collapse
Affiliation(s)
- Liang Chen
- College of Computer and Information Engineering, Tianjin Normal University, 393 Binshui W Ave, Tianjin, Tianjin 300387, China
| | - Madison Dautle
- Department of Biological and Biomedical Sciences, Rowan University, 201 Mullica Hill Road, Glassboro, NJ 08028, United States
| | - Ruoying Gao
- College of Computer and Information Engineering, Tianjin Normal University, 393 Binshui W Ave, Tianjin, Tianjin 300387, China
| | - Shaoqiang Zhang
- College of Computer and Information Engineering, Tianjin Normal University, 393 Binshui W Ave, Tianjin, Tianjin 300387, China
| | - Yong Chen
- Department of Biological and Biomedical Sciences, Rowan University, 201 Mullica Hill Road, Glassboro, NJ 08028, United States
| |
Collapse
|
11
|
Stock M, Losert C, Zambon M, Popp N, Lubatti G, Hörmanseder E, Heinig M, Scialdone A. Leveraging prior knowledge to infer gene regulatory networks from single-cell RNA-sequencing data. Mol Syst Biol 2025; 21:214-230. [PMID: 39939367 PMCID: PMC11876610 DOI: 10.1038/s44320-025-00088-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2024] [Revised: 01/29/2025] [Accepted: 01/30/2025] [Indexed: 02/14/2025] Open
Abstract
Many studies have used single-cell RNA sequencing (scRNA-seq) to infer gene regulatory networks (GRNs), which are crucial for understanding complex cellular regulation. However, the inherent noise and sparsity of scRNA-seq data present significant challenges to accurate GRN inference. This review explores one promising approach that has been proposed to address these challenges: integrating prior knowledge into the inference process to enhance the reliability of the inferred networks. We categorize common types of prior knowledge, such as experimental data and curated databases, and discuss methods for representing priors, particularly through graph structures. In addition, we classify recent GRN inference algorithms based on their ability to incorporate these priors and assess their performance in different contexts. Finally, we propose a standardized benchmarking framework to evaluate algorithms more fairly, ensuring biologically meaningful comparisons. This review provides guidance for researchers selecting GRN inference methods and offers insights for developers looking to improve current approaches and foster innovation in the field.
Collapse
Affiliation(s)
- Marco Stock
- Helmholtz Center Munich Institute of Epigenetics und Stem Cells, Munich, Germany
- Helmholtz Center Munich Institute of Computational Biology, Munich, Germany
- Helmholtz Center Munich Institute of Functional Epigenetics, Munich, Germany
- TUM School of Life Sciences Weihenstephan, Technical University of Munich, Freising, Germany
| | - Corinna Losert
- Helmholtz Center Munich Institute of Computational Biology, Munich, Germany
- Department of Computer Science, TUM School of Computation, Information and Technology, Technical University of Munich, Garching, Germany
| | - Matteo Zambon
- Helmholtz Center Munich Institute of Epigenetics und Stem Cells, Munich, Germany
- Helmholtz Center Munich Institute of Computational Biology, Munich, Germany
- Helmholtz Center Munich Institute of Functional Epigenetics, Munich, Germany
| | - Niclas Popp
- Helmholtz Center Munich Institute of Epigenetics und Stem Cells, Munich, Germany
- Helmholtz Center Munich Institute of Computational Biology, Munich, Germany
- Helmholtz Center Munich Institute of Functional Epigenetics, Munich, Germany
| | - Gabriele Lubatti
- Helmholtz Center Munich Institute of Epigenetics und Stem Cells, Munich, Germany
- Helmholtz Center Munich Institute of Computational Biology, Munich, Germany
- Helmholtz Center Munich Institute of Functional Epigenetics, Munich, Germany
| | - Eva Hörmanseder
- Helmholtz Center Munich Institute of Epigenetics und Stem Cells, Munich, Germany
| | - Matthias Heinig
- Helmholtz Center Munich Institute of Computational Biology, Munich, Germany
- Department of Computer Science, TUM School of Computation, Information and Technology, Technical University of Munich, Garching, Germany
- German Centre for Cardiovascular Research (DZHK), Munich Heart Association, Partner Site Munich, Berlin, Germany
| | - Antonio Scialdone
- Helmholtz Center Munich Institute of Epigenetics und Stem Cells, Munich, Germany.
- Helmholtz Center Munich Institute of Computational Biology, Munich, Germany.
- Helmholtz Center Munich Institute of Functional Epigenetics, Munich, Germany.
| |
Collapse
|
12
|
Wang C, Liu ZP. Diffusion-based generation of gene regulatory networks from scRNA-seq data with DigNet. Genome Res 2025; 35:340-354. [PMID: 39694856 PMCID: PMC11874984 DOI: 10.1101/gr.279551.124] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2024] [Accepted: 12/10/2024] [Indexed: 12/20/2024]
Abstract
A gene regulatory network (GRN) intricately encodes the interconnectedness of identities and functionalities of genes within cells, ultimately shaping cellular specificity. Despite decades of endeavors, reverse engineering of GRNs from gene expression profiling data remains a profound challenge, particularly when it comes to reconstructing cell-specific GRNs that are tailored to precise cellular and genetic contexts. Here, we propose a discrete diffusion generation model, called DigNet, capable of generating corresponding GRNs from high-throughput single-cell RNA sequencing (scRNA-seq) data. DigNet embeds the network generation process into a multistep recovery procedure with Markov properties. Each intermediate step has a specific model to recover a portion of the gene regulatory architectures. It thus can ensure compatibility between global network structures and regulatory modules through the unique multistep diffusion procedure. Furthermore, through iMetacell integration and non-Euclidean discrete space modeling, DigNet is robust to the presence of noise in scRNA-seq data and the sparsity of GRNs. Benchmark evaluation results against more than a dozen state-of-the-art network inference methods demonstrate that DigNet achieves superior performance across various single-cell GRN reconstruction experiments. Furthermore, DigNet provides unique insights into the immune response in breast cancer, derived from differential gene regulation identified in T cells. As an open-source software, DigNet offers a powerful and effective tool for generating cell-specific GRNs from scRNA-seq data.
Collapse
Affiliation(s)
- Chuanyuan Wang
- Department of Biomedical Engineering, School of Control Science and Engineering, Shandong University, Jinan, Shandong 250061, China
| | - Zhi-Ping Liu
- Department of Biomedical Engineering, School of Control Science and Engineering, Shandong University, Jinan, Shandong 250061, China
| |
Collapse
|
13
|
Sun Y, Gao J. HGATLink: single-cell gene regulatory network inference via the fusion of heterogeneous graph attention networks and transformer. BMC Bioinformatics 2025; 26:49. [PMID: 39934680 PMCID: PMC11817978 DOI: 10.1186/s12859-025-06071-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2024] [Accepted: 01/29/2025] [Indexed: 02/13/2025] Open
Abstract
BACKGROUND Gene regulatory networks (GRNs) involve complex regulatory relationships between genes and play important roles in the study of various biological systems and diseases. The introduction of single-cell sequencing (scRNA-seq) technology has allowed gene regulation studies to be carried out on specific cell types, providing the opportunity to accurately infer gene regulatory networks. However, the sparsity and noise problems of single-cell sequencing data pose challenges for gene regulatory network inference, and although many gene regulatory network inference methods have been proposed, they often fail to eliminate transitive interactions or do not address multilevel relationships and nonlinear features in the graph data well. RESULTS On the basis of the above limitations, we propose a gene regulatory network inference framework named HGATLink. HGATLink combines the heterogeneous graph attention network and simplified transformer to capture complex interactions effectively between genes in low-dimensional space via matrix decomposition techniques, which not only enhances the ability to model complex heterogeneous graph structures and alleviate transitive interactions, but also effectively captures the long-range dependencies between genes to ensure more accurate prediction. CONCLUSIONS Compared with 10 state-of-the-art GRN inference methods on 14 scRNA-seq datasets under two metrics, AUROC and AUPRC, HGATLink shows good stability and accuracy in gene regulatory network inference tasks.
Collapse
Affiliation(s)
- Yao Sun
- Department of Computer Science and Technology, College of Computer and Information Engineering, Inner Mongolia Agricultural University, Hohhot, 010011, Inner Mongolia, China
- Inner Mongolia Autonomous Region Key Laboratory of Big Data Research, Hohhot, 010018, Inner Mongolia, China
| | - Jing Gao
- Department of Computer Science and Technology, College of Computer and Information Engineering, Inner Mongolia Agricultural University, Hohhot, 010011, Inner Mongolia, China.
- Inner Mongolia Autonomous Region Key Laboratory of Big Data Research, Hohhot, 010018, Inner Mongolia, China.
| |
Collapse
|
14
|
Yuan Q, Duren Z. Inferring gene regulatory networks from single-cell multiome data using atlas-scale external data. Nat Biotechnol 2025; 43:247-257. [PMID: 38609714 PMCID: PMC11825371 DOI: 10.1038/s41587-024-02182-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2023] [Accepted: 02/26/2024] [Indexed: 04/14/2024]
Abstract
Existing methods for gene regulatory network (GRN) inference rely on gene expression data alone or on lower resolution bulk data. Despite the recent integration of chromatin accessibility and RNA sequencing data, learning complex mechanisms from limited independent data points still presents a daunting challenge. Here we present LINGER (Lifelong neural network for gene regulation), a machine-learning method to infer GRNs from single-cell paired gene expression and chromatin accessibility data. LINGER incorporates atlas-scale external bulk data across diverse cellular contexts and prior knowledge of transcription factor motifs as a manifold regularization. LINGER achieves a fourfold to sevenfold relative increase in accuracy over existing methods and reveals a complex regulatory landscape of genome-wide association studies, enabling enhanced interpretation of disease-associated variants and genes. Following the GRN inference from reference single-cell multiome data, LINGER enables the estimation of transcription factor activity solely from bulk or single-cell gene expression data, leveraging the abundance of available gene expression data to identify driver regulators from case-control studies.
Collapse
Affiliation(s)
- Qiuyue Yuan
- Center for Human Genetics, Department of Genetics and Biochemistry, Clemson University, Greenwood, SC, USA
| | - Zhana Duren
- Center for Human Genetics, Department of Genetics and Biochemistry, Clemson University, Greenwood, SC, USA.
| |
Collapse
|
15
|
Shin D, Gong J, Jeong SD, Cho Y, Kim H, Kim T, Cho K. Attractor Landscape Analysis Reveals a Reversion Switch in the Transition of Colorectal Tumorigenesis. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2412503. [PMID: 39840939 PMCID: PMC11848608 DOI: 10.1002/advs.202412503] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/07/2024] [Revised: 11/27/2024] [Indexed: 01/23/2025]
Abstract
A cell fate change such as tumorigenesis incurs critical transition. It remains a longstanding challenge whether the underlying mechanism can be unraveled and a molecular switch that can reverse such transition is found. Here a systems framework, REVERT, is presented with which can reconstruct the core molecular regulatory network model and a reversion switch based on single-cell transcriptome data over the transition process is identified. The usefulness of REVERT is demonstrated by applying it to single-cell transcriptome of patient-derived matched organoids of colon cancer and normal colon. REVERT is a generic framework that can be applied to investigate various cell fate transition phenomena.
