1
|
Zhang H, Li X, Song D, Yukselen O, Nanda S, Kucukural A, Li JJ, Garber M, Walhout AJM. Worm Perturb-Seq: massively parallel whole-animal RNAi and RNA-seq. Nat Commun 2025; 16:4785. [PMID: 40404656 PMCID: PMC12098853 DOI: 10.1038/s41467-025-60154-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2024] [Accepted: 05/15/2025] [Indexed: 05/24/2025] Open
Abstract
Transcriptomes provide highly informative molecular phenotypes that, combined with gene perturbation, can connect genotype to phenotype. An ultimate goal is to perturb every gene and measure transcriptome changes, however, this is challenging, especially in whole animals. Here, we present 'Worm Perturb-Seq (WPS)', a method that provides high-resolution RNA-sequencing profiles for hundreds of replicate perturbations at a time in living animals. WPS introduces multiple experimental advances combining strengths of Caenhorhabditis elegans genetics and multiplexed RNA-sequencing with a novel analytical framework, EmpirDE. EmpirDE leverages the unique power of large transcriptomic datasets and improves statistical rigor by using gene-specific empirical null distributions to identify DEGs. We apply WPS to 103 nuclear hormone receptors (NHRs) and find a striking 'pairwise modularity' in which pairs of NHRs regulate shared target genes. We envision the advances of WPS to be useful not only for C. elegans, but broadly for other models, including human cells.
Collapse
Affiliation(s)
- Hefei Zhang
- Department of Systems Biology, University of Massachusetts Chan Medical School, Worcester, MA, USA
| | - Xuhang Li
- Department of Systems Biology, University of Massachusetts Chan Medical School, Worcester, MA, USA
| | - Dongyuan Song
- Bioinformatics Interdepartmental Ph.D. Program, University of California, Los Angeles, CA, USA
- Department of Genetics and Genome Sciences, University of Connecticut Health Center, Farmington, CT, USA
| | | | - Shivani Nanda
- Department of Systems Biology, University of Massachusetts Chan Medical School, Worcester, MA, USA
- Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Alper Kucukural
- Via Scientific Inc., Cambridge, MA, USA
- Department of Genomics and Computational Biology, University of Massachusetts Chan Medical School, Worcester, MA, USA
| | - Jingyi Jessica Li
- Bioinformatics Interdepartmental Ph.D. Program, University of California, Los Angeles, CA, USA
- Department of Statistics and Data Science, Department of Biostatistics, Department of Computational Medicine, and Department of Human Genetics, University of California, Los Angeles, CA, USA
| | - Manuel Garber
- Department of Genomics and Computational Biology, University of Massachusetts Chan Medical School, Worcester, MA, USA.
| | - Albertha J M Walhout
- Department of Systems Biology, University of Massachusetts Chan Medical School, Worcester, MA, USA.
| |
Collapse
|
2
|
Panagopoulos A, Stout M, Kilic S, Leary P, Vornberger J, Pasti V, Galarreta A, Lezaja A, Kirschenbühler K, Imhof R, Rehrauer H, Ziegler U, Altmeyer M. Multigenerational cell tracking of DNA replication and heritable DNA damage. Nature 2025:10.1038/s41586-025-08986-0. [PMID: 40399682 DOI: 10.1038/s41586-025-08986-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2024] [Accepted: 04/04/2025] [Indexed: 05/23/2025]
Abstract
Cell heterogeneity is a universal feature of life. Although biological processes affected by cell-to-cell variation are manifold, from developmental plasticity to tumour heterogeneity and differential drug responses, the sources of cell heterogeneity remain largely unclear1,2. Mutational and epigenetic signatures from cancer (epi)genomics are powerful for deducing processes that shaped cancer genome evolution3-5. However, retrospective analyses face difficulties in resolving how cellular heterogeneity emerges and is propagated to subsequent cell generations. Here, we used multigenerational single-cell tracking based on endogenously labelled proteins and custom-designed computational tools to elucidate how oncogenic perturbations induce sister cell asymmetry and phenotypic heterogeneity. Dual CRISPR-based genome editing enabled simultaneous tracking of DNA replication patterns and heritable endogenous DNA lesions. Cell lineage trees of up to four generations were tracked in asynchronously growing cells, and time-resolved lineage analyses were combined with end-point measurements of cell cycle and DNA damage markers through iterative staining. Besides revealing replication and repair dynamics, damage inheritance and emergence of sister cell heterogeneity across multiple cell generations, through combination with single-cell transcriptomics, we delineate how common oncogenic events trigger multiple routes towards polyploidization with distinct outcomes for genome integrity. Our study provides a framework to dissect phenotypic plasticity at the single-cell level and sheds light onto cellular processes that may resemble early events during cancer development.
Collapse
Affiliation(s)
- Andreas Panagopoulos
- Department of Molecular Mechanisms of Disease, University of Zurich, Zurich, Switzerland
| | - Merula Stout
- Department of Molecular Mechanisms of Disease, University of Zurich, Zurich, Switzerland
| | - Sinan Kilic
- Department of Molecular Mechanisms of Disease, University of Zurich, Zurich, Switzerland
- The Novo Nordisk Foundation Center for Protein Research, University of Copenhagen, Copenhagen, Denmark
| | - Peter Leary
- Functional Genomics Center Zurich, ETH Zurich and University of Zurich, Zurich, Switzerland
| | - Julia Vornberger
- Department of Molecular Mechanisms of Disease, University of Zurich, Zurich, Switzerland
| | - Virginia Pasti
- Department of Molecular Mechanisms of Disease, University of Zurich, Zurich, Switzerland
| | - Antonio Galarreta
- Department of Molecular Mechanisms of Disease, University of Zurich, Zurich, Switzerland
| | - Aleksandra Lezaja
- Department of Molecular Mechanisms of Disease, University of Zurich, Zurich, Switzerland
| | - Kyra Kirschenbühler
- Department of Molecular Mechanisms of Disease, University of Zurich, Zurich, Switzerland
- NEXUS Personalized Health, ETH Zurich, Schlieren, Switzerland
| | - Ralph Imhof
- Department of Molecular Mechanisms of Disease, University of Zurich, Zurich, Switzerland
| | - Hubert Rehrauer
- Functional Genomics Center Zurich, ETH Zurich and University of Zurich, Zurich, Switzerland
| | - Urs Ziegler
- Center for Microscopy and Image Analysis, University of Zurich, Zurich, Switzerland
| | - Matthias Altmeyer
- Department of Molecular Mechanisms of Disease, University of Zurich, Zurich, Switzerland.
| |
Collapse
|
3
|
Pandey AC, Bezney J, DeAscanis D, Kirsch EB, Ahmed F, Crinklaw A, Choudhary KS, Mandala T, Deason J, Hamidi JS, Siddique A, Ranganathan S, Brown K, Armstrong J, Head S, Ordoukhanian P, Steinmetz LM, Topol EJ. A CRISPR/Cas9-based enhancement of high-throughput single-cell transcriptomics. Nat Commun 2025; 16:4664. [PMID: 40389438 PMCID: PMC12089397 DOI: 10.1038/s41467-025-59880-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2024] [Accepted: 05/03/2025] [Indexed: 05/21/2025] Open
Abstract
Single-cell RNA-seq (scRNAseq) struggles to capture the cellular heterogeneity of transcripts within individual cells due to the prevalence of highly abundant and ubiquitous transcripts, which can obscure the detection of biologically distinct transcripts expressed up to several orders of magnitude lower levels. To address this challenge, here we introduce single-cell CRISPRclean (scCLEAN), a molecular method that globally recomposes scRNAseq libraries, providing a benefit that cannot be recapitulated with deeper sequencing. scCLEAN utilizes the programmability of CRISPR/Cas9 to target and remove less than 1% of the transcriptome while redistributing approximately half of reads, shifting the focus toward less abundant transcripts. We experimentally apply scCLEAN to both heterogeneous immune cells and homogenous vascular smooth muscle cells to demonstrate its ability to uncover biological signatures in different biological contexts. We further emphasize scCLEAN's versatility by applying it to a third-generation sequencing method, single-cell MAS-Seq, to increase transcript-level detection and discovery. Here we show the possible utility of scCLEAN across a wide array of human tissues and cell types, indicating which contexts this technology proves beneficial and those in which its application is not advisable.
Collapse
Affiliation(s)
- Amitabh C Pandey
- Section of Cardiology, Tulane Heart and Vascular Institute, Department of Medicine, Tulane University School of Medicine, New Orleans, LA, USA.
- Southeast Louisiana Veterans Health Care System, New Orleans, LA, USA.
- Department of Molecular Medicine, Scripps Research Translational Institute, The Scripps Research Institute, La Jolla, CA, USA.
| | - Jon Bezney
- Genomics Core Facility, The Scripps Research Institute, La Jolla, CA, USA
- Jumpcode Genomics, San Diego, CA, USA
- Department of Genetics, Stanford University School of Medicine, Stanford, CA, USA
| | | | - Ethan B Kirsch
- Department of Molecular Medicine, Scripps Research Translational Institute, The Scripps Research Institute, La Jolla, CA, USA
| | - Farin Ahmed
- Genomics Core Facility, The Scripps Research Institute, La Jolla, CA, USA
| | | | | | - Tony Mandala
- Genomics Core Facility, The Scripps Research Institute, La Jolla, CA, USA
| | | | - Jasmin S Hamidi
- Department of Molecular Medicine, Scripps Research Translational Institute, The Scripps Research Institute, La Jolla, CA, USA
| | | | | | | | | | - Steven Head
- Genomics Core Facility, The Scripps Research Institute, La Jolla, CA, USA
| | | | - Lars M Steinmetz
- Department of Genetics, Stanford University School of Medicine, Stanford, CA, USA
- Stanford Genome Technology Center, Palo Alto, CA, USA
- European Molecular Biology Laboratory (EMBL), Genome Biology Unit, Heidelberg, Germany
| | - Eric J Topol
- Department of Molecular Medicine, Scripps Research Translational Institute, The Scripps Research Institute, La Jolla, CA, USA
| |
Collapse
|
4
|
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
|
5
|
Hu H, Fan Y, Wang J, Zhang J, Lyu Y, Hou X, Cui J, Zhang Y, Gao J, Zhang T, Nan K. Single-cell technology for cell-based drug delivery and pharmaceutical research. J Control Release 2025; 381:113587. [PMID: 40032008 DOI: 10.1016/j.jconrel.2025.113587] [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: 10/16/2024] [Revised: 02/25/2025] [Accepted: 02/26/2025] [Indexed: 03/05/2025]
Abstract
Leveraging the capacity to precisely manipulate and analyze individual cells, single-cell technology has rapidly become an indispensable tool in the advancement of cell-based drug delivery systems and innovative cell therapies. This technology offers powerful means to address cellular heterogeneity and significantly enhance therapeutic efficacy. Recent breakthroughs in techniques such as single-cell electroporation, mechanical perforation, and encapsulation, particularly when integrated with microfluidics and bioelectronics, have led to remarkable improvements in drug delivery efficiency, reductions in cytotoxicity, and more precise targeting of therapeutic effects. Moreover, single-cell analyses, including advanced sequencing and high-resolution sensing, offer profound insights into complex disease mechanisms, the development of drug resistance, and the intricate processes of stem cell differentiation. This review summarizes the most significant applications of these single-cell technologies, highlighting their impact on the landscape of modern biomedicine. Furthermore, it provides a forward-looking perspective on future research directions aimed at further optimizing drug delivery strategies and enhancing therapeutic outcomes in the treatment of various diseases.
