1
|
Arya A, Tripathi P, Dubey N, Aier I, Kumar Varadwaj P. Navigating single-cell RNA-sequencing: protocols, tools, databases, and applications. Genomics Inform 2025; 23:13. [PMID: 40382658 DOI: 10.1186/s44342-025-00044-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2025] [Accepted: 04/07/2025] [Indexed: 05/20/2025] Open
Abstract
Single-cell RNA-sequencing (scRNA-seq) technology brought about a revolutionary change in the transcriptomic world, paving the way for comprehensive analysis of cellular heterogeneity in complex biological systems. It enabled researchers to see how different cells behaved at single-cell levels, providing new insights into the process. However, despite all these advancements, scRNA-seq also experiences challenges related to the complexity of data analysis, interpretation, and multi-omics data integration. In this review, these complications were discussed in detail, directly pointing at the optimization of scRNA-seq approaches and understanding the world of single-cell and its dynamics. Different protocols and currently functional single-cell databases were also covered. This review highlights different tools for the analysis of scRNA-seq and their methodologies, emphasizing innovative techniques that enhance resolution and accuracy at a single-cell level. Various applications were explored across domains including drug discovery, tumor microenvironment (TME), biomarker discovery, and microbial profiling, and case studies were discussed to explain the importance of scRNA-seq by uncovering novel and rare cell types and their identification. This review underlines a crucial aspect of scRNA-seq in the advancement of personalized medicine and highlights its potential to understand the complexity of biological systems.
Collapse
Affiliation(s)
- Ankish Arya
- Department of Applied Sciences, Indian Institute of Information Technology Allahabad, Jhalwa, Prayagraj, 211015, Uttar Pradesh, India
| | - Prabhat Tripathi
- Department of Applied Sciences, Indian Institute of Information Technology Allahabad, Jhalwa, Prayagraj, 211015, Uttar Pradesh, India
| | - Nidhi Dubey
- Department of Applied Sciences, Indian Institute of Information Technology Allahabad, Jhalwa, Prayagraj, 211015, Uttar Pradesh, India
| | - Imlimaong Aier
- Department of Applied Sciences, Indian Institute of Information Technology Allahabad, Jhalwa, Prayagraj, 211015, Uttar Pradesh, India
| | - Pritish Kumar Varadwaj
- Department of Applied Sciences, Indian Institute of Information Technology Allahabad, Jhalwa, Prayagraj, 211015, Uttar Pradesh, India.
| |
Collapse
|
2
|
Gaspard-Boulinc LC, Gortana L, Walter T, Barillot E, Cavalli FMG. Cell-type deconvolution methods for spatial transcriptomics. Nat Rev Genet 2025:10.1038/s41576-025-00845-y. [PMID: 40369312 DOI: 10.1038/s41576-025-00845-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/14/2025] [Indexed: 05/16/2025]
Abstract
Spatial transcriptomics is a powerful method for studying the spatial organization of cells, which is a critical feature in the development, function and evolution of multicellular life. However, sequencing-based spatial transcriptomics has not yet achieved cellular-level resolution, so advanced deconvolution methods are needed to infer cell-type contributions at each location in the data. Recent progress has led to diverse tools for cell-type deconvolution that are helping to describe tissue architectures in health and disease. In this Review, we describe the varied types of cell-type deconvolution methods for spatial transcriptomics, contrast their capabilities and summarize them in a web-based, interactive table to enable more efficient method selection.
Collapse
Affiliation(s)
- Lucie C Gaspard-Boulinc
- Institut Curie, PSL University, Paris, France
- Institut National de la Santé et de la Recherche Médicale (INSERM), U1331, Paris, France
- Mines Paris, PSL University, CBIO - Centre for Computational Biology, Paris, France
| | - Luca Gortana
- Institut Curie, PSL University, Paris, France
- Institut National de la Santé et de la Recherche Médicale (INSERM), U1331, Paris, France
- Mines Paris, PSL University, CBIO - Centre for Computational Biology, Paris, France
| | - Thomas Walter
- Institut Curie, PSL University, Paris, France
- Institut National de la Santé et de la Recherche Médicale (INSERM), U1331, Paris, France
- Mines Paris, PSL University, CBIO - Centre for Computational Biology, Paris, France
| | - Emmanuel Barillot
- Institut Curie, PSL University, Paris, France
- Institut National de la Santé et de la Recherche Médicale (INSERM), U1331, Paris, France
- Mines Paris, PSL University, CBIO - Centre for Computational Biology, Paris, France
| | - Florence M G Cavalli
- Institut Curie, PSL University, Paris, France.
- Institut National de la Santé et de la Recherche Médicale (INSERM), U1331, Paris, France.
- Mines Paris, PSL University, CBIO - Centre for Computational Biology, Paris, France.
| |
Collapse
|
3
|
Ramirez A, Orcutt-Jahns BT, Pascoe S, Abraham A, Remigio B, Thomas N, Meyer AS. Integrative, high-resolution analysis of single-cell gene expression across experimental conditions with PARAFAC2-RISE. Cell Syst 2025:101294. [PMID: 40378843 DOI: 10.1016/j.cels.2025.101294] [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: 08/26/2024] [Revised: 02/20/2025] [Accepted: 04/22/2025] [Indexed: 05/19/2025]
Abstract
Effective exploration and analysis tools are vital for the extraction of insights from single-cell data. However, current techniques for modeling single-cell studies performed across experimental conditions (e.g., samples) require restrictive assumptions or do not adequately deconvolute condition-to-condition variation from cell-to-cell variation. Here, we report that reduction and insight in single-cell exploration (RISE), an adaptation of the tensor decomposition method PARAFAC2, enables the dimensionality reduction and analysis of single-cell data across conditions. We demonstrate the benefits of RISE across distinct examples of single-cell RNA-sequencing experiments of peripheral immune cells: pharmacologic drug perturbations and systemic lupus erythematosus patient samples. RISE enables associations of gene variation patterns with patients or perturbations while connecting each coordinated change to single cells without requiring cell-type annotations. The theoretical grounding of RISE suggests a unified framework for many single-cell data modeling tasks while providing an intuitive dimensionality reduction approach for multi-sample single-cell studies across biological contexts. A record of this paper's transparent peer review process is included in the supplemental information.
Collapse
Affiliation(s)
- Andrew Ramirez
- Department of Bioengineering, University of California, Los Angeles (UCLA), Los Angeles, CA 90095, USA
| | - Brian T Orcutt-Jahns
- Department of Bioengineering, University of California, Los Angeles (UCLA), Los Angeles, CA 90095, USA
| | - Sean Pascoe
- Department of Bioengineering, University of California, Los Angeles (UCLA), Los Angeles, CA 90095, USA; Department of Molecular Biosciences, Northwestern University, Evanston, IL 60208, USA
| | - Armaan Abraham
- Department of Bioengineering, University of California, Los Angeles (UCLA), Los Angeles, CA 90095, USA
| | - Breanna Remigio
- Computational and Systems Biology, UCLA, Los Angeles, CA 90095, USA
| | - Nathaniel Thomas
- Department of Computer Science, UCLA, Los Angeles, CA 90095, USA
| | - Aaron S Meyer
- Department of Bioengineering, University of California, Los Angeles (UCLA), Los Angeles, CA 90095, USA; Jonsson Comprehensive Cancer Center, UCLA, Los Angeles, CA 90095, USA; Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, UCLA, Los Angeles, CA 90095, USA.
| |
Collapse
|
4
|
Zeng J, Zhou H, Wan H, Yang J. Single-cell omics: moving towards a new era in ischemic stroke research. Eur J Pharmacol 2025; 1000:177725. [PMID: 40350018 DOI: 10.1016/j.ejphar.2025.177725] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2024] [Revised: 05/08/2025] [Accepted: 05/09/2025] [Indexed: 05/14/2025]
Abstract
Ischemic stroke (IS) is a highly complex and heterogeneous disease involving multiple pathophysiological events. A better understanding of the pathophysiology of IS will enhance preventive, diagnostic and therapeutic strategies. Despite significant advances in modern medicine, the molecular mechanisms of IS are still largely unknown. The high-throughput omics approach opens new avenues for identifying IS biomarkers and elucidating disease pathogenesis mechanisms. Single-cell omics enables a more thorough and in-depth analysis of the cellular interactions and properties in IS. This will lead to a better understanding of the onset, treatment and prognosis of IS. In this paper, we first reviewed the disease signatures and mechanisms research of IS. Subsequently, the use of single-cell omics to comprehend the mechanisms of IS was discussed, along with some recent developments in the field. To further delineate the upstream pathogenic alterations and downstream molecular impacts of IS, we also discussed the current use of machine learning approaches to single-cell omics data analysis. Particularly, single-cell omics is being used to inform risk assessment, early patient diagnosis and treatment strategies, and their potential impact on precision medicine. Thus, we summarized the role of single-cell omics in precision medicine. Despite the relative youth of the field, the development of single-cell omics promises to provide a powerful tool for elucidating the pathogenesis of IS.
Collapse
Affiliation(s)
- Jieqiong Zeng
- School of Basic Medicine Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, China; School of Ecological and Environmental, Hubei Industrial Polytechnic, Shiyan, 442000, China
| | - Huifen Zhou
- School of Basic Medicine Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, China
| | - Haitong Wan
- School of Basic Medicine Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, China.
| | - Jiehong Yang
- School of Basic Medicine Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, China.
| |
Collapse
|
5
|
Zhou Y, Sheng Q, Jin S. Integrating single-cell data with biological variables. Proc Natl Acad Sci U S A 2025; 122:e2416516122. [PMID: 40294274 PMCID: PMC12067276 DOI: 10.1073/pnas.2416516122] [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: 03/30/2025] [Indexed: 04/30/2025] Open
Abstract
Constructing single-cell atlases requires preserving differences attributable to biological variables, such as cell types, tissue origins, and disease states, while eliminating batch effects. However, existing methods are inadequate in explicitly modeling these biological variables. Here, we introduce SIGNAL, a general framework that leverages biological variables to disentangle biological and technical effects, thereby linking these metadata to data integration. SIGNAL employs a variant of principal component analysis to align multiple batches, enabling the integration of 1 million cells in approximately 2 min. SIGNAL, despite its computational simplicity, surpasses state-of-the-art methods across multiple integration scenarios: 1) heterogeneous datasets, 2) cross-species datasets, 3) simulated datasets, 4) integration on low-quality cell annotations, and 5) reference-based integration. Furthermore, we demonstrate that SIGNAL accurately transfers knowledge from reference to query datasets. Notably, we propose a self-adjustment strategy to restore annotated cell labels potentially distorted during integration. Finally, we apply SIGNAL to multiple large-scale atlases, including a human heart cell atlas containing 2.7 million cells, identifying tissue- and developmental stage-specific subtypes, as well as condition-specific cell states. This underscores SIGNAL's exceptional capability in multiscale analysis.
Collapse
Affiliation(s)
- Yang Zhou
- School of Mathematics, Harbin Institute of Technology, Harbin150001, China
- Zhengzhou Research Institute, Harbin Institute of Technology, Zhengzhou450000, China
| | - Qiongyu Sheng
- School of Mathematics, Harbin Institute of Technology, Harbin150001, China
- Zhengzhou Research Institute, Harbin Institute of Technology, Zhengzhou450000, China
| | - Shuilin Jin
- School of Mathematics, Harbin Institute of Technology, Harbin150001, China
- Zhengzhou Research Institute, Harbin Institute of Technology, Zhengzhou450000, China
| |
Collapse
|
6
|
Leduc A, Xu Y, Shipkovenska G, Dou Z, Slavov N. Limiting the impact of protein leakage in single-cell proteomics. Nat Commun 2025; 16:4169. [PMID: 40324992 PMCID: PMC12053607 DOI: 10.1038/s41467-025-56736-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2024] [Accepted: 01/28/2025] [Indexed: 05/07/2025] Open
Abstract
Limiting artifacts during sample preparation can significantly increase data quality in single-cell proteomics experiments. Towards this goal, we characterize the impact of protein leakage by analyzing thousands of primary single cells from mouse trachea. The cells were prepared either fresh immediately after dissociation or first cryopreserved and prepared at a later date. We directly identify permeabilized cells by imaging a cell permeable dye and use the data to define a signature for protein leakage. This signature is similar across diverse cell types and reflects increased leakage propensities for cytosolic and nuclear proteins compared to membrane and mitochondrial proteins. A classifier based on the signature allowed for the accurate identification of permeabilized cells across cell types and species. The classifier is integrated into QuantQC ( scp.slavovlab.net/QuantQC ) to support its application to diverse samples and workflows.
Collapse
Affiliation(s)
- Andrew Leduc
- Departments of Bioengineering, Biology, Chemistry and Chemical Biology, Single Cell Proteomics Center, Northeastern University, Boston, MA, USA.
| | - Yanxin Xu
- Center for Regenerative Medicine, Massachusetts General Hospital, Boston, MA, USA
| | - Gergana Shipkovenska
- Center for Regenerative Medicine, Massachusetts General Hospital, Boston, MA, USA
| | - Zhixun Dou
- Center for Regenerative Medicine, Massachusetts General Hospital, Boston, MA, USA
| | - Nikolai Slavov
- Departments of Bioengineering, Biology, Chemistry and Chemical Biology, Single Cell Proteomics Center, Northeastern University, Boston, MA, USA.
- Parallel Squared Technology Institute, Watertown, MA, USA.
| |
Collapse
|
7
|
Lee DI, Roy S. Examining the dynamics of three-dimensional genome organization with multitask matrix factorization. Genome Res 2025; 35:1179-1193. [PMID: 40113262 PMCID: PMC12047540 DOI: 10.1101/gr.279930.124] [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: 08/15/2024] [Accepted: 02/20/2025] [Indexed: 03/22/2025]
Abstract
Three-dimensional (3D) genome organization, which determines how the DNA is packaged inside the nucleus, has emerged as a key component of the gene regulation machinery. High-throughput chromosome conformation data sets, such as Hi-C, have become available across multiple conditions and time points, offering a unique opportunity to examine changes in 3D genome organization and link them to phenotypic changes in normal and disease processes. However, systematic detection of higher-order structural changes across multiple Hi-C data sets remains a major challenge. Existing computational methods either do not model higher-order structural units or cannot model dynamics across more than two conditions of interest. We address these limitations with tree-guided integrated factorization (TGIF), a generalizable multitask nonnegative matrix factorization (NMF) approach that can be applied to time series or hierarchically related biological conditions. TGIF can identify large-scale changes at the compartment or subcompartment levels, as well as local changes at boundaries of topologically associated domains (TADs). Based on benchmarking in simulated and real Hi-C data, TGIF boundaries are more accurate and reproducible across differential levels of noise and sources of technical artifacts, and are more enriched in CTCF. Application to three multisample mammalian data sets shows that TGIF can detect differential regions at compartment, subcompartment, and boundary levels that are associated with significant changes in regulatory signals and gene expression enriched in tissue-specific processes. Finally, we leverage TGIF boundaries to prioritize sequence variants for multiple phenotypes from the NHGRI GWAS catalog. Taken together, TGIF is a flexible tool to examine 3D genome organization dynamics across disease and developmental processes.
Collapse
Affiliation(s)
- Da-Inn Lee
- Department of Biostatistics and Medical Informatics, University of Wisconsin-Madison, Madison, Wisconsin 53715, USA
| | - Sushmita Roy
- Department of Biostatistics and Medical Informatics, University of Wisconsin-Madison, Madison, Wisconsin 53715, USA;
- Wisconsin Institute for Discovery, Madison, Wisconsin 53715, USA
| |
Collapse
|
8
|
Gao L, Liu Y, Zou J, Deng F, Liu Z, Zhang Z, Zhao X, Chen L, Tong HHY, Ji Y, Le H, Zou X, Hao J. Deep scSTAR: leveraging deep learning for the extraction and enhancement of phenotype-associated features from single-cell RNA sequencing and spatial transcriptomics data. Brief Bioinform 2025; 26:bbaf160. [PMID: 40315434 PMCID: PMC12047704 DOI: 10.1093/bib/bbaf160] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2024] [Revised: 02/28/2025] [Accepted: 03/19/2025] [Indexed: 05/04/2025] Open
Abstract
Single-cell sequencing has advanced our understanding of cellular heterogeneity and disease pathology, offering insights into cellular behavior and immune mechanisms. However, extracting meaningful phenotype-related features is challenging due to noise, batch effects, and irrelevant biological signals. To address this, we introduce Deep scSTAR (DscSTAR), a deep learning-based tool designed to enhance phenotype-associated features. DscSTAR identified HSP+ FKBP4+ T cells in CD8+ T cells, which linked to immune dysfunction and resistance to immune checkpoint blockade in non-small cell lung cancer. It has also enhanced spatial transcriptomics analysis of renal cell carcinoma, revealing interactions between cancer cells, CD8+ T cells, and tumor-associated macrophages that may promote immune suppression and affect outcomes. In hepatocellular carcinoma, it highlighted the role of S100A12+ neutrophils and cancer-associated fibroblasts in forming tumor immune barriers and potentially contributing to immunotherapy resistance. These findings demonstrate DscSTAR's capacity to model and extract phenotype-specific information, advancing our understanding of disease mechanisms and therapy resistance.