Collapse
Affiliation(s)
- Dongkwan Shin
- Department of Bio and Brain EngineeringKorea Advanced Institute of Science and Technology (KAIST)Daejeon34141Republic of Korea
- Research InstituteNational Cancer CenterGoyang10408Republic of Korea
- Department of Cancer Biomedical ScienceNational Cancer Center Graduate School of Cancer Science and PolicyGoyang10408Republic of Korea
| | - Jeong‐Ryeol Gong
- Department of Bio and Brain EngineeringKorea Advanced Institute of Science and Technology (KAIST)Daejeon34141Republic of Korea
| | - Seoyoon D. Jeong
- Department of Bio and Brain EngineeringKorea Advanced Institute of Science and Technology (KAIST)Daejeon34141Republic of Korea
| | - Youngwon Cho
- Department of Molecular Medicine and Biopharmaceutical SciencesGraduate School of Convergence Science and TechnologySeoul National UniversitySeoul03080Republic of Korea
| | - Hwang‐Phill Kim
- Department of Molecular Medicine and Biopharmaceutical SciencesGraduate School of Convergence Science and TechnologySeoul National UniversitySeoul03080Republic of Korea
| | - Tae‐You Kim
- Department of Molecular Medicine and Biopharmaceutical SciencesGraduate School of Convergence Science and TechnologySeoul National UniversitySeoul03080Republic of Korea
| | - Kwang‐Hyun Cho
- Department of Bio and Brain EngineeringKorea Advanced Institute of Science and Technology (KAIST)Daejeon34141Republic of Korea
| |
Collapse
|
16
|
Yang S, Bussing A, Marra G, Brinkmeier ML, Camper SA, Davis SW, Ho YY. TIME-CoExpress: Temporal Trajectory Modeling of Dynamic Gene Co-expression Patterns Using Single-Cell Transcriptomics Data. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.01.23.634392. [PMID: 39896591 PMCID: PMC11785143 DOI: 10.1101/2025.01.23.634392] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/04/2025]
Abstract
The rapid advancements of single-cell RNA sequencing (scRNAseq) technology provide high-resolution views of transcriptomic activity within a single cell. Most routine analyses of scRNAseq data focus on individual genes; however, the one-gene-at-a-time analysis is likely to miss meaningful genetic interactions. Gene co-expression analysis addresses this issue by identifying coordinated gene expression changes in response to cellular conditions, such as developmental or temporal trajectory. Identifying differential co-expression gene combinations along the cell temporal trajectory using scRNAseq data can provide deeper insight into the biological processes. Existing approaches for gene co-expression analysis assume a restrictive linear change of gene co-expression. In this paper, we propose a copula-based approach with proper data-driven smoothing functions to model non-linear gene co-expression changes along cellular temporal trajectories. Our proposed approach provides flexibility to incorporate characteristics such as over-dispersion and zero-inflation rate observed in scRNAseq data into the modeling framework. We conducted a series of simulation analyses to evaluate the performance of the proposed algorithm. We demonstrate the implementation of the proposed algorithm using a scRNAseq dataset and identify differential co-expression gene pairs along cell temporal trajectory in pituitary embryonic development comparing Nxn - / - mutated versus wild-type mice.
Collapse
|
17
|
Jung S. Advances in modeling cellular state dynamics: integrating omics data and predictive techniques. Anim Cells Syst (Seoul) 2025; 29:72-83. [PMID: 39807350 PMCID: PMC11727055 DOI: 10.1080/19768354.2024.2449518] [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: 10/15/2024] [Revised: 12/19/2024] [Accepted: 12/29/2024] [Indexed: 01/16/2025] Open
Abstract
Dynamic modeling of cellular states has emerged as a pivotal approach for understanding complex biological processes such as cell differentiation, disease progression, and tissue development. This review provides a comprehensive overview of current approaches for modeling cellular state dynamics, focusing on techniques ranging from dynamic or static biomolecular network models to deep learning models. We highlight how these approaches integrated with various omics data such as transcriptomics, and single-cell RNA sequencing could be used to capture and predict cellular behavior and transitions. We also discuss applications of these modeling approaches in predicting gene knockout effects, designing targeted interventions, and simulating organ development. This review emphasizes the importance of selecting appropriate modeling strategies based on scalability and resolution requirements, which vary according to the complexity and size of biological systems under study. By evaluating strengths, limitations, and recent advancements of these methodologies, we aim to guide future research in developing more robust and interpretable models for understanding and manipulating cellular state dynamics in various biological contexts, ultimately advancing therapeutic strategies and precision medicine.
Collapse
Affiliation(s)
- Sungwon Jung
- Department of Genome Medicine and Science, Gachon University College of Medicine, Incheon, Republic of Korea
- Gachon Institute of Genome Medicine and Science, Gachon University Gil Medical Center, Incheon, Republic of Korea
| |
Collapse
|
18
|
Cao G, Chen D. Unveiling Long Non-coding RNA Networks from Single-Cell Omics Data Through Artificial Intelligence. Methods Mol Biol 2025; 2883:257-279. [PMID: 39702712 DOI: 10.1007/978-1-0716-4290-0_11] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2024]
Abstract
Single-cell omics technologies have revolutionized the study of long non-coding RNAs (lncRNAs), offering unprecedented resolution in elucidating their expression dynamics, cell-type specificity, and associated gene regulatory networks (GRNs). Concurrently, the integration of artificial intelligence (AI) methodologies has significantly advanced our understanding of lncRNA functions and its implications in disease pathogenesis. This chapter discusses the progress in single-cell omics data analysis, emphasizing its pivotal role in unraveling the molecular mechanisms underlying cellular heterogeneity and the associated regulatory networks involving lncRNAs. Additionally, we provide a summary of single-cell omics resources and AI models for constructing single-cell gene regulatory networks (scGRNs). Finally, we explore the challenges and prospects of exploring scGRNs in the context of lncRNA biology.
Collapse
Affiliation(s)
- Guangshuo Cao
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing, China
| | - Dijun Chen
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing, China.
| |
Collapse
|
19
|
Weng G, Martin P, Kim H, Won KJ. Integrating Prior Knowledge Using Transformer for Gene Regulatory Network Inference. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2409990. [PMID: 39605181 PMCID: PMC11744656 DOI: 10.1002/advs.202409990] [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: 08/31/2024] [Revised: 10/23/2024] [Indexed: 11/29/2024]
Abstract
Gene regulatory network (GRN) inference, a process of reconstructing gene regulatory rules from experimental data, has the potential to discover new regulatory rules. However, existing methods often struggle to generalize across diverse cell types and account for unseen regulators. Here, this work presents GRNPT, a novel Transformer-based framework that integrates large language model (LLM) embeddings from publicly accessible biological data and a temporal convolutional network (TCN) autoencoder to capture regulatory patterns from single-cell RNA sequencing (scRNA-seq) trajectories. GRNPT significantly outperforms both supervised and unsupervised methods in inferring GRNs, particularly when training data is limited. Notably, GRNPT exhibits exceptional generalizability, accurately predicting regulatory relationships in previously unseen cell types and even regulators. By combining LLMs ability to distillate biological knowledge from text and deep learning methodologies capturing complex patterns in gene expression data, GRNPT overcomes the limitations of traditional GRN inference methods and enables more accurate and comprehensive understanding of gene regulatory dynamics.
Collapse
Affiliation(s)
- Guangzheng Weng
- Biotech Research and Innovation Centre (BRIC)University of CopenhagenOle Maaløes Vej 5Copenhagen2200Denmark
| | - Patrick Martin
- Department of Computational BiomedicineCedars‐Sinai Medical CenterLos AngelesCA90069USA
| | - Hyobin Kim
- Department of Computational BiomedicineCedars‐Sinai Medical CenterLos AngelesCA90069USA
| | - Kyoung Jae Won
- Department of Computational BiomedicineCedars‐Sinai Medical CenterLos AngelesCA90069USA
| |
Collapse
|
20
|
Cui W, Long Q, Liu W, Fang C, Wang X, Wang P, Zhou Y. Hierarchical Graph Transformer With Contrastive Learning for Gene Regulatory Network Inference. IEEE J Biomed Health Inform 2025; 29:690-699. [PMID: 39401117 DOI: 10.1109/jbhi.2024.3476490] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/05/2025]
Abstract
Gene regulatory networks (GRNs) are crucial for understanding gene regulation and cellular processes. Inferring GRNs helps uncover regulatory pathways, shedding light on the regulation and development of cellular processes. With the rise of high-throughput sequencing and advancements in computational technology, computational models have emerged as cost-effective alternatives to traditional experimental studies. Moreover, the surge in ChIP-seq data for TF-DNA binding has catalyzed the development of graph neural network (GNN)-based methods, greatly advancing GRN inference capabilities. However, most existing GNN-based methods suffer from the inability to capture long-distance structural semantic correlations due to transitive interactions. In this paper, we introduce a novel GNN-based model named Hierarchical Graph Transformer with Contrastive Learning for GRN (HGTCGRN) inference. HGTCGRN excels at capturing structural semantics using a hierarchical graph Transformer, which introduces a series of gene family nodes representing gene functions as virtual nodes to interact with nodes in the GRNS. These semantic-aware virtual-node embeddings are aggregated to produce node representations with varying emphasis. Additionally, we leverage gene ontology information to construct gene interaction networks for contrastive learning optimization of GRNs. Experimental results demonstrate that HGTCGRN achieves superior performance in GRN inference.
Collapse
|
21
|
Yachimura T, Wang H, Imoto Y, Yoshida M, Tasaki S, Kojima Y, Yabuta Y, Saitou M, Hiraoka Y. scEGOT: single-cell trajectory inference framework based on entropic Gaussian mixture optimal transport. BMC Bioinformatics 2024; 25:388. [PMID: 39710672 DOI: 10.1186/s12859-024-05988-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2024] [Accepted: 11/13/2024] [Indexed: 12/24/2024] Open
Abstract
BACKGROUND Time-series scRNA-seq data have opened a door to elucidate cell differentiation, and in this context, the optimal transport theory has been attracting much attention. However, there remain critical issues in interpretability and computational cost. RESULTS We present scEGOT, a comprehensive framework for single-cell trajectory inference, as a generative model with high interpretability and low computational cost. Applied to the human primordial germ cell-like cell (PGCLC) induction system, scEGOT identified the PGCLC progenitor population and bifurcation time of segregation. Our analysis shows TFAP2A is insufficient for identifying PGCLC progenitors, requiring NKX1-2. Additionally, MESP1 and GATA6 are also crucial for PGCLC/somatic cell segregation. CONCLUSIONS These findings shed light on the mechanism that segregates PGCLC from somatic lineages. Notably, not limited to scRNA-seq, scEGOT's versatility can extend to general single-cell data like scATAC-seq, and hence has the potential to revolutionize our understanding of such datasets and, thereby also, developmental biology.