Collapse
Affiliation(s)
- Huihui Hu
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310000, China
| | - Yunlong Fan
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310000, China; MicroTech Medical (Hangzhou) Co., Hangzhou 311100, China
| | - Jiawen Wang
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310000, China
| | - Jialu Zhang
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310000, China
| | - Yidan Lyu
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310000, China
| | - Xiaoqi Hou
- School of Chemistry and Materials Science, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou 310024, China
| | - Jizhai Cui
- Department of Materials Science, Fudan University, Shanghai 200438, China; International Institute of Intelligent Nanorobots and Nanosystems, Fudan University, Shanghai 200438, China
| | - Yamin Zhang
- Department of Chemical and Biomolecular Engineering, National University of Singapore, 117585, Singapore
| | - Jianqing Gao
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310000, China
| | - Tianyuan Zhang
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310000, China.
| | - Kewang Nan
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310000, China.
| |
Collapse
|
6
|
Slenders L, Wesseling M, Wei S, Boltjes A, Kapteijn DMC, van de Kraak P, Depuydt MAC, Prange KHM, van den Dungen NAM, Benavente ED, de Kleijn DPV, de Borst GJ, de Winther MPJ, den Ruijter HM, Owens GK, Pasterkamp G, Mokry M. Endothelial-to-mesenchymal transition gene signature derived from single-cell transcriptomics of human atherosclerotic tissue associates with stable plaque histological characteristics. Vascul Pharmacol 2025; 159:107498. [PMID: 40318741 DOI: 10.1016/j.vph.2025.107498] [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: 03/19/2025] [Revised: 04/28/2025] [Accepted: 04/29/2025] [Indexed: 05/07/2025]
Abstract
BACKGROUND Endothelial cells within atherosclerotic plaques can differentiate into a mesenchymal-like phenotype through endothelial-to-mesenchymal transition (EndoMT). Our understanding of the molecular mechanisms underlying EndoMT in human atherosclerosis remains limited. Current gene expression signatures are often derived from in vitro experiments or animal studies and typically reflect genes upregulated in fully differentiated mesenchymal cell states, while genes upregulated during the process are omitted. To address this knowledge gap, we utilized in silico lineage tracing in single-cell transcriptomic (scRNA-seq) data from human plaque tissues to identify the EndoMT gene expression signature. METHODS AND RESULTS We constructed three candidate EndoMT lineages across subpopulations of ECs and SMCs in human carotid scRNA-seq data (n = 46). We examined gene expression over the course of these lineages and identified a core signature of 73 genes upregulated in EndoMT. Upregulation of those genes was confirmed in EndoMT trajectories of other human datasets derived from plaque tissue and in Cdh5-CreERT2 Rosa-eYFP apoE-/- lineage-traced mice. Analysis of human carotid plaque bulk RNA-seq data (632 patients) found the association of core gene signature with fibrous and more stable histological phenotypes. CONCLUSION This study defines the core gene signature of EndoMT in human atherosclerotic plaques, which can serve as a reference for future studies and gene set enrichment analysis.
Collapse
Affiliation(s)
- Lotte Slenders
- Central Diagnostics Laboratory, Department of Laboratories, Pharmacy and Biomedical Genetics, University Medical Center Utrecht, University Utrecht, Utrecht, the Netherlands
| | - Marian Wesseling
- Central Diagnostics Laboratory, Department of Laboratories, Pharmacy and Biomedical Genetics, University Medical Center Utrecht, University Utrecht, Utrecht, the Netherlands
| | - Siting Wei
- Central Diagnostics Laboratory, Department of Laboratories, Pharmacy and Biomedical Genetics, University Medical Center Utrecht, University Utrecht, Utrecht, the Netherlands
| | - Arjan Boltjes
- Central Diagnostics Laboratory, Department of Laboratories, Pharmacy and Biomedical Genetics, University Medical Center Utrecht, University Utrecht, Utrecht, the Netherlands
| | - Daniek M C Kapteijn
- Laboratory of Experimental Cardiology, Department of Cardiology, University Medical Center Utrecht, University Utrecht, Utrecht, the Netherlands
| | - Petra van de Kraak
- Pathology department, University Medical Center Utrecht, University Utrecht, Utrecht, the Netherlands
| | - Marie A C Depuydt
- Leiden Academic Centre for Drug Research, Division of BioTherapeutics, Leiden University, Leiden, the Netherlands
| | - Koen H M Prange
- Amsterdam University Medical Centers - location AMC, University of Amsterdam, Experimental Vascular Biology, Department of Medical Biochemistry, Amsterdam Cardiovascular Sciences, Amsterdam, the Netherlands
| | - Noortje A M van den Dungen
- Central Diagnostics Laboratory, Department of Laboratories, Pharmacy and Biomedical Genetics, University Medical Center Utrecht, University Utrecht, Utrecht, the Netherlands
| | - Ernest D Benavente
- Laboratory of Experimental Cardiology, Department of Cardiology, University Medical Center Utrecht, University Utrecht, Utrecht, the Netherlands
| | - Dominique P V de Kleijn
- Department of Vascular Surgery, University Medical Center Utrecht, University Utrecht, Utrecht, the Netherlands
| | - Gert J de Borst
- Department of Vascular Surgery, University Medical Center Utrecht, University Utrecht, Utrecht, the Netherlands
| | - Menno P J de Winther
- Amsterdam University Medical Centers - location AMC, University of Amsterdam, Experimental Vascular Biology, Department of Medical Biochemistry, Amsterdam Cardiovascular Sciences, Amsterdam, the Netherlands
| | - Hester M den Ruijter
- Laboratory of Experimental Cardiology, Department of Cardiology, University Medical Center Utrecht, University Utrecht, Utrecht, the Netherlands
| | - Gary K Owens
- Robert M. Berne Cardiovascular Research Center, University of Virginia, Charlottesville, USA; Department of Molecular Physiology and Biological Physics, University of Virginia, Charlottesville, USA
| | - Gerard Pasterkamp
- Central Diagnostics Laboratory, Department of Laboratories, Pharmacy and Biomedical Genetics, University Medical Center Utrecht, University Utrecht, Utrecht, the Netherlands.
| | - Michal Mokry
- Central Diagnostics Laboratory, Department of Laboratories, Pharmacy and Biomedical Genetics, University Medical Center Utrecht, University Utrecht, Utrecht, the Netherlands; Laboratory of Experimental Cardiology, Department of Cardiology, University Medical Center Utrecht, University Utrecht, Utrecht, the Netherlands.
| |
Collapse
|
7
|
Wang J, Ye F, Chai H, Jiang Y, Wang T, Ran X, Xia Q, Xu Z, Fu Y, Zhang G, Wu H, Guo G, Guo H, Ruan Y, Wang Y, Xing D, Xu X, Zhang Z. Advances and applications in single-cell and spatial genomics. SCIENCE CHINA. LIFE SCIENCES 2025; 68:1226-1282. [PMID: 39792333 DOI: 10.1007/s11427-024-2770-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/20/2024] [Accepted: 10/10/2024] [Indexed: 01/12/2025]
Abstract
The applications of single-cell and spatial technologies in recent times have revolutionized the present understanding of cellular states and the cellular heterogeneity inherent in complex biological systems. These advancements offer unprecedented resolution in the examination of the functional genomics of individual cells and their spatial context within tissues. In this review, we have comprehensively discussed the historical development and recent progress in the field of single-cell and spatial genomics. We have reviewed the breakthroughs in single-cell multi-omics technologies, spatial genomics methods, and the computational strategies employed toward the analyses of single-cell atlas data. Furthermore, we have highlighted the advances made in constructing cellular atlases and their clinical applications, particularly in the context of disease. Finally, we have discussed the emerging trends, challenges, and opportunities in this rapidly evolving field.