Collapse
Affiliation(s)
- Lianchong Gao
- Shanghai Center for Systems Biomedicine, Key Laboratory of Systems Biomedicine (Ministry of Education), Shanghai Jiao Tong University, 800# Dong Chuan Road, Minhang District, Shanghai 200240, China
| | - Yujun Liu
- Department of Radiation Oncology, Fudan University Shanghai Cancer Center, Fudan University, Shanghai 200433, China
| | - Jiawei Zou
- Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai 200031, China
| | - Fulan Deng
- Centre for Artificial Intelligence Driven Drug Discovery, Faculty of Applied Sciences, Macao Polytechnic University, Macao SAR, China
| | - Zheqi Liu
- Department of Oral and Maxillofacial Surgery, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Zhen Zhang
- Department of Oral and Maxillofacial-Head and Neck Oncology, Ninth People’s Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200011, China
| | - Xinran Zhao
- Department of Oral and Maxillofacial-Head and Neck Oncology, Ninth People’s Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200011, China
| | - Lei Chen
- Renji Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200127, China
| | - Henry H Y Tong
- Centre for Artificial Intelligence Driven Drug Discovery, Faculty of Applied Sciences, Macao Polytechnic University, Macao SAR, China
| | - Yuan Ji
- Molecular Pathology Center, Dept. Pathology, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Huangying Le
- Shanghai Center for Systems Biomedicine, Key Laboratory of Systems Biomedicine (Ministry of Education), Shanghai Jiao Tong University, 800# Dong Chuan Road, Minhang District, Shanghai 200240, China
| | - Xin Zou
- Digital Diagnosis and Treatment Innovation Center for Cancer, Institute of Translational Medicine, Shanghai Jiao Tong University, 800# Dong Chuan Road, Shanghai 200240, China
| | - Jie Hao
- Shanghai Key Laboratory of Plant Functional Genomics and Resources, Shanghai Chenshan Botanical Garden, Chen Hua Road, Songjiang District, Shanghai 201602, China
- Institute of Clinical Science, Zhongshan Hospital, Fudan University, No.180 Fenglin Road, Xuhui District, Shanghai 200032, China
| |
Collapse
|
9
|
Ly A, Hotchkiss H, Prévost ED, Pelletier JM, Deming MA, Murib L, Root DH. Assessing the role of BNST GABA neurons in backward conditioned suppression. Neurobiol Learn Mem 2025; 219:108058. [PMID: 40318802 DOI: 10.1016/j.nlm.2025.108058] [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: 12/31/2024] [Revised: 04/11/2025] [Accepted: 04/26/2025] [Indexed: 05/07/2025]
Abstract
Conditioned suppression is a useful paradigm for measuring learned avoidance. In most conditioned suppression studies, forward conditioning is used where a cue predicts an aversive stimulus. However, backward conditioning, in which an aversive stimulus predicts a cue, provides unique insights into learned avoidance due to its influence on both conditioned excitation and inhibition. We trained mice to consume sucrose in context A, associated an aversive stimulus in context B to few or many forward or backwards paired cues (CS + ), and then tested for conditioned suppression in context A in response to the CS + . We found that few or many forward CS + and few backward CS + produced conditioned suppression, but many backwards cues did not. Administration of diazepam, a positive allosteric modulator of the GABAA receptor, prevented conditioned suppression to the backward CS + but not to the forward CS + . Furthermore, freezing behavior was observed in response to the forward CS + but not the backward CS+, and diazepam had no effect on freezing or locomotion. We next examined BNST GABA neurons for potential sensitivity to backwards cues and conditioned suppression. VGaT BNST signaling increased in response to sucrose licks during the backward CS + but not to licks outside the CS + and not to the backward CS + onset or offset. Using designer receptors, we found that BNST VGaT neuron activation, but not its inhibition, prevented backward conditioned suppression expression. We conclude that backward conditioned suppression is dependent on both positive allosteric modulation of GABA on GABAA receptors by diazepam and BNST GABA neurons.
Collapse
Affiliation(s)
- Annie Ly
- Department of Psychology and Neuroscience, University of Colorado Boulder, 2860 Wilderness Pl, Boulder, CO 80301, USA
| | - Hayden Hotchkiss
- Department of Psychology and Neuroscience, University of Colorado Boulder, 2860 Wilderness Pl, Boulder, CO 80301, USA
| | - Emily D Prévost
- Department of Psychology and Neuroscience, University of Colorado Boulder, 2860 Wilderness Pl, Boulder, CO 80301, USA
| | - Julianne M Pelletier
- Department of Psychology and Neuroscience, University of Colorado Boulder, 2860 Wilderness Pl, Boulder, CO 80301, USA
| | - Melissa A Deming
- Department of Psychology and Neuroscience, University of Colorado Boulder, 2860 Wilderness Pl, Boulder, CO 80301, USA
| | - Luma Murib
- Department of Psychology and Neuroscience, University of Colorado Boulder, 2860 Wilderness Pl, Boulder, CO 80301, USA
| | - David H Root
- Department of Psychology and Neuroscience, University of Colorado Boulder, 2860 Wilderness Pl, Boulder, CO 80301, USA.
| |
Collapse
|
10
|
Li H, Zhang Z, Squires M, Chen X, Zhang X. scMultiSim: simulation of single-cell multi-omics and spatial data guided by gene regulatory networks and cell-cell interactions. Nat Methods 2025; 22:982-993. [PMID: 40247122 DOI: 10.1038/s41592-025-02651-0] [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: 08/27/2023] [Accepted: 03/07/2025] [Indexed: 04/19/2025]
Abstract
Simulated single-cell data are essential for designing and evaluating computational methods in the absence of experimental ground truth. Here we present scMultiSim, a comprehensive simulator that generates multimodal single-cell data encompassing gene expression, chromatin accessibility, RNA velocity and spatial cell locations while accounting for the relationships between modalities. Unlike existing tools that focus on limited biological factors, scMultiSim simultaneously models cell identity, gene regulatory networks, cell-cell interactions and chromatin accessibility while incorporating technical noise. Moreover, it allows users to adjust each factor's effect easily. Here we show that scMultiSim generates data with expected biological effects, and demonstrate its applications by benchmarking a wide range of computational tasks, including multimodal and multi-batch data integration, RNA velocity estimation, gene regulatory network inference and cell-cell interaction inference using spatially resolved gene expression data. Compared to existing simulators, scMultiSim can benchmark a much broader range of existing computational problems and even new potential tasks.
Collapse
Affiliation(s)
- Hechen Li
- Georgia Institute of Technology, Atlanta, GA, USA
| | - Ziqi Zhang
- Georgia Institute of Technology, Atlanta, GA, USA
| | | | - Xi Chen
- Southern University of Science and Technology, Shenzhen, China
| | - Xiuwei Zhang
- Georgia Institute of Technology, Atlanta, GA, USA.
| |
Collapse
|
11
|
Wan Q, Wu X, Zhou J, Wu W, Cao Y, Sun C, Li Z, Gong Z, Tang H, Li Q, Chu J, Wang Q, Cui K, Lu X. The Hypoxia-Associated High-Risk Cell Subpopulation Distinctly Enhances the Progression of Glioma. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2416231. [PMID: 40047299 PMCID: PMC12061283 DOI: 10.1002/advs.202416231] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/04/2024] [Revised: 02/20/2025] [Indexed: 05/10/2025]
Abstract
Less-aggressive lower-grade gliomas (LGGs) frequently transform into glioblastoma (GBM). Most previous studies of gliomas have not focused on LGG-original high-risk subpopulations, which may be one of the most critical hallmarks of glioma progression. In this study, LGG samples are collected to perform single-cell sequencing (scRNA-seq) and identify a unique cell subpopulation marked by CDC20, KIF20A and PTTG1, correlating with poor survival in multiple cohorts. Importantly, the CDC20+KIF20A+PTTG1+ cell subpopulation is strongly associated with transforming LGG to GBM according to scRNA-seq and multiplexed immunofluorescence staining assays. In vitro, ex vivo and in vivo investigations further hint that this cell subpopulation is critical to the proliferation and growth of gliomas, and is associated with the hypoxia core activation. Pharmaceutically and therapeutically, the inhibition of this cell subpopulation showed significant anti-tumor effects and effective enhancement of the Temozolomide treatment efficiency. These findings provide insights into the therapeutic strategies of glioma progression, highlighting promising ways to avoid early-stage gliomas developing into advanced gliomas.
Collapse
Affiliation(s)
- Quan Wan
- Department of Neurosurgery and Emergency MedicineJiangnan University Medical Center (Wuxi No.2 People's Hospital)WuxiJiangsu214002China
- Neuroscience CenterWuxi School of MedicineJiangnan UniversityWuxiJiangsu214122China
- Wuxi Neurosurgical InstituteWuxiJiangsu214043China
| | - Xuechao Wu
- Department of Neurosurgery and Emergency MedicineJiangnan University Medical Center (Wuxi No.2 People's Hospital)WuxiJiangsu214002China
- Neuroscience CenterWuxi School of MedicineJiangnan UniversityWuxiJiangsu214122China
- Wuxi Neurosurgical InstituteWuxiJiangsu214043China
| | - Jinxu Zhou
- Department of NeurosurgeryThe 904th Hospital of Joint Logistic Support Force of PLAWuxiJiangsu214044China
| | - Weiqi Wu
- Department of NeurosurgeryBeijing Tiantan HospitalCapital Medical UniversityBeijing100070China
| | - Yuanliang Cao
- Neuroscience CenterWuxi School of MedicineJiangnan UniversityWuxiJiangsu214122China
| | - Cuiping Sun
- Neuroscience CenterWuxi School of MedicineJiangnan UniversityWuxiJiangsu214122China
| | - Zheng Li
- Department of Neurosurgery and Emergency MedicineJiangnan University Medical Center (Wuxi No.2 People's Hospital)WuxiJiangsu214002China
| | - Zhicheng Gong
- Wuxi Cancer InstituteAffiliated Hospital of Jiangnan UniversityWuxiJiangsu214062China
| | - Hong Tang
- Neuroscience CenterWuxi School of MedicineJiangnan UniversityWuxiJiangsu214122China
- Department of PathologyJiangnan University Medical Center (Wuxi No.2 People's Hospital)WuxiJiangsu214002China
| | - Qilin Li
- Computer Vision LabDepartment of Electrical EngineeringCalifornia Institute of TechnologyPasadenaCalifornia91125USA
| | - Junsheng Chu
- Department of NeurosurgeryBeijing Tiantan HospitalCapital Medical UniversityBeijing100070China
| | - Qing Wang
- Department of Neurosurgery and Emergency MedicineJiangnan University Medical Center (Wuxi No.2 People's Hospital)WuxiJiangsu214002China
- Neuroscience CenterWuxi School of MedicineJiangnan UniversityWuxiJiangsu214122China
- Wuxi Neurosurgical InstituteWuxiJiangsu214043China
| | - Kaisa Cui
- Neuroscience CenterWuxi School of MedicineJiangnan UniversityWuxiJiangsu214122China
- Wuxi Cancer InstituteAffiliated Hospital of Jiangnan UniversityWuxiJiangsu214062China
| | - Xiaojie Lu
- Department of Neurosurgery and Emergency MedicineJiangnan University Medical Center (Wuxi No.2 People's Hospital)WuxiJiangsu214002China
- Neuroscience CenterWuxi School of MedicineJiangnan UniversityWuxiJiangsu214122China
- Wuxi Neurosurgical InstituteWuxiJiangsu214043China
| |
Collapse
|
12
|
Almasaad JM, Bataineh ZM, Zaqout S. Neuronal diversity in the caudate nucleus: A comparative study between camel and human brains. Anat Rec (Hoboken) 2025; 308:1410-1424. [PMID: 39118384 PMCID: PMC11967514 DOI: 10.1002/ar.25555] [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: 03/20/2024] [Revised: 07/05/2024] [Accepted: 07/18/2024] [Indexed: 08/10/2024]
Abstract
Caudate nucleus (CN) neurons in camels and humans were examined using modified Golgi impregnation methods. Neurons were classified based on soma morphology, dendritic characteristics, and spine distribution. Three primary neuron types were identified in both species: rich-spiny (Type I), sparsely-spiny (Type II), and aspiny (Type III), each comprising subtypes with specific features. Comparative analysis revealed significant differences in soma size, dendritic morphology, and spine distribution between camels and humans. The study contributes to our understanding of structural diversity in CN neurons and provides insights into evolutionary neural adaptations.
Collapse
Affiliation(s)
- Juman M. Almasaad
- Department of Basic Medical Sciences, College of MedicineKing Saud Bin Abdul Aziz University for Health Sciences (KSAU‐HS)JeddahSaudi Arabia
- King Abdullah International Medical Research Centre (KIAMRC)King Abdulaziz Medical CityJeddahSaudi Arabia
| | - Ziad M. Bataineh
- Department of Anatomy, Faculty of MedicineJordan University of Science & TechnologyIrbidJordan
| | - Sami Zaqout
- Department of Basic Medical Sciences, College of Medicine, QU HealthQatar UniversityDohaQatar
| |
Collapse
|
13
|
Marand AP, Jiang L, Gomez-Cano F, Minow MAA, Zhang X, Mendieta JP, Luo Z, Bang S, Yan H, Meyer C, Schlegel L, Johannes F, Schmitz RJ. The genetic architecture of cell type-specific cis regulation in maize. Science 2025; 388:eads6601. [PMID: 40245149 DOI: 10.1126/science.ads6601] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2024] [Accepted: 02/04/2025] [Indexed: 04/19/2025]
Abstract
Gene expression and complex phenotypes are determined by the activity of cis-regulatory elements. However, an understanding of how extant genetic variants affect cis regulation remains limited. Here, we investigated the consequences of cis-regulatory diversity using single-cell genomics of more than 0.7 million nuclei across 172 Zea mays (maize) inbreds. Our analyses pinpointed cis-regulatory elements distinct to domesticated maize and revealed how historical transposon activity has shaped the cis-regulatory landscape. Leveraging population genetics principles, we fine-mapped about 22,000 chromatin accessibility-associated genetic variants with widespread cell type-specific effects. Variants in TEOSINTE BRANCHED1/CYCLOIDEA/PROLIFERATING CELL FACTOR-binding sites were the most prevalent determinants of chromatin accessibility. Finally, integrating chromatin accessibility-associated variants, organismal trait variation, and population differentiation revealed how local adaptation has rewired regulatory networks in unique cellular contexts to alter maize flowering.
Collapse
Affiliation(s)
| | - Luguang Jiang
- Department of Molecular, Cellular, and Developmental Biology, University of Michigan, Ann Arbor, MI, USA
| | - Fabio Gomez-Cano
- Department of Molecular, Cellular, and Developmental Biology, University of Michigan, Ann Arbor, MI, USA
| | - Mark A A Minow
- Department of Genetics, University of Georgia, Athens, GA, USA
| | - Xuan Zhang
- Department of Genetics, University of Georgia, Athens, GA, USA
| | - John P Mendieta
- Department of Genetics, University of Georgia, Athens, GA, USA
| | - Ziliang Luo
- Department of Genetics, University of Georgia, Athens, GA, USA
| | - Sohyun Bang
- Institute of Bioinformatics, University of Georgia, Athens, GA, USA
| | - Haidong Yan
- Department of Genetics, University of Georgia, Athens, GA, USA
| | - Cullan Meyer
- Department of Genetics, University of Georgia, Athens, GA, USA
| | - Luca Schlegel
- Plant Epigenomics, TUM School of Life Sciences Weihenstephan, Technical University of Munich, Munich, Germany
| | - Frank Johannes
- Plant Epigenomics, TUM School of Life Sciences Weihenstephan, Technical University of Munich, Munich, Germany
| | | |
Collapse
|
14
|
Menon AV, Song B, Chao L, Sriram D, Chansky P, Bakshi I, Ulianova J, Li W. Unraveling the future of genomics: CRISPR, single-cell omics, and the applications in cancer and immunology. Front Genome Ed 2025; 7:1565387. [PMID: 40292231 PMCID: PMC12021818 DOI: 10.3389/fgeed.2025.1565387] [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: 01/23/2025] [Accepted: 03/26/2025] [Indexed: 04/30/2025] Open
Abstract
The CRISPR system has transformed many research areas, including cancer and immunology, by providing a simple yet effective genome editing system. Its simplicity has facilitated large-scale experiments to assess gene functionality across diverse biological contexts, generating extensive datasets that boosted the development of computational methods and machine learning/artificial intelligence applications. Integrating CRISPR with single-cell technologies has further advanced our understanding of genome function and its role in many biological processes, providing unprecedented insights into human biology and disease mechanisms. This powerful combination has accelerated AI-driven analyses, enhancing disease diagnostics, risk prediction, and therapeutic innovations. This review provides a comprehensive overview of CRISPR-based genome editing systems, highlighting their advancements, current progress, challenges, and future opportunities, especially in cancer and immunology.
Collapse
Affiliation(s)
- A. Vipin Menon
- Center for Genetic Medicine Research, Children’s National Hospital, Washington, DC, DC, United States
- Department of Genomics and Precision Medicine, George Washington University, Washington, DC, DC, United States
| | - Bicna Song
- Center for Genetic Medicine Research, Children’s National Hospital, Washington, DC, DC, United States
- Department of Genomics and Precision Medicine, George Washington University, Washington, DC, DC, United States
| | - Lumen Chao
- Center for Genetic Medicine Research, Children’s National Hospital, Washington, DC, DC, United States
- Department of Genomics and Precision Medicine, George Washington University, Washington, DC, DC, United States
| | - Diksha Sriram
- The George Washington University, Washington, DC, DC, United States
| | - Pamela Chansky
- Center for Genetic Medicine Research, Children’s National Hospital, Washington, DC, DC, United States
- Integrated Biomedical Sciences (IBS) Program, The George Washington University, Washington, DC, DC, United States
| | - Ishnoor Bakshi
- The George Washington University, Washington, DC, DC, United States
| | - Jane Ulianova
- Integrated Biomedical Sciences (IBS) Program, The George Washington University, Washington, DC, DC, United States
| | - Wei Li
- Center for Genetic Medicine Research, Children’s National Hospital, Washington, DC, DC, United States
- Department of Genomics and Precision Medicine, George Washington University, Washington, DC, DC, United States
| |
Collapse
|
15
|
Zhu Q, Jiang Z, Thomson M, Gartner Z. Revealing a coherent cell state landscape across single cell datasets with CONCORD. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.03.13.643146. [PMID: 40161827 PMCID: PMC11952503 DOI: 10.1101/2025.03.13.643146] [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: 04/02/2025]
Abstract
Batch integration, denoising, and dimensionality reduction remain fundamental challenges in single-cell data analysis. While many machine learning tools aim to overcome these challenges by engineering model architectures, we use a different strategy, building on the insight that optimized mini-batch sampling during training can profoundly influence learning outcomes. We present CONCORD, a self-supervised learning approach that implements a unified, probabilistic data sampling scheme combining neighborhood-aware and dataset-aware sampling: the former enhancing resolution while the latter removing batch effects. Using only a minimalist one-hidden-layer neural network and contrastive learning, CONCORD achieves state-of-the-art performance without relying on deep architectures, auxiliary losses, or supervision. It generates high-resolution cell atlases that seamlessly integrate data across batches, technologies, and species, without relying on prior assumptions about data structure. The resulting latent representations are denoised, interpretable, and biologically meaningful-capturing gene co-expression programs, resolving subtle cellular states, and preserving both local geometric relationships and global topological organization. We demonstrate CONCORD's broad applicability across diverse datasets, establishing it as a general-purpose framework for learning unified, high-fidelity representations of cellular identity and dynamics.