Collapse
Affiliation(s)
- Toshiaki Yachimura
- Mathematical Science Center for Co-creative Society, Tohoku University, Sendai, 980-0845, Japan.
| | - Hanbo Wang
- Department of Anatomy and Cell Biology, Graduate School of Medicine, Kyoto University, Yoshida-Konoe-cho, Sakyo-ku, Kyoto, 606-8501, Japan
| | - Yusuke Imoto
- Institute for the Advanced Study of Human Biology, Kyoto University Institute for Advanced Study, Kyoto University, Yoshida-Konoe-cho, Sakyo-ku, Kyoto, 606-8501, Japan
| | - Momoko Yoshida
- Faculty of Science, Kyoto University, Kitashirakawa Oiwake-cho, Sakyo-ku, Kyoto, 606-8502, Japan
| | - Sohei Tasaki
- Department of Mathematics, Faculty of Science, Hokkaido University, Sapporo, 060-0810, Japan
| | - Yoji Kojima
- Center for iPS Cell Research and Application, Kyoto University, 53 Kawahara-cho, Shogoin, Sakyo-ku, Kyoto, 606-8507, Japan
| | - Yukihiro Yabuta
- Department of Anatomy and Cell Biology, Graduate School of Medicine, Kyoto University, Yoshida-Konoe-cho, Sakyo-ku, Kyoto, 606-8501, Japan
- Institute for the Advanced Study of Human Biology, Kyoto University Institute for Advanced Study, Kyoto University, Yoshida-Konoe-cho, Sakyo-ku, Kyoto, 606-8501, Japan
| | - Mitinori Saitou
- Department of Anatomy and Cell Biology, Graduate School of Medicine, Kyoto University, Yoshida-Konoe-cho, Sakyo-ku, Kyoto, 606-8501, Japan
- Institute for the Advanced Study of Human Biology, Kyoto University Institute for Advanced Study, Kyoto University, Yoshida-Konoe-cho, Sakyo-ku, Kyoto, 606-8501, Japan
- Center for iPS Cell Research and Application, Kyoto University, 53 Kawahara-cho, Shogoin, Sakyo-ku, Kyoto, 606-8507, Japan
| | - Yasuaki Hiraoka
- Institute for the Advanced Study of Human Biology, Kyoto University Institute for Advanced Study, Kyoto University, Yoshida-Konoe-cho, Sakyo-ku, Kyoto, 606-8501, Japan.
| |
Collapse
|
22
|
Manchel A, Gee M, Vadigepalli R. From sampling to simulating: Single-cell multiomics in systems pathophysiological modeling. iScience 2024; 27:111322. [PMID: 39628578 PMCID: PMC11612781 DOI: 10.1016/j.isci.2024.111322] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/06/2024] Open
Abstract
As single-cell omics data sampling and acquisition methods have accumulated at an unprecedented rate, various data analysis pipelines have been developed for the inference of cell types, cell states and their distribution, state transitions, state trajectories, and state interactions. This presents a new opportunity in which single-cell omics data can be utilized to generate high-resolution, high-fidelity computational models. In this review, we discuss how single-cell omics data can be used to build computational models to simulate biological systems at various scales. We propose that single-cell data can be integrated with physiological information to generate organ-specific models, which can then be assembled to generate multi-organ systems pathophysiological models. Finally, we discuss how generic multi-organ models can be brought to the patient-specific level thus permitting their use in the clinical setting.
Collapse
Affiliation(s)
- Alexandra Manchel
- Daniel Baugh Institute of Functional Genomics/Computational Biology, Department of Pathology and Genomic Medicine, Thomas Jefferson University, Philadelphia, PA, USA
| | - Michelle Gee
- Daniel Baugh Institute of Functional Genomics/Computational Biology, Department of Pathology and Genomic Medicine, Thomas Jefferson University, Philadelphia, PA, USA
- Department of Chemical and Biomolecular Engineering, University of Delaware, Newark, DE, USA
| | - Rajanikanth Vadigepalli
- Daniel Baugh Institute of Functional Genomics/Computational Biology, Department of Pathology and Genomic Medicine, Thomas Jefferson University, Philadelphia, PA, USA
| |
Collapse
|
23
|
Khullar S, Huang X, Ramesh R, Svaren J, Wang D. NetREm: Network Regression Embeddings reveal cell-type transcription factor coordination for gene regulation. BIOINFORMATICS ADVANCES 2024; 5:vbae206. [PMID: 40260118 PMCID: PMC12011367 DOI: 10.1093/bioadv/vbae206] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 06/05/2024] [Revised: 10/22/2024] [Accepted: 12/18/2024] [Indexed: 04/23/2025]
Abstract
Motivation Transcription factor (TF) coordination plays a key role in gene regulation via direct and/or indirect protein-protein interactions (PPIs) and co-binding to regulatory elements on DNA. Single-cell technologies facilitate gene expression measurement for individual cells and cell-type identification, yet the connection between TF-TF coordination and target gene (TG) regulation of various cell types remains unclear. Results To address this, we introduce our innovative computational approach, Network Regression Embeddings (NetREm), to reveal cell-type TF-TF coordination activities for TG regulation. NetREm leverages network-constrained regularization, using prior knowledge of PPIs among TFs, to analyze single-cell gene expression data, uncovering cell-type coordinating TFs and identifying revolutionary TF-TG candidate regulatory network links. NetREm's performance is validated using simulation studies and benchmarked across several datasets in humans, mice, yeast. Further, we showcase NetREm's ability to prioritize valid novel human TF-TF coordination links in 9 peripheral blood mononuclear and 42 immune cell sub-types. We apply NetREm to examine cell-type networks in central and peripheral nerve systems (e.g. neuronal, glial, Schwann cells) and in Alzheimer's disease versus Controls. Top predictions are validated with experimental data from rat, mouse, and human models. Additional functional genomics data helps link genetic variants to our TF-TG regulatory and TF-TF coordination networks. Availability and implementation https://github.com/SaniyaKhullar/NetREm.
Collapse
Affiliation(s)
- Saniya Khullar
- Waisman Center, University of Wisconsin-Madison, Madison, WI 53705, United States
- Department of Biostatistics and Medical Informatics, University of Wisconsin-Madison, Madison, WI 53076, United States
| | - Xiang Huang
- Waisman Center, University of Wisconsin-Madison, Madison, WI 53705, United States
| | - Raghu Ramesh
- Waisman Center, University of Wisconsin-Madison, Madison, WI 53705, United States
- Comparative Biomedical Sciences Training Program, University of Wisconsin-Madison, Madison, WI 53706, United States
| | - John Svaren
- Waisman Center, University of Wisconsin-Madison, Madison, WI 53705, United States
- Department of Comparative Biosciences, School of Veterinary Medicine, University of Wisconsin-Madison, Madison, WI 53706, United States
| | - Daifeng Wang
- Waisman Center, University of Wisconsin-Madison, Madison, WI 53705, United States
- Department of Biostatistics and Medical Informatics, University of Wisconsin-Madison, Madison, WI 53076, United States
- Department of Computer Sciences, University of Wisconsin-Madison, Madison, WI 53706, United States
| |
Collapse
|
24
|
Yang B, Li J, Li X, Liu S. Gene regulatory network inference based on novel ensemble method. Brief Funct Genomics 2024; 23:866-878. [PMID: 39324652 DOI: 10.1093/bfgp/elae036] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2024] [Revised: 08/09/2024] [Accepted: 09/06/2024] [Indexed: 09/27/2024] Open
Abstract
Gene regulatory networks (GRNs) contribute toward understanding the function of genes and the development of cancer or the impact of key genes on diseases. Hence, this study proposes an ensemble method based on 13 basic classification methods and a flexible neural tree (FNT) to improve GRN identification accuracy. The primary classification methods contain ridge classification, stochastic gradient descent, Gaussian process classification, Bernoulli Naive Bayes, adaptive boosting, gradient boosting decision tree, hist gradient boosting classification, eXtreme gradient boosting (XGBoost), multilayer perceptron, light gradient boosting machine, random forest, support vector machine, and k-nearest neighbor algorithm, which are regarded as the input variable set of FNT model. Additionally, a hybrid evolutionary algorithm based on a gene programming variant and particle swarm optimization is developed to search for the optimal FNT model. Experiments on three simulation datasets and three real single-cell RNA-seq datasets demonstrate that the proposed ensemble feature outperforms 13 supervised algorithms, seven unsupervised algorithms (ARACNE, CLR, GENIE3, MRNET, PCACMI, GENECI, and EPCACMI) and four single cell-specific methods (SCODE, BiRGRN, LEAP, and BiGBoost) based on the area under the receiver operating characteristic curve, area under the precision-recall curve, and F1 metrics.
Collapse
Affiliation(s)
- Bin Yang
- School of Information Science and Engineering, Zaozhuang University, No. 1 Beian Road, Zaozhuang 277160, China
| | - Jing Li
- School of Information Science and Engineering, Zaozhuang University, No. 1 Beian Road, Zaozhuang 277160, China
| | - Xiang Li
- Information Department, Qingdao Eighth People's Hospital, No. 84 Fengshan Road, Qingdao 266121, China
| | - Sanrong Liu
- School of Information Science and Engineering, Zaozhuang University, No. 1 Beian Road, Zaozhuang 277160, China
| |
Collapse
|
25
|
Bonev B, Castelo-Branco G, Chen F, Codeluppi S, Corces MR, Fan J, Heiman M, Harris K, Inoue F, Kellis M, Levine A, Lotfollahi M, Luo C, Maynard KR, Nitzan M, Ramani V, Satijia R, Schirmer L, Shen Y, Sun N, Green GS, Theis F, Wang X, Welch JD, Gokce O, Konopka G, Liddelow S, Macosko E, Ali Bayraktar O, Habib N, Nowakowski TJ. Opportunities and challenges of single-cell and spatially resolved genomics methods for neuroscience discovery. Nat Neurosci 2024; 27:2292-2309. [PMID: 39627587 PMCID: PMC11999325 DOI: 10.1038/s41593-024-01806-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2023] [Accepted: 09/23/2024] [Indexed: 12/13/2024]
Abstract
Over the past decade, single-cell genomics technologies have allowed scalable profiling of cell-type-specific features, which has substantially increased our ability to study cellular diversity and transcriptional programs in heterogeneous tissues. Yet our understanding of mechanisms of gene regulation or the rules that govern interactions between cell types is still limited. The advent of new computational pipelines and technologies, such as single-cell epigenomics and spatially resolved transcriptomics, has created opportunities to explore two new axes of biological variation: cell-intrinsic regulation of cell states and expression programs and interactions between cells. Here, we summarize the most promising and robust technologies in these areas, discuss their strengths and limitations and discuss key computational approaches for analysis of these complex datasets. We highlight how data sharing and integration, documentation, visualization and benchmarking of results contribute to transparency, reproducibility, collaboration and democratization in neuroscience, and discuss needs and opportunities for future technology development and analysis.
Collapse
Affiliation(s)
- Boyan Bonev
- Helmholtz Pioneer Campus, Helmholtz Zentrum München, Neuherberg, Germany
- Physiological Genomics, Biomedical Center, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Gonçalo Castelo-Branco
- Laboratory of Molecular Neurobiology, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden
| | - Fei Chen
- The Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | | | - M Ryan Corces
- Gladstone Institute of Neurological Disease, San Francisco, CA, USA
- Gladstone Institute of Data Science and Biotechnology, San Francisco, CA, USA
- Department of Neurology, University of California, San Francisco, San Francisco, CA, USA
| | - Jean Fan
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD, USA
| | - Myriam Heiman
- Department of Brain and Cognitive Sciences, MIT, Cambridge, MA, USA
- The Picower Institute for Learning and Memory, MIT, Cambridge, MA, USA
| | - Kenneth Harris
- UCL Queen Square Institute of Neurology, University College London, London, UK
| | - Fumitaka Inoue
- Institute for the Advanced Study of Human Biology (WPI-ASHBi), Kyoto University, Kyoto, Japan
| | - Manolis Kellis
- The Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Computer Science and Artificial Intelligence Lab, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Ariel Levine
- Spinal Circuits and Plasticity Unit, National Institute of Neurological Disorders and Stroke, Bethesda, MD, USA
| | - Mo Lotfollahi
- Institute of Computational Biology, Helmholtz Center Munich - German Research Center for Environmental Health, Neuherberg, Germany
- Wellcome Sanger Institute, Wellcome Genome Campus, Cambridge, UK
| | - Chongyuan Luo
- Department of Human Genetics, University of California, Los Angeles, Los Angeles, CA, USA
| | - Kristen R Maynard
- Lieber Institute for Brain Development, Baltimore, MD, USA
- Department of Psychiatry, Johns Hopkins School of Medicine, Baltimore, MD, USA
- Department of Neuroscience, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Mor Nitzan
- School of Computer Science and Engineering, The Hebrew University of Jerusalem, Jerusalem, Israel
- Racah Institute of Physics, The Hebrew University of Jerusalem, Jerusalem, Israel
- Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Vijay Ramani
- Gladstone Institute of Data Science and Biotechnology, San Francisco, CA, USA
- Department of Biochemistry and Biophysics, University of California, San Francisco, San Francisco, CA, USA
- Bakar Computational Health Sciences Institute, San Francisco, CA, USA
| | - Rahul Satijia
- New York Genome Center, New York, NY, USA
- Center for Genomics and Systems Biology, New York University, New York, NY, USA
| | - Lucas Schirmer
- Department of Neurology, Mannheim Center for Translational Neuroscience, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Yin Shen
- Department of Neurology, University of California, San Francisco, San Francisco, CA, USA
- Institute for Human Genetics, University of California, San Francisco, San Francisco, CA, USA
- Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA, USA
| | - Na Sun
- The Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Computer Science and Artificial Intelligence Lab, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Gilad S Green
- The Edmond and Lily Safra Center for Brain Sciences, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Fabian Theis
- Wellcome Sanger Institute, Wellcome Genome Campus, Cambridge, UK
- Department of Human Genetics, University of California, Los Angeles, Los Angeles, CA, USA
| | - Xiao Wang
- The Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Department of Chemistry, Massachusetts Institute of Technology, Cambridge, MA, USA
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Joshua D Welch
- Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, MI, USA
| | - Ozgun Gokce
- German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany.