Collapse
Affiliation(s)
- Jingjing Wang
- Bone Marrow Transplantation Center of the First Affiliated Hospital & Liangzhu Laboratory, Zhejiang University School of Medicine, Hangzhou, 310058, China
| | - Fang Ye
- Bone Marrow Transplantation Center of the First Affiliated Hospital & Liangzhu Laboratory, Zhejiang University School of Medicine, Hangzhou, 310058, China
| | - Haoxi Chai
- Life Sciences Institute and The Second Affiliated Hospital, Zhejiang University, Hangzhou, 310058, China
| | - Yujia Jiang
- BGI Research, Shenzhen, 518083, China
- BGI Research, Hangzhou, 310030, China
| | - Teng Wang
- Biomedical Pioneering Innovation Center (BIOPIC) and School of Life Sciences, Peking University, Beijing, 100871, China
- Academy for Advanced Interdisciplinary Studies, Peking University, Beijing, 100871, China
| | - Xia Ran
- Bone Marrow Transplantation Center of the First Affiliated Hospital & Liangzhu Laboratory, Zhejiang University School of Medicine, Hangzhou, 310058, China
- Institute of Hematology, Zhejiang University, Hangzhou, 310000, China
| | - Qimin Xia
- Biomedical Pioneering Innovation Center (BIOPIC) and School of Life Sciences, Peking University, Beijing, 100871, China
| | - Ziye Xu
- Department of Laboratory Medicine of The First Affiliated Hospital & Liangzhu Laboratory, Zhejiang University School of Medicine, Hangzhou, 310058, China
| | - Yuting Fu
- Bone Marrow Transplantation Center of the First Affiliated Hospital & Liangzhu Laboratory, Zhejiang University School of Medicine, Hangzhou, 310058, China
- Center for Stem Cell and Regenerative Medicine, Zhejiang University School of Medicine, Hangzhou, 310058, China
| | - Guodong Zhang
- Bone Marrow Transplantation Center of the First Affiliated Hospital & Liangzhu Laboratory, Zhejiang University School of Medicine, Hangzhou, 310058, China
- Center for Stem Cell and Regenerative Medicine, Zhejiang University School of Medicine, Hangzhou, 310058, China
| | - Hanyu Wu
- Bone Marrow Transplantation Center of the First Affiliated Hospital & Liangzhu Laboratory, Zhejiang University School of Medicine, Hangzhou, 310058, China
- Center for Stem Cell and Regenerative Medicine, Zhejiang University School of Medicine, Hangzhou, 310058, China
| | - Guoji Guo
- Bone Marrow Transplantation Center of the First Affiliated Hospital & Liangzhu Laboratory, Zhejiang University School of Medicine, Hangzhou, 310058, China.
- Center for Stem Cell and Regenerative Medicine, Zhejiang University School of Medicine, Hangzhou, 310058, China.
- Zhejiang Provincial Key Lab for Tissue Engineering and Regenerative Medicine, Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cell and Regenerative Medicine, Hangzhou, 310058, China.
- Institute of Hematology, Zhejiang University, Hangzhou, 310000, China.
| | - Hongshan Guo
- Bone Marrow Transplantation Center of the First Affiliated Hospital & Liangzhu Laboratory, Zhejiang University School of Medicine, Hangzhou, 310058, China.
- Institute of Hematology, Zhejiang University, Hangzhou, 310000, China.
| | - Yijun Ruan
- Life Sciences Institute and The Second Affiliated Hospital, Zhejiang University, Hangzhou, 310058, China.
| | - Yongcheng Wang
- Department of Laboratory Medicine of The First Affiliated Hospital & Liangzhu Laboratory, Zhejiang University School of Medicine, Hangzhou, 310058, China.
| | - Dong Xing
- Biomedical Pioneering Innovation Center (BIOPIC) and School of Life Sciences, Peking University, Beijing, 100871, China.
- Beijing Advanced Innovation Center for Genomics (ICG), Peking University, Beijing, 100871, China.
| | - Xun Xu
- BGI Research, Shenzhen, 518083, China.
- BGI Research, Hangzhou, 310030, China.
- Guangdong Provincial Key Laboratory of Genome Read and Write, BGI Research, Shenzhen, 518083, China.
| | - Zemin Zhang
- Biomedical Pioneering Innovation Center (BIOPIC) and School of Life Sciences, Peking University, Beijing, 100871, China.
| |
Collapse
|
8
|
Zhu X, Zhao L, Teng F, Meng S, Xie M. ScAGCN: Graph Convolutional Network with Adaptive Aggregation Mechanism for scRNA-seq Data Dimensionality Reduction. Interdiscip Sci 2025:10.1007/s12539-025-00702-w. [PMID: 40281370 DOI: 10.1007/s12539-025-00702-w] [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: 10/17/2024] [Revised: 03/10/2025] [Accepted: 03/12/2025] [Indexed: 04/29/2025]
Abstract
With the development of single-cell RNA-sequencing (scRNA-seq) technology, scRNA-seq data analysis suffers huge challenges due to large scale, high dimensionality, high noise, and high sparsity. To achieve accurately embedded representation in the large-scale scRNA-seq data, we try to design a novel graph convolutional network with an adaptive aggregation mechanism. Based on the assumption that the aggregation order of different cells would be different, a graph convolutional network with an adaptive aggregation-based dimensionality reduction algorithm for scRNA-seq data is developed, named scAGCN. In scAGCN, a preprocessing consisting of quality control and feature selection is implemented. Then, an approximate nearest neighbor graph is rapidly constructed. Finally, a graph convolutional network with an adaptive aggregation mechanism is constructed, in which the neighborhood selection strategy based on node distribution and similarity boxplots is designed, and the aggregation function is optimized by defining a similarity measurement between neighborhood nodes and the central node. The results show that scAGCN outperforms existing dimensionality reduction methods on 15 real scRNA-seq datasets, especially in 10 large-scale scRNA-seq datasets.
Collapse
Affiliation(s)
- Xiaoshu Zhu
- School of Computer and Information Security, Guangxi Key Laboratory of Trusted Software, Guilin University of Electronic Technology, Guilin, 541004, China.
| | - Liquan Zhao
- School of Computer and Information Security, Guangxi Key Laboratory of Trusted Software, Guilin University of Electronic Technology, Guilin, 541004, China
| | - Fei Teng
- School of Computer and Information Security, Guangxi Key Laboratory of Trusted Software, Guilin University of Electronic Technology, Guilin, 541004, China
| | - Shuang Meng
- School of Computer Science and Engineering, Guangxi Normal University, Guilin, 541006, China
| | - Miao Xie
- School of Computer Science and Engineering, Yulin Normal University, Yulin, 537000, China.
| |
Collapse
|
9
|
Meena D, Huang J, Zare M, Hasbani NR, Romuald BOUAP, Mustafa R, van der Laan SW, Xu H, Terry JG, Bis JC, Jain D, Palmer ND, Heard-Costa N, Min YI, Guo X, Yao J, Taylor KD, Tan J, Peralta J, Pereira AC, Khan A, Choudhury A, Newman AB, Xiang AH, Hingorani A, Freedman BI, O’Donnell CJ, Giambartolomei C, Herrington DM, Jacobs DR, Klarin D, Wang FF, Heiss G, Doddapaneni H, Hodis HN, Broome J, Wilson JG, Brandenburg JT, Blangero J, Krieger JE, Smith JD, Viaud-Martinez KA, Ryan KA, Lange LA, Montasser ME, Mahaney MC, Mokry M, Fornage M, Munroe P, Gibbs RA, Tracy RP, Kim RW, Damrauer SM, Rich SS, Hsueh WA, Chen YDI, NHLBI Trans-Omics for Precision Medicine (TOPMed) Consortium, The Million Veteran Program (MVP), TOPMed Atherosclerosis Working Group, Morrison AC, Mitchell BD, Carr JJ, Psaty BM, Bowden DW, Vasan RS, Correa A, Post WS, Goodarzi MO, Raffel LJ, Curran JE, Ramsay M, Rotter JI, Elliott P, Franceschini N, de Vries PS, Tzoulaki I, Dehghan A. Genome-wide association study and multi-ancestry meta-analysis identify common variants associated with carotid artery intima-media thickness. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2025:2025.04.11.25325582. [PMID: 40321265 PMCID: PMC12047956 DOI: 10.1101/2025.04.11.25325582] [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: 05/12/2025]
Abstract
Carotid artery intima-media thickness (cIMT) is a measurement of subclinical atherosclerosis that predicts future cardiovascular events, including stroke and myocardial infarction. Genome-wide association studies (GWAS) have identified only a fraction of the genetic variants associated with cIMT. We performed the largest GWAS for cIMT involving up to 131,000 individuals. For the first time, we meta-analysed a diverse range of ancestries including populations with African, Asian (Chinese), Brazilian, European, and Hispanic ancestries. Our study identified 59 independent loci (53 loci from the multi-ancestry single variant analysis of which 19 are novel, P<5×10-8; 6 novel in gene-based analysis from single variant analysis, P=2.6×10-6, 2 novel in meta-regression) associated with cIMT. Gene-based, tissue-expression and gene-set enrichment analyses revealed novel genes of potential interest and highlighted significant relationships between vascular tissues (aorta, coronary and tibial arteries) and genetic associations. We found that circulatory levels of seven proteins, including ACAN, BCAM, DUT, ERI1, APOE, FN1, and GLRX were potentially causally associated with cIMT levels. We found a strong genome-wide correlation between cIMT and various cardiometabolic, smoking phenotypes, and lipid traits. Using Mendelian randomisation, our analyses provide robust evidence for causal associations between cIMT and several clinically relevant traits, including lipids, blood pressure, and waist circumference. Our study extends our genetic knowledge of atherosclerosis and highlights potential causal relations between risk factors, atherosclerosis and clinical diagnoses.