Collapse
Affiliation(s)
- Qin Zhu
- Department of Pharmaceutical Chemistry, University of California San Francisco; San Francisco, CA 94158, USA
| | - Zuzhi Jiang
- Tetrad Graduate Program, University of California San Francisco; San Francisco, CA 94158, USA
| | - Matt Thomson
- Division of Biology and Biological Engineering, California Institute of Technology; Pasadena, CA 91125, USA
| | - Zev Gartner
- Department of Pharmaceutical Chemistry, University of California San Francisco; San Francisco, CA 94158, USA
- Chan Zuckerberg Biohub; San Francisco, CA 94158, USA
- Center for Cellular Construction, University of California San Francisco; San Francisco, CA 94158, USA
| |
Collapse
|
16
|
Luo L, Jing W, Guo Y, Liu D, He A, Lu Y. A cell-type-specific circuit of somatostatin neurons in the habenula encodes antidepressant action in male mice. Nat Commun 2025; 16:3417. [PMID: 40210897 PMCID: PMC11985912 DOI: 10.1038/s41467-025-58591-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: 08/12/2024] [Accepted: 03/27/2025] [Indexed: 04/12/2025] Open
Abstract
Major depression is characterized by an array of negative experiences, including hopelessness and anhedonia. We hypothesize that inhibition of negative experiences or aversion may generate antidepressant action. To directly test this hypothesis, we perform multimodal behavioral screenings in male mice and identify somatostatin (SST)-expressing neurons in the region X (HBX) between the lateral and medial habenula as a specific type of antidepressant neuron. SST neuronal activity modulation dynamically regulates antidepressant induction and relief. We also explore the circuit basis for encoding these modulations using single-unit recordings. We find that SST neurons receive inhibitory synaptic inputs directly from cholecystokinin-expressing neurons in the bed nucleus of the stria terminalis and project excitatory axon terminals onto proenkephalin-expressing neurons in the interpeduncular nucleus. This study reveals a cell-type-specific circuit of SST neurons in the HBX that encodes antidepressant action, and the control of the circuit may contribute to improving well-being.
Collapse
Affiliation(s)
- Lingli Luo
- Innovation Center of Brain Medical Sciences, Ministry of Education of the People's Republic of China, Huazhong University of Science and Technology, Wuhan, China
- Department of Pathophysiology, School of Basic Medicine and Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Wei Jing
- Innovation Center of Brain Medical Sciences, Ministry of Education of the People's Republic of China, Huazhong University of Science and Technology, Wuhan, China
- Department of Anatomy, School of Basic Medicine and Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yiqing Guo
- Innovation Center of Brain Medical Sciences, Ministry of Education of the People's Republic of China, Huazhong University of Science and Technology, Wuhan, China
- Department of Pathophysiology, School of Basic Medicine and Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Dan Liu
- Innovation Center of Brain Medical Sciences, Ministry of Education of the People's Republic of China, Huazhong University of Science and Technology, Wuhan, China.
- Department of Medical Genetics, School of Basic Medicine and Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| | - Aodi He
- Innovation Center of Brain Medical Sciences, Ministry of Education of the People's Republic of China, Huazhong University of Science and Technology, Wuhan, China.
- Department of Pathophysiology, School of Basic Medicine and Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
- Department of Anatomy, School of Basic Medicine and Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| | - Youming Lu
- Innovation Center of Brain Medical Sciences, Ministry of Education of the People's Republic of China, Huazhong University of Science and Technology, Wuhan, China.
- Department of Pathophysiology, School of Basic Medicine and Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
- Department of Physiology, School of Basic Medicine and Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| |
Collapse
|
17
|
Ma M, Luo Q, Chen L, Liu F, Yin L, Guan B. Novel insights into kidney disease: the scRNA-seq and spatial transcriptomics approaches: a literature review. BMC Nephrol 2025; 26:181. [PMID: 40200175 DOI: 10.1186/s12882-025-04103-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2024] [Accepted: 03/28/2025] [Indexed: 04/10/2025] Open
Abstract
Over the past decade, single-cell RNA sequencing (scRNA-seq) and spatial transcriptomics (ST) have revolutionized biomedical research, particularly in understanding cellular heterogeneity in kidney diseases. This review summarizes the application and development of scRNA-seq combined with ST in the context of kidney disease. By dissecting cellular heterogeneity at an unprecedented resolution, these advanced techniques have identified novel cell subpopulations and their dynamic interactions within the renal microenvironment. The integration of scRNA-seq with ST has been instrumental in elucidating the cellular and molecular mechanisms underlying kidney development, homeostasis, and disease progression. This approach has not only identified key cellular players in renal pathophysiology but also revealed the spatial organization of cells within the kidney, which is crucial for understanding their functional specialization. This paper highlights the transformative impact of these techniques on renal research that have paved the way for targeted therapeutic interventions and personalized medicine in the management of kidney disease.
Collapse
Affiliation(s)
- Mingming Ma
- Institute of Nephrology and Blood Purification, The First Affiliated Hospital of Jinan University, Jinan University, No. 613, West Huangpu Avenue, Guangzhou, 510632, China
| | - Qiao Luo
- Institute of Nephrology and Blood Purification, The First Affiliated Hospital of Jinan University, Jinan University, No. 613, West Huangpu Avenue, Guangzhou, 510632, China
| | - Liangmei Chen
- Institute of Nephrology and Blood Purification, The First Affiliated Hospital of Jinan University, Jinan University, No. 613, West Huangpu Avenue, Guangzhou, 510632, China
| | - Fanna Liu
- Institute of Nephrology and Blood Purification, The First Affiliated Hospital of Jinan University, Jinan University, No. 613, West Huangpu Avenue, Guangzhou, 510632, China
| | - Lianghong Yin
- Institute of Nephrology and Blood Purification, The First Affiliated Hospital of Jinan University, Jinan University, No. 613, West Huangpu Avenue, Guangzhou, 510632, China.
| | - Baozhang Guan
- Institute of Nephrology and Blood Purification, The First Affiliated Hospital of Jinan University, Jinan University, No. 613, West Huangpu Avenue, Guangzhou, 510632, China.
| |
Collapse
|
18
|
Chen J, Richardson PR, Kirby C, Eddy SR, Hoekstra HE. Cellular evolution of the hypothalamic preoptic area of behaviorally divergent deer mice. eLife 2025; 13:RP103109. [PMID: 40191998 PMCID: PMC11975375 DOI: 10.7554/elife.103109] [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] [Indexed: 04/09/2025] Open
Abstract
Genetic variation is known to contribute to the variation of animal social behavior, but the molecular mechanisms that lead to behavioral differences are still not fully understood. Here, we investigate the cellular evolution of the hypothalamic preoptic area (POA), a brain region that plays a critical role in social behavior, across two sister species of deer mice (Peromyscus maniculatus and P. polionotus) with divergent social systems. These two species exhibit large differences in mating and parental care behavior across species and sex. Using single-nucleus RNA-sequencing, we build a cellular atlas of the POA for males and females of both Peromyscus species. We identify four cell types that are differentially abundant across species, two of which may account for species differences in parental care behavior based on known functions of these cell types. Our data further implicate two sex-biased cell types to be important for the evolution of sex-specific behavior. Finally, we show a remarkable reduction of sex-biased gene expression in P. polionotus, a monogamous species that also exhibits reduced sexual dimorphism in parental care behavior. Our POA atlas is a powerful resource to investigate how molecular neuronal traits may be evolving to give rise to innate differences in social behavior across animal species.
Collapse
Affiliation(s)
- Jenny Chen
- Department of Molecular & Cellular Biology, Harvard UniversityCambridgeUnited States
- Department of Organismic and Evolutionary Biology, Harvard UniversityCambridgeUnited States
| | - Phoebe R Richardson
- Department of Organismic and Evolutionary Biology, Harvard UniversityCambridgeUnited States
| | - Christopher Kirby
- Department of Organismic and Evolutionary Biology, Harvard UniversityCambridgeUnited States
| | - Sean R Eddy
- Department of Molecular & Cellular Biology, Harvard UniversityCambridgeUnited States
- Howard Hughes Medical Institute, Harvard UniversityCambridgeUnited States
| | - Hopi E Hoekstra
- Department of Molecular & Cellular Biology, Harvard UniversityCambridgeUnited States
- Department of Organismic and Evolutionary Biology, Harvard UniversityCambridgeUnited States
| |
Collapse
|
19
|
Sertbas M, Ulgen KO. Exploring Human Brain Metabolism via Genome-Scale Metabolic Modeling with Highlights on Multiple Sclerosis. ACS Chem Neurosci 2025; 16:1346-1360. [PMID: 40091499 PMCID: PMC11969529 DOI: 10.1021/acschemneuro.5c00006] [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: 01/06/2025] [Revised: 02/18/2025] [Accepted: 03/03/2025] [Indexed: 03/19/2025] Open
Abstract
Cerebral dysfunctions give rise to a wide range of neurological diseases due to the structural and functional complexity of the human brain stemming from the interactive cellular metabolism of its specific cells, including neurons and glial cells. In parallel with advances in isolation and measurement technologies, genome-scale metabolic models (GEMs) have become a powerful tool in the studies of systems biology to provide critical insights into the understanding of sophisticated eukaryotic systems. In this study, brain cell-specific GEMs were reconstructed for neurons, astrocytes, microglia, oligodendrocytes, and oligodendrocyte precursor cells by integrating single-cell RNA-seq data and global Human1 via a task-driven integrative network inference for tissues (tINIT) algorithm. Then, intercellular reactions among neurons, astrocytes, microglia, and oligodendrocytes were added to generate a combined brain model, iHumanBrain2690. This brain network was used in the prediction of metabolic alterations in glucose, ketone bodies, oxygen change, and reporter metabolites. Glucose supplementation increased the subsystems' activities in glycolysis, and ketone bodies elevated those in the TCA cycle and oxidative phosphorylation. Reporter metabolite analysis identified L-carnitine and arachidonate as the top reporter metabolites in gray and white matter microglia in multiple sclerosis (MS), respectively. Carbamoyl-phosphate was found to be the top reporter metabolite in primary progressive MS. Taken together, single and integrated iHumanBrain2690 metabolic networks help us elucidate complex metabolism in brain physiology and homeostasis in health and disease.
Collapse
Affiliation(s)
- Mustafa Sertbas
- Department
of Chemical Engineering, Bogazici University, 34342 Istanbul, Turkey
- Department
of Chemical Engineering, Istanbul Technical
University, 34469 Istanbul, Turkey
| | - Kutlu O. Ulgen
- Department
of Chemical Engineering, Bogazici University, 34342 Istanbul, Turkey
| |
Collapse
|
20
|
Ernst IVS, Lehtonen L, Nilsson SM, Nielsen FL, Marcher AB, Mandrup S, Madsen JGS. Single Nucleus Multiome Analysis Reveals Early Inflammatory Response to High-Fat Diet in Mouse Pancreatic Islets. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.04.01.646568. [PMID: 40236154 PMCID: PMC11996447 DOI: 10.1101/2025.04.01.646568] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/17/2025]
Abstract
In periods of sustained hyper-nutrition, pancreatic β-cells undergo functional compensation through transcriptional upregulation of gene programs driving insulin secretion. This adaptation is essential for maintaining systemic glucose homeostasis and metabolic health. Using single nuclei multiomics, we have mapped the early transcriptional compensation mechanisms in murine islets of Langerhans exposed to high-fat diet (HFD) for one and three weeks. We show that β-cells exhibit the largest transcriptional response to HFD, characterized by early activation of proinflammatory eRegulons and downregulation of β-cell identity genes, particularly in a distinct subset of β-cells. Our observations translate to humans, as we observe an increase in the inflammatory gene signatures in human β-cells in pre-diabetes and diabetes. Collectively, these observations point to cellular cross-talk through proinflammatory signaling as a central and early driver of β-cell dysfunction that limits the compensatory capacity of β-cells, which is closely linked to the development of diabetes.
Collapse
|
21
|
Yang J, Zheng Z, Jiao Y, Yu K, Bhatara S, Yang X, Natarajan S, Zhang J, Pan Q, Easton J, Yan KK, Peng J, Liu K, Yu J. Spotiphy enables single-cell spatial whole transcriptomics across an entire section. Nat Methods 2025; 22:724-736. [PMID: 40074951 PMCID: PMC11978521 DOI: 10.1038/s41592-025-02622-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2024] [Accepted: 01/29/2025] [Indexed: 03/14/2025]
Abstract
Spatial transcriptomics (ST) has advanced our understanding of tissue regionalization by enabling the visualization of gene expression within whole-tissue sections, but current approaches remain plagued by the challenge of achieving single-cell resolution without sacrificing whole-genome coverage. Here we present Spotiphy (spot imager with pseudo-single-cell-resolution histology), a computational toolkit that transforms sequencing-based ST data into single-cell-resolved whole-transcriptome images. Spotiphy delivers the most precise cellular proportions in extensive benchmarking evaluations. Spotiphy-derived inferred single-cell profiles reveal astrocyte and disease-associated microglia regional specifications in Alzheimer's disease and healthy mouse brains. Spotiphy identifies multiple spatial domains and alterations in tumor-tumor microenvironment interactions in human breast ST data. Spotiphy bridges the information gap and enables visualization of cell localization and transcriptomic profiles throughout entire sections, offering highly informative outputs and an innovative spatial analysis pipeline for exploring complex biological systems.
Collapse
Affiliation(s)
- Jiyuan Yang
- Department of Computational Biology, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Ziqian Zheng
- Department of Industrial & Systems Engineering, University of Wisconsin-Madison, Madison, WI, USA
| | - Yun Jiao
- Department of Structural Biology, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Kaiwen Yu
- Center of Proteomics and Metabolomics, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Sheetal Bhatara
- Department of Computational Biology, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Xu Yang
- Department of Computational Biology, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Sivaraman Natarajan
- Department of Computational Biology, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Jiahui Zhang
- Department of Industrial & Systems Engineering, University of Wisconsin-Madison, Madison, WI, USA
| | - Qingfei Pan
- Department of Computational Biology, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - John Easton
- Department of Computational Biology, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Koon-Kiu Yan
- Department of Computational Biology, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Junmin Peng
- Department of Structural Biology, St. Jude Children's Research Hospital, Memphis, TN, USA.
| | - Kaibo Liu
- Department of Industrial & Systems Engineering, University of Wisconsin-Madison, Madison, WI, USA.
| | - Jiyang Yu
- Department of Computational Biology, St. Jude Children's Research Hospital, Memphis, TN, USA.
| |
Collapse
|
22
|
Li Y, Hu M, Zhang Z, Wu B, Zheng J, Zhang F, Hao J, Xue T, Li Z, Zhu C, Liu Y, Zhao L, Xu W, Xin P, Feng C, Wang W, Zhao Y, Qiu Q, Wang K. Origin and stepwise evolution of vertebrate lungs. Nat Ecol Evol 2025; 9:672-691. [PMID: 39953253 DOI: 10.1038/s41559-025-02642-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2023] [Accepted: 01/15/2025] [Indexed: 02/17/2025]
Abstract
Lungs are essential respiratory organs in terrestrial vertebrates, present in most bony fishes but absent in cartilaginous fishes, making them an ideal model for studying organ evolution. Here we analysed single-cell RNA sequencing data from adult and developing lungs across vertebrate species, revealing significant similarities in cell composition, developmental trajectories and gene expression patterns. Surprisingly, a large proportion of lung-related genes, coexpression patterns and many lung enhancers are present in cartilaginous fishes despite their lack of lungs, suggesting that a substantial genetic foundation for lung development existed in the last common ancestor of jawed vertebrates. In addition, the 1,040 enhancers that emerged since the last common ancestor of bony fishes probably contain lung-specific elements that led to the development of lungs. We further identified alveolar type 1 cells as a mammal-specific alveolar cell type, along with several mammal-specific genes, including ager and sfta2, that are highly expressed in lungs. Functional validation showed that deletion of sfta2 in mice leads to severe respiratory defects, highlighting its critical role in mammalian lung features. Our study provides comprehensive insights into the evolution of vertebrate lungs, demonstrating how both regulatory network modifications and the emergence of new genes have shaped lung development and specialization across species.
Collapse
Affiliation(s)
- Ye Li
- Shaanxi Key Laboratory of Qinling Ecological Intelligent Monitoring and Protection, School of Ecology and Environment, Northwestern Polytechnical University, Xi'an, China
| | - Mingliang Hu
- Shaanxi Key Laboratory of Qinling Ecological Intelligent Monitoring and Protection, School of Ecology and Environment, Northwestern Polytechnical University, Xi'an, China
| | - Zhigang Zhang
- State Key Laboratory of Cancer Biology and Department of Physiology and Pathophysiology, Fourth Military Medical University, Xi'an, China
| | - Baosheng Wu
- Guangdong Key Laboratory of Animal Conservation and Resource Utilization, Institute of Zoology, Guangdong Academy of Sciences, Guangzhou, China
| | - Jiangmin Zheng
- Shaanxi Key Laboratory of Qinling Ecological Intelligent Monitoring and Protection, School of Ecology and Environment, Northwestern Polytechnical University, Xi'an, China
| | - Fenghua Zhang
- Shaanxi Key Laboratory of Qinling Ecological Intelligent Monitoring and Protection, School of Ecology and Environment, Northwestern Polytechnical University, Xi'an, China
| | - Jiaqi Hao
- Shaanxi Key Laboratory of Qinling Ecological Intelligent Monitoring and Protection, School of Ecology and Environment, Northwestern Polytechnical University, Xi'an, China
| | - Tingfeng Xue
- Shaanxi Key Laboratory of Qinling Ecological Intelligent Monitoring and Protection, School of Ecology and Environment, Northwestern Polytechnical University, Xi'an, China
| | - Zhaohong Li
- Shenzhen Branch, Guangdong Laboratory for Lingnan Modern Agriculture, Key Laboratory of Livestock and Poultry Multi-Omics of MARA, Agricultural Genomics Institute at Shenzhen, Chinese Academy of Agricultural Sciences, Shenzhen, China
| | - Chenglong Zhu
- Shaanxi Key Laboratory of Qinling Ecological Intelligent Monitoring and Protection, School of Ecology and Environment, Northwestern Polytechnical University, Xi'an, China
| | - Yuxuan Liu
- Shaanxi Key Laboratory of Qinling Ecological Intelligent Monitoring and Protection, School of Ecology and Environment, Northwestern Polytechnical University, Xi'an, China
| | - Lei Zhao
- Shaanxi Key Laboratory of Qinling Ecological Intelligent Monitoring and Protection, School of Ecology and Environment, Northwestern Polytechnical University, Xi'an, China
| | - Wenjie Xu
- Shaanxi Key Laboratory of Qinling Ecological Intelligent Monitoring and Protection, School of Ecology and Environment, Northwestern Polytechnical University, Xi'an, China
| | - Peidong Xin
- Shaanxi Key Laboratory of Qinling Ecological Intelligent Monitoring and Protection, School of Ecology and Environment, Northwestern Polytechnical University, Xi'an, China
| | - Chenguang Feng
- Shaanxi Key Laboratory of Qinling Ecological Intelligent Monitoring and Protection, School of Ecology and Environment, Northwestern Polytechnical University, Xi'an, China.
| | - Wen Wang
- Shaanxi Key Laboratory of Qinling Ecological Intelligent Monitoring and Protection, School of Ecology and Environment, Northwestern Polytechnical University, Xi'an, China.