- Department of Neurodegenerative Diseases and Geriatric Psychiatry, University Hospital Bonn, Bonn, Germany.
| | - Genevieve Konopka
- Department of Neuroscience, UT Southwestern Medical Center, Dallas, TX, USA.
- Peter O'Donnell Jr. Brain Institute, UT Southwestern Medical Center, Dallas, TX, USA.
| | - Shane Liddelow
- Neuroscience Institute, NYU Grossman School of Medicine, New York, NY, USA.
- Department of Neuroscience & Physiology, NYU Grossman School of Medicine, New York, NY, USA.
- Parekh Center for Interdisciplinary Neurology, NYU Grossman School of Medicine, New York, NY, USA.
- Department of Ophthalmology, NYU Grossman School of Medicine, New York, NY, USA.
| | - Evan Macosko
- The Broad Institute of MIT and Harvard, Cambridge, MA, USA.
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA.
- Department of Neurobiology, Harvard Medical School, Boston, MA, USA.
- Department of Psychiatry, Massachusetts General Hospital, Boston, MA, USA.
| | | | - Naomi Habib
- The Edmond and Lily Safra Center for Brain Sciences, The Hebrew University of Jerusalem, Jerusalem, Israel.
| | - Tomasz J Nowakowski
- Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA, USA.
- Department of Neurological Surgery, University of California, San Francisco, San Francisco, CA, USA.
- Department of Anatomy, University of California, San Francisco, San Francisco, CA, USA.
- Department of Psychiatry and Behavioral Sciences, University of California, San Francisco, San Francisco, CA, USA.
- The Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California, San Francisco, San Francisco, CA, USA.
| |
Collapse
|
26
|
Liu W, Teng Z, Li Z, Chen J. CVGAE: A Self-Supervised Generative Method for Gene Regulatory Network Inference Using Single-Cell RNA Sequencing Data. Interdiscip Sci 2024; 16:990-1004. [PMID: 38778003 DOI: 10.1007/s12539-024-00633-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2023] [Revised: 04/07/2024] [Accepted: 04/09/2024] [Indexed: 05/25/2024]
Abstract
Gene regulatory network (GRN) inference based on single-cell RNA sequencing data (scRNAseq) plays a crucial role in understanding the regulatory mechanisms between genes. Various computational methods have been employed for GRN inference, but their performance in terms of network accuracy and model generalization is not satisfactory, and their poor performance is caused by high-dimensional data and network sparsity. In this paper, we propose a self-supervised method for gene regulatory network inference using single-cell RNA sequencing data (CVGAE). CVGAE uses graph neural network for inductive representation learning, which merges gene expression data and observed topology into a low-dimensional vector space. The well-trained vectors will be used to calculate mathematical distance of each gene, and further predict interactions between genes. In overall framework, FastICA is implemented to relief computational complexity caused by high dimensional data, and CVGAE adopts multi-stacked GraphSAGE layers as an encoder and an improved decoder to overcome network sparsity. CVGAE is evaluated on several single cell datasets containing four related ground-truth networks, and the result shows that CVGAE achieve better performance than comparative methods. To validate learning and generalization capabilities, CVGAE is applied in few-shot environment by change the ratio of train set and test set. In condition of few-shot, CVGAE obtains comparable or superior performance.
Collapse
Affiliation(s)
- Wei Liu
- School of Computer Science, Xiangtan University, Xiangtan, 411105, China.
| | - Zhijie Teng
- School of Computer Science, Xiangtan University, Xiangtan, 411105, China
| | - Zejun Li
- School of Computer Science and Engineering, Hunan Institute of Technology, Hengyang, 412002, China
| | - Jing Chen
- School of Electronic and Information Engineering, Suzhou University of Science and Technology, Suzhou, 215009, China.
| |
Collapse
|
27
|
Peng D, Cahan P. OneSC: a computational platform for recapitulating cell state transitions. Bioinformatics 2024; 40:btae703. [PMID: 39570626 PMCID: PMC11630913 DOI: 10.1093/bioinformatics/btae703] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2024] [Revised: 11/13/2024] [Accepted: 11/19/2024] [Indexed: 11/22/2024] Open
Abstract
MOTIVATION Computational modeling of cell state transitions has been a great interest of many in the field of developmental biology, cancer biology, and cell fate engineering because it enables performing perturbation experiments in silico more rapidly and cheaply than could be achieved in a lab. Recent advancements in single-cell RNA-sequencing (scRNA-seq) allow the capture of high-resolution snapshots of cell states as they transition along temporal trajectories. Using these high-throughput datasets, we can train computational models to generate in silico "synthetic" cells that faithfully mimic the temporal trajectories. RESULTS Here we present OneSC, a platform that can simulate cell state transitions using systems of stochastic differential equations govern by a regulatory network of core transcription factors (TFs). Different from many current network inference methods, OneSC prioritizes on generating Boolean network that produces faithful cell state transitions and terminal cell states that mimic real biological systems. Applying OneSC to real data, we inferred a core TF network using a mouse myeloid progenitor scRNA-seq dataset and showed that the dynamical simulations of that network generate synthetic single-cell expression profiles that faithfully recapitulate the four myeloid differentiation trajectories going into differentiated cell states (erythrocytes, megakaryocytes, granulocytes, and monocytes). Finally, through the in silico perturbations of the mouse myeloid progenitor core network, we showed that OneSC can accurately predict cell fate decision biases of TF perturbations that closely match with previous experimental observations. AVAILABILITY AND IMPLEMENTATION OneSC is implemented as a Python package on GitHub (https://github.com/CahanLab/oneSC) and on Zenodo (https://zenodo.org/records/14052421).
Collapse
Affiliation(s)
- Da Peng
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD 21205, United States
| | - Patrick Cahan
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD 21205, United States
- Institute for Cell Engineering, Johns Hopkins University, Baltimore, MD 21205, United States
- Department of Molecular Biology and Genetics, Johns Hopkins University, Baltimore, MD 21205, United States
| |
Collapse
|
28
|
Li R, Wu J, Li G, Liu J, Liu J, Xuan J, Deng Z. SIGRN: Inferring Gene Regulatory Network with Soft Introspective Variational Autoencoders. Int J Mol Sci 2024; 25:12741. [PMID: 39684451 DOI: 10.3390/ijms252312741] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2024] [Revised: 11/21/2024] [Accepted: 11/25/2024] [Indexed: 12/18/2024] Open
Abstract
Gene regulatory networks (GRNs) exhibit the complex regulatory relationships among genes, which are essential for understanding developmental biology and uncovering the fundamental aspects of various biological phenomena. It is an effective and economical way to infer GRNs from single-cell RNA sequencing (scRNA-seq) with computational methods. Recent researches have been done on the problem by using variational autoencoder (VAE) and structural equation model (SEM). Due to the shortcoming of VAE generating poor-quality data, in this paper, a soft introspective adversarial gene regulatory network unsupervised inference model, called SIGRN, is proposed by introducing adversarial mechanism in building a variational autoencoder model. SIGRN applies "soft" introspective adversarial mode to avoid training additional neural networks and adding additional training parameters. It demonstrates superior inference accuracy across most benchmark datasets when compared to nine leading-edge methods. In addition, method SIGRN also achieves better performance on representing cells and generating scRNA-seq data in most datasets. All of which have been verified via substantial experiments. The SIGRN method shows promise for generating scRNA-seq data and inferring GRNs.
Collapse
Affiliation(s)
- Rongyuan Li
- Key Lab of Education Blockchain and Intelligent Technology, Ministry of Education, Guangxi Normal University, Guilin 541004, China
- Guangxi Key Lab of Multi-Source Information Mining & Security, Guangxi Normal University, Guilin 541004, China
- School of Computer Science and Engineering, Guangxi Normal University, Guilin 541004, China
| | - Jingli Wu
- Key Lab of Education Blockchain and Intelligent Technology, Ministry of Education, Guangxi Normal University, Guilin 541004, China
- Guangxi Key Lab of Multi-Source Information Mining & Security, Guangxi Normal University, Guilin 541004, China
- School of Computer Science and Engineering, Guangxi Normal University, Guilin 541004, China
| | - Gaoshi Li
- Key Lab of Education Blockchain and Intelligent Technology, Ministry of Education, Guangxi Normal University, Guilin 541004, China
- Guangxi Key Lab of Multi-Source Information Mining & Security, Guangxi Normal University, Guilin 541004, China
- School of Computer Science and Engineering, Guangxi Normal University, Guilin 541004, China
| | - Jiafei Liu
- Key Lab of Education Blockchain and Intelligent Technology, Ministry of Education, Guangxi Normal University, Guilin 541004, China
- Guangxi Key Lab of Multi-Source Information Mining & Security, Guangxi Normal University, Guilin 541004, China
- School of Computer Science and Engineering, Guangxi Normal University, Guilin 541004, China
| | - Jinlu Liu
- Key Lab of Education Blockchain and Intelligent Technology, Ministry of Education, Guangxi Normal University, Guilin 541004, China
- Guangxi Key Lab of Multi-Source Information Mining & Security, Guangxi Normal University, Guilin 541004, China
- School of Computer Science and Engineering, Guangxi Normal University, Guilin 541004, China
| | - Junbo Xuan
- Key Lab of Education Blockchain and Intelligent Technology, Ministry of Education, Guangxi Normal University, Guilin 541004, China
- Guangxi Key Lab of Multi-Source Information Mining & Security, Guangxi Normal University, Guilin 541004, China
- School of Computer Science and Engineering, Guangxi Normal University, Guilin 541004, China
| | - Zheng Deng
- Key Lab of Education Blockchain and Intelligent Technology, Ministry of Education, Guangxi Normal University, Guilin 541004, China
- Guangxi Key Lab of Multi-Source Information Mining & Security, Guangxi Normal University, Guilin 541004, China
- School of Computer Science and Engineering, Guangxi Normal University, Guilin 541004, China
| |
Collapse
|
29
|
Karamveer, Uzun Y. Approaches for Benchmarking Single-Cell Gene Regulatory Network Methods. Bioinform Biol Insights 2024; 18:11779322241287120. [PMID: 39502448 PMCID: PMC11536393 DOI: 10.1177/11779322241287120] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2024] [Accepted: 09/10/2024] [Indexed: 11/08/2024] Open
Abstract
Gene regulatory networks are powerful tools for modeling genetic interactions that control the expression of genes driving cell differentiation, and single-cell sequencing offers a unique opportunity to build these networks with high-resolution genomic data. There are many proposed computational methods to build these networks using single-cell data, and different approaches are used to benchmark these methods. However, a comprehensive discussion specifically focusing on benchmarking approaches is missing. In this article, we lay the GRN terminology, present an overview of common gold-standard studies and data sets, and define the performance metrics for benchmarking network construction methodologies. We also point out the advantages and limitations of different benchmarking approaches, suggest alternative ground truth data sets that can be used for benchmarking, and specify additional considerations in this context.