Collapse
Affiliation(s)
- Devendra Meena
- Department of Epidemiology and Biostatistics, Imperial College London School of Public Health, London, UK
| | - Jian Huang
- Department of Epidemiology and Biostatistics, Imperial College London School of Public Health, London, UK
| | - Marjan Zare
- Maternal-fetal medicine Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Natalie R. Hasbani
- Human Genetics Center, Department of Epidemiology, Human Genetics, and Environmental Sciences, School of Public Health, The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - BOUA Palwendé Romuald
- Clinical Research Unit of Nanoro, Institut de Recherche en Sciences de la Santé, CNRST, Burkina Faso
| | - Rima Mustafa
- Department of Epidemiology and Biostatistics, Imperial College London School of Public Health, London, UK
| | - Sander W van der Laan
- Central Diagnostics Laboratory, Division Laboratories, Pharmacy, and Biomedical genetics, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands
| | - Huichun Xu
- Division of Endocrinology, Diabetes and Nutrition, Department of Medicine, University of Maryland School of Medicine, Baltimore, MD, USA
| | - James G. Terry
- Department of Radiology, Vanderbilt Translational and Clinical Cardiovascular Research Center, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Joshua C. Bis
- Cardiovascular Health Research Unit, Department of Medicine, University of Washington, Seattle, WA, USA
| | - Deepti Jain
- Genetic Analysis Center, Department of Biostatistics, School of Public Health, University of Washington, Seattle, WA, USA
| | - Nicholette D. Palmer
- Department of Biochemistry, Wake Forest School of Medicine, Winston-Salem, NC, USA
| | - Nancy Heard-Costa
- Boston University School of Medicine, Boston, MA, USA
- National Heart, Lung, and Blood Institute’s Framingham Heart Study, Framingham, MA, USA
| | - Yuan-I Min
- Jackson Heart Study, Department of Medicine, University of Mississippi Medical Center, Jackson, MS, USA
| | - Xiuqing Guo
- The Institute for Translational Genomics and Population Sciences, Department of Pediatrics, The Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, CA, USA
| | - Jie Yao
- The Institute for Translational Genomics and Population Sciences, Department of Pediatrics, The Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, CA, USA
| | - Kent D. Taylor
- The Institute for Translational Genomics and Population Sciences, Department of Pediatrics, The Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, CA, USA
| | - Jingyi Tan
- The Institute for Translational Genomics and Population Sciences, Department of Pediatrics, The Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, CA, USA
| | - Juan Peralta
- Department of Human Genetics, University of Texas Rio Grande Valley School of Medicine, Brownsville, TX, USA
- South Texas Diabetes and Obesity Institute, University of Texas Rio Grande Valley School of Medicine, Brownsville, TX, USA
| | - Alexandre C. Pereira
- Department of Genetics, Harvard Medical School, Boston, MA, USA
- Laboratory of Genetics and Molecular Cardiology, Heart Institute, University of S√£o Paulo, S√£o Paulo, Brazil
| | - Alyna Khan
- Genetic Analysis Center, Department of Biostatistics, School of Public Health, University of Washington, Seattle, WA, USA
| | - Ananyo Choudhury
- Division of Human Genetics at the National Health Laboratory Service, University of the Witwatersrand, Johannesburg, South Africa
| | - Anne B. Newman
- Department of Epidemiology, Graduate School of Public Health, University of Pittsburgh, Pittsburgh, PA, USA
| | - Anny H. Xiang
- Department of Research and Evaluation, Kaiser Permanente Southern California, Pasadena, CA USA
| | - Aroon Hingorani
- Institute of Cardiovascular Science, Faculty of Population Health, University College London, London, United Kingdom
| | - Barry I. Freedman
- Section on Nephrology, Department of Internal Medicine, Wake Forest School of Medicine, Winston-Salem, NC, USA
| | - Christopher J. O’Donnell
- VA Boston Healthcare System, West Roxbury VA Medical Center, Cardiology Section, Boston, MA, USA
| | - Claudia Giambartolomei
- Department of Pathology and Laboratory Medicine, University of California (UCLA), Los Angeles, Los Angeles, CA, USA
| | - David M. Herrington
- Department of Internal Medicine, Section of Cardiovascular Medicine, Wake Forest School of Medicine, Winston-Salem, NC, USA
| | - David R. Jacobs
- Division of Epidemiology and Community Health, University of Minnesota School of Public Health, Minneapolis, MN, USA
| | - Derek Klarin
- VA Palo Alto Healthcare System, Palo Alto, CA
- Department of Surgery, Stanford University Medical Center, Stanford, CA
| | - Fei Fei Wang
- Genetic Analysis Center, Department of Biostatistics, School of Public Health, University of Washington, Seattle, WA, USA
| | - Gerardo Heiss
- Department of Epidemiology, Gillings School of Global Public Health, University of North Carolina, Chapel Hill, NC, USA
| | | | - Howard N. Hodis
- Department of Preventive Medicine, Department of Medicine, Atherosclerosis Research Unit, Keck School of Medicine, University of Southern California, Los Angeles, CA USA
| | - Jai Broome
- Genetic Analysis Center, Department of Biostatistics, School of Public Health, University of Washington, Seattle, WA, USA
| | - James G. Wilson
- Division of Cardiology, Beth Israel Deaconess Medical Center, Boston, MA, USA
| | - Jean-Tristan Brandenburg
- Division of Human Genetics at the National Health Laboratory Service, University of the Witwatersrand, Johannesburg, South Africa
| | - John Blangero
- Department of Human Genetics, University of Texas Rio Grande Valley School of Medicine, Brownsville, TX, USA
- South Texas Diabetes and Obesity Institute, University of Texas Rio Grande Valley School of Medicine, Brownsville, TX, USA
| | - Jose E Krieger
- Laboratory of Genetics and Molecular Cardiology, Heart Institute, University of S√£o Paulo, S√£o Paulo, Brazil
| | - Josh D. Smith
- Department of Genome Sciences, University of Washington, Seattle, WA, USA
- Northwest Genomics Center, University of Washington, Seattle, WA, USA
| | | | - Kathleen A. Ryan
- Division of Endocrinology, Diabetes and Nutrition, Department of Medicine, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Leslie A. Lange
- Department of Medicine, University of Colorado Denver, Anschutz Medical Campus, Aurora, CO, USA
| | - May E. Montasser
- Division of Endocrinology, Diabetes and Nutrition, Department of Medicine, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Michael C. Mahaney
- Department of Human Genetics, University of Texas Rio Grande Valley School of Medicine, Brownsville, TX, USA
- South Texas Diabetes and Obesity Institute, University of Texas Rio Grande Valley School of Medicine, Brownsville, TX, USA
| | - Michal Mokry
- Laboratory of Experimental Cardiology, Department of Cardiology, University Medical Center Utrecht, University Utrecht, Utrecht, The Netherlands
| | - Myriam Fornage
- Institute of Molecular Medicine, The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Patricia Munroe
- Clinical Pharmacology, William Harvey Research Institute, Queen Mary University of London, Charterhouse Square, London, UK
- NIHR Barts Biomedical Research Centre, Queen Mary University of London, Charterhouse Square, London, UK
| | - Richard A. Gibbs
- Human Genome Sequencing Center, Baylor College of Medicine, Houston, TX, USA
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA
| | - Russell P. Tracy
- Department of Pathology and Laboratory Medicine, Robert Larner, M.D. College of Medicine, University of Vermont, Burlington, VT, USA
| | | | - Scott M. Damrauer
- Division of Vascular Surgery, Hospital of the University of Pennsylvania, Philadelphia, PA
| | - Stephen S. Rich
- Center for Public Health Genomics, University of Virginia School of Medicine, Charlottesville, VA, USA
| | - Willa A. Hsueh
- Department of Internal Medicine, The Ohio State University Wexner Medical Center, Columbus, OH USA
| | - Yii-Der Ida Chen
- The Institute for Translational Genomics and Population Sciences, Department of Pediatrics, The Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, CA, USA
| | | | - Alanna C. Morrison
- Human Genetics Center, Department of Epidemiology, Human Genetics, and Environmental Sciences, School of Public Health, The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Braxton D. Mitchell
- Division of Endocrinology, Diabetes and Nutrition, Department of Medicine, University of Maryland School of Medicine, Baltimore, MD, USA
- Geriatrics Research and Education Clinical Center, Baltimore Veterans Administration Medical Center, Baltimore, MD, USA
| | - John Jeffrey Carr
- Department of Radiology, Vanderbilt Translational and Clinical Cardiovascular Research Center, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Bruce M. Psaty
- Cardiovascular Health Research Unit, Department of Medicine, University of Washington, Seattle, WA, USA
- Department of Epidemiology, University of Washington, Seattle, WA, USA
- Department of Health Systems and Population Health, University of Washington, Seattle, WA, USA
| | - Donald W. Bowden
- Department of Biochemistry, Wake Forest School of Medicine, Winston-Salem, NC, USA
| | - Ramachandran S. Vasan
- National Heart, Lung, and Blood Institute’s Framingham Heart Study, Framingham, MA, USA
- Department of Medicine, Boston University School of Medicine, Boston, MA, USA
- Department of Epidemiology, Boston University School of Public Health, Boston, MA, USA
| | - Adolfo Correa
- Jackson Heart Study, Department of Medicine, University of Mississippi Medical Center, Jackson, MS, USA
- Department of Population Health Science, University of Mississippi Medical Center, Jackson, MS, USA
| | - Wendy S. Post
- Division of Cardiology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Mark O. Goodarzi
- Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA USA
| | - Leslie J. Raffel
- Division of Genetic and Genomic Medicine, Department of Pediatrics, University of California, Irvine, CA USA
| | - Joanne E. Curran
- Department of Human Genetics, University of Texas Rio Grande Valley School of Medicine, Brownsville, TX, USA
- South Texas Diabetes and Obesity Institute, University of Texas Rio Grande Valley School of Medicine, Brownsville, TX, USA
| | - Michele Ramsay
- Division of Human Genetics at the National Health Laboratory Service, University of the Witwatersrand, Johannesburg, South Africa
| | - Jerome I. Rotter
- The Institute for Translational Genomics and Population Sciences, Department of Pediatrics, The Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, CA, USA
| | - Paul Elliott
- MRC Centre for Environment and Health, Department of Epidemiology and Biostatistics, School of Public Health, Faculty of Medicine, Imperial College London, London, UK
- UK Dementia Research Institute, Imperial College London, London UK
- National Institute for Health Research Imperial College Biomedical Research Centre, Imperial College London, London, UK
| | - Nora Franceschini
- Department of Epidemiology, Gillings School of Global Public Health, University of North Carolina, Chapel Hill, NC, USA
| | - Paul S. de Vries
- Human Genetics Center, Department of Epidemiology, Human Genetics, and Environmental Sciences, School of Public Health, The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Ioanna Tzoulaki
- Department of Epidemiology and Biostatistics, Imperial College London School of Public Health, London, UK
- British Heart Foundation Centre of Excellence, Imperial College London, London, UK
- Department of Hygiene and Epidemiology, University of Ioannina, Ioannina, Greece
| | - Abbas Dehghan
- Department of Epidemiology and Biostatistics, Imperial College London School of Public Health, London, UK
| |
Collapse
|
10
|
Hirano K, Nakabayashi C, Sasaki M, Suzuki M, Aoyagi Y, Tanaka K, Murakami A, Tsuchiya M, Umemoto E, Takabayashi S, Kitajima Y, Ono Y, Matsukawa T, Matsushita M, Ohkawa Y, Mori Y, Hara Y. Mg 2+ influx mediated by TRPM7 triggers the initiation of muscle stem cell activation. SCIENCE ADVANCES 2025; 11:eadu0601. [PMID: 40184450 PMCID: PMC11970462 DOI: 10.1126/sciadv.adu0601] [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: 10/22/2024] [Accepted: 02/28/2025] [Indexed: 04/06/2025]
Abstract
Muscle satellite cells (MuSCs) respond immediately to environmental cues upon skeletal muscle injuries. Despite decades of research into muscle regeneration, the specific molecular factors that trigger the transition of MuSCs from a quiescent to an active state remain largely unidentified. Here, we identify transient receptor potential melastatin 7 (TRPM7), an Mg2+-permeable ion channel, as a critical regulator of MuSC activation. Trpm7 deletion in MuSCs reduced Mg2+ influx, impairing myofiber regeneration and leading to decreased MuSC numbers and cell cycle arrest during regeneration. These changes were linked to disrupted mTOR signaling, which drives the transition of MuSCs from G0 to GAlert phase. In addition, Trpm7-deficient MuSCs exhibited impaired early responses, including quiescent projection retraction and AP-1 induction. Mg2+ supplementation rescued these defects, restoring normal MuSC activation. Our findings reveal a previously unrecognized mechanism where Mg2+ permeation through TRPM7 is essential for MuSC activation and efficient skeletal muscle regeneration, highlighting TRPM7 as a critical regulator of muscle repair.