- New Cornerstone Science Laboratory, Xi'an, China.
| | - Yilin Zhao
- State Key Laboratory of Cancer Biology and Department of Physiology and Pathophysiology, Fourth Military Medical University, Xi'an, China.
| | - Qiang Qiu
- Shaanxi Key Laboratory of Qinling Ecological Intelligent Monitoring and Protection, School of Ecology and Environment, Northwestern Polytechnical University, Xi'an, China.
| | - Kun Wang
- Shaanxi Key Laboratory of Qinling Ecological Intelligent Monitoring and Protection, School of Ecology and Environment, Northwestern Polytechnical University, Xi'an, China.
- Laboratory for Marine Biology and Biotechnology, Qingdao Marine Science and Technology Center, Qingdao, China.
| |
Collapse
|
23
|
Marco Salas S, Kuemmerle LB, Mattsson-Langseth C, Tismeyer S, Avenel C, Hu T, Rehman H, Grillo M, Czarnewski P, Helgadottir S, Tiklova K, Andersson A, Rafati N, Chatzinikolaou M, Theis FJ, Luecken MD, Wählby C, Ishaque N, Nilsson M. Optimizing Xenium In Situ data utility by quality assessment and best-practice analysis workflows. Nat Methods 2025; 22:813-823. [PMID: 40082609 PMCID: PMC11978515 DOI: 10.1038/s41592-025-02617-2] [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: 02/13/2023] [Accepted: 02/04/2025] [Indexed: 03/16/2025]
Abstract
The Xenium In Situ platform is a new spatial transcriptomics product commercialized by 10x Genomics, capable of mapping hundreds of genes in situ at subcellular resolution. Given the multitude of commercially available spatial transcriptomics technologies, recommendations in choice of platform and analysis guidelines are increasingly important. Herein, we explore 25 Xenium datasets generated from multiple tissues and species, comparing scalability, resolution, data quality, capacities and limitations with eight other spatially resolved transcriptomics technologies and commercial platforms. In addition, we benchmark the performance of multiple open-source computational tools, when applied to Xenium datasets, in tasks including preprocessing, cell segmentation, selection of spatially variable features and domain identification. This study serves as an independent analysis of the performance of Xenium, and provides best practices and recommendations for analysis of such datasets.
Collapse
Affiliation(s)
- Sergio Marco Salas
- Science for Life Laboratory, Department of Biochemistry and Biophysics, Stockholm University, Stockholm, Sweden.
- Institute of Computational Biology, Computational Health Center, Helmholtz Munich, Munich, Germany.
| | - Louis B Kuemmerle
- Institute of Computational Biology, Computational Health Center, Helmholtz Munich, Munich, Germany
- Institute for Tissue Engineering and Regenerative Medicine (iTERM), Helmholtz Zentrum München, Munich, Germany
| | | | - Sebastian Tismeyer
- Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Digital Health Center, Berlin, Germany
| | - Christophe Avenel
- Department of Information Technology and SciLifeLab BioImage Informatics Facility, Uppsala University, Uppsala, Sweden
| | - Taobo Hu
- Science for Life Laboratory, Department of Biochemistry and Biophysics, Stockholm University, Stockholm, Sweden
| | - Habib Rehman
- Institute of Computational Biology, Computational Health Center, Helmholtz Munich, Munich, Germany
- Department of Bioengineering, University of Illinois at Urbana-Champaign, Champaign, IL, USA
| | - Marco Grillo
- Science for Life Laboratory, Department of Biochemistry and Biophysics, Stockholm University, Stockholm, Sweden
| | - Paulo Czarnewski
- Science for Life Laboratory, Department of Gene Technology, KTH Royal Institute of Technology, Stockholm, Sweden
- Science for Life Laboratory, Department of Biochemistry and Biophysics, National Bioinformatics Infrastructure Sweden, Stockholm University, Stockholm, Sweden
| | - Saga Helgadottir
- Science for Life Laboratory, Department of Biochemistry and Biophysics, Stockholm University, Stockholm, Sweden
| | - Katarina Tiklova
- Science for Life Laboratory, Department of Biochemistry and Biophysics, Stockholm University, Stockholm, Sweden
| | - Axel Andersson
- Department of Information Technology and SciLifeLab BioImage Informatics Facility, Uppsala University, Uppsala, Sweden
| | - Nima Rafati
- National Bioinformatics Infrastructure Sweden, Uppsala University, SciLifeLab, Department of Medical Biochemistry and Microbiology, Uppsala, Sweden
| | - Maria Chatzinikolaou
- Science for Life Laboratory, Department of Biochemistry and Biophysics, Stockholm University, Stockholm, Sweden
| | - Fabian J Theis
- Institute of Computational Biology, Computational Health Center, Helmholtz Munich, Munich, Germany
| | - Malte D Luecken
- Institute of Computational Biology, Computational Health Center, Helmholtz Munich, Munich, Germany
- Institute of Lung Health & Immunity, Helmholtz Munich; Member of the German Center for Lung Research (DZL), Munich, Germany
| | - Carolina Wählby
- Department of Information Technology and SciLifeLab BioImage Informatics Facility, Uppsala University, Uppsala, Sweden
| | - Naveed Ishaque
- Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Digital Health Center, Berlin, Germany
| | - Mats Nilsson
- Science for Life Laboratory, Department of Biochemistry and Biophysics, Stockholm University, Stockholm, Sweden.
| |
Collapse
|
24
|
Ding DY, Tang Z, Zhu B, Ren H, Shalek AK, Tibshirani R, Nolan GP. Quantitative characterization of tissue states using multiomics and ecological spatial analysis. Nat Genet 2025; 57:910-921. [PMID: 40169791 PMCID: PMC11985343 DOI: 10.1038/s41588-025-02119-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2024] [Accepted: 02/05/2025] [Indexed: 04/03/2025]
Abstract
The spatial organization of cells in tissues underlies biological function, and recent advances in spatial profiling technologies have enhanced our ability to analyze such arrangements to study biological processes and disease progression. We propose MESA (multiomics and ecological spatial analysis), a framework drawing inspiration from ecological concepts to delineate functional and spatial shifts across tissue states. MESA introduces metrics to systematically quantify spatial diversity and identify hot spots, linking spatial patterns to phenotypic outcomes, including disease progression. Furthermore, MESA integrates spatial and single-cell multiomics data to facilitate an in-depth, molecular understanding of cellular neighborhoods and their spatial interactions within tissue microenvironments. Applying MESA to diverse datasets demonstrates additional insights it brings over prior methods, including newly identified spatial structures and key cell populations linked to disease states. Available as a Python package, MESA offers a versatile framework for quantitative decoding of tissue architectures in spatial omics across health and disease.
Collapse
Affiliation(s)
- Daisy Yi Ding
- Department of Biomedical Data Science, Stanford University, Stanford, CA, USA
| | - Zeyu Tang
- Weill Cornell Graduate School of Medical Sciences, Weill Cornell Medicine, New York, NY, USA
| | - Bokai Zhu
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Hongyu Ren
- Department of Computer Science, Stanford University, Stanford, CA, USA
| | - Alex K Shalek
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Institute for Medical Engineering and Science, MIT, Cambridge, MA, USA
- Department of Chemistry, MIT, Cambridge, MA, USA
- Koch Institute for Integrative Cancer Research, MIT, Cambridge, MA, USA
| | - Robert Tibshirani
- Department of Biomedical Data Science, Stanford University, Stanford, CA, USA
- Department of Statistics, Stanford University, Stanford, CA, USA
| | - Garry P Nolan
- Department of Pathology, Stanford University, Stanford, CA, USA.
| |
Collapse
|
25
|
Llera-Oyola J, Pérez-Moraga R, Parras M, Rosón B. How to view the female reproductive tract through single-cell looking glasses. Am J Obstet Gynecol 2025; 232:S21-S43. [PMID: 40253081 DOI: 10.1016/j.ajog.2024.08.040] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2023] [Revised: 07/04/2024] [Accepted: 08/24/2024] [Indexed: 04/21/2025]
Abstract
Single-cell technologies have emerged as an unprecedented tool for biologists and clinicians, allowing them to assess organs and tissues at the level of individual cells. In the field of women's reproductive biology, single-cell studies have provided insights into the cellular and molecular processes that regulate reproductive and obstetrical functions in health and disease. The knowledge that these studies generate is helping clinicians to improve the understanding and diagnosis of infertility related issues or pregnancy complications and to find new avenues for their treatment. However, navigating the expansive landscape of this type of transcriptomic data analysis represents a pivotal challenge in current research. Single cell RNA sequencing involves isolating cells into droplets, reverse transcribing RNA to generate complementary DNA, with each droplet content uniquely labeled by a barcode. Upon sequencing the complementary DNAs, the barcodes enable the reassignment of sequencing reads to individual droplets, facilitating the reconstruction of the cellular landscape of the sample obtained from a tissue or organ and beyond. Researchers, equipped with the metaphorical 'single-cell glasses,' must adequately choose from a plethora of strategies to dissect and interpret cellular information. Sophisticated algorithms and the decision-making process are often underestimated, resulting in artefactual or cumbersome interpreted results. Computational biologists apply and innovate computational tools designed to process, model, and interpret expansive datasets. The ramifications of their work extend far beyond the realm of data processing; they give shape to the outcome of analyses, playing a pivotal role in drawing meaningful conclusions from the wealth of information garnered. In this review, we describe the wide variety of approaches and analytical steps available with enough detail to gain a concise picture of what a complete examination of a single-cell dataset would be. We commence with a discussion on key points in experimental design, highlighting crucial questions one should consider. Following this, we delve into the various preprocessing and quality control steps essential for any single-cell dataset. The subsequent section offers a detailed guide on constructing a single-cell atlas, exploring nuances such as differential characteristics in visualization and clustering techniques, as well as strategies for assigning identity to cell populations through gene marker annotations. Moving beyond the creation of an atlas, we explore methods for investigating pathological conditions. This involves conducting cell population comparison tests between conditions and analyzing specific cell-to-cell communications and cellular differentiation trajectories in both health and disease scenarios. This work aims to furnish a newcomer researcher and/or clinician with essential guidelines to embark on a single-cell adventure without succumbing to common pitfalls. By bridging the gap between theory and practice, it facilitates the translation of single-cell technologies into clinically relevant applications. Throughout the manuscript, practical examples of its usage in women's reproductive health studies are provided. Various sections delve into specific clinical scenarios, demonstrating how these guidelines can be instrumental in unraveling the molecular landscapes of diseases and physiological processes related to women's reproduction.
Collapse
Affiliation(s)
- Jaime Llera-Oyola
- Carlos Simon Foundation, INCLIVA Health Research Institute, Valencia, Spain
| | - Raúl Pérez-Moraga
- Carlos Simon Foundation, INCLIVA Health Research Institute, Valencia, Spain; R&D Department, Igenomix, Valencia, Spain
| | - Marcos Parras
- Carlos Simon Foundation, INCLIVA Health Research Institute, Valencia, Spain
| | - Beatriz Rosón
- Carlos Simon Foundation, INCLIVA Health Research Institute, Valencia, Spain.
| |
Collapse
|
26
|
Rodríguez-Durán A, Andrade-Silva V, Numan M, Waldman J, Ali A, Logullo C, da Silva Vaz Junior I, Parizi LF. Multi-Omics Technologies Applied to Improve Tick Research. Microorganisms 2025; 13:795. [PMID: 40284631 PMCID: PMC12029647 DOI: 10.3390/microorganisms13040795] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2025] [Revised: 03/10/2025] [Accepted: 03/27/2025] [Indexed: 04/29/2025] Open
Abstract
The advancement of multi-omics technologies is crucial to deepen knowledge on tick biology. These approaches, used to study diverse phenomena, are applied to experiments that aim to understand changes in gene transcription, protein function, cellular processes, and prediction of systems at global biological levels. This review addressed the application of omics data to investigate and elucidate tick physiological processes, such as feeding, digestion, reproduction, neuronal, endocrine systems, understanding population dynamics, transmitted pathogens, control, and identifying new vaccine targets. Furthermore, new therapeutic perspectives using tick bioactive molecules, such as anti-inflammatory, analgesic, and antitumor, were summarized. Taken together, the application of omics technologies can help to understand the protein functions and biological behavior of ticks, as well as the identification of potential new antigens influencing the development of alternative control strategies and, consequently, the tick-borne disease prevention in veterinary and public health contexts. Finally, tick population dynamics have been determined through a combination of environmental factors, host availability, and genetic adaptations, and recent advances in omics technologies have improved our understanding of their ecological resilience and resistance mechanisms. Future directions point to the integration of spatial omics and artificial intelligence to further unravel tick biology and improve control strategies.
Collapse
Affiliation(s)
- Arlex Rodríguez-Durán
- Programa de Pós-Graduação em Ciências Veterinária, Universidade Federal do Rio Grande do Sul, Avenida Bento Gonçalves, 9090, Porto Alegre 91540-000, RS, Brazil; (A.R.-D.); (M.N.)
- Centro de Biotecnologia, Universidade Federal do Rio Grande do Sul, Avenida Bento Gonçalves, 9500, Porto Alegre 91501-970, RS, Brazil; (V.A.-S.); (J.W.); (I.d.S.V.J.)
- Grupo de Investigación Parasitología Veterinaria, Laboratorio de Parasitología Veterinaria, Universidad Nacional de Colombia (UNAL), Carrera 30 No 45-03, Bogotá 110111, Colombia
| | - Vinícius Andrade-Silva
- Centro de Biotecnologia, Universidade Federal do Rio Grande do Sul, Avenida Bento Gonçalves, 9500, Porto Alegre 91501-970, RS, Brazil; (V.A.-S.); (J.W.); (I.d.S.V.J.)
| | - Muhammad Numan
- Programa de Pós-Graduação em Ciências Veterinária, Universidade Federal do Rio Grande do Sul, Avenida Bento Gonçalves, 9090, Porto Alegre 91540-000, RS, Brazil; (A.R.-D.); (M.N.)
- Centro de Biotecnologia, Universidade Federal do Rio Grande do Sul, Avenida Bento Gonçalves, 9500, Porto Alegre 91501-970, RS, Brazil; (V.A.-S.); (J.W.); (I.d.S.V.J.)
| | - Jéssica Waldman
- Centro de Biotecnologia, Universidade Federal do Rio Grande do Sul, Avenida Bento Gonçalves, 9500, Porto Alegre 91501-970, RS, Brazil; (V.A.-S.); (J.W.); (I.d.S.V.J.)
| | - Abid Ali
- Department of Zoology, Abdul Wali Khan University Mardan, Mardan 23200, Khyber Pakhtunkhwa, Pakistan;
| | - Carlos Logullo
- Instituto de Bioquímica Médica Leopoldo de Meis, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-902, RJ, Brazil;
- Instituto Nacional de Ciência e Tecnologia em Entomologia Molecular (INCT-EM), Rio de Janeiro 21941-853, RJ, Brazil
| | - Itabajara da Silva Vaz Junior
- Centro de Biotecnologia, Universidade Federal do Rio Grande do Sul, Avenida Bento Gonçalves, 9500, Porto Alegre 91501-970, RS, Brazil; (V.A.-S.); (J.W.); (I.d.S.V.J.)
- Instituto Nacional de Ciência e Tecnologia em Entomologia Molecular (INCT-EM), Rio de Janeiro 21941-853, RJ, Brazil
- Faculdade de Veterinária, Universidade Federal do Rio Grande do Sul, Avenida Bento Gonçalves, 9090, Porto Alegre 91540-000, RS, Brazil
| | - Luís Fernando Parizi
- Centro de Biotecnologia, Universidade Federal do Rio Grande do Sul, Avenida Bento Gonçalves, 9500, Porto Alegre 91501-970, RS, Brazil; (V.A.-S.); (J.W.); (I.d.S.V.J.)
- Faculdade de Veterinária, Universidade Federal do Rio Grande do Sul, Avenida Bento Gonçalves, 9090, Porto Alegre 91540-000, RS, Brazil
| |
Collapse
|
27
|
Liu J, Wang Y, Li C, Gu Y, Ono N, Welch J. CytoSimplex: visualizing single-cell fates and transitions on a simplex. Bioinformatics 2025; 41:btaf119. [PMID: 40119904 PMCID: PMC11992338 DOI: 10.1093/bioinformatics/btaf119] [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: 12/10/2023] [Revised: 01/23/2025] [Accepted: 03/20/2025] [Indexed: 03/25/2025] Open
Abstract
SUMMARY Cells differentiate to their final fates along unique trajectories, often involving multi-potent progenitors that can produce multiple terminally differentiated cell types. Recent developments in single-cell transcriptomic and epigenomic measurement provide tremendous opportunities for mapping these trajectories. The visualization of single-cell data often relies on dimension reduction methods such as UMAP to simplify high-dimensional single-cell data down to an understandable 2D form. However, these dimension reduction methods are not constructed to allow direct interpretation of the reduced dimensions in terms of cell differentiation. To address these limitations, we developed a new approach that places each cell from a single-cell dataset within a simplex whose vertices correspond to terminally differentiated cell types. Our approach can quantify and visualize current cell fate commitment and future cell potential. We developed CytoSimplex, a standalone open-source package implemented in R and Python that provides simple and intuitive visualizations of cell differentiation in 2D ternary and 3D quaternary plots. We believe that CytoSimplex can help researchers gain a better understanding of cell type transitions in specific tissues and characterize developmental processes. AVAILABILITY AND IMPLEMENTATION The R version of CytoSimplex is available on Github at https://github.com/welch-lab/CytoSimplex. The Python version of CytoSimplex is available on Github at https://github.com/welch-lab/pyCytoSimplex.