Collapse
Affiliation(s)
- Karamveer
- Department of Pediatrics, The Pennsylvania State University College of Medicine, Hershey, PA, USA
| | - Yasin Uzun
- Department of Pediatrics, The Pennsylvania State University College of Medicine, Hershey, PA, USA
- Department of Biochemistry and Molecular Biology, The Pennsylvania State University College of Medicine, Hershey, PA, USA
- Penn State Cancer Institute, The Pennsylvania State University College of Medicine, Hershey, PA, USA
| |
Collapse
|
30
|
Zhu L, Kang X, Li C, Zheng J. TMELand: An End-to-End Pipeline for Quantification and Visualization of Waddington's Epigenetic Landscape Based on Gene Regulatory Network. IEEE/ACM TRANSACTIONS ON COMPUTATIONAL BIOLOGY AND BIOINFORMATICS 2024; 21:1604-1612. [PMID: 37310837 DOI: 10.1109/tcbb.2023.3285395] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
Waddington's epigenetic landscape is a framework depicting the processes of cell differentiation and reprogramming under the control of a gene regulatory network (GRN). Traditional model-driven methods for landscape quantification focus on the Boolean network or differential equation-based models of GRN, which need sophisticated prior knowledge and hence hamper their practical applications. To resolve this problem, we combine data-driven methods for inferring GRNs from gene expression data with model-driven approach to the landscape mapping. Specifically, we build an end-to-end pipeline to link data-driven and model-driven methods and develop a software tool named TMELand for GRN inference, visualizing Waddington's epigenetic landscape, and calculating state transition paths between attractors to uncover the intrinsic mechanism of cellular transition dynamics. By integrating GRN inference from real transcriptomic data with landscape modeling, TMELand can facilitate studies of computational systems biology, such as predicting cellular states and visualizing the dynamical trends of cell fate determination and transition dynamics from single-cell transcriptomic data.
Collapse
|
31
|
Wang Y, Zheng P, Cheng YC, Wang Z, Aravkin A. WENDY: Covariance dynamics based gene regulatory network inference. Math Biosci 2024; 377:109284. [PMID: 39168402 DOI: 10.1016/j.mbs.2024.109284] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2024] [Revised: 06/25/2024] [Accepted: 08/16/2024] [Indexed: 08/23/2024]
Abstract
Determining gene regulatory network (GRN) structure is a central problem in biology, with a variety of inference methods available for different types of data. For a widely prevalent and challenging use case, namely single-cell gene expression data measured after intervention at multiple time points with unknown joint distributions, there is only one known specifically developed method, which does not fully utilize the rich information contained in this data type. We develop an inference method for the GRN in this case, netWork infErence by covariaNce DYnamics, dubbed WENDY. The core idea of WENDY is to model the dynamics of the covariance matrix, and solve this dynamics as an optimization problem to determine the regulatory relationships. To evaluate its effectiveness, we compare WENDY with other inference methods using synthetic data and experimental data. Our results demonstrate that WENDY performs well across different data sets.
Collapse
Affiliation(s)
- Yue Wang
- Irving Institute for Cancer Dynamics and Department of Statistics, Columbia University, New York, 10027, NY, USA.
| | - Peng Zheng
- Institute for Health Metrics and Evaluation, Seattle, 98195, WA, USA; Department of Health Metrics Sciences, University of Washington, Seattle, 98195, WA, USA
| | - Yu-Chen Cheng
- Department of Data Science, Dana-Farber Cancer Institute, Boston, 02215, MA, USA; Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, 02115, MA, USA; Center for Cancer Evolution, Dana-Farber Cancer Institute, Boston, 02215, MA, USA; Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, 02138, MA, USA
| | - Zikun Wang
- Laboratory of Genetics, The Rockefeller University, New York, 10065, NY, USA
| | - Aleksandr Aravkin
- Department of Applied Mathematics, University of Washington, Seattle, 98195, WA, USA
| |
Collapse
|
32
|
Zhu W, Du Z, Xu Z, Yang D, Chen M, Song Q. SCRN: Single-Cell Gene Regulatory Network Identification in Alzheimer's Disease. IEEE/ACM TRANSACTIONS ON COMPUTATIONAL BIOLOGY AND BIOINFORMATICS 2024; 21:1886-1896. [PMID: 38976461 DOI: 10.1109/tcbb.2024.3424400] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/10/2024]
Abstract
Alzheimer's disease (AD) is the most common neurodegenerative disease, and it consumes considerable medical resources with increasing number of patients every year. Mounting evidence show that the regulatory disruptions altering the intrinsic activity of genes in brain cells contribute to AD pathogenesis. To gain insights into the underlying gene regulation in AD, we proposed a graph learning method, Single-Cell based Regulatory Network (SCRN), to identify the regulatory mechanisms based on single-cell data. SCRN implements the γ-decaying heuristic link prediction based on graph neural networks and can identify reliable gene regulatory networks using locally closed subgraphs. In this work, we first performed UMAP dimension reduction analysis on single-cell RNA sequencing (scRNA-seq) data of AD and normal samples. Then we used SCRN to construct the gene regulatory network based on three well-recognized AD genes (APOE, CX3CR1, and P2RY12). Enrichment analysis of the regulatory network revealed significant pathways including NGF signaling, ERBB2 signaling, and hemostasis. These findings demonstrate the feasibility of using SCRN to uncover potential biomarkers and therapeutic targets related to AD.
Collapse
|
33
|
Dong J, Li J, Wang F. Deep Learning in Gene Regulatory Network Inference: A Survey. IEEE/ACM TRANSACTIONS ON COMPUTATIONAL BIOLOGY AND BIOINFORMATICS 2024; 21:2089-2101. [PMID: 39137088 DOI: 10.1109/tcbb.2024.3442536] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/15/2024]
Abstract
Understanding the intricate regulatory relationships among genes is crucial for comprehending the development, differentiation, and cellular response in living systems. Consequently, inferring gene regulatory networks (GRNs) based on observed data has gained significant attention as a fundamental goal in biological applications. The proliferation and diversification of available data present both opportunities and challenges in accurately inferring GRNs. Deep learning, a highly successful technique in various domains, holds promise in aiding GRN inference. Several GRN inference methods employing deep learning models have been proposed; however, the selection of an appropriate method remains a challenge for life scientists. In this survey, we provide a comprehensive analysis of 12 GRN inference methods that leverage deep learning models. We trace the evolution of these major methods and categorize them based on the types of applicable data. We delve into the core concepts and specific steps of each method, offering a detailed evaluation of their effectiveness and scalability across different scenarios. These insights enable us to make informed recommendations. Moreover, we explore the challenges faced by GRN inference methods utilizing deep learning and discuss future directions, providing valuable suggestions for the advancement of data scientists in this field.
Collapse
|
34
|
Cingiz MÖ. k- Strong Inference Algorithm: A Hybrid Information Theory Based Gene Network Inference Algorithm. Mol Biotechnol 2024; 66:3213-3225. [PMID: 37950851 DOI: 10.1007/s12033-023-00929-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2023] [Accepted: 10/05/2023] [Indexed: 11/13/2023]
Abstract
Gene networks allow researchers to understand the underlying mechanisms between diseases and genes while reducing the need for wet lab experiments. Numerous gene network inference (GNI) algorithms have been presented in the literature to infer accurate gene networks. We proposed a hybrid GNI algorithm, k-Strong Inference Algorithm (ksia), to infer more reliable and robust gene networks from omics datasets. To increase reliability, ksia integrates Pearson correlation coefficient (PCC) and Spearman rank correlation coefficient (SCC) scores to determine mutual information scores between molecules to increase diversity of relation predictions. To infer a more robust gene network, ksia applies three different elimination steps to remove redundant and spurious relations between genes. The performance of ksia was evaluated on microbe microarrays database in the overlap analysis with other GNI algorithms, namely ARACNE, C3NET, CLR, and MRNET. Ksia inferred less number of relations due to its strict elimination steps. However, ksia generally performed better on Escherichia coli (E.coli) and Saccharomyces cerevisiae (yeast) gene expression datasets due to F- measure and precision values. The integration of association estimator scores and three elimination stages slightly increases the performance of ksia based gene networks. Users can access ksia R package and user manual of package via https://github.com/ozgurcingiz/ksia .
Collapse
Affiliation(s)
- Mustafa Özgür Cingiz
- Computer Engineering Department, Faculty of Engineering and Natural Sciences, Bursa Technical University, Mimar Sinan Campus, Yildirim, 16310, Bursa, Turkey.
| |
Collapse
|
35
|
Wang W, Wang Y, Lyu R, Grün D. Scalable identification of lineage-specific gene regulatory networks from metacells with NetID. Genome Biol 2024; 25:275. [PMID: 39425176 PMCID: PMC11488259 DOI: 10.1186/s13059-024-03418-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2023] [Accepted: 10/08/2024] [Indexed: 10/21/2024] Open
Abstract
The identification of gene regulatory networks (GRNs) is crucial for understanding cellular differentiation. Single-cell RNA sequencing data encode gene-level covariations at high resolution, yet data sparsity and high dimensionality hamper accurate and scalable GRN reconstruction. To overcome these challenges, we introduce NetID leveraging homogenous metacells while avoiding spurious gene-gene correlations. Benchmarking demonstrates superior performance of NetID compared to imputation-based methods. By incorporating cell fate probability information, NetID facilitates the prediction of lineage-specific GRNs and recovers known network motifs governing bone marrow hematopoiesis, making it a powerful toolkit for deciphering gene regulatory control of cellular differentiation from large-scale single-cell transcriptome data.
Collapse
Affiliation(s)
- Weixu Wang
- Human Phenome Institute, Fudan University, Shanghai, China
- Institute of Computational Biology, Helmholtz Center Munich, Munich, Germany
| | - Yichen Wang
- Cancer, Ageing and Somatic Mutation, Wellcome Sanger Institute, Hinxton, UK
| | - Ruiqi Lyu
- School of Computer Science, Carnegie Mellon University, Pittsburgh, USA
| | - Dominic Grün
- Würzburg Institute of Systems Immunology, Julius-Maximilians-Universität Würzburg, Würzburg, Germany.
- CAIDAS - Center for Artificial Intelligence and Data Science, Würzburg, Germany.
| |
Collapse
|
36
|
Graham J, Zhang Y, He L, Gonzalez-Fernandez T. CRISPR-GEM: A Novel Machine Learning Model for CRISPR Genetic Target Discovery and Evaluation. ACS Synth Biol 2024; 13:3413-3429. [PMID: 39375864 PMCID: PMC11494708 DOI: 10.1021/acssynbio.4c00473] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2024] [Revised: 09/17/2024] [Accepted: 09/27/2024] [Indexed: 10/09/2024]
Abstract
CRISPR gene editing strategies are shaping cell therapies through precise and tunable control over gene expression. However, limitations in safely delivering high quantities of CRISPR machinery demand careful target gene selection to achieve reliable therapeutic effects. Informed target gene selection requires a thorough understanding of the involvement of target genes in gene regulatory networks (GRNs) and thus their impact on cell phenotype. Effective decoding of these complex networks has been achieved using machine learning models, but current techniques are limited to single cell types and focus mainly on transcription factors, limiting their applicability to CRISPR strategies. To address this, we present CRISPR-GEM, a multilayer perceptron (MLP) based synthetic GRN constructed to accurately predict the downstream effects of CRISPR gene editing. First, input and output nodes are identified as differentially expressed genes between defined experimental and target cell/tissue types, respectively. Then, MLP training learns regulatory relationships in a black-box approach allowing accurate prediction of output gene expression using only input gene expression. Finally, CRISPR-mimetic perturbations are made to each input gene individually, and the resulting model predictions are compared to those for the target group to score and assess each input gene as a CRISPR candidate. The top scoring genes provided by CRISPR-GEM therefore best modulate experimental group GRNs to motivate transcriptomic shifts toward a target group phenotype. This machine learning model is the first of its kind for predicting optimal CRISPR target genes and serves as a powerful tool for enhanced CRISPR strategies across a range of cell therapies.