Collapse
Affiliation(s)
- Kotaro Hirano
- School of Pharmaceutical Sciences, University of Shizuoka, Shizuoka 422-8526, Japan
| | - Chika Nakabayashi
- School of Pharmaceutical Sciences, University of Shizuoka, Shizuoka 422-8526, Japan
| | - Mao Sasaki
- School of Pharmaceutical Sciences, University of Shizuoka, Shizuoka 422-8526, Japan
| | - Miki Suzuki
- School of Pharmaceutical Sciences, University of Shizuoka, Shizuoka 422-8526, Japan
- Faculty of Pharmacy, Laboratory of Hygienic Chemistry, Juntendo University, Chiba 279-0013, Japan
| | - Yuta Aoyagi
- School of Pharmaceutical Sciences, University of Shizuoka, Shizuoka 422-8526, Japan
| | - Kaori Tanaka
- Department of Medicine and Bioregulatory Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka 812-8582, Japan
| | - Akira Murakami
- School of Pharmaceutical Sciences, University of Shizuoka, Shizuoka 422-8526, Japan
| | - Masaki Tsuchiya
- School of Pharmaceutical Sciences, University of Shizuoka, Shizuoka 422-8526, Japan
- PRESTO, JST, Kawaguchi, Saitama 332-0012, Japan
| | - Eiji Umemoto
- School of Pharmaceutical Sciences, University of Shizuoka, Shizuoka 422-8526, Japan
| | - Shuji Takabayashi
- Institute of Photonics Medicine, Hamamatsu University School of Medicine, Shizuoka 431-3192, Japan
| | - Yasuo Kitajima
- Department of Immunology, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima 734-8551, Japan
| | - Yusuke Ono
- Department of Muscle Development and Regeneration, Institute of Molecular Embryology and Genetics, Kumamoto University, Kumamoto 860-0811, Japan
| | - Takehisa Matsukawa
- Faculty of Pharmacy, Laboratory of Hygienic Chemistry, Juntendo University, Chiba 279-0013, Japan
| | - Masayuki Matsushita
- Department of Molecular and Cellular Physiology, Graduate School of Medicine, University of the Ryukyus, Okinawa 903-0213, Japan
| | - Yasuyuki Ohkawa
- Department of Medicine and Bioregulatory Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka 812-8582, Japan
| | - Yasuo Mori
- Department of Synthetic Chemistry and Biological Chemistry, Graduate School of Engineering, Kyoto University, Kyoto 615-8510, Japan
| | - Yuji Hara
- School of Pharmaceutical Sciences, University of Shizuoka, Shizuoka 422-8526, Japan
| |
Collapse
|
11
|
Blotenburg M, Suurenbroek L, Bax D, de Visser J, Bhardwaj V, Braccioli L, de Wit E, van Boxtel A, Marks H, Zeller P. Stem cell culture conditions affect in vitro differentiation potential and mouse gastruloid formation. PLoS One 2025; 20:e0317309. [PMID: 40138371 PMCID: PMC11940422 DOI: 10.1371/journal.pone.0317309] [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: 05/23/2024] [Accepted: 12/24/2024] [Indexed: 03/29/2025] Open
Abstract
Aggregating low numbers of mouse embryonic stem cells (mESCs) and inducing Wnt signalling generates 'gastruloids', self-organising complex structures that display an anteroposterior organisation of cell types derived from all three germ layers. Current gastruloid protocols display considerable heterogeneity between experiments in terms of morphology, elongation efficiency, and cell type composition. We therefore investigated whether altering the mESC pluripotency state would provide more consistent results. By growing three mESC lines from two different genetic backgrounds in different intervals of ESLIF and 2i medium the pluripotency state of cells was modulated, and mESC culture as well as the resulting gastruloids were analysed. Microscopic analysis showed a pre-culture-specific effect on gastruloid formation, in terms of aspect ratio and reproducibility. RNA-seq analysis of the mESC start population confirmed that short-term pulses of 2i and ESLIF modulate the pluripotency state, and result in different cellular states. Since multiple epigenetic regulators were detected among the top differentially expressed genes, we further analysed genome-wide DNA methylation and H3K27me3 distributions. We observed epigenetic differences between conditions, most dominantly in the promoter regions of developmental regulators. Lastly, when we investigated the cell type composition of gastruloids grown from these different pre-cultures, we observed that mESCs subjected to 2i-ESLIF preceding aggregation generated gastruloids more consistently, including more complex mesodermal contributions as compared to the ESLIF-only control. These results indicate that optimisation of the mESCs pluripotency state allows the modulation of cell differentiation during gastruloid formation.
Collapse
Affiliation(s)
- Marloes Blotenburg
- Hubrecht Institute-KNAW (Royal Netherlands Academy of Arts and Sciences), Oncode Institute, Utrecht, The Netherlands
- University Medical Center Utrecht, Utrecht, The Netherlands
| | - Lianne Suurenbroek
- Hubrecht Institute-KNAW (Royal Netherlands Academy of Arts and Sciences), Oncode Institute, Utrecht, The Netherlands
- University Medical Center Utrecht, Utrecht, The Netherlands
| | - Danique Bax
- Department of Molecular Biology, Faculty of Science, Radboud Institute for Molecular Life Sciences (RIMLS), Radboud University Nijmegen, Nijmegen, The Netherlands
| | - Joëlle de Visser
- Developmental, Stem Cell and Cancer Biology, Swammerdam Institute for Life Sciences, University of Amsterdam, Amsterdam, The Netherlands
| | - Vivek Bhardwaj
- Hubrecht Institute-KNAW (Royal Netherlands Academy of Arts and Sciences), Oncode Institute, Utrecht, The Netherlands
- University Medical Center Utrecht, Utrecht, The Netherlands
| | - Luca Braccioli
- The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Elzo de Wit
- The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Antonius van Boxtel
- Developmental, Stem Cell and Cancer Biology, Swammerdam Institute for Life Sciences, University of Amsterdam, Amsterdam, The Netherlands
| | - Hendrik Marks
- Department of Molecular Biology, Faculty of Science, Radboud Institute for Molecular Life Sciences (RIMLS), Radboud University Nijmegen, Nijmegen, The Netherlands
| | - Peter Zeller
- Hubrecht Institute-KNAW (Royal Netherlands Academy of Arts and Sciences), Oncode Institute, Utrecht, The Netherlands
- University Medical Center Utrecht, Utrecht, The Netherlands
- Department of Molecular Biology and Genetics, Aarhus University, Aarhus, Denmark
| |
Collapse
|
12
|
Cisternino F, Song Y, Peters TS, Westerman R, de Borst GJ, Diez Benavente E, van den Dungen NA, Homoed-van der Kraak P, de Kleijn DP, Mekke J, Mokry M, Pasterkamp G, den Ruijter HM, Velema E, Miller CL, Glastonbury CA, van der Laan S. Intraplaque haemorrhage quantification and molecular characterisation using attention based multiple instance learning. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2025:2025.03.04.25323316. [PMID: 40093230 PMCID: PMC11908327 DOI: 10.1101/2025.03.04.25323316] [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: 03/19/2025]
Abstract
Intraplaque haemorrhage (IPH) represents a critical feature of plaque vulnerability as it is robustly associated with adverse cardiovascular events, including stroke and myocardial infarction. How IPH drives plaque instability is unknown. However, its identification and quantification in atherosclerotic plaques is currently performed manually, with high interobserver variability, limiting its accurate assessment in large cohorts. Leveraging the Athero-Express biobank, an ongoing study comprising a comprehensive dataset of histological, transcriptional, and clinical information from 2,595 carotid endarterectomy patients, we developed an attention-based additive multiple instance learning (MIL) framework to automate the detection and quantification of IPH across whole-slide images of nine distinct histological stains. We demonstrate that routinely available Haematoxylin and Eosin (H&E) staining outperformed all other plaque relevant Immunohistochemistry (IHC) stains tested (AUROC = 0.86), underscoring its utility in quantifying IPH. When combining stains through ensemble models, we see that H&E + CD68 (a macrophage marker) as well as H&E + Verhoeff-Van Gieson elastic fibers staining (EVG) leads to a substantial improvement (AUROC = 0.92). Using our model, we could derive IPH area from the MIL-derived patch-level attention scores, enabling not only classification but precise localisation and quantification of IPH area in each plaque, facilitating downstream analyses of its association and cellular composition with clinical outcomes. By doing so, we demonstrate that IPH presence and area are the most significant predictors of both preoperative symptom presentation and major adverse cardiovascular events (MACE), outperforming manual scoring methods. Automating IPH detection also allowed us to characterise IPH on a molecular level at scale. Pairing IPH measurements with single-cell transcriptomic analyses revealed key molecular pathways involved in IPH, including TNF-α signalling, extracellular matrix remodelling and the presence of foam cells. This study represents the largest effort in the cardiovascular field to integrate digital pathology, machine learning, and molecular data to predict and characterize IPH which leads to better understanding how it drives symptoms and MACE. Our model provides a scalable, interpretable, and reproducible method for plaque phenotyping, enabling the derivation of plaque phenotypes for predictive modelling of MACE outcomes.