Collapse
Affiliation(s)
- Jialin Liu
- Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, MI, 48109, United States
| | - Yichen Wang
- Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, MI, 48109, United States
| | - Chen Li
- Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, MI, 48109, United States
| | - Yichen Gu
- Department of Electrical and Computer Engineering, University of Michigan, Ann Arbor, MI, 48109, United States
| | - Noriaki Ono
- University of Texas Health Science Center at Houston School of Dentistry, Houston, TX, 77030, United States
| | - Joshua Welch
- Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, MI, 48109, United States
- Department of Computer Science and Engineering, University of Michigan, Ann Arbor, MI, 48109, United States
| |
Collapse
|
28
|
Sun Z, Zhu H, He X, Lendemeijer B, Wang Z, Fan J, Sun Y, Zhang Z, Markx S, Kushner SA, Xu B, Gogos JA. Genomic and Transcriptomic Signatures of SETD1A Disruption in Human Excitatory Neuron Development and Psychiatric Disease Risk. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.03.26.645419. [PMID: 40196527 PMCID: PMC11974865 DOI: 10.1101/2025.03.26.645419] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 04/09/2025]
Abstract
Genetic disruption of SETD1A markedly increases the risk for schizophrenia. To elucidate the underlying mechanisms, we generated isogenic organoid models of the developing human cerebral cortex harboring a SETD1A loss-of-function schizophrenia risk mutation. Employing chromatin profiling combined with RNA sequencing, we identified high-confidence SETD1A target genes, analyzed the impact of the mutation on SETD1A binding and transcriptional regulation and validated key findings with orthogonal approaches. Disruption of SETD1A function disturbs the finely tuned temporal gene expression in the excitatory neuron lineage, yielding an aberrant transcriptional program that compromises key regulatory and metabolic pathways essential for neurodevelopmental transitions. Although overall SETD1A binding remains unchanged in mutant neurons, we identified localized alterations in SETD1A binding that correlate with shifts in H3K4me3 levels and gene expression. These changes are enriched at enhancer regions, suggesting that enhancer-regulated genes are especially vulnerable to SETD1A reduction. Notably, target genes with enhancer-bound SETD1A are primarily linked to neuronal functions while those with promoter-bound SETD1A are enriched for basic cellular functions. By mapping the SETD1A binding landscape in excitatory neurons of the human fetal frontal cortex and integrating multimodal neuroimaging and genetic datasets, we demonstrate that the genomic context of SETD1A binding differentially correlates with macroscale brain organization and establish a link between SETD1A-bound enhancers, schizophrenia-associated brain alterations and genetic susceptibility. Our study advances our understanding of the role of SETD1A binding patterns in schizophrenia pathogenesis, offering insights that may guide future therapeutic strategies.
Collapse
Affiliation(s)
- Zhixiong Sun
- Mortimer B. Zuckerman Mind Brain and Behavior Institute, Columbia University, New York, NY 10027, USA
- Department of Psychiatry, Columbia University & New York State Psychiatric Institute, New York, NY 10032, USA
- Stavros Niarchos Foundation Center for Precision Psychiatry and Mental Health, New York, NY 10032, USA
| | - Huixiang Zhu
- Department of Psychiatry, Columbia University & New York State Psychiatric Institute, New York, NY 10032, USA
- Stavros Niarchos Foundation Center for Precision Psychiatry and Mental Health, New York, NY 10032, USA
| | - Xiaofu He
- Department of Psychiatry, Columbia University & New York State Psychiatric Institute, New York, NY 10032, USA
| | - Bas Lendemeijer
- Stavros Niarchos Foundation Center for Precision Psychiatry and Mental Health, New York, NY 10032, USA
| | - Zanxu Wang
- Department of Psychiatry, Columbia University & New York State Psychiatric Institute, New York, NY 10032, USA
| | - Jack Fan
- Department of Psychiatry, Columbia University & New York State Psychiatric Institute, New York, NY 10032, USA
| | - Yan Sun
- Department of Psychiatry, Columbia University & New York State Psychiatric Institute, New York, NY 10032, USA
- Stavros Niarchos Foundation Center for Precision Psychiatry and Mental Health, New York, NY 10032, USA
| | - Zhiguo Zhang
- Department of Genetics and Development, Columbia University, New York, NY 10032, USA
| | - Sander Markx
- Department of Psychiatry, Columbia University & New York State Psychiatric Institute, New York, NY 10032, USA
- Stavros Niarchos Foundation Center for Precision Psychiatry and Mental Health, New York, NY 10032, USA
| | - Steven A. Kushner
- Department of Psychiatry, Columbia University & New York State Psychiatric Institute, New York, NY 10032, USA
- Stavros Niarchos Foundation Center for Precision Psychiatry and Mental Health, New York, NY 10032, USA
| | - Bin Xu
- Department of Psychiatry, Columbia University & New York State Psychiatric Institute, New York, NY 10032, USA
- Stavros Niarchos Foundation Center for Precision Psychiatry and Mental Health, New York, NY 10032, USA
| | - Joseph A. Gogos
- Mortimer B. Zuckerman Mind Brain and Behavior Institute, Columbia University, New York, NY 10027, USA
- Department of Psychiatry, Columbia University & New York State Psychiatric Institute, New York, NY 10032, USA
- Stavros Niarchos Foundation Center for Precision Psychiatry and Mental Health, New York, NY 10032, USA
- Department of Physiology and Cellular Biophysics, Columbia University, New York, NY 10032, USA
- Department of Neuroscience, Columbia University, New York, NY 10032, USA
| |
Collapse
|
29
|
Liao X, Li Y, Li S, Wen L, Li X, Yu B. Enhanced Integration of Single-Cell Multi-Omics Data Using Graph Attention Networks. ACS Synth Biol 2025; 14:931-942. [PMID: 39888834 DOI: 10.1021/acssynbio.4c00864] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2025]
Abstract
The continuous advancement of single-cell multimodal omics (scMulti-omics) technologies offers unprecedented opportunities to measure various modalities, including RNA expression, protein abundance, gene perturbation, DNA methylation, and chromatin accessibility at single-cell resolution. These advances hold significant potential for breakthroughs by integrating diverse omics modalities. However, the data generated from different omics layers often face challenges due to high dimensionality, heterogeneity, and sparsity, which can adversely impact the accuracy and efficiency of data integration analyses. To address these challenges, we propose a high-precision analysis method called scMGAT (single-cell multiomics data analysis based on multihead graph attention networks). This method effectively coordinates reliable information across multiomics data sets using a multihead attention mechanism, allowing for better management of the heterogeneous characteristics inherent in scMulti-omics data. We evaluated scMGAT's performance on eight sets of real scMulti-omics data, including samples from both human and mouse. The experimental results demonstrate that scMGAT significantly enhances the quality of multiomics data and improves the accuracy of cell-type annotation compared to state-of-the-art methods. scMGAT is now freely accessible at https://github.com/Xingyu-Liao/scMGAT.
Collapse
Affiliation(s)
- Xingyu Liao
- School of Computer Science, Northwestern Polytechnical University (NPU), Chang'an Campus, Xi'an, Shaanxi 710072, P.R. China
| | - Yanyan Li
- School of Computer Science, Northwestern Polytechnical University (NPU), Chang'an Campus, Xi'an, Shaanxi 710072, P.R. China
| | - Shuangyi Li
- School of Data Science, Qingdao University of Science and Technology, Qingdao 266061, P.R. China
| | - Long Wen
- School of Computer Science, Northwestern Polytechnical University (NPU), Chang'an Campus, Xi'an, Shaanxi 710072, P.R. China
| | - Xingyi Li
- School of Computer Science, Northwestern Polytechnical University (NPU), Chang'an Campus, Xi'an, Shaanxi 710072, P.R. China
| | - Bin Yu
- School of Data Science, Qingdao University of Science and Technology, Qingdao 266061, P.R. China
| |
Collapse
|
30
|
Liu T, Lin Y, Luo X, Sun Y, Zhao H. VISTA Uncovers Missing Gene Expression and Spatial-induced Information for Spatial Transcriptomic Data Analysis. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2024.08.26.609718. [PMID: 40166134 PMCID: PMC11957009 DOI: 10.1101/2024.08.26.609718] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 04/02/2025]
Abstract
Characterizing cell activities within a spatially resolved context is essential to enhance our understanding of spatially-induced cellular states and features. While single-cell RNA-seq (scRNA-seq) offers comprehensive profiling of cells within a tissue, it fails to capture spatial context. Conversely, subcellular spatial transcriptomics (SST) technologies provide high-resolution spatial profiles of gene expression, yet their utility is constrained by the limited number of genes they can simultaneously profile. To address this limitation, we introduce VISTA, a novel approach designed to predict the expression levels of unobserved genes specifically tailored for SST data. VISTA jointly models scRNA-seq data and SST data based on variational inference and geometric deep learning, and incorporates uncertainty quantification. Using four SST datasets, we demonstrate VISTA's superior performance in imputation and in analyzing large-scale SST datasets with satisfactory time efficiency and memory consumption. The imputation of VISTA enables a multitude of downstream applications, including the detection of new spatially variable genes, the discovery of novel ligand-receptor interactions, the inference of spatial RNA velocity, the generation for spatial transcriptomics with in-silico perturbation, and an improved decomposition of spatial and intrinsic variations.
Collapse
Affiliation(s)
- Tianyu Liu
- Interdepartmental Program in Computational Biology & Bioinformatics, Yale University, New Haven, 06511, CT, USA
| | - Yingxin Lin
- Department of Biostatistics, Yale University, New Haven, 06511, CT, USA
| | - Xiao Luo
- Department of Computer Science, University of California, Los Angeles, Los Angeles, 90095, CA, USA
| | - Yizhou Sun
- Department of Computer Science, University of California, Los Angeles, Los Angeles, 90095, CA, USA
| | - Hongyu Zhao
- Interdepartmental Program in Computational Biology & Bioinformatics, Yale University, New Haven, 06511, CT, USA
- Department of Biostatistics, Yale University, New Haven, 06511, CT, USA
| |
Collapse
|
31
|
Jeon EY, Kwak Y, Kang H, Kim H, Jin SY, Park S, Kim RG, Ko D, Won JK, Cho A, Jung I, Lee CH, Park J, Kim HY, Chae JH, Choi M. Inhibiting EZH2 complements steroid effects in Duchenne muscular dystrophy. SCIENCE ADVANCES 2025; 11:eadr4443. [PMID: 40085707 PMCID: PMC11908487 DOI: 10.1126/sciadv.adr4443] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/02/2024] [Accepted: 02/10/2025] [Indexed: 03/16/2025]
Abstract
Duchenne muscular dystrophy (DMD) is a devastating X-linked disorder caused by dystrophin gene mutations. Despite recent advances in understanding the disease etiology and applying emerging treatment methodologies, glucocorticoid derivatives remain the only general therapeutic option that can slow disease development. However, the precise molecular mechanism of glucocorticoid action remains unclear, and there is still need for additional remedies to complement the treatment. Here, using single-nucleus RNA sequencing and spatial transcriptome analyses of human and mouse muscles, we investigated pathogenic features in patients with DMD and palliative effects of glucocorticoids. Our approach further illuminated the importance of proliferating satellite cells and revealed increased activity of a signal transduction pathway involving EZH2 in the patient cells. Subsequent administration of EZH2 inhibitors to Dmd mutant mice resulted in improved muscle phenotype through maintaining the immune-suppressing effect but overriding the muscle weakness and fibrogenic effects exerted by glucocorticoids. Our analysis reveals pathogenic mechanisms that can be readily targeted by extant therapeutic options for DMD.
Collapse
MESH Headings
- Muscular Dystrophy, Duchenne/drug therapy
- Muscular Dystrophy, Duchenne/metabolism
- Muscular Dystrophy, Duchenne/genetics
- Muscular Dystrophy, Duchenne/pathology
- Animals
- Humans
- Enhancer of Zeste Homolog 2 Protein/antagonists & inhibitors
- Enhancer of Zeste Homolog 2 Protein/metabolism
- Enhancer of Zeste Homolog 2 Protein/genetics
- Mice
- Glucocorticoids/pharmacology
- Glucocorticoids/therapeutic use
- Male
- Muscle, Skeletal/metabolism
- Muscle, Skeletal/drug effects
- Muscle, Skeletal/pathology
- Disease Models, Animal
- Satellite Cells, Skeletal Muscle/metabolism
- Satellite Cells, Skeletal Muscle/drug effects
- Signal Transduction/drug effects
- Steroids/pharmacology
Collapse
Affiliation(s)
- Eun Young Jeon
- Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Yejin Kwak
- Department of Information Convergence Engineering, Pusan National University, Yangsan, Republic of Korea
| | - Hyeji Kang
- Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul, Republic of Korea
- Department of Pharmacology, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Hanbyeol Kim
- Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul, Republic of Korea
- Department of Pharmacology, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Se Young Jin
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, Republic of Korea
| | - Soojin Park
- Department of Genomic Medicine, Seoul National University Hospital, Seoul, Republic of Korea
| | - Ryeo Gyeong Kim
- Department of Pediatrics, Rare Disease Center, Seoul National University Bundang Hospital, Gyeonggi-do, Republic of Korea
| | - Dayoung Ko
- Department of Pediatric Surgery, Seoul National University Children’s Hospital, Seoul, Republic of Korea
| | - Jae-Kyung Won
- Department of Pathology, Seoul National University Hospital, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Anna Cho
- Department of Pediatrics, Rare Disease Center, Seoul National University Bundang Hospital, Gyeonggi-do, Republic of Korea
| | - Inkyung Jung
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, Republic of Korea
| | - Chul-Hwan Lee
- Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul, Republic of Korea
- Department of Pharmacology, Seoul National University College of Medicine, Seoul, Republic of Korea
- Cancer Research Institute, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Jeongbin Park
- Department of Information Convergence Engineering, Pusan National University, Yangsan, Republic of Korea
- School of Biomedical Convergence Engineering, Pusan National University, Yangsan, Republic of Korea
| | - Hyun-Young Kim
- Department of Pediatric Surgery, Seoul National University Children’s Hospital, Seoul, Republic of Korea
| | - Jong-Hee Chae
- Department of Genomic Medicine, Seoul National University Hospital, Seoul, Republic of Korea
- Department of Pediatrics, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Murim Choi
- Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul, Republic of Korea
| |
Collapse
|
32
|
Guan A, Quek C. Single-Cell Multi-Omics: Insights into Therapeutic Innovations to Advance Treatment in Cancer. Int J Mol Sci 2025; 26:2447. [PMID: 40141092 PMCID: PMC11942442 DOI: 10.3390/ijms26062447] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2025] [Revised: 03/04/2025] [Accepted: 03/07/2025] [Indexed: 03/28/2025] Open
Abstract
Advances in single-cell multi-omics technologies have deepened our understanding of cancer biology by integrating genomic, transcriptomic, epigenomic, and proteomic data at single-cell resolution. These single-cell multi-omics technologies provide unprecedented insights into tumour heterogeneity, tumour microenvironment, and mechanisms of therapeutic resistance, enabling the development of precision medicine strategies. The emerging field of single-cell multi-omics in genomic medicine has improved patient outcomes. However, most clinical applications still depend on bulk genomic approaches, which fail to directly capture the genomic variations driving cellular heterogeneity. In this review, we explore the common single-cell multi-omics platforms and discuss key analytical steps for data integration. Furthermore, we highlight emerging knowledge in therapeutic resistance and immune evasion, and the potential of new therapeutic innovations informed by single-cell multi-omics. Finally, we discuss the future directions of the application of single-cell multi-omics technologies. By bridging the gap between technological advancements and clinical implementation, this review provides a roadmap for leveraging single-cell multi-omics to improve cancer treatment and patient outcomes.
Collapse
Affiliation(s)
- Angel Guan
- Melanoma Institute Australia, The University of Sydney, Sydney, NSW 2065, Australia;
- Faculty of Medicine and Health, The University of Sydney, Sydney, NSW 2006, Australia
- Charles Perkins Centre, The University of Sydney, Sydney, NSW 2006, Australia
| | - Camelia Quek
- Melanoma Institute Australia, The University of Sydney, Sydney, NSW 2065, Australia;
- Faculty of Medicine and Health, The University of Sydney, Sydney, NSW 2006, Australia
- Charles Perkins Centre, The University of Sydney, Sydney, NSW 2006, Australia
| |
Collapse
|
33
|
Ge S, Sun S, Xu H, Cheng Q, Ren Z. Deep learning in single-cell and spatial transcriptomics data analysis: advances and challenges from a data science perspective. Brief Bioinform 2025; 26:bbaf136. [PMID: 40185158 PMCID: PMC11970898 DOI: 10.1093/bib/bbaf136] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2024] [Revised: 02/17/2025] [Accepted: 03/05/2025] [Indexed: 04/07/2025] Open
Abstract
The development of single-cell and spatial transcriptomics has revolutionized our capacity to investigate cellular properties, functions, and interactions in both cellular and spatial contexts. Despite this progress, the analysis of single-cell and spatial omics data remains challenging. First, single-cell sequencing data are high-dimensional and sparse, and are often contaminated by noise and uncertainty, obscuring the underlying biological signal. Second, these data often encompass multiple modalities, including gene expression, epigenetic modifications, metabolite levels, and spatial locations. Integrating these diverse data modalities is crucial for enhancing prediction accuracy and biological interpretability. Third, while the scale of single-cell sequencing has expanded to millions of cells, high-quality annotated datasets are still limited. Fourth, the complex correlations of biological tissues make it difficult to accurately reconstruct cellular states and spatial contexts. Traditional feature engineering approaches struggle with the complexity of biological networks, while deep learning, with its ability to handle high-dimensional data and automatically identify meaningful patterns, has shown great promise in overcoming these challenges. Besides systematically reviewing the strengths and weaknesses of advanced deep learning methods, we have curated 21 datasets from nine benchmarks to evaluate the performance of 58 computational methods. Our analysis reveals that model performance can vary significantly across different benchmark datasets and evaluation metrics, providing a useful perspective for selecting the most appropriate approach based on a specific application scenario. We highlight three key areas for future development, offering valuable insights into how deep learning can be effectively applied to transcriptomic data analysis in biological, medical, and clinical settings.