Collapse
Affiliation(s)
- Joshua
P. Graham
- Department
of Bioengineering, Lehigh University, Bethlehem, Pennsylvania 18015, United States
| | - Yu Zhang
- Department
of Bioengineering, Lehigh University, Bethlehem, Pennsylvania 18015, United States
- Department
of Electrical and Computer Engineering, Lehigh University, Bethlehem, Pennsylvania 18015, United States
| | - Lifang He
- Department
of Computer Science and Engineering, Lehigh
University, Bethlehem, Pennsylvania 18015, United States
| | | |
Collapse
|
37
|
Yuan L, Zhao L, Jiang Y, Shen Z, Zhang Q, Zhang M, Zheng CH, Huang DS. scMGATGRN: a multiview graph attention network-based method for inferring gene regulatory networks from single-cell transcriptomic data. Brief Bioinform 2024; 25:bbae526. [PMID: 39417321 PMCID: PMC11484520 DOI: 10.1093/bib/bbae526] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2024] [Revised: 07/09/2024] [Accepted: 10/03/2024] [Indexed: 10/19/2024] Open
Abstract
The gene regulatory network (GRN) plays a vital role in understanding the structure and dynamics of cellular systems, revealing complex regulatory relationships, and exploring disease mechanisms. Recently, deep learning (DL)-based methods have been proposed to infer GRNs from single-cell transcriptomic data and achieved impressive performance. However, these methods do not fully utilize graph topological information and high-order neighbor information from multiple receptive fields. To overcome those limitations, we propose a novel model based on multiview graph attention network, namely, scMGATGRN, to infer GRNs. scMGATGRN mainly consists of GAT, multiview, and view-level attention mechanism. GAT can extract essential features of the gene regulatory network. The multiview model can simultaneously utilize local feature information and high-order neighbor feature information of nodes in the gene regulatory network. The view-level attention mechanism dynamically adjusts the relative importance of node embedding representations and efficiently aggregates node embedding representations from two views. To verify the effectiveness of scMGATGRN, we compared its performance with 10 methods (five shallow learning algorithms and five state-of-the-art DL-based methods) on seven benchmark single-cell RNA sequencing (scRNA-seq) datasets from five cell lines (two in human and three in mouse) with four different kinds of ground-truth networks. The experimental results not only show that scMGATGRN outperforms competing methods but also demonstrate the potential of this model in inferring GRNs. The code and data of scMGATGRN are made freely available on GitHub (https://github.com/nathanyl/scMGATGRN).
Collapse
Affiliation(s)
- Lin Yuan
- Key Laboratory of Computing Power Network and Information Security, Ministry of Education, Shandong Computer Science Center, Qilu University of Technology (Shandong Academy of Sciences), 3501 Daxue Road, 250353, Shandong, China
- Shandong Engineering Research Center of Big Data Applied Technology, Faculty of Computer Science and Technology, Qilu University of Technology (Shandong Academy of Sciences), 3501 Daxue Road, 250353, Shandong, China
- Shandong Provincial Key Laboratory of Industrial Network and Information System Security, Shandong Fundamental Research Center for Computer Science, 3501 Daxue Road, 250353, Shandong, China
| | - Ling Zhao
- Key Laboratory of Computing Power Network and Information Security, Ministry of Education, Shandong Computer Science Center, Qilu University of Technology (Shandong Academy of Sciences), 3501 Daxue Road, 250353, Shandong, China
- Shandong Engineering Research Center of Big Data Applied Technology, Faculty of Computer Science and Technology, Qilu University of Technology (Shandong Academy of Sciences), 3501 Daxue Road, 250353, Shandong, China
- Shandong Provincial Key Laboratory of Industrial Network and Information System Security, Shandong Fundamental Research Center for Computer Science, 3501 Daxue Road, 250353, Shandong, China
| | - Yufeng Jiang
- Key Laboratory of Computing Power Network and Information Security, Ministry of Education, Shandong Computer Science Center, Qilu University of Technology (Shandong Academy of Sciences), 3501 Daxue Road, 250353, Shandong, China
- Shandong Engineering Research Center of Big Data Applied Technology, Faculty of Computer Science and Technology, Qilu University of Technology (Shandong Academy of Sciences), 3501 Daxue Road, 250353, Shandong, China
- Shandong Provincial Key Laboratory of Industrial Network and Information System Security, Shandong Fundamental Research Center for Computer Science, 3501 Daxue Road, 250353, Shandong, China
| | - Zhen Shen
- School of Computer and Software, Nanyang Institute of Technology, 80 Changjiang Road, 473004, Henan, China
| | - Qinhu Zhang
- Ningbo Institute of Digital Twin, Eastern Institute of Technology, 568 Tongxin Road, 315201, Zhejiang, China
| | - Ming Zhang
- Department of Pediatrics, Zhongshan Hospital Xiamen University, 201 Hubinnan Road, 361004, Fujian, China
| | - Chun-Hou Zheng
- Key Lab of Intelligent Computing and Signal Processing of Ministry of Education, School of Artificial Intelligence, Anhui University, 111 Jiulong Road, 230601, Anhui, China
| | - De-Shuang Huang
- Ningbo Institute of Digital Twin, Eastern Institute of Technology, 568 Tongxin Road, 315201, Zhejiang, China
- Institute for Regenerative Medicine, Medical Innovation Center and State Key Laboratory of Cardiology, Shanghai East Hospital, School of Life Sciences and Technology, Tongji University, 1239 Siping Road, 200123, Shanghai, China
| |
Collapse
|
38
|
Zhao W, Larschan E, Sandstede B, Singh R. Optimal transport reveals dynamic gene regulatory networks via gene velocity estimation. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.09.12.612590. [PMID: 39345416 PMCID: PMC11429941 DOI: 10.1101/2024.09.12.612590] [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: 10/01/2024]
Abstract
Inferring gene regulatory networks from gene expression data is an important and challenging problem in the biology community. We propose OTVelo, a methodology that takes time-stamped single-cell gene expression data as input and predicts gene regulation across two time points. It is known that the rate of change of gene expression, which we will refer to as gene velocity, provides crucial information that enhances such inference; however, this information is not always available due to the limitations in sequencing depth. Our algorithm overcomes this limitation by estimating gene velocities using optimal transport. We then infer gene regulation using time-lagged correlation and Granger causality via regularized linear regression. Instead of providing an aggregated network across all time points, our method uncovers the underlying dynamical mechanism across time points. We validate our algorithm on 13 simulated datasets with both synthetic and curated networks and demonstrate its efficacy on 4 experimental data sets.
Collapse
Affiliation(s)
- Wenjun Zhao
- Division of Applied Mathematics, Brown University, Providence, RI 02912, USA
| | - Erica Larschan
- Department of Molecular Biology, Cell Biology and Biochemistry, Center for Computational Molecular Biology, Brown University, Providence, RI 02912, USA
| | - Björn Sandstede
- Division of Applied Mathematics , Brown University, Providence, RI 02912, USA
| | - Ritambhara Singh
- Department of Computer Science, Center for Computational Molecular Biology, Brown University, Providence, RI 02912, USA
| |
Collapse
|
39
|
Zhang L, Sagan A, Qin B, Kim E, Hu B, Osmanbeyoglu HU. STAN, a computational framework for inferring spatially informed transcription factor activity. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.06.26.600782. [PMID: 38979296 PMCID: PMC11230390 DOI: 10.1101/2024.06.26.600782] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/10/2024]
Abstract
Transcription factors (TFs) drive significant cellular changes in response to environmental cues and intercellular signaling. Neighboring cells influence TF activity and, consequently, cellular fate and function. Spatial transcriptomics (ST) captures mRNA expression patterns across tissue samples, enabling characterization of the local microenvironment. However, these datasets have not been fully leveraged to systematically estimate TF activity governing cell identity. Here, we present STAN ( S patially informed T ranscription factor A ctivity N etwork), a linear mixed-effects computational method that predicts spot-specific, spatially informed TF activities by integrating curated TF-target gene priors, mRNA expression, spatial coordinates, and morphological features from corresponding imaging data. We tested STAN using lymph node, breast cancer, and glioblastoma ST datasets to demonstrate its applicability by identifying TFs associated with specific cell types, spatial domains, pathological regions, and ligand‒receptor pairs. STAN augments the utility of STs to reveal the intricate interplay between TFs and spatial organization across a spectrum of cellular contexts.
Collapse
|
40
|
Zhang J, Larschan E, Bigness J, Singh R. scNODE : generative model for temporal single cell transcriptomic data prediction. Bioinformatics 2024; 40:ii146-ii154. [PMID: 39230694 PMCID: PMC11373355 DOI: 10.1093/bioinformatics/btae393] [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] [Indexed: 09/05/2024] Open
Abstract
SUMMARY Measurement of single-cell gene expression at different timepoints enables the study of cell development. However, due to the resource constraints and technical challenges associated with the single-cell experiments, researchers can only profile gene expression at discrete and sparsely sampled timepoints. This missing timepoint information impedes downstream cell developmental analyses. We propose scNODE, an end-to-end deep learning model that can predict in silico single-cell gene expression at unobserved timepoints. scNODE integrates a variational autoencoder with neural ordinary differential equations to predict gene expression using a continuous and nonlinear latent space. Importantly, we incorporate a dynamic regularization term to learn a latent space that is robust against distribution shifts when predicting single-cell gene expression at unobserved timepoints. Our evaluations on three real-world scRNA-seq datasets show that scNODE achieves higher predictive performance than state-of-the-art methods. We further demonstrate that scNODE's predictions help cell trajectory inference under the missing timepoint paradigm and the learned latent space is useful for in silico perturbation analysis of relevant genes along a developmental cell path. AVAILABILITY AND IMPLEMENTATION The data and code are publicly available at https://github.com/rsinghlab/scNODE.
Collapse
Affiliation(s)
- Jiaqi Zhang
- Department of Computer Science, Brown University, Providence, RI 02906, United States
| | - Erica Larschan
- Center for Computational Molecular Biology, Brown University, Providence, RI 02912, United States
- Department of Molecular Biology, Cell Biology and Biochemistry, Brown University, Providence, RI 02912, United States
| | - Jeremy Bigness
- Center for Computational Molecular Biology, Brown University, Providence, RI 02912, United States
| | - Ritambhara Singh
- Department of Computer Science, Brown University, Providence, RI 02906, United States
- Center for Computational Molecular Biology, Brown University, Providence, RI 02912, United States
| |
Collapse
|
41
|
Loers JU, Vermeirssen V. A single-cell multimodal view on gene regulatory network inference from transcriptomics and chromatin accessibility data. Brief Bioinform 2024; 25:bbae382. [PMID: 39207727 PMCID: PMC11359808 DOI: 10.1093/bib/bbae382] [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: 04/05/2024] [Revised: 06/27/2024] [Accepted: 07/23/2024] [Indexed: 09/04/2024] Open
Abstract
Eukaryotic gene regulation is a combinatorial, dynamic, and quantitative process that plays a vital role in development and disease and can be modeled at a systems level in gene regulatory networks (GRNs). The wealth of multi-omics data measured on the same samples and even on the same cells has lifted the field of GRN inference to the next stage. Combinations of (single-cell) transcriptomics and chromatin accessibility allow the prediction of fine-grained regulatory programs that go beyond mere correlation of transcription factor and target gene expression, with enhancer GRNs (eGRNs) modeling molecular interactions between transcription factors, regulatory elements, and target genes. In this review, we highlight the key components for successful (e)GRN inference from (sc)RNA-seq and (sc)ATAC-seq data exemplified by state-of-the-art methods as well as open challenges and future developments. Moreover, we address preprocessing strategies, metacell generation and computational omics pairing, transcription factor binding site detection, and linear and three-dimensional approaches to identify chromatin interactions as well as dynamic and causal eGRN inference. We believe that the integration of transcriptomics together with epigenomics data at a single-cell level is the new standard for mechanistic network inference, and that it can be further advanced with integrating additional omics layers and spatiotemporal data, as well as with shifting the focus towards more quantitative and causal modeling strategies.