Collapse
Affiliation(s)
| | - Yipei Song
- Department of Genome Sciences, University of Virginia, Charlottesville, VA, USA
- Department of Computer Engineering, University of Virginia, Charlottesville, VA, USA
| | - Tim S. Peters
- Central Diagnostic Laboratory, Division Laboratories, Pharmacy, and Biomedical genetics, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands
| | - Roderick Westerman
- Central Diagnostic Laboratory, Division Laboratories, Pharmacy, and Biomedical genetics, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands
| | - Gert J. de Borst
- Vascular surgery, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands
| | - Ernest Diez Benavente
- Central Diagnostic Laboratory, Division Laboratories, Pharmacy, and Biomedical genetics, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands
| | - Noortje A.M. van den Dungen
- Central Diagnostic Laboratory, Division Laboratories, Pharmacy, and Biomedical genetics, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands
| | | | - Dominique P.V. de Kleijn
- Vascular surgery, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands
| | - Joost Mekke
- Vascular surgery, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands
| | - Michal Mokry
- Central Diagnostic Laboratory, Division Laboratories, Pharmacy, and Biomedical genetics, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands
| | - Gerard Pasterkamp
- Central Diagnostic Laboratory, Division Laboratories, Pharmacy, and Biomedical genetics, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands
| | - Hester M. den Ruijter
- Central Diagnostic Laboratory, Division Laboratories, Pharmacy, and Biomedical genetics, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands
- Experimental Cardiology, Department Cardiology, Division Heart & Lungs, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands
| | - Evelyn Velema
- Experimental Cardiology, Department Cardiology, Division Heart & Lungs, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands
| | - Clint L. Miller
- Department of Genome Sciences, University of Virginia, Charlottesville, VA, USA
- Department of Public Health Sciences, University of Virginia, Charlottesville, VA, USA
- Department of Biochemistry and Molecular Genetics, University of Virginia, Charlottesville, VA, USA
| | - Craig A. Glastonbury
- Human Technopole, Viale Rita Levi-Montalcini 1, 20157, Milan, Italy
- Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - S.W. van der Laan
- Department of Genome Sciences, University of Virginia, Charlottesville, VA, USA
- Central Diagnostic Laboratory, Division Laboratories, Pharmacy, and Biomedical genetics, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands
| |
Collapse
|
13
|
Yang SY, Han SM, Lee JY, Kim KS, Lee JE, Lee DW. Advancing Gut Microbiome Research: The Shift from Metagenomics to Multi-Omics and Future Perspectives. J Microbiol Biotechnol 2025; 35:e2412001. [PMID: 40223273 PMCID: PMC12010094 DOI: 10.4014/jmb.2412.12001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2024] [Revised: 02/14/2025] [Accepted: 02/24/2025] [Indexed: 04/15/2025]
Abstract
The gut microbiome, a dynamic and integral component of human health, has co-evolved with its host, playing essential roles in metabolism, immunity, and disease prevention. Traditional microbiome studies, primarily focused on microbial composition, have provided limited insights into the functional and mechanistic interactions between microbiota and their host. The advent of multi-omics technologies has transformed microbiome research by integrating genomics, transcriptomics, proteomics, and metabolomics, offering a comprehensive, systems-level understanding of microbial ecology and host-microbiome interactions. These advances have propelled innovations in personalized medicine, enabling more precise diagnostics and targeted therapeutic strategies. This review highlights recent breakthroughs in microbiome research, demonstrating how these approaches have elucidated microbial functions and their implications for health and disease. Additionally, it underscores the necessity of standardizing multi-omics methodologies, conducting large-scale cohort studies, and developing novel platforms for mechanistic studies, which are critical steps toward translating microbiome research into clinical applications and advancing precision medicine.
Collapse
Affiliation(s)
- So-Yeon Yang
- Department of Biotechnology, Yonsei University, Seoul 03722, Republic of Korea
| | - Seung Min Han
- Department of Biotechnology, Yonsei University, Seoul 03722, Republic of Korea
| | - Ji-Young Lee
- Department of Biotechnology, Yonsei University, Seoul 03722, Republic of Korea
| | - Kyoung Su Kim
- Department of Biotechnology, Yonsei University, Seoul 03722, Republic of Korea
| | - Jae-Eun Lee
- Department of Biotechnology, Yonsei University, Seoul 03722, Republic of Korea
| | - Dong-Woo Lee
- Department of Biotechnology, Yonsei University, Seoul 03722, Republic of Korea
| |
Collapse
|
14
|
Theunis K, Vanuytven S, Claes I, Geurts J, Rambow F, Brown D, Van Der Haegen M, Marin-Bejar O, Rogiers A, Van Raemdonck N, Leucci E, Demeulemeester J, Sifrim A, Marine JC, Voet T. Single-cell genome and transcriptome sequencing without upfront whole-genome amplification reveals cell state plasticity of melanoma subclones. Nucleic Acids Res 2025; 53:gkaf173. [PMID: 40138718 PMCID: PMC11941470 DOI: 10.1093/nar/gkaf173] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2023] [Revised: 02/07/2025] [Accepted: 02/21/2025] [Indexed: 03/29/2025] Open
Abstract
Single-cell multi-omics methods enable the study of cell state diversity, which is largely determined by the interplay of the genome, epigenome, and transcriptome. Here, we describe Gtag&T-seq, a genome-and-transcriptome sequencing (G&T-seq) protocol of the same single cells that omits whole-genome amplification (WGA) by using direct genomic tagmentation (Gtag). Gtag drastically decreases the cost and improves coverage uniformity at single-cell and pseudo-bulk levels compared to WGA-based G&T-seq. We also show that transcriptome-based DNA copy number inference has limited resolution and accuracy, underlining the importance of affordable multi-omic approaches. Applying Gtag&T-seq to a melanoma xenograft model before treatment and at minimal residual disease revealed differential cell state plasticity and treatment response between cancer subclones. In summary, Gtag&T-seq is a low-cost and accurate single-cell multi-omics method that explores genetic alterations and their functional consequences in single cells at scale.
Collapse
Affiliation(s)
- Koen Theunis
- Laboratory of Reproductive Genomics, Department of Human Genetics, KU Leuven, 3000 Leuven, Belgium
| | - Sebastiaan Vanuytven
- Laboratory of Reproductive Genomics, Department of Human Genetics, KU Leuven, 3000 Leuven, Belgium
| | - Irene Claes
- Laboratory of Reproductive Genomics, Department of Human Genetics, KU Leuven, 3000 Leuven, Belgium
- KU Leuven Institute for Single Cell Omics (LISCO), KU Leuven, 3000 Leuven, Belgium
| | - Jarne Geurts
- Laboratory of Reproductive Genomics, Department of Human Genetics, KU Leuven, 3000 Leuven, Belgium
- KU Leuven Institute for Single Cell Omics (LISCO), KU Leuven, 3000 Leuven, Belgium
| | - Florian Rambow
- Laboratory for Molecular Cancer Biology, Department of Oncology, KU Leuven, 3000 Leuven, Belgium
- VIB Center for Cancer Biology, VIB, 3000 Leuven, Belgium
| | - Daniel Brown
- Advanced Technology and Biology Division, The Walter and Eliza Hall Institute of Medical Research, 3052 Parkville, Australia
| | - Michiel Van Der Haegen
- Laboratory of Reproductive Genomics, Department of Human Genetics, KU Leuven, 3000 Leuven, Belgium
| | - Oskar Marin-Bejar
- Laboratory for Molecular Cancer Biology, Department of Oncology, KU Leuven, 3000 Leuven, Belgium
- VIB Center for Cancer Biology, VIB, 3000 Leuven, Belgium
| | - Aljosja Rogiers
- Laboratory for Molecular Cancer Biology, Department of Oncology, KU Leuven, 3000 Leuven, Belgium
- VIB Center for Cancer Biology, VIB, 3000 Leuven, Belgium
| | - Nina Van Raemdonck
- Laboratory for Molecular Cancer Biology, Department of Oncology, KU Leuven, 3000 Leuven, Belgium
- VIB Center for Cancer Biology, VIB, 3000 Leuven, Belgium
| | - Eleonora Leucci
- Laboratory for RNA Cancer Biology, Department of Oncology, KU Leuven, 3000 Leuven, Belgium
- Trace, Leuven Cancer Institute, Department of Oncology, KU Leuven, 3000 Leuven, Belgium
| | - Jonas Demeulemeester
- Laboratory of Reproductive Genomics, Department of Human Genetics, KU Leuven, 3000 Leuven, Belgium
- KU Leuven Institute for Single Cell Omics (LISCO), KU Leuven, 3000 Leuven, Belgium
| | - Alejandro Sifrim
- Laboratory of Multi-omic Integrative Bioinformatics, Department of Human Genetics, KU Leuven, 3000 Leuven, Belgium
- KU Leuven Institute for Single Cell Omics (LISCO), KU Leuven, 3000 Leuven, Belgium
| | - Jean-Christophe Marine
- Laboratory for Molecular Cancer Biology, Department of Oncology, KU Leuven, 3000 Leuven, Belgium
- VIB Center for Cancer Biology, VIB, 3000 Leuven, Belgium
| | - Thierry Voet
- Laboratory of Reproductive Genomics, Department of Human Genetics, KU Leuven, 3000 Leuven, Belgium
- KU Leuven Institute for Single Cell Omics (LISCO), KU Leuven, 3000 Leuven, Belgium
| |
Collapse
|
15
|
Sharma A, Steger RF, Li JM, Baude JA, Heom KA, Dey SS, Stowers RS. Sp1 mechanotransduction regulates breast cancer cell invasion in response to multiple tumor-mimicking extracellular matrix cues. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.03.18.643983. [PMID: 40166320 PMCID: PMC11957027 DOI: 10.1101/2025.03.18.643983] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 04/02/2025]
Abstract
Breast cancer progression is marked by extracellular matrix (ECM) remodeling, including increased stiffness, faster stress relaxation, and elevated collagen levels. In vitro experiments have revealed a role for each of these factors to individually promote malignant behavior, but their combined effects remain unclear. To address this, we developed alginate-collagen hydrogels with independently tunable stiffness, stress relaxation, and collagen density. We show that these combined tumor-mimicking ECM cues reinforced invasive morphologies and promoted spheroid invasion in breast cancer and mammary epithelial cells. High stiffness and low collagen density in slow-relaxing matrices led to the greatest cell migration speed and displacement. RNA-seq revealed Sp1 target gene enrichment in response to both individual and combined ECM cues, with a greater enrichment observed under multiple cues. Notably, high expression of Sp1 target genes upregulated by fast stress relaxation correlated with poor patient survival. Mechanistically, we found that phosphorylated-Sp1 (T453) was increasingly located in the nucleus in stiff and/or fast relaxing matrices, which was regulated by PI3K and ERK1/2 signaling, as well as actomyosin contractility. This study emphasizes how multiple ECM cues in complex microenvironments reinforce malignant traits and supports an emerging role for Sp1 as a mechanoresponsive transcription factor.