Collapse
Affiliation(s)
- Shuang Ge
- Shenzhen International Graduate School, Tsinghua University, 2279 Lishui Road, Nanshan District, Shenzhen 518055, Guangdong, China
- Pengcheng Laboratory, 6001 Shahe West Road, Nanshan District, Shenzhen 518055, Guangdong, China
| | - Shuqing Sun
- Shenzhen International Graduate School, Tsinghua University, 2279 Lishui Road, Nanshan District, Shenzhen 518055, Guangdong, China
| | - Huan Xu
- School of Public Health, Anhui University of Science and Technology, 15 Fengxia Road, Changfeng County, Hefei 231131, Anhui, China
| | - Qiang Cheng
- Department of Computer Science, University of Kentucky, 329 Rose Street, Lexington 40506, Kentucky, USA
- Institute for Biomedical Informatics, University of Kentucky, 800 Rose Street, Lexington 40506, Kentucky, USA
| | - Zhixiang Ren
- Pengcheng Laboratory, 6001 Shahe West Road, Nanshan District, Shenzhen 518055, Guangdong, China
| |
Collapse
|
34
|
Wei X, Chen T, Wang X, Shen W, Liu C, Wu S, Wong HS. COME: contrastive mapping learning for spatial reconstruction of single-cell RNA sequencing data. Bioinformatics 2025; 41:btaf083. [PMID: 39992219 PMCID: PMC11897431 DOI: 10.1093/bioinformatics/btaf083] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2024] [Revised: 02/12/2025] [Accepted: 02/20/2025] [Indexed: 02/25/2025] Open
Abstract
MOTIVATION Single-cell RNA sequencing (scRNA-seq) enables high-throughput transcriptomic profiling at single-cell resolution. The inherent spatial location is crucial for understanding how single cells orchestrate multicellular functions and drive diseases. However, spatial information is often lost during tissue dissociation. Spatial transcriptomic (ST) technologies can provide precise spatial gene expression atlas, while their practicality is constrained by the number of genes they can assay or the associated costs at a larger scale and the fine-grained cell-type annotation. By transferring knowledge between scRNA-seq and ST data through cell correspondence learning, it is possible to recover the spatial properties inherent in scRNA-seq datasets. RESULTS In this study, we introduce COME, a COntrastive Mapping lEarning approach that learns mapping between ST and scRNA-seq data to recover the spatial information of scRNA-seq data. Extensive experiments demonstrate that the proposed COME method effectively captures precise cell-spot relationships and outperforms previous methods in recovering spatial location for scRNA-seq data. More importantly, our method is capable of precisely identifying biologically meaningful information within the data, such as the spatial structure of missing genes, spatial hierarchical patterns, and the cell-type compositions for each spot. These results indicate that the proposed COME method can help to understand the heterogeneity and activities among cells within tissue environments. AVAILABILITY AND IMPLEMENTATION The COME is freely available in GitHub (https://github.com/cindyway/COME).
Collapse
Affiliation(s)
- Xindian Wei
- Department of Computer Science, City University of Hong Kong, Kowloon 999077, Hong Kong
| | - Tianyi Chen
- Department of Computer Science, City University of Hong Kong, Kowloon 999077, Hong Kong
| | - Xibiao Wang
- Department of Computer Science, Shantou University, Shantou 515063, China
| | - Wenjun Shen
- Department of Bioinformatics, Shantou University Medical College, Shantou 515041, China
| | - Cheng Liu
- Department of Computer Science, Shantou University, Shantou 515063, China
| | - Si Wu
- Department of Computer Science and Engineering, South China University of Technology, Guangzhou, Guangdong 510006, China
| | - Hau-San Wong
- Department of Computer Science, City University of Hong Kong, Kowloon 999077, Hong Kong
| |
Collapse
|
35
|
Chen J, Min W. sTPLS: identifying common and specific correlated patterns under multiple biological conditions. Brief Bioinform 2025; 26:bbaf195. [PMID: 40285361 PMCID: PMC12031727 DOI: 10.1093/bib/bbaf195] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2024] [Revised: 03/19/2025] [Accepted: 04/07/2025] [Indexed: 04/29/2025] Open
Abstract
The rapidly emerging large-scale data in diverse biological research fields present valuable opportunities to explore the underlying mechanisms of tissue development and disease progression. However, few existing methods can simultaneously capture common and condition-specific association between different types of features across different biological conditions, such as cancer types or cell populations. Therefore, we developed the sparse tensor-based partial least squares (sTPLS) method, which integrates multiple pairs of datasets containing two types of features but derived from different biological conditions. We demonstrated the effectiveness and versatility of sTPLS through simulation study and three biological applications. By integrating the pairwise pharmacogenomic data, sTPLS identified 11 gene-drug comodules with high biological functional relevance specific for seven cancer types and two comodules that shared across multi-type cancers, such as breast, ovarian, and colorectal cancers. When applied to single-cell data, it uncovered nine gene-peak comodules representing transcriptional regulatory relationships specific for five cell types and three comodules shared across similar cell types, such as intermediate and naïve B cells. Furthermore, sTPLS can be directly applied to tensor-structured data, successfully revealing shared and distinct cell communication patterns mediated by the MK signaling pathway in coronavirus disease 2019 patients and healthy controls. These results highlight the effectiveness of sTPLS in identifying biologically meaningful relationships across diverse conditions, making it useful for multi-omics integrative analysis.
Collapse
Affiliation(s)
- Jinyu Chen
- School of Mathematics, Statistics and Mechanics, Beijing University of Technology, 100 Pingleyuan, Chaoyang District, Beijing 100124, China
| | - Wenwen Min
- School of Information Science and Engineering, Yunnan University, East Outer Ring Road, Chenggong District, Kunming 650500, China
| |
Collapse
|
36
|
Liu C, Li X, Hu Q, Jia Z, Ye Q, Wang X, Zhao K, Liu L, Wang M. Decoding the blueprints of embryo development with single-cell and spatial omics. Semin Cell Dev Biol 2025; 167:22-39. [PMID: 39889540 DOI: 10.1016/j.semcdb.2025.01.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2023] [Revised: 01/18/2025] [Accepted: 01/18/2025] [Indexed: 02/03/2025]
Abstract
Embryonic development is a complex and intricately regulated process that encompasses precise control over cell differentiation, morphogenesis, and the underlying gene expression changes. Recent years have witnessed a remarkable acceleration in the development of single-cell and spatial omic technologies, enabling high-throughput profiling of transcriptomic and other multi-omic information at the individual cell level. These innovations offer fresh and multifaceted perspectives for investigating the intricate cellular and molecular mechanisms that govern embryonic development. In this review, we provide an in-depth exploration of the latest technical advancements in single-cell and spatial multi-omic methodologies and compile a systematic catalog of their applications in the field of embryonic development. We deconstruct the research strategies employed by recent studies that leverage single-cell sequencing techniques and underscore the unique advantages of spatial transcriptomics. Furthermore, we delve into both the current applications, data analysis algorithms and the untapped potential of these technologies in advancing our understanding of embryonic development. With the continuous evolution of multi-omic technologies, we anticipate their widespread adoption and profound contributions to unraveling the intricate molecular foundations underpinning embryo development in the foreseeable future.
Collapse
Affiliation(s)
- Chang Liu
- BGI Research, Hangzhou 310030, China; BGI Research, Shenzhen 518083, China; Shanxi Medical University-BGI Collaborative Center for Future Medicine, Shanxi Medical University, Taiyuan 030001, China; Shenzhen Proof-of-Concept Center of Digital Cytopathology, BGI Research, Shenzhen 518083, China
| | | | - Qinan Hu
- Shenzhen Key Laboratory of Gene Regulation and Systems Biology, School of Life Sciences, Southern University of Science and Technology, Shenzhen 518005, China; Department of Pharmacology, School of Medicine, Southern University of Science and Technology, Shenzhen 518005, China
| | - Zihan Jia
- BGI Research, Hangzhou 310030, China; College of Life Sciences, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Qing Ye
- BGI Research, Hangzhou 310030, China; China Jiliang University, Hangzhou 310018, China
| | | | - Kaichen Zhao
- College of Biomedicine and Health, College of Life Science and Technology, Huazhong Agricultural University, Wuhan, Hubei 430070, China
| | - Longqi Liu
- BGI Research, Hangzhou 310030, China; Shanxi Medical University-BGI Collaborative Center for Future Medicine, Shanxi Medical University, Taiyuan 030001, China.
| | - Mingyue Wang
- BGI Research, Hangzhou 310030, China; Key Laboratory of Spatial Omics of Zhejiang Province, BGI Research, Hangzhou 310030, China.
| |
Collapse
|
37
|
Liang M, Chu L, Yue Z. New Multiomic Studies Shed Light on Cellular Diversity and Neuronal Susceptibility in Parkinson's Disease. Mov Disord 2025; 40:431-437. [PMID: 39812497 DOI: 10.1002/mds.30097] [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: 11/13/2024] [Revised: 12/03/2024] [Accepted: 12/11/2024] [Indexed: 01/16/2025] Open
Abstract
Parkinson's disease is a complex neurodegenerative disorder characterized by degeneration of dopaminergic neurons, with patients manifesting varying motor and nonmotor symptoms. Previous studies using single-cell RNA sequencing in rodent models and humans have identified distinct heterogeneity of neurons and glial cells with differential vulnerability. Recent studies have increasingly leveraged multiomics approaches, including spatial transcriptomics, epigenomics, and proteomics, in the study of Parkinson's disease, providing new insights into pathogenic mechanisms. Continued advancements in experimental technologies and sophisticated computational tools will be essential in uncovering a network of neuronal vulnerability and prioritizing disease modifiers for novel therapeutics development. © 2025 International Parkinson and Movement Disorder Society.
Collapse
Affiliation(s)
- Marianna Liang
- Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, New York, USA
- Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, New York, USA
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, New York, USA
- Center for Parkinson's Disease Neurobiology, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Linh Chu
- Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, New York, USA
- Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, New York, USA
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, New York, USA
- Center for Parkinson's Disease Neurobiology, Icahn School of Medicine at Mount Sinai, New York, New York, USA
- Department of Genetics & Genomic Sciences, Mount Sinai Center for Transformative Disease Modeling, Icahn Genomics Institute, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Zhenyu Yue
- Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, New York, USA
- Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, New York, USA
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, New York, USA
- Center for Parkinson's Disease Neurobiology, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| |
Collapse
|
38
|
Zhang H, Patrick MT, Zhao J, Zhai X, Liu J, Li Z, Gu Y, Welch J, Zhou X, Modlin RL, Tsoi LC, Gudjonsson JE. Techniques and analytic workflow for spatial transcriptomics and its application to allergy and inflammation. J Allergy Clin Immunol 2025; 155:678-687. [PMID: 39837466 PMCID: PMC11875981 DOI: 10.1016/j.jaci.2025.01.009] [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: 08/02/2024] [Revised: 01/02/2025] [Accepted: 01/14/2025] [Indexed: 01/23/2025]
Abstract
Spatial profiling, through single-cell gene-level expression data paired with cell localization, offers unprecedented biologic insights within the intact spatial context of cells in healthy and diseased tissue, adding a novel dimension to data interpretation. This review summarizes recent developments in this field, its application to allergy and inflammation, and recent single-cell resolution platforms designed for spatial transcriptomics with a focus on data processing and analyses for efficient biologic interpretation of data. By preserving spatial context, these technologies provide critical insights into tissue architecture and cellular interactions that are unattainable with traditional transcriptomics methods, such as revealing localized inflammatory cell network in atopic dermatitis and T-cell interactions in the lung in chronic obstructive pulmonary disease. Spatial profiling offers opportunities for discovering novel biomarkers, defining compartmentalization of immune responses within tissues and individual diseases, and accelerating novel discoveries toward a greater understanding of fundamental disease mechanisms and, eventually, toward the development of future targeted therapies.
Collapse
Affiliation(s)
- Haihan Zhang
- Department of Biostatistics, University of Michigan, Ann Arbor, Mich
| | - Matthew T Patrick
- Department of Dermatology, University of Michigan Medical School, Ann Arbor, Mich
| | - Jingyu Zhao
- Department of Biostatistics, University of Michigan, Ann Arbor, Mich
| | - Xintong Zhai
- Department of Biostatistics, University of Michigan, Ann Arbor, Mich
| | - Jialin Liu
- Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, Mich
| | - Zheng Li
- Department of Biostatistics, University of Michigan, Ann Arbor, Mich
| | - Yiqian Gu
- Department of Internal Medicine, Division of Dermatology, UCLA, Los Angeles, Calif
| | - Joshua Welch
- Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, Mich; Department of Electrical Engineering and Computer Science, University of Michigan, Ann Arbor, Mich
| | - Xiang Zhou
- Department of Biostatistics, University of Michigan, Ann Arbor, Mich
| | - Robert L Modlin
- Department of Internal Medicine, Division of Dermatology, UCLA, Los Angeles, Calif
| | - Lam C Tsoi
- Department of Biostatistics, University of Michigan, Ann Arbor, Mich; Department of Dermatology, University of Michigan Medical School, Ann Arbor, Mich; Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, Mich.
| | - Johann E Gudjonsson
- Department of Dermatology, University of Michigan Medical School, Ann Arbor, Mich; Taubman Medical Research Institute, University of Michigan Medical School, Ann Arbor, Mich.
| |
Collapse
|
39
|
Ortega-Batista A, Jaén-Alvarado Y, Moreno-Labrador D, Gómez N, García G, Guerrero EN. Single-Cell Sequencing: Genomic and Transcriptomic Approaches in Cancer Cell Biology. Int J Mol Sci 2025; 26:2074. [PMID: 40076700 PMCID: PMC11901077 DOI: 10.3390/ijms26052074] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2024] [Revised: 02/18/2025] [Accepted: 02/24/2025] [Indexed: 03/14/2025] Open
Abstract
This article reviews the impact of single-cell sequencing (SCS) on cancer biology research. SCS has revolutionized our understanding of cancer and tumor heterogeneity, clonal evolution, and the complex interplay between cancer cells and tumor microenvironment. SCS provides high-resolution profiling of individual cells in genomic, transcriptomic, and epigenomic landscapes, facilitating the detection of rare mutations, the characterization of cellular diversity, and the integration of molecular data with phenotypic traits. The integration of SCS with multi-omics has provided a multidimensional view of cellular states and regulatory mechanisms in cancer, uncovering novel regulatory mechanisms and therapeutic targets. Advances in computational tools, artificial intelligence (AI), and machine learning have been crucial in interpreting the vast amounts of data generated, leading to the identification of new biomarkers and the development of predictive models for patient stratification. Furthermore, there have been emerging technologies such as spatial transcriptomics and in situ sequencing, which promise to further enhance our understanding of tumor microenvironment organization and cellular interactions. As SCS and its related technologies continue to advance, they are expected to drive significant advances in personalized cancer diagnostics, prognosis, and therapy, ultimately improving patient outcomes in the era of precision oncology.
Collapse
Affiliation(s)
- Ana Ortega-Batista
- Faculty of Science and Technology, Technological University of Panama, Ave Justo Arosemena, Entre Calle 35 y 36, Corregimiento de Calidonia, Panama City, Panama; (A.O.-B.)
| | - Yanelys Jaén-Alvarado
- Faculty of Science and Technology, Technological University of Panama, Ave Justo Arosemena, Entre Calle 35 y 36, Corregimiento de Calidonia, Panama City, Panama; (A.O.-B.)
- Gorgas Memorial Institute for Health Studies, Ave Justo Arosemena, Entre Calle 35 y 36, Corregimiento de Calidonia, Panama City, Panama
| | - Dilan Moreno-Labrador
- Faculty of Science and Technology, Technological University of Panama, Ave Justo Arosemena, Entre Calle 35 y 36, Corregimiento de Calidonia, Panama City, Panama; (A.O.-B.)
| | - Natasha Gómez
- Faculty of Science and Technology, Technological University of Panama, Ave Justo Arosemena, Entre Calle 35 y 36, Corregimiento de Calidonia, Panama City, Panama; (A.O.-B.)
| | - Gabriela García
- Faculty of Science and Technology, Technological University of Panama, Ave Justo Arosemena, Entre Calle 35 y 36, Corregimiento de Calidonia, Panama City, Panama; (A.O.-B.)
| | - Erika N. Guerrero
- Gorgas Memorial Institute for Health Studies, Ave Justo Arosemena, Entre Calle 35 y 36, Corregimiento de Calidonia, Panama City, Panama
- Sistema Nacional de Investigación, Secretaria Nacional de Ciencia y Tecnología, Edificio 205, Ciudad del Saber, Panama City, Panama
| |
Collapse
|
40
|
Hashikawa K, Hashikawa Y, Briones B, Ishii K, Liu Y, Rossi MA, Basiri ML, Chen JY, Ahmad OR, Mukundan RV, Johnston NL, Simon RC, Soetedjo JC, Siputro JR, McHenry JA, Palmiter RD, Rubinow DR, Zweifel LS, Stuber GD. Esr1-Dependent Signaling and Transcriptional Maturation in the Medial Preoptic Area of the Hypothalamus Shapes the Development of Mating Behavior during Adolescence. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.02.26.640339. [PMID: 40060480 PMCID: PMC11888408 DOI: 10.1101/2025.02.26.640339] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 03/27/2025]
Abstract
Mating and other behaviors emerge during adolescence through the coordinated actions of steroid hormone signaling throughout the nervous system and periphery. In this study, we investigated the transcriptional dynamics of the medial preoptic area (MPOA), a critical region for reproductive behavior, using single-cell RNA sequencing (scRNAseq) and in situ hybridization techniques in male and female mice throughout adolescence development. Our findings reveal that estrogen receptor 1 (Esr1) plays a pivotal role in the transcriptional maturation of GABAergic neurons within the MPOA during adolescence. Deletion of the estrogen receptor gene, Esr1, in GABAergic neurons (Vgat+) disrupted the developmental progression of mating behaviors in both sexes, while its deletion in glutamatergic neurons (Vglut2+) had no observable effect. In males and females, these neurons displayed distinct transcriptional trajectories, with hormone-dependent gene expression patterns emerging throughout adolescence and regulated by Esr1. Esr1 deletion in MPOA GABAergic neurons, prior to adolescence, arrested adolescent transcriptional progression of these cells and uncovered sex-specific gene-regulatory networks associated with Esr1 signaling. Our results underscore the critical role of Esr1 in orchestrating sex-specific transcriptional dynamics during adolescence, revealing gene regulatory networks implicated in the development of hypothalamic controlled reproductive behaviors.