Collapse
Affiliation(s)
- Jens Uwe Loers
- Lab for Computational Biology, Integromics and Gene Regulation (CBIGR), Cancer Research Institute Ghent (CRIG), Corneel Heymanslaan 10, 9000 Ghent, Belgium
- Department of Biomedical Molecular Biology, Ghent University, Zwijnaarde-Technologiepark 71, 9052 Ghent, Belgium
- Department of Biomolecular Medicine, Ghent University, Corneel Heymanslaan 10, 9000 Ghent, Belgium
| | - Vanessa Vermeirssen
- Lab for Computational Biology, Integromics and Gene Regulation (CBIGR), Cancer Research Institute Ghent (CRIG), Corneel Heymanslaan 10, 9000 Ghent, Belgium
- Department of Biomedical Molecular Biology, Ghent University, Zwijnaarde-Technologiepark 71, 9052 Ghent, Belgium
- Department of Biomolecular Medicine, Ghent University, Corneel Heymanslaan 10, 9000 Ghent, Belgium
| |
Collapse
|
42
|
Chang Z, Xu Y, Dong X, Gao Y, Wang C. Single-cell and spatial multiomic inference of gene regulatory networks using SCRIPro. Bioinformatics 2024; 40:btae466. [PMID: 39024032 PMCID: PMC11288411 DOI: 10.1093/bioinformatics/btae466] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2024] [Revised: 06/05/2024] [Accepted: 07/17/2024] [Indexed: 07/20/2024] Open
Abstract
MOTIVATION The burgeoning generation of single-cell or spatial multiomic data allows for the characterization of gene regulation networks (GRNs) at an unprecedented resolution. However, the accurate reconstruction of GRNs from sparse and noisy single-cell or spatial multiomic data remains challenging. RESULTS Here, we present SCRIPro, a comprehensive computational framework that robustly infers GRNs for both single-cell and spatial multi-omics data. SCRIPro first improves sample coverage through a density clustering approach based on multiomic and spatial similarities. Additionally, SCRIPro scans transcriptional regulator (TR) importance by performing chromatin reconstruction and in silico deletion analyses using a comprehensive reference covering 1,292 human and 994 mouse TRs. Finally, SCRIPro combines TR-target importance scores derived from multiomic data with TR-target expression levels to ensure precise GRN reconstruction. We benchmarked SCRIPro on various datasets, including single-cell multiomic data from human B-cell lymphoma, mouse hair follicle development, Stereo-seq of mouse embryos, and Spatial-ATAC-RNA from mouse brain. SCRIPro outperforms existing motif-based methods and accurately reconstructs cell type-specific, stage-specific, and region-specific GRNs. Overall, SCRIPro emerges as a streamlined and fast method capable of reconstructing TR activities and GRNs for both single-cell and spatial multi-omic data. AVAILABILITY SCRIPro is available at https://github.com/wanglabtongji/SCRIPro. SUPPLEMENTARY INFORMATION Supplementary data are available at Bioinformatics online.
Collapse
Affiliation(s)
- Zhanhe Chang
- Key Laboratory of Spine and Spinal Cord Injury Repair and Regeneration of Ministry of Education, Department of Orthopedics, Tongji Hospital, School of Life Sciences and Technology, Tongji University, Shanghai 200092, China
- Frontier Science Center for Stem Cell Research, Tongji University, Shanghai, China
- Institute for Regenerative Medicine, Shanghai East Hospital, Shanghai Key Laboratory of Signaling and Disease Research, School of Life Sciences and Technology, Tongji University, Shanghai, China
| | - Yunfan Xu
- Key Laboratory of Spine and Spinal Cord Injury Repair and Regeneration of Ministry of Education, Department of Orthopedics, Tongji Hospital, School of Life Sciences and Technology, Tongji University, Shanghai 200092, China
- Frontier Science Center for Stem Cell Research, Tongji University, Shanghai, China
| | - Xin Dong
- Key Laboratory of Spine and Spinal Cord Injury Repair and Regeneration of Ministry of Education, Department of Orthopedics, Tongji Hospital, School of Life Sciences and Technology, Tongji University, Shanghai 200092, China
- Frontier Science Center for Stem Cell Research, Tongji University, Shanghai, China
| | - Yawei Gao
- Frontier Science Center for Stem Cell Research, Tongji University, Shanghai, China
- Institute for Regenerative Medicine, Shanghai East Hospital, Shanghai Key Laboratory of Signaling and Disease Research, School of Life Sciences and Technology, Tongji University, Shanghai, China
| | - Chenfei Wang
- Key Laboratory of Spine and Spinal Cord Injury Repair and Regeneration of Ministry of Education, Department of Orthopedics, Tongji Hospital, School of Life Sciences and Technology, Tongji University, Shanghai 200092, China
- Frontier Science Center for Stem Cell Research, Tongji University, Shanghai, China
- National Key Laboratory of Autonomous Intelligent Unmanned Systems, Tongji University, Shanghai 200120, China
- Frontier Science Center for Intelligent Autonomous Systems, Tongji University, Shanghai 200120, China
| |
Collapse
|
43
|
Graham JP, Zhang Y, He L, Gonzalez-Fernandez T. CRISPR-GEM: A Novel Machine Learning Model for CRISPR Genetic Target Discovery and Evaluation. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.07.01.601587. [PMID: 39005295 PMCID: PMC11244939 DOI: 10.1101/2024.07.01.601587] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/16/2024]
Abstract
CRISPR gene editing strategies are shaping cell therapies through precise and tunable control over gene expression. However, achieving reliable therapeutic effects with improved safety and efficacy requires informed target gene selection. This depends on a thorough understanding of the involvement of target genes in gene regulatory networks (GRNs) that regulate cell phenotype and function. Machine learning models have been previously used for GRN reconstruction using RNA-seq data, but current techniques are limited to single cell types and focus mainly on transcription factors. This restriction overlooks many potential CRISPR target genes, such as those encoding extracellular matrix components, growth factors, and signaling molecules, thus limiting the applicability of these models for CRISPR strategies. To address these limitations, we have developed CRISPR-GEM, a multi-layer perceptron (MLP)-based synthetic GRN constructed to accurately predict the downstream effects of CRISPR gene editing. First, input and output nodes are identified as differentially expressed genes between defined experimental and target cell/tissue types respectively. Then, MLP training learns regulatory relationships in a black-box approach allowing accurate prediction of output gene expression using only input gene expression. Finally, CRISPR-mimetic perturbations are made to each input gene individually and the resulting model predictions are compared to those for the target group to score and assess each input gene as a CRISPR candidate. The top scoring genes provided by CRISPR-GEM therefore best modulate experimental group GRNs to motivate transcriptomic shifts towards a target group phenotype. This machine learning model is the first of its kind for predicting optimal CRISPR target genes and serves as a powerful tool for enhanced CRISPR strategies across a range of cell therapies.
Collapse
Affiliation(s)
- Josh P Graham
- Department of Bioengineering, Lehigh University, Bethlehem, PA, USA
| | - Yu Zhang
- Department of Bioengineering, Lehigh University, Bethlehem, PA, USA
- Department of Electrical and Computer Engineering, Lehigh University, Bethlehem, PA, USA
| | - Lifang He
- Department of Computer Science and Engineering, Lehigh University, Bethlehem, PA, USA
| | | |
Collapse
|
44
|
Zhou X, Pan J, Chen L, Zhang S, Chen Y. DeepIMAGER: Deeply Analyzing Gene Regulatory Networks from scRNA-seq Data. Biomolecules 2024; 14:766. [PMID: 39062480 PMCID: PMC11274664 DOI: 10.3390/biom14070766] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2024] [Revised: 06/22/2024] [Accepted: 06/25/2024] [Indexed: 07/28/2024] Open
Abstract
Understanding the dynamics of gene regulatory networks (GRNs) across diverse cell types poses a challenge yet holds immense value in unraveling the molecular mechanisms governing cellular processes. Current computational methods, which rely solely on expression changes from bulk RNA-seq and/or scRNA-seq data, often result in high rates of false positives and low precision. Here, we introduce an advanced computational tool, DeepIMAGER, for inferring cell-specific GRNs through deep learning and data integration. DeepIMAGER employs a supervised approach that transforms the co-expression patterns of gene pairs into image-like representations and leverages transcription factor (TF) binding information for model training. It is trained using comprehensive datasets that encompass scRNA-seq profiles and ChIP-seq data, capturing TF-gene pair information across various cell types. Comprehensive validations on six cell lines show DeepIMAGER exhibits superior performance in ten popular GRN inference tools and has remarkable robustness against dropout-zero events. DeepIMAGER was applied to scRNA-seq datasets of multiple myeloma (MM) and detected potential GRNs for TFs of RORC, MITF, and FOXD2 in MM dendritic cells. This technical innovation, combined with its capability to accurately decode GRNs from scRNA-seq, establishes DeepIMAGER as a valuable tool for unraveling complex regulatory networks in various cell types.
Collapse
Affiliation(s)
- Xiguo Zhou
- College of Computer and Information Engineering, Tianjin Normal University, Tianjin 300387, China; (X.Z.); (J.P.); (L.C.)
| | - Jingyi Pan
- College of Computer and Information Engineering, Tianjin Normal University, Tianjin 300387, China; (X.Z.); (J.P.); (L.C.)
| | - Liang Chen
- College of Computer and Information Engineering, Tianjin Normal University, Tianjin 300387, China; (X.Z.); (J.P.); (L.C.)
| | - Shaoqiang Zhang
- College of Computer and Information Engineering, Tianjin Normal University, Tianjin 300387, China; (X.Z.); (J.P.); (L.C.)
| | - Yong Chen
- Department of Biological and Biomedical Sciences, Rowan University, Glassboro, NJ 08028, USA
| |
Collapse
|
45
|
Huo Q, Song R, Ma Z. Recent advances in exploring transcriptional regulatory landscape of crops. FRONTIERS IN PLANT SCIENCE 2024; 15:1421503. [PMID: 38903438 PMCID: PMC11188431 DOI: 10.3389/fpls.2024.1421503] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 04/22/2024] [Accepted: 05/23/2024] [Indexed: 06/22/2024]
Abstract
Crop breeding entails developing and selecting plant varieties with improved agronomic traits. Modern molecular techniques, such as genome editing, enable more efficient manipulation of plant phenotype by altering the expression of particular regulatory or functional genes. Hence, it is essential to thoroughly comprehend the transcriptional regulatory mechanisms that underpin these traits. In the multi-omics era, a large amount of omics data has been generated for diverse crop species, including genomics, epigenomics, transcriptomics, proteomics, and single-cell omics. The abundant data resources and the emergence of advanced computational tools offer unprecedented opportunities for obtaining a holistic view and profound understanding of the regulatory processes linked to desirable traits. This review focuses on integrated network approaches that utilize multi-omics data to investigate gene expression regulation. Various types of regulatory networks and their inference methods are discussed, focusing on recent advancements in crop plants. The integration of multi-omics data has been proven to be crucial for the construction of high-confidence regulatory networks. With the refinement of these methodologies, they will significantly enhance crop breeding efforts and contribute to global food security.