Collapse
Affiliation(s)
- Abhishek Sharma
- Department of Mechanical Engineering, University of California, Santa Barbara, Santa Barbara, CA, USA
| | - Rowan F Steger
- Department of Chemical Engineering, University of California, Santa Barbara, Santa Barbara, CA, USA
| | - Jen M Li
- Department of Molecular, Cellular, and Developmental Biology, University of California, Santa Barbara, Santa Barbara, CA, USA
| | - Jane A Baude
- Department of Molecular, Cellular, and Developmental Biology, University of California, Santa Barbara, Santa Barbara, CA, USA
| | - Kellie A Heom
- Department of Chemical Engineering, University of California, Santa Barbara, Santa Barbara, CA, USA
| | - Siddharth S Dey
- Department of Chemical Engineering, University of California, Santa Barbara, Santa Barbara, CA, USA
- Department of Bioengineering, University of California, Santa Barbara, Santa Barbara, CA, USA
| | - Ryan S Stowers
- Department of Mechanical Engineering, University of California, Santa Barbara, Santa Barbara, CA, USA
- Department of Bioengineering, University of California, Santa Barbara, Santa Barbara, CA, USA
| |
Collapse
|
16
|
Apostolou S, Donega V. Embracing the heterogeneity of neural stem cells in the subventricular zone. Stem Cell Reports 2025:102452. [PMID: 40118056 DOI: 10.1016/j.stemcr.2025.102452] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2024] [Revised: 02/20/2025] [Accepted: 02/21/2025] [Indexed: 03/23/2025] Open
Abstract
Neural stem cells (NSCs) of the subventricular zone (SVZ) could be a potential source for brain repair. These are heterogeneous cells with distinct activation states. To identify NSCs in the SVZ, different markers are used, including Gfap, Nestin, and Sox2. A comparison of these different methods to assess if the NSC marker used is selective toward specific NSC states is currently lacking. Here, we integrated six previously published single-cell RNA sequencing datasets from the adult mouse SVZ, where different methods were used to identify NSCs. Our data show that the approach used to isolate NSCs favors certain cell states over others. Our analyses underscore the importance of enriching for the NSC population of interest to increase data granularity. We also observed that cells with lower gene expression can be assigned incorrectly to clusters. We provide a framework for choosing the most optimal approach to enrich for NSC states of interest.
Collapse
Affiliation(s)
- Stefania Apostolou
- Amsterdam UMC location Vrije Universiteit Amsterdam, department of Anatomy and Neurosciences, De Boelelaan 1117, Amsterdam, the Netherlands
| | - Vanessa Donega
- Amsterdam UMC location Vrije Universiteit Amsterdam, department of Anatomy and Neurosciences, De Boelelaan 1117, Amsterdam, the Netherlands; Amsterdam Neuroscience, Cellular and Molecular Mechanisms, Amsterdam, the Netherlands.
| |
Collapse
|
17
|
Tjalsma SJD, Rinzema NJ, Verstegen MJAM, Robers MJ, Nieto-Aliseda A, Gremmen RA, Allahyar A, Muraro MJ, Krijger PHL, de Laat W. Long-range enhancer-controlled genes are hypersensitive to regulatory factor perturbations. CELL GENOMICS 2025; 5:100778. [PMID: 40010352 PMCID: PMC11960515 DOI: 10.1016/j.xgen.2025.100778] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/31/2024] [Revised: 12/18/2024] [Accepted: 01/29/2025] [Indexed: 02/28/2025]
Abstract
Cell-type-specific gene activation is regulated by enhancers, sometimes located at large genomic distances from target gene promoters. Whether distal enhancers require specific factors to orchestrate gene regulation remains unclear. Here, we used enhancer distance-controlled reporter screens to find candidate factors. We depleted them and employed activity-by-contact predictions to genome-wide classify genes based on enhancer distance. Predicted distal enhancers typically control tissue-restricted genes and often are strong enhancers. We find cohesin, but also mediator, most specifically required for long-range activation, with cohesin repressing short-range gene activation and prioritizing distal over proximal HBB genes competing for shared enhancers. Long-range controlled genes are also most sensitive to perturbations of other regulatory proteins and to BET inhibitor JQ1, this being more a consequence of their distinct enhancer features than distance. Our work predicts that lengthening of intervening sequences can help limit the expression of target genes to specialized cells with optimal trans-factor environments.
Collapse
Affiliation(s)
- Sjoerd J D Tjalsma
- Oncode Institute, Hubrecht Institute-KNAW and University Medical Center Utrecht, Utrecht, the Netherlands
| | - Niels J Rinzema
- Oncode Institute, Hubrecht Institute-KNAW and University Medical Center Utrecht, Utrecht, the Netherlands
| | - Marjon J A M Verstegen
- Oncode Institute, Hubrecht Institute-KNAW and University Medical Center Utrecht, Utrecht, the Netherlands
| | - Michelle J Robers
- Oncode Institute, Hubrecht Institute-KNAW and University Medical Center Utrecht, Utrecht, the Netherlands
| | - Andrea Nieto-Aliseda
- Oncode Institute, Hubrecht Institute-KNAW and University Medical Center Utrecht, Utrecht, the Netherlands
| | - Richard A Gremmen
- Oncode Institute, Hubrecht Institute-KNAW and University Medical Center Utrecht, Utrecht, the Netherlands
| | - Amin Allahyar
- Oncode Institute, Hubrecht Institute-KNAW and University Medical Center Utrecht, Utrecht, the Netherlands
| | | | - Peter H L Krijger
- Oncode Institute, Hubrecht Institute-KNAW and University Medical Center Utrecht, Utrecht, the Netherlands
| | - Wouter de Laat
- Oncode Institute, Hubrecht Institute-KNAW and University Medical Center Utrecht, Utrecht, the Netherlands.
| |
Collapse
|
18
|
Snabel RR, Cofiño-Fabrés C, Baltissen M, Schwach V, Passier R, Veenstra GJC. Cardiac differentiation roadmap for analysis of plasticity and balanced lineage commitment. Stem Cell Reports 2025; 20:102422. [PMID: 40020683 PMCID: PMC11960529 DOI: 10.1016/j.stemcr.2025.102422] [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: 11/06/2024] [Revised: 01/27/2025] [Accepted: 01/28/2025] [Indexed: 03/03/2025] Open
Abstract
Stem cell-based models of human heart tissue and cardiac differentiation employ monolayer and 3D organoid cultures with different properties, cell type composition, and maturity. Here we show how cardiac monolayer, embryoid body, and engineered heart tissue trajectories compare in a single-cell roadmap of atrial and ventricular differentiation conditions. Using a multiomic approach and gene-regulatory network inference, we identified regulators of the epicardial, atrial, and ventricular cardiomyocyte lineages. We identified ZNF711 as a regulatory switch and safeguard for cardiomyocyte commitment. We show that ZNF711 ablation prevents cardiomyocyte differentiation in the absence of retinoic acid, causing progenitors to be diverted more prominently to epicardial and other lineages. Retinoic acid rescues this shift in lineage commitment and promotes atrial cardiomyocyte differentiation by regulation of shared and complementary target genes, showing interplay between ZNF711 and retinoic acid in cardiac lineage commitment.