Collapse
Affiliation(s)
- Koichi Hashikawa
- Center for the Neurobiology of Addiction, Pain, and Emotion, Department of Anesthesiology and Pain Medicine, Department of Pharmacology, University of Washington, Seattle, WA 98195
| | - Yoshiko Hashikawa
- Center for the Neurobiology of Addiction, Pain, and Emotion, Department of Anesthesiology and Pain Medicine, Department of Pharmacology, University of Washington, Seattle, WA 98195
| | - Brandy Briones
- Center for the Neurobiology of Addiction, Pain, and Emotion, Department of Anesthesiology and Pain Medicine, Department of Pharmacology, University of Washington, Seattle, WA 98195
| | - Kentaro Ishii
- Center for the Neurobiology of Addiction, Pain, and Emotion, Department of Anesthesiology and Pain Medicine, Department of Pharmacology, University of Washington, Seattle, WA 98195
| | - Yuejia Liu
- Center for the Neurobiology of Addiction, Pain, and Emotion, Department of Anesthesiology and Pain Medicine, Department of Pharmacology, University of Washington, Seattle, WA 98195
| | - Mark A. Rossi
- Center for the Neurobiology of Addiction, Pain, and Emotion, Department of Anesthesiology and Pain Medicine, Department of Pharmacology, University of Washington, Seattle, WA 98195
| | - Marcus L. Basiri
- Center for the Neurobiology of Addiction, Pain, and Emotion, Department of Anesthesiology and Pain Medicine, Department of Pharmacology, University of Washington, Seattle, WA 98195
- University of North Carolina, Chapel Hill, NC 27599
| | - Jane Y. Chen
- Department of Biochemistry, University of Washington, Seattle, WA 98195
| | - Omar R. Ahmad
- Center for the Neurobiology of Addiction, Pain, and Emotion, Department of Anesthesiology and Pain Medicine, Department of Pharmacology, University of Washington, Seattle, WA 98195
| | - Rishi V. Mukundan
- Center for the Neurobiology of Addiction, Pain, and Emotion, Department of Anesthesiology and Pain Medicine, Department of Pharmacology, University of Washington, Seattle, WA 98195
| | - Nathan L. Johnston
- Center for the Neurobiology of Addiction, Pain, and Emotion, Department of Anesthesiology and Pain Medicine, Department of Pharmacology, University of Washington, Seattle, WA 98195
| | - Rhiana C. Simon
- Center for the Neurobiology of Addiction, Pain, and Emotion, Department of Anesthesiology and Pain Medicine, Department of Pharmacology, University of Washington, Seattle, WA 98195
| | - James C. Soetedjo
- Center for the Neurobiology of Addiction, Pain, and Emotion, Department of Anesthesiology and Pain Medicine, Department of Pharmacology, University of Washington, Seattle, WA 98195
| | - Jason R. Siputro
- Center for the Neurobiology of Addiction, Pain, and Emotion, Department of Anesthesiology and Pain Medicine, Department of Pharmacology, University of Washington, Seattle, WA 98195
| | - Jenna A. McHenry
- Department of Psychology & Neuroscience, Duke University, Durham, NC 27708
| | - Richard D. Palmiter
- Department of Biochemistry, University of Washington, Seattle, WA 98195
- Howard Hughes Medical Institute, University of Washington, Seattle, WA 98195
| | - David R. Rubinow
- Department of Psychiatry, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599
| | - Larry S. Zweifel
- Department of Psychiatry and Behavioral Sciences, University of Washington, Seattle, WA 98195
- Department of Pharmacology, University of Washington, Seattle, WA 98195
| | - Garret D. Stuber
- Center for the Neurobiology of Addiction, Pain, and Emotion, Department of Anesthesiology and Pain Medicine, Department of Pharmacology, University of Washington, Seattle, WA 98195
| |
Collapse
|
41
|
Saddic L, Kaneda G, Momenzadeh A, Zilberberg L, Song Y, Mastali M, Kreimer S, Hutton A, Haghani A, Meyer J, Parker S. Single Cell Proteomics Reveals Novel Cell Phenotypes in Marfan Mouse Aneurysm. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.02.15.638465. [PMID: 40027651 PMCID: PMC11870452 DOI: 10.1101/2025.02.15.638465] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/05/2025]
Abstract
Background Single-cell omics technology is a powerful tool in biomedical research. However, single cell proteomics has lagged due to an inability to amplify peptides in a similar fashion to nucleotide strings. Single cell proteomics is important because proteins are the main functional unit in cells, and they often poorly correlate with mRNA quantities. In this paper we describe the first single cell proteomic analysis of complex tissue, comparing aneurysmal and normal mouse aorta from males and females. We also compare and integrate our single cell proteomic profiles with a matching single cell transcriptomics dataset. Methods We compared single cell proteomes between male and female, wild-type and Fbn1 C1041G/+ Marfan mice (N=3 per group). Individual cells from mouse aortic root single cell suspensions were deposited in 384 well plates and subjected to ultra-sensitive nanoflow liquid chromatography-ion mobility-time of flight-mass spectrometry. The data were analyzed with leiden clustering to identify cell types. Statistical analyses were performed to detect differential proteins within cell types and multi-omics analysis integrated single cell proteomics with published single cell RNA-seq. Results We identified all major aortic cell types including 7 distinct smooth muscle cell subtypes. The proportion of these cells varied based on sex and the Fbn1 C1041G/+ genotype. Differentially expressed proteins between male and female in addition to wild-type and Marfan samples uncovered enhanced endothelial to mesenchymal transition patterns in endothelial cells from male Marfan mice. Comparisons between single cell RNA and single cell proteomic profiles showed similarities in major subtypes but not smooth muscle cell subtypes. Multi-omics analysis of these two single cell platforms demonstrated a potential novel role for smooth muscle cell derived angiotensin signaling in the Marfan phenotype. Conclusions Single cell proteomics identified new subpopulations of vascular smooth muscles cells and novel cell type specific protein signatures related to sex differences and aneurysm formation. Abbreviations Next generation sequencing (NGS), Mass spectrometer (MS), Single cell proteomics by Mass Spectrometry (ScOPE-MS), Marfan's syndrome (MFS), Fibrillin 1 (FBN1), Transforming growth factor β (TGFβ), Smooth muscle cell (SMC), Single cell proteomic (scProteomic), Differentially expressed proteins (DEPs), Wild-type (WT), Hanks' balanced salt solution (HBSS), Fetal bovine serum (FBS), Dulbecco's Modified Eagle Medium (DMEM), Data-independent acquisition parallel accumulation-serial fragmentation (DIA-PASEF), Magnetic assisted cell sorted (MACS), Single Cell Analysis in Python (Scanpy), Kyoto Encyclopedia of Genes and Genomes (KEGG), Principal component analysis (PCA), Uniform manifold projection (UMAP), Single cell transcriptomic (scTranscriptomic), Smoothelin (Smtn), Transgelin (Tagln), Myosin heavy chain 11 (Myh11), Platelet endothelial cell adhesion molecule 1 (Pecam1), Dipeptidase 1 (Dpep1), Uncoupling protein 1 (Ucp1), Low-density lipoprotein receptor-related protein (Lrp1), DNA ligase 3 (Lig3), Capsaicin channel transient receptor potential vanilloid 1 (Trpv1), Endothelial to mesenchymal transition (endMT), Intercellular adhesion molecule 1 (Icam1), Intercellular adhesion molecule 2 (Icam2), Endothelial cell-selective adhesion molecule (Esam), Calponin 1 (Cnn1), Vimentin (Vim), Zinc finger E-box-binding homeobox 1 (Zeb1), Snail family transcriptional repressor 1 (Snai1), Tropomyosin alpha-4 chain (Tpm4), Angiotensin converting enzyme (Ace).
Collapse
|
42
|
Wu J, Wan C, Ji Z, Zhou Y, Hou W. EpiFoundation: A Foundation Model for Single-Cell ATAC-seq via Peak-to-Gene Alignment. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.02.05.636688. [PMID: 39975086 PMCID: PMC11839112 DOI: 10.1101/2025.02.05.636688] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/21/2025]
Abstract
Foundation models exhibit strong capabilities for downstream tasks by learning generalized representations through self-supervised pre-training on large datasets. While several foundation models have been developed for single-cell RNA-seq (scRNA-seq) data, there is still a lack of models specifically tailored for single-cell ATAC-seq (scATAC-seq), which measures epigenetic information in individual cells. The principal challenge in developing such a model lies in the vast number of scATAC peaks and the significant sparsity of the data, which complicates the formulation of peak-to-peak correlations. To address this challenge, we introduce EpiFoundation, a foundation model for learning cell representations from the high-dimensional and sparse space of peaks. EpiFoundation relies on an innovative cross-modality pre-training procedure with two key technical innovations. First, EpiFoundation exclusively processes the non-zero peak set, thereby enhancing the density of cell-specific information within the input data. Second, EpiFoundation utilizes dense gene expression information to supervise the pre-training process, aligning peak-to-gene correlations. EpiFoundation can handle various types of downstream tasks, including cell-type annotation, batch correction, and gene expression prediction. To train and validate EpiFoundation, we curated MiniAtlas, a dataset of 100,000+ single cells with paired scRNA-seq and scATAC-seq data, along with diverse test sets spanning various tissues and cell types for robust evaluation. EpiFoundation demonstrates state-of-the-art performance across multiple tissues and diverse downstream tasks.
Collapse
Affiliation(s)
- Juncheng Wu
- Department of Computer Science and Engineering, UC Santa Cruz
| | - Changxin Wan
- Department of Biostatistics and Bioinformatics, Duke University
| | - Zhicheng Ji
- Department of Biostatistics and Bioinformatics, Duke University
| | - Yuyin Zhou
- Department of Computer Science and Engineering, UC Santa Cruz
| | - Wenpin Hou
- Department of Biostatistics, Mailman School of Public Health, Columbia University
| |
Collapse
|
43
|
Wilson AM, Jacobs MM, Lambert TY, Valada A, Meloni G, Gilmore E, Murray J, Morgello S, Akbarian S. Transcriptional impacts of substance use disorder and HIV on human ventral midbrain neurons and microglia. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.02.05.636667. [PMID: 39974894 PMCID: PMC11838593 DOI: 10.1101/2025.02.05.636667] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/21/2025]
Abstract
For people with HIV (PWH), substance use disorders (SUDs) are a prominent neurological risk factor, and the impacts of both on dopaminergic pathways are a potential point of deleterious convergence. Here, we profile, at single nucleus resolution, the substantia nigra (SN) transcriptomes of 90 postmortem donors in the context of chronic HIV and opioid/cocaine SUD, including 67 prospectively characterized PWH. We report altered microglial expression for hundreds of pro- and anti-inflammatory regulators attributable to HIV, and separately, to SUD. Stepwise, progressive microglial dysregulation, coupled to altered SN dopaminergic and GABAergic signaling, was associated with SUD/HIV dual diagnosis and further with lack of viral suppression in blood. In virologically suppressed donors, SUD comorbidity was associated with microglial transcriptional changes permissive for HIV infection. We report HIV-related downregulation of monoamine reuptake transporters specifically in dopaminergic neurons regardless of SUD status or viral load, and additional transcriptional signatures consistent with selective vulnerability of SN dopamine neurons.
Collapse
Affiliation(s)
- Alyssa M. Wilson
- Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Michelle M. Jacobs
- Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Tova Y. Lambert
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Aditi Valada
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Gregory Meloni
- Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Evan Gilmore
- Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Jacinta Murray
- Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Susan Morgello
- Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Department of Pathology, Molecular and Cell Based Medicine, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Schahram Akbarian
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| |
Collapse
|
44
|
Zheng L, Gu M, Li X, Hu X, Chen C, Kang Y, Pan B, Chen W, Xian G, Wu X, Li C, Wang C, Li Z, Guan M, Zhou G, Mobasheri A, Song W, Peng S, Sheng P, Zhang Z. ITGA5 + synovial fibroblasts orchestrate proinflammatory niche formation by remodelling the local immune microenvironment in rheumatoid arthritis. Ann Rheum Dis 2025; 84:232-252. [PMID: 39919897 DOI: 10.1136/ard-2024-225778] [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: 03/08/2024] [Accepted: 10/17/2024] [Indexed: 11/04/2024]
Abstract
OBJECTIVES To investigate the phenotypic heterogeneity of tissue-resident synovial fibroblasts and their role in inflammatory response in rheumatoid arthritis (RA). METHODS We used single-cell and spatial transcriptomics to profile synovial cells and spatial gene expressions of synovial tissues to identify phenotypic changes in patients with osteoarthritis, RA in sustained remission and active state. Immunohistology, multiplex immunofluorescence and flow cytometry were used to identify synovial fibroblasts subsets. Deconvolution methods further validated our findings in two cohorts (PEAC and R4RA) with treatment response. Cell coculture was used to access the potential cell-cell interactions. Adoptive transfer of synovial cells in collagen-induced arthritis (CIA) mice and bulk RNA sequencing of synovial joints further validate the cellular functions. RESULTS We identified a novel tissue-remodelling CD45-CD31-PDPN+ITGA5+ synovial fibroblast population with unique transcriptome of POSTN, COL3A1, CCL5 and TGFB1, and enriched in immunoregulatory pathways. This subset was upregulated in active and lympho-myeloid type of RA, associated with an increased risk of multidrug resistance. Transforming growth factor (TGF)-β1 might participate in the differentiation of this subset. Moreover, ITGA5+ synovial fibroblasts might occur in early stage of inflammation and induce the differentiation of CXCL13hiPD-1hi peripheral helper T cells (TPHs) from naïve CD4+ T cells, by secreting TGF-β1. Intra-articular injection of ITGA5+ synovial fibroblasts exacerbates RA development and upregulates TPHs in CIA mice. CONCLUSIONS We demonstrate that ITGA5+ synovial fibroblasts might regulate the RA progression by inducing the differentiation of CXCL13hiPD-1hi TPHs and remodelling the proinflammatory microenvironments. Therapeutic modulation of this subpopulation could therefore be a potential treatment strategy for RA.
Collapse
Affiliation(s)
- Linli Zheng
- Department of Joint Surgery, Sun Yat-sen University First Affiliated Hospital, Guangzhou, Guangdong, China; Guangdong Provincial Key Laboratory of Orthopaedics and Traumatology, Sun Yat-sen University First Affiliated Hospital, Guangzhou, Guangdong, China
| | - Minghui Gu
- Department of Joint Surgery, Sun Yat-sen University First Affiliated Hospital, Guangzhou, Guangdong, China; Guangdong Provincial Key Laboratory of Orthopaedics and Traumatology, Sun Yat-sen University First Affiliated Hospital, Guangzhou, Guangdong, China
| | - Xiang Li
- Guangdong Provincial Key Laboratory of Orthopaedics and Traumatology, Sun Yat-sen University First Affiliated Hospital, Guangzhou, Guangdong, China; Department of Spine Surgery, Sun Yat-sen University First Affiliated Hospital, Guangzhou, Guangdong, China
| | - Xuantao Hu
- Department of Joint Surgery, Sun Yat-sen University First Affiliated Hospital, Guangzhou, Guangdong, China; Guangdong Provincial Key Laboratory of Orthopaedics and Traumatology, Sun Yat-sen University First Affiliated Hospital, Guangzhou, Guangdong, China; Department of Spine Surgery, Sun Yat-sen University Third Affiliated Hospital, Guangzhou, Guangdong, China
| | - Chen Chen
- Trauma Orthopedics, Foot and Ankle Surgery, Sun Yat-sen Memorial Hostpial, Guangzhou, Guangdong, China; Institute of Precision Medicine, Sun Yat-sen University First Affiliated Hospital, Guangzhou, Guangdong, China
| | - Yunze Kang
- Department of Joint Surgery, Sun Yat-sen University First Affiliated Hospital, Guangzhou, Guangdong, China; Guangdong Provincial Key Laboratory of Orthopaedics and Traumatology, Sun Yat-sen University First Affiliated Hospital, Guangzhou, Guangdong, China
| | - Baiqi Pan
- Department of Joint Surgery, Sun Yat-sen University First Affiliated Hospital, Guangzhou, Guangdong, China; Guangdong Provincial Key Laboratory of Orthopaedics and Traumatology, Sun Yat-sen University First Affiliated Hospital, Guangzhou, Guangdong, China
| | - Weishen Chen
- Department of Joint Surgery, Sun Yat-sen University First Affiliated Hospital, Guangzhou, Guangdong, China; Guangdong Provincial Key Laboratory of Orthopaedics and Traumatology, Sun Yat-sen University First Affiliated Hospital, Guangzhou, Guangdong, China
| | | | - Xiaoyu Wu
- Department of Joint Surgery, Sun Yat-sen University First Affiliated Hospital, Guangzhou, Guangdong, China; Guangdong Provincial Key Laboratory of Orthopaedics and Traumatology, Sun Yat-sen University First Affiliated Hospital, Guangzhou, Guangdong, China
| | - Chengxin Li
- Department of Joint Surgery, Sun Yat-sen University First Affiliated Hospital, Guangzhou, Guangdong, China; Guangdong Provincial Key Laboratory of Orthopaedics and Traumatology, Sun Yat-sen University First Affiliated Hospital, Guangzhou, Guangdong, China
| | - Chao Wang
- Department of Joint Surgery, Sun Yat-sen University First Affiliated Hospital, Guangzhou, Guangdong, China; Guangdong Provincial Key Laboratory of Orthopaedics and Traumatology, Sun Yat-sen University First Affiliated Hospital, Guangzhou, Guangdong, China
| | - Zhiwen Li
- Department of Joint Surgery, Sun Yat-sen University First Affiliated Hospital, Guangzhou, Guangdong, China; Guangdong Provincial Key Laboratory of Orthopaedics and Traumatology, Sun Yat-sen University First Affiliated Hospital, Guangzhou, Guangdong, China
| | - Mingqiang Guan
- Department of Orthopedics and Traumatology, Foshan Hospital of Traditional Chinese Medicine, Foshan, Guangdong, China
| | - Guanming Zhou
- Department of Orthopedics and Traumatology, Foshan Hospital of Traditional Chinese Medicine, Foshan, Guangdong, China
| | - Ali Mobasheri
- Research Unit of Medical Imaging, Physics and Technology, Faculty of Medicine, University of Oulu, Oulu, Finland; Department of Regenerative Medicine, State Research Institute Centre for Innovative Medicine, Vilnius, Lithuania; Public Health Aspects of Musculoskeletal Health and Aging, World Health Organization Collaborating Centre, Liege, Belgium
| | - Weidong Song
- Trauma Orthopedics, Foot and Ankle Surgery, Sun Yat-sen Memorial Hostpial, Guangzhou, Guangdong, China
| | - Sui Peng
- Institute of Precision Medicine, Sun Yat-sen University First Affiliated Hospital, Guangzhou, Guangdong, China; Clinical Trials Unit, Sun Yat-sen University First Affiliated Hospital, Guangzhou, Guangdong, China; Department of Gastroenterology and Hepatology, Sun Yat-sen University First Affiliated Hospital, Guangzhou, Guangdong, China.