Collapse
Affiliation(s)
| | | | - Zeyang Ma
- State Key Laboratory of Maize Bio-breeding, Frontiers Science Center for Molecular Design Breeding, Joint International Research Laboratory of Crop Molecular Breeding, National Maize Improvement Center, College of Agronomy and Biotechnology, China Agricultural University, Beijing, China
| |
Collapse
|
46
|
Peng D, Cahan P. OneSC: A computational platform for recapitulating cell state transitions. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.05.31.596831. [PMID: 38895453 PMCID: PMC11185539 DOI: 10.1101/2024.05.31.596831] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/21/2024]
Abstract
Computational modelling of cell state transitions has been a great interest of many in the field of developmental biology, cancer biology and cell fate engineering because it enables performing perturbation experiments in silico more rapidly and cheaply than could be achieved in a wet lab. Recent advancements in single-cell RNA sequencing (scRNA-seq) allow the capture of high-resolution snapshots of cell states as they transition along temporal trajectories. Using these high-throughput datasets, we can train computational models to generate in silico 'synthetic' cells that faithfully mimic the temporal trajectories. Here we present OneSC, a platform that can simulate synthetic cells across developmental trajectories using systems of stochastic differential equations govern by a core transcription factors (TFs) regulatory network. Different from the current network inference methods, OneSC prioritizes on generating Boolean network that produces faithful cell state transitions and steady cell states that mimic real biological systems. Applying OneSC to real data, we inferred a core TF network using a mouse myeloid progenitor scRNA-seq dataset and showed that the dynamical simulations of that network generate synthetic single-cell expression profiles that faithfully recapitulate the four myeloid differentiation trajectories going into differentiated cell states (erythrocytes, megakaryocytes, granulocytes and monocytes). Finally, through the in-silico perturbations of the mouse myeloid progenitor core network, we showed that OneSC can accurately predict cell fate decision biases of TF perturbations that closely match with previous experimental observations.
Collapse
Affiliation(s)
- Da Peng
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, Maryland, 21205, USA
| | - Patrick Cahan
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, Maryland, 21205, USA
- Institute for Cell Engineering, Johns Hopkins University, Baltimore, Maryland, 21205, USA
- Department of Molecular Biology and Genetics, Johns Hopkins University, Baltimore, Maryland, 21205, USA
| |
Collapse
|
47
|
Wu S, Jin K, Tang M, Xia Y, Gao W. Inference of Gene Regulatory Networks Based on Multi-view Hierarchical Hypergraphs. Interdiscip Sci 2024; 16:318-332. [PMID: 38342857 DOI: 10.1007/s12539-024-00604-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2023] [Revised: 11/26/2023] [Accepted: 01/03/2024] [Indexed: 02/13/2024]
Abstract
Since gene regulation is a complex process in which multiple genes act simultaneously, accurately inferring gene regulatory networks (GRNs) is a long-standing challenge in systems biology. Although graph neural networks can formally describe intricate gene expression mechanisms, current GRN inference methods based on graph learning regard only transcription factor (TF)-target gene interactions as pairwise relationships, and cannot model the many-to-many high-order regulatory patterns that prevail among genes. Moreover, these methods often rely on limited prior regulatory knowledge, ignoring the structural information of GRNs in gene expression profiles. Therefore, we propose a multi-view hierarchical hypergraphs GRN (MHHGRN) inference model. Specifically, multiple heterogeneous biological information is integrated to construct multi-view hierarchical hypergraphs of TFs and target genes, using hypergraph convolution networks to model higher order complex regulatory relationships. Meanwhile, the coupled information diffusion mechanism and the cross-domain messaging mechanism facilitate the information sharing between genes to optimise gene embedding representations. Finally, a unique channel attention mechanism is used to adaptively learn feature representations from multiple views for GRN inference. Experimental results show that MHHGRN achieves better results than the baseline methods on the E. coli and S. cerevisiae benchmark datasets of the DREAM5 challenge, and it has excellent cross-species generalization, achieving comparable or better performance on scRNA-seq datasets from five mouse and two human cell lines.
Collapse
Affiliation(s)
- Songyang Wu
- School of Information Science and Technology, Yunnan Normal University, Kunming, 650500, China
| | - Kui Jin
- School of Information Science and Technology, Yunnan Normal University, Kunming, 650500, China
| | - Mingjing Tang
- School of Life Science, Yunnan Normal University, Kunming, 650500, China.
- Engineering Research Center of Sustainable Development and Utilization of Biomass Energy, Ministry of Education, Yunnan Normal University, Kunming, 650500, China.
| | - Yuelong Xia
- School of Information Science and Technology, Yunnan Normal University, Kunming, 650500, China
| | - Wei Gao
- School of Information Science and Technology, Yunnan Normal University, Kunming, 650500, China
| |
Collapse
|
48
|
Wan R, Zhang Y, Peng Y, Tian F, Gao G, Tang F, Jia J, Ge H. Unveiling gene regulatory networks during cellular state transitions without linkage across time points. Sci Rep 2024; 14:12355. [PMID: 38811747 PMCID: PMC11137113 DOI: 10.1038/s41598-024-62850-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2024] [Accepted: 05/22/2024] [Indexed: 05/31/2024] Open
Abstract
Time-stamped cross-sectional data, which lack linkage across time points, are commonly generated in single-cell transcriptional profiling. Many previous methods for inferring gene regulatory networks (GRNs) driving cell-state transitions relied on constructing single-cell temporal ordering. Introducing COSLIR (COvariance restricted Sparse LInear Regression), we presented a direct approach to reconstructing GRNs that govern cell-state transitions, utilizing only the first and second moments of samples between two consecutive time points. Simulations validated COSLIR's perfect accuracy in the oracle case and demonstrated its robust performance in real-world scenarios. When applied to single-cell RT-PCR and RNAseq datasets in developmental biology, COSLIR competed favorably with existing methods. Notably, its running time remained nearly independent of the number of cells. Therefore, COSLIR emerges as a promising addition to GRN reconstruction methods under cell-state transitions, bypassing the single-cell temporal ordering to enhance accuracy and efficiency in single-cell transcriptional profiling.
Collapse
Affiliation(s)
- Ruosi Wan
- Beijing International Center for Mathematical Research, Peking University, Beijing, China
| | - Yuhao Zhang
- Biomedical Pioneering Innovation Center, Peking University, Beijing, China
| | - Yongli Peng
- Beijing International Center for Mathematical Research, Peking University, Beijing, China
| | - Feng Tian
- Biomedical Pioneering Innovation Center, Peking University, Beijing, China
| | - Ge Gao
- Biomedical Pioneering Innovation Center, Peking University, Beijing, China
- Beijing Advanced Innovation Center for Genomics, Peking University, Beijing, China
| | - Fuchou Tang
- Biomedical Pioneering Innovation Center, Peking University, Beijing, China
- Beijing Advanced Innovation Center for Genomics, Peking University, Beijing, China
| | - Jinzhu Jia
- School of Public Health and Center for Statistical Science, Peking University, Beijing, China.
| | - Hao Ge
- Beijing International Center for Mathematical Research, Peking University, Beijing, China.
- Biomedical Pioneering Innovation Center, Peking University, Beijing, China.
| |
Collapse
|
49
|
Lei Y, Huang XT, Guo X, Hang Katie Chan K, Gao L. DeepGRNCS: deep learning-based framework for jointly inferring gene regulatory networks across cell subpopulations. Brief Bioinform 2024; 25:bbae334. [PMID: 38980373 PMCID: PMC11232306 DOI: 10.1093/bib/bbae334] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2024] [Revised: 06/03/2024] [Accepted: 07/01/2024] [Indexed: 07/10/2024] Open
Abstract
Inferring gene regulatory networks (GRNs) allows us to obtain a deeper understanding of cellular function and disease pathogenesis. Recent advances in single-cell RNA sequencing (scRNA-seq) technology have improved the accuracy of GRN inference. However, many methods for inferring individual GRNs from scRNA-seq data are limited because they overlook intercellular heterogeneity and similarities between different cell subpopulations, which are often present in the data. Here, we propose a deep learning-based framework, DeepGRNCS, for jointly inferring GRNs across cell subpopulations. We follow the commonly accepted hypothesis that the expression of a target gene can be predicted based on the expression of transcription factors (TFs) due to underlying regulatory relationships. We initially processed scRNA-seq data by discretizing data scattering using the equal-width method. Then, we trained deep learning models to predict target gene expression from TFs. By individually removing each TF from the expression matrix, we used pre-trained deep model predictions to infer regulatory relationships between TFs and genes, thereby constructing the GRN. Our method outperforms existing GRN inference methods for various simulated and real scRNA-seq datasets. Finally, we applied DeepGRNCS to non-small cell lung cancer scRNA-seq data to identify key genes in each cell subpopulation and analyzed their biological relevance. In conclusion, DeepGRNCS effectively predicts cell subpopulation-specific GRNs. The source code is available at https://github.com/Nastume777/DeepGRNCS.
Collapse
Affiliation(s)
- Yahui Lei
- School of Computer Science and Technology, Xidian University, Xi’an 710071, Shaanxi, China
| | - Xiao-Tai Huang
- School of Computer Science and Technology, Xidian University, Xi’an 710071, Shaanxi, China
| | - Xingli Guo
- School of Computer Science and Technology, Xidian University, Xi’an 710071, Shaanxi, China
| | - Kei Hang Katie Chan
- Department of Electrical Engineering, City University of Hong Kong, Hong Kong SAR, China
- Department of Biomedical Sciences, City University of Hong Kong, Hong Kong SAR, China
- Department of Epidemiology and Center for Global Cardiometabolic Health, Brown University, Providence, RI, United States
| | - Lin Gao
- School of Computer Science and Technology, Xidian University, Xi’an 710071, Shaanxi, China
| |
Collapse
|
50
|
Singh R, Wu AP, Mudide A, Berger B. Causal gene regulatory analysis with RNA velocity reveals an interplay between slow and fast transcription factors. Cell Syst 2024; 15:462-474.e5. [PMID: 38754366 DOI: 10.1016/j.cels.2024.04.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2023] [Revised: 08/25/2023] [Accepted: 04/18/2024] [Indexed: 05/18/2024]
Abstract
Single-cell expression dynamics, from differentiation trajectories or RNA velocity, have the potential to reveal causal links between transcription factors (TFs) and their target genes in gene regulatory networks (GRNs). However, existing methods either overlook these expression dynamics or necessitate that cells be ordered along a linear pseudotemporal axis, which is incompatible with branching trajectories. We introduce Velorama, an approach to causal GRN inference that represents single-cell differentiation dynamics as a directed acyclic graph of cells, constructed from pseudotime or RNA velocity measurements. Additionally, Velorama enables the estimation of the speed at which TFs influence target genes. Applying Velorama, we uncover evidence that the speed of a TF's interactions is tied to its regulatory function. For human corticogenesis, we find that slow TFs are linked to gliomas, while fast TFs are associated with neuropsychiatric diseases. We expect Velorama to become a critical part of the RNA velocity toolkit for investigating the causal drivers of differentiation and disease.
Collapse
Affiliation(s)
- Rohit Singh
- Computer Science and Artificial Intelligence Laboratory, MIT, Cambridge, MA 02139, USA.
| | - Alexander P Wu
- Computer Science and Artificial Intelligence Laboratory, MIT, Cambridge, MA 02139, USA
| | - Anish Mudide
- Phillips Exeter Academy, Exeter, NH 03883, USA; Computer Science and Artificial Intelligence Laboratory and Department of Mathematics, MIT, Cambridge, MA 02139, USA
| | - Bonnie Berger
- Computer Science and Artificial Intelligence Laboratory and Department of Mathematics, MIT, Cambridge, MA 02139, USA.
| |
Collapse
|