Collapse
Affiliation(s)
- Rebecca R Snabel
- Department of Molecular Developmental Biology, Radboud Institute for Molecular Life Sciences, Faculty of Science, Radboud University, Nijmegen, the Netherlands
| | - Carla Cofiño-Fabrés
- Department of Bioengineering Technologies, Applied Stem Cell Technologies Group, TechMed Centre, University of Twente, Enschede, the Netherlands
| | - Marijke Baltissen
- Department of Molecular Biology, Radboud Institute for Molecular Life Sciences, Faculty of Science, Oncode Institute, Radboud University, Nijmegen, the Netherlands
| | - Verena Schwach
- Department of Bioengineering Technologies, Applied Stem Cell Technologies Group, TechMed Centre, University of Twente, Enschede, the Netherlands
| | - Robert Passier
- Department of Bioengineering Technologies, Applied Stem Cell Technologies Group, TechMed Centre, University of Twente, Enschede, the Netherlands.
| | - Gert Jan C Veenstra
- Department of Molecular Developmental Biology, Radboud Institute for Molecular Life Sciences, Faculty of Science, Radboud University, Nijmegen, the Netherlands.
| |
Collapse
|
19
|
Cheung S, Bredikhin D, Gerber T, Steenbergen PJ, Basu S, Bailleul R, Hansen P, Paix A, Benton MA, Korswagen HC, Arendt D, Stegle O, Ikmi A. Systemic coordination of whole-body tissue remodeling during local regeneration in sea anemones. Dev Cell 2025; 60:780-793.e7. [PMID: 39615481 DOI: 10.1016/j.devcel.2024.11.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2024] [Revised: 08/06/2024] [Accepted: 11/01/2024] [Indexed: 03/14/2025]
Abstract
The complexity of regeneration extends beyond local wound responses, eliciting systemic processes across the entire organism. However, the functional relevance and coordination of distant molecular processes remain unclear. In the cnidarian Nematostella vectensis, we show that local regeneration triggers a systemic homeostatic response, leading to coordinated whole-body remodeling. Leveraging spatial transcriptomics, endogenous protein tagging, and live imaging, we comprehensively dissect this systemic response at the organismal scale. We identify proteolysis as a critical process driven by both local and systemic upregulation of metalloproteases. We show that metalloproteinase expression levels and activity scale with the extent of tissue loss. This proportional response drives long-range tissue and extracellular matrix movement. Our findings demonstrate the adaptive nature of the systematic response in regeneration, enabling the organism to maintain shape homeostasis while coping with a wide range of injuries.
Collapse
Affiliation(s)
- Stephanie Cheung
- European Molecular Biology Laboratory (EMBL), Developmental Biology Unit, Heidelberg 69117, Germany
| | - Danila Bredikhin
- European Molecular Biology Laboratory (EMBL), Genome Biology Unit, Heidelberg 69117, Germany; Division of Computational Genomics and Systems Genetics, German Cancer Research Center (DKFZ), Heidelberg 69120, Germany
| | - Tobias Gerber
- European Molecular Biology Laboratory (EMBL), Developmental Biology Unit, Heidelberg 69117, Germany; European Molecular Biology Laboratory (EMBL), Genome Biology Unit, Heidelberg 69117, Germany
| | - Petrus J Steenbergen
- European Molecular Biology Laboratory (EMBL), Developmental Biology Unit, Heidelberg 69117, Germany
| | - Soham Basu
- European Molecular Biology Laboratory (EMBL), Developmental Biology Unit, Heidelberg 69117, Germany
| | - Richard Bailleul
- European Molecular Biology Laboratory (EMBL), Developmental Biology Unit, Heidelberg 69117, Germany
| | - Pauline Hansen
- European Molecular Biology Laboratory (EMBL), Developmental Biology Unit, Heidelberg 69117, Germany
| | - Alexandre Paix
- European Molecular Biology Laboratory (EMBL), Developmental Biology Unit, Heidelberg 69117, Germany
| | - Matthew A Benton
- European Molecular Biology Laboratory (EMBL), Developmental Biology Unit, Heidelberg 69117, Germany
| | - Hendrik C Korswagen
- Hubrecht Institute, Royal Netherlands Academy of Arts and Sciences and University Medical Center Utrecht, Uppsalalaan 8, 3584 CT Utrecht, the Netherlands
| | - Detlev Arendt
- European Molecular Biology Laboratory (EMBL), Developmental Biology Unit, Heidelberg 69117, Germany
| | - Oliver Stegle
- European Molecular Biology Laboratory (EMBL), Genome Biology Unit, Heidelberg 69117, Germany; Division of Computational Genomics and Systems Genetics, German Cancer Research Center (DKFZ), Heidelberg 69120, Germany; Faculty of Biosciences, Heidelberg University, Heidelberg, Germany; Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton CB10 1SA, Cambridgeshire, UK.
| | - Aissam Ikmi
- European Molecular Biology Laboratory (EMBL), Developmental Biology Unit, Heidelberg 69117, Germany.
| |
Collapse
|
20
|
Ma L, Luan Y, Lu L. Analyze the Diversity and Function of Immune Cells in the Tumor Microenvironment From the Perspective of Single-Cell RNA Sequencing. Cancer Med 2025; 14:e70622. [PMID: 40062730 PMCID: PMC11891933 DOI: 10.1002/cam4.70622] [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: 08/29/2024] [Revised: 12/14/2024] [Accepted: 01/09/2025] [Indexed: 05/13/2025] Open
Abstract
BACKGROUND Cancer development is closely associated with complex alterations in the tumor microenvironment (TME). Among these, immune cells within the TME play a huge role in personalized tumor diagnosis and treatment. OBJECTIVES This review aims to summarize the diversity of immune cells in the TME, their impact on patient prognosis and treatment response, and the contributions of single-cell RNA sequencing (scRNA-seq) in understanding their functional heterogeneity. METHODS We analyzed recent studies utilizing scRNA-seq to investigate immune cell populations in the TME, focusing on their interactions and regulatory mechanisms. RESULTS ScRNA-seq reveals the functional heterogeneity of immune cells, enhances our understanding of their role in tumor antibody responses, and facilitates the construction of immune cell interaction networks. These insights provide guidance for the development of cancer immunotherapies and personalized treatment approaches. CONCLUSION Applying scRNA-seq to immune cell analysis in the TME offers a novel pathway for personalized cancer treatment. Despite its promise, several challenges remain, highlighting the need for further advancements to fully integrate scRNA-seq into clinical applications.
Collapse
Affiliation(s)
- Lujuan Ma
- Department of Medical Oncology, Guangzhou First People's Hospital, School of MedicineSouth China University of TechnologyGuangzhouGuangdongChina
| | - Yu Luan
- Department of Medical Oncology, Guangzhou First People's Hospital, School of MedicineSouth China University of TechnologyGuangzhouGuangdongChina
| | - Lin Lu
- Department of Medical Oncology, Guangzhou First People's Hospital, School of MedicineSouth China University of TechnologyGuangzhouGuangdongChina
| |
Collapse
|
21
|
Huang Y, Zhu S, Yao S, Zhai H, Liu C, Han JDJ. Unraveling aging from transcriptomics. Trends Genet 2025; 41:218-235. [PMID: 39424502 DOI: 10.1016/j.tig.2024.09.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2024] [Revised: 09/19/2024] [Accepted: 09/19/2024] [Indexed: 10/21/2024]
Abstract
Research into aging constitutes a pivotal endeavor aimed at elucidating the underlying biological mechanisms governing aging and age-associated diseases, as well as promoting healthy longevity. Recent advances in transcriptomic technologies, such as bulk RNA sequencing (RNA-seq), single-cell transcriptomics, and spatial transcriptomics, have revolutionized our ability to study aging at unprecedented resolution and scale. These technologies present novel opportunities for the discovery of biomarkers, elucidation of molecular pathways, and development of targeted therapeutic strategies for age-related disorders. This review surveys recent breakthroughs in different types of transcripts on aging, such as mRNA, long noncoding (lnc)RNA, tRNA, and miRNA, highlighting key findings and discussing their potential implications for future studies in this field.
Collapse
Affiliation(s)
- Yuanfang Huang
- Peking-Tsinghua Center for Life Sciences, Center for Quantitative Biology (CQB), Academy for Advanced Interdisciplinary Studies, Peking University, Beijing 100871, China
| | - Shouxuan Zhu
- Peking-Tsinghua Center for Life Sciences, Center for Quantitative Biology (CQB), Academy for Advanced Interdisciplinary Studies, Peking University, Beijing 100871, China
| | - Shuai Yao
- Peking-Tsinghua Center for Life Sciences, Center for Quantitative Biology (CQB), Academy for Advanced Interdisciplinary Studies, Peking University, Beijing 100871, China
| | - Haotian Zhai
- Peking-Tsinghua Center for Life Sciences, Center for Quantitative Biology (CQB), Academy for Advanced Interdisciplinary Studies, Peking University, Beijing 100871, China
| | - Chenyang Liu
- Peking-Tsinghua Center for Life Sciences, Center for Quantitative Biology (CQB), Academy for Advanced Interdisciplinary Studies, Peking University, Beijing 100871, China
| | - Jing-Dong J Han
- Peking-Tsinghua Center for Life Sciences, Center for Quantitative Biology (CQB), Academy for Advanced Interdisciplinary Studies, Peking University, Beijing 100871, China; Peking University Chengdu Academy for Advanced Interdisciplinary Biotechnologies, Chengdu, China.
| |
Collapse
|
22
|
Sullivan DK, Min KHJ, Hjörleifsson KE, Luebbert L, Holley G, Moses L, Gustafsson J, Bray NL, Pimentel H, Booeshaghi AS, Melsted P, Pachter L. kallisto, bustools and kb-python for quantifying bulk, single-cell and single-nucleus RNA-seq. Nat Protoc 2025; 20:587-607. [PMID: 39390263 DOI: 10.1038/s41596-024-01057-0] [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: 01/18/2024] [Accepted: 07/29/2024] [Indexed: 10/12/2024]
Abstract
The term 'RNA-seq' refers to a collection of assays based on sequencing experiments that involve quantifying RNA species from bulk tissue, single cells or single nuclei. The kallisto, bustools and kb-python programs are free, open-source software tools for performing this analysis that together can produce gene expression quantification from raw sequencing reads. The quantifications can be individualized for multiple cells, multiple samples or both. Additionally, these tools allow gene expression values to be classified as originating from nascent RNA species or mature RNA species, making this workflow amenable to both cell-based and nucleus-based assays. This protocol describes in detail how to use kallisto and bustools in conjunction with a wrapper, kb-python, to preprocess RNA-seq data. Execution of this protocol requires basic familiarity with a command line environment. With this protocol, quantification of a moderately sized RNA-seq dataset can be completed within minutes.
Collapse
Affiliation(s)
- Delaney K Sullivan
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, USA
- UCLA-Caltech Medical Scientist Training Program, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA
| | | | | | - Laura Luebbert
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, USA
| | | | - Lambda Moses
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, USA
| | | | | | - Harold Pimentel
- Department of Computer Science, University of California, Los Angeles, Los Angeles