| | - Puyi Sheng
- Department of Joint Surgery, Sun Yat-sen University First Affiliated Hospital, Guangzhou, Guangdong, China; Guangdong Provincial Key Laboratory of Orthopaedics and Traumatology, Sun Yat-sen University First Affiliated Hospital, Guangzhou, Guangdong, China.
| | - Ziji Zhang
- Department of Joint Surgery, Sun Yat-sen University First Affiliated Hospital, Guangzhou, Guangdong, China; Guangdong Provincial Key Laboratory of Orthopaedics and Traumatology, Sun Yat-sen University First Affiliated Hospital, Guangzhou, Guangdong, China.
| |
Collapse
|
45
|
Yang P, Jin K, Yao Y, Jin L, Shao X, Li C, Lu X, Fan X. Spatial integration of multi-omics single-cell data with SIMO. Nat Commun 2025; 16:1265. [PMID: 39893194 PMCID: PMC11787318 DOI: 10.1038/s41467-025-56523-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2024] [Accepted: 01/16/2025] [Indexed: 02/04/2025] Open
Abstract
Technical limitations in spatial and single-cell omics sequencing pose challenges for capturing and describing multimodal information at the spatial scale. To address this, we develop SIMO, a computational method designed for the Spatial Integration of Multi-Omics datasets through probabilistic alignment. Unlike previous tools, SIMO not only integrates spatial transcriptomics with single-cell RNA-seq but expands beyond, enabling integration across multiple single-cell modalities, such as chromatin accessibility and DNA methylation, which have not been co-profiled spatially before. We benchmark SIMO on simulated datasets, demonstrating its high accuracy and robustness. Further application on biological datasets reveals SIMO's ability to detect topological patterns of cells and their regulatory modes across multiple omics layers. Through comprehensive analysis of real-world data, SIMO uncovers multimodal spatial heterogeneity, offering deeper insights into the spatial organization and regulation of biological molecules. These findings position SIMO as a powerful tool for advancing spatial biology by revealing previously inaccessible multimodal insights.
Collapse
Affiliation(s)
- Penghui Yang
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China
- National Key Laboratory of Chinese Medicine Modernization, Innovation Center of Yangtze River Delta, Zhejiang University, Jiaxing, 314103, China
| | - Kaiyu Jin
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China
| | - Yue Yao
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China
| | - Lijun Jin
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China
- National Key Laboratory of Chinese Medicine Modernization, Innovation Center of Yangtze River Delta, Zhejiang University, Jiaxing, 314103, China
| | - Xin Shao
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China
- National Key Laboratory of Chinese Medicine Modernization, Innovation Center of Yangtze River Delta, Zhejiang University, Jiaxing, 314103, China
| | - Chengyu Li
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China
- National Key Laboratory of Chinese Medicine Modernization, Innovation Center of Yangtze River Delta, Zhejiang University, Jiaxing, 314103, China
| | - Xiaoyan Lu
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China.
- National Key Laboratory of Chinese Medicine Modernization, Innovation Center of Yangtze River Delta, Zhejiang University, Jiaxing, 314103, China.
| | - Xiaohui Fan
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China.
- National Key Laboratory of Chinese Medicine Modernization, Innovation Center of Yangtze River Delta, Zhejiang University, Jiaxing, 314103, China.
- The Joint-laboratory of clinical multi-omics research between Zhejiang University and Ningbo Municipal Hospital of TCM, Ningbo Municipal Hospital of TCM, Ningbo, 315012, China.
- College of Chemistry & Chemical Engineering, Shaoxing University, Shaoxing, PR China.
| |
Collapse
|
46
|
Li B, Tang Z, Budhkar A, Liu X, Zhang T, Yang B, Su J, Song Q. SpaIM: Single-cell Spatial Transcriptomics Imputation via Style Transfer. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.01.24.634756. [PMID: 39975319 PMCID: PMC11838188 DOI: 10.1101/2025.01.24.634756] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 02/21/2025]
Abstract
Spatial transcriptomics (ST) technologies have revolutionized our understanding of cellular ecosystems. However, these technologies face challenges such as sparse gene signals and limited gene detection capacities, which hinder their ability to fully capture comprehensive spatial gene expression profiles. To address these limitations, we propose leveraging single-cell RNA sequencing (scRNA-seq), which provides comprehensive gene expression data but lacks spatial context, to enrich ST profiles. Herein, we introduce SpaIM, an innovative style transfer learning model that utilizes scRNA-seq information to predict unmeasured gene expressions in ST data, thereby improving gene coverage and expressions. SpaIM segregates scRNA-seq and ST data into data-agnostic contents and data-specific styles, with the contents capture the commonalities between the two data types, while the styles highlight their unique differences. By integrating the strengths of scRNA-seq and ST, SpaIM overcomes data sparsity and limited gene coverage issues, making significant advancements over 12 existing methods. This improvement is demonstrated across 53 diverse ST datasets, spanning sequencing- and imaging-based spatial technologies in various tissue types. Additionally, SpaIM enhances downstream analyses, including the detection of ligand-receptor interactions, spatial domain characterization, and identification of differentially expressed genes. Released as open-source software, SpaIM increases accessibility for spatial transcriptomics analysis. In summary, SpaIM represents a pioneering approach to enrich spatial transcriptomics using scRNA-seq data, enabling precise gene expression imputation and advancing the field of spatial transcriptomics research.
Collapse
Affiliation(s)
- Bo Li
- Department of Computer and Information Science, University of Macau, Taipa, Macau SAR, China
| | - Ziyang Tang
- Department of Computer and Information Technology, Purdue University, Indiana, USA
| | - Aishwarya Budhkar
- Department of Biostatistics and Health Data Science, Indiana University School of Medicine, Indiana, USA
| | - Xiang Liu
- Department of Biostatistics and Health Data Science, Indiana University School of Medicine, Indiana, USA
| | - Tonglin Zhang
- Department of Statistics, Purdue University, Indiana, USA
| | - Baijian Yang
- Department of Computer and Information Technology, Purdue University, Indiana, USA
| | - Jing Su
- Department of Biostatistics and Health Data Science, Indiana University School of Medicine, Indiana, USA
| | - Qianqian Song
- Department of Cancer Biology, Wake Forest University School of Medicine, North Carolina, USA
- Department of Health Outcomes and Biomedical Informatics, College of Medicine, University of Florida, Florida, USA
| |
Collapse
|
47
|
Zhao M, Jankovic D, Link VM, Souza COS, Hornick KM, Oyesola O, Belkaid Y, Lack J, Loke P. Genetic variation in IL-4 activated tissue resident macrophages determines strain-specific synergistic responses to LPS epigenetically. Nat Commun 2025; 16:1030. [PMID: 39863579 PMCID: PMC11762786 DOI: 10.1038/s41467-025-56379-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2024] [Accepted: 01/16/2025] [Indexed: 01/27/2025] Open
Abstract
How macrophages in the tissue environment integrate multiple stimuli depends on the genetic background of the host, but this is still poorly understood. We investigate IL-4 activation of male C57BL/6 and BALB/c strain specific in vivo tissue-resident macrophages (TRMs) from the peritoneal cavity. C57BL/6 TRMs are more transcriptionally responsive to IL-4 stimulation, with induced genes associated with more super enhancers, induced enhancers, and topologically associating domains (TAD) boundaries. IL-4-directed epigenomic remodeling reveals C57BL/6 specific enrichment of NF-κB, IRF, and STAT motifs. Additionally, IL-4-activated C57BL/6 TRMs demonstrate an augmented synergistic response upon in vitro lipopolysaccharide (LPS) exposure, despite naïve BALB/c TRMs displaying a more robust transcriptional response to LPS. Single-cell RNA sequencing (scRNA-seq) analysis of mixed bone marrow chimeras indicates that transcriptional differences and synergy are cell intrinsic within the same tissue environment. Hence, genetic variation alters IL-4-induced cell intrinsic epigenetic reprogramming resulting in strain specific synergistic responses to LPS exposure.
Collapse
Affiliation(s)
- Mingming Zhao
- Type 2 Immunity Section, Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Bethesda, MD, USA
| | - Dragana Jankovic
- Type 2 Immunity Section, Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Bethesda, MD, USA
| | - Verena M Link
- Metaorganism Immunity Section, Laboratory of Host Immunity and Microbiome, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Camila Oliveira Silva Souza
- Type 2 Immunity Section, Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Bethesda, MD, USA
| | - Katherine M Hornick
- NIAID Collaborative Bioinformatics Resource, Integrated Data Sciences Section, Research Technology Branch, Division of Intramural Research, NIAID, NIH, Bethesda, MD, USA
| | - Oyebola Oyesola
- Type 2 Immunity Section, Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Bethesda, MD, USA
| | - Yasmine Belkaid
- Metaorganism Immunity Section, Laboratory of Host Immunity and Microbiome, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Justin Lack
- NIAID Collaborative Bioinformatics Resource, Integrated Data Sciences Section, Research Technology Branch, Division of Intramural Research, NIAID, NIH, Bethesda, MD, USA
| | - Png Loke
- Type 2 Immunity Section, Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Bethesda, MD, USA.
| |
Collapse
|
48
|
Zhang X, Luo Z, Marand AP, Yan H, Jang H, Bang S, Mendieta JP, Minow MAA, Schmitz RJ. A spatially resolved multi-omic single-cell atlas of soybean development. Cell 2025; 188:550-567.e19. [PMID: 39742806 DOI: 10.1016/j.cell.2024.10.050] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2024] [Revised: 09/26/2024] [Accepted: 10/31/2024] [Indexed: 01/04/2025]
Abstract
Cis-regulatory elements (CREs) precisely control spatiotemporal gene expression in cells. Using a spatially resolved single-cell atlas of gene expression with chromatin accessibility across ten soybean tissues, we identified 103 distinct cell types and 303,199 accessible chromatin regions (ACRs). Nearly 40% of the ACRs showed cell-type-specific patterns and were enriched for transcription factor (TF) motifs defining diverse cell identities. We identified de novo enriched TF motifs and explored the conservation of gene regulatory networks underpinning legume symbiotic nitrogen fixation. With comprehensive developmental trajectories for endosperm and embryo, we uncovered the functional transition of the three sub-cell types of endosperm, identified 13 sucrose transporters sharing the DNA binding with one finger 11 (DOF11) motif that were co-upregulated in late peripheral endosperm, and identified key embryo cell-type specification regulators during embryogenesis, including a homeobox TF that promotes cotyledon parenchyma identity. This resource provides a valuable foundation for analyzing gene regulatory programs in soybean cell types across tissues and life stages.
Collapse
Affiliation(s)
- Xuan Zhang
- Department of Genetics, University of Georgia, Athens, GA, USA
| | - Ziliang Luo
- Department of Genetics, University of Georgia, Athens, GA, USA
| | - Alexandre P Marand
- Department of Molecular, Cellular, and Development Biology, University of Michigan, Ann Arbor, MI, USA
| | - Haidong Yan
- Department of Genetics, University of Georgia, Athens, GA, USA
| | - Hosung Jang
- Department of Genetics, University of Georgia, Athens, GA, USA
| | - Sohyun Bang
- Institute of Bioinformatics, University of Georgia, Athens, GA, USA
| | - John P Mendieta
- Department of Genetics, University of Georgia, Athens, GA, USA
| | - Mark A A Minow
- Department of Genetics, University of Georgia, Athens, GA, USA
| | | |
Collapse
|
49
|
Chen J, Richardson PR, Kirby C, Eddy SR, Hoekstra HE. Cellular evolution of the hypothalamic preoptic area of behaviorally divergent deer mice. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2024.08.22.608850. [PMID: 39253506 PMCID: PMC11383002 DOI: 10.1101/2024.08.22.608850] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 09/11/2024]
Abstract
Genetic variation is known to contribute to the variation of animal social behavior, but the molecular mechanisms that lead to behavioral differences are still not fully understood. Here, we investigate the cellular evolution of the hypothalamic preoptic area (POA), a brain region that plays a critical role in social behavior, across two sister species of deer mice (Peromyscus maniculatus and P. polionotus) with divergent social systems. These two species exhibit large differences in mating and parental care behavior across species and sex. Using single-nucleus RNA-sequencing, we build a cellular atlas of the POA for males and females of both Peromyscus species. We identify four cell types that are differentially abundant across species, two of which may account for species differences in parental care behavior based on known functions of these cell types. Our data further implicate two sex-biased cell types to be important for the evolution of sex-specific behavior. Finally, we show a remarkable reduction of sex-biased gene expression in P. polionotus, a monogamous species that also exhibits reduced sexual dimorphism in parental care behavior. Our POA atlas is a powerful resource to investigate how molecular neuronal traits may be evolving to give rise to innate differences in social behavior across animal species.
Collapse
Affiliation(s)
- Jenny Chen
- Department of Molecular & Cellular Biology, Harvard University, Cambridge, Massachusetts, USA
- Department of Organismic and Evolutionary Biology, Harvard University, Cambridge, Massachusetts, USA
| | - Phoebe R Richardson
- Department of Organismic and Evolutionary Biology, Harvard University, Cambridge, Massachusetts, USA
| | - Christopher Kirby
- Department of Organismic and Evolutionary Biology, Harvard University, Cambridge, Massachusetts, USA
| | - Sean R Eddy
- Department of Molecular & Cellular Biology, Harvard University, Cambridge, Massachusetts, USA
- Howard Hughes Medical Institute, Harvard University, Cambridge, Massachusetts, USA
| | - Hopi E Hoekstra
- Department of Molecular & Cellular Biology, Harvard University, Cambridge, Massachusetts, USA
- Department of Organismic and Evolutionary Biology, Harvard University, Cambridge, Massachusetts, USA
| |
Collapse
|
50
|
Ranga V, Dakal TC, Maurya PK, Johnson MS, Sharma NK, Kumar A. Role of RGD-binding Integrins in ovarian cancer progression, metastasis and response to therapy. Integr Biol (Camb) 2025; 17:zyaf003. [PMID: 39916547 DOI: 10.1093/intbio/zyaf003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2024] [Revised: 09/12/2024] [Accepted: 01/29/2025] [Indexed: 05/08/2025]
Abstract
Integrins are transmembrane receptors that play a crucial role in cell adhesion and signaling by connecting the extracellular environment to the intracellular cytoskeleton. After binding with specific ligands in the extracellular matrix (ECM), integrins undergo conformational changes that transmit signals across the cell membrane. The integrin-mediated bidirectional signaling triggers various cellular responses, such as changes in cell shape, migration and proliferation. Irregular integrin expression and activity are closely linked to tumor initiation, angiogenesis, cell motility, invasion, and metastasis. Thus, understanding the intricate regulatory mechanism is essential for slowing cancer progression and preventing carcinogenesis. Among the four classes of integrins, the arginine-glycine-aspartic acid (RGD)-binding integrins stand out as the most crucial integrin receptor subfamily in cancer and its metastasis. Dysregulation of almost all RGD-binding integrins promotes ECM degradation in ovarian cancer, resulting in ovarian carcinoma progression and resistance to therapy. Preclinical studies have demonstrated that targeting these integrins with therapeutic antibodies and ligands, such as RGD-containing peptides and their derivatives, can enhance the precision of these therapeutic agents in treating ovarian cancer. Therefore, the development of novel therapeutic agents is essential for treating ovarian cancer. This review mainly discusses genes and their importance across different ovarian cancer subtypes, the involvement of RGD motif-containing ECM proteins in integrin-mediated signaling in ovarian carcinoma, ongoing, completed, partially completed, and unsuccessful clinical trials of therapeutic agents, as well as existing limitations and challenges, advancements made so far, potential strategies, and directions for future research in the field. Insight Box Integrin-mediated signaling regulates cell migration, proliferation and differentiation. Dysregulated integrin expression and activity promote tumor growth and dissemination. Thus, a proper understanding of this complex regulatory mechanism is essential to delay cancer progression and prevent carcinogenesis. Notably, integrins binding to RGD motifs play an important role in tumor initiation, evolution, and metastasis. Preclinical studies have demonstrated that therapeutic agents, such as antibodies and small molecules with RGD motifs, target RGD-binding integrins and disrupt their interactions with the ECM, thereby inhibiting ovarian cancer proliferation and migration. Altogether, this review highlights the potential of RGD-binding integrins in providing new insights into the progression and metastasis of ovarian cancer and how these integrins have been utilized to develop effective treatment plans.
Collapse
Affiliation(s)
- Vipin Ranga
- DBT-North East Centre for Agricultural Biotechnology (DBT-NECAB), Assam Agricultural University, Agriculture University Road, Jorhat, Assam 785013, India
| | - Tikam Chand Dakal
- Genome and Computational Biology Laboratory, Department of Biotechnology, Mohanlal Sukhadia University, University Road, Udaipur, Rajasthan 313001, India
| | - Pawan Kumar Maurya
- Department of Biochemistry, Central University of Haryana, Central University of Haryana Road, Mahendergarh, Haryana 123031, India
| | - Mark S Johnson
- Structural Bioinformatics Laboratory and InFLAMES Research Flagship Center, Biochemistry, Faculty of Science and Engineering, Åbo Akademi University, Tykistökatu 6A, Turku 20520, Finland
| | - Narendra Kumar Sharma
- Department of Bioscience and Biotechnology, Banasthali Vidyapith, Vanasthali Road, Tonk, Rajasthan 304022, India
| | - Abhishek Kumar
- Manipal Academy of Higher Education (MAHE), Tiger Circle Road, Manipal, Karnataka 576104, India
- Institute of Bioinformatics, Discoverer Building, International Technology Park, Whitefield, Bangalore, Karnataka 560006, India
| |
Collapse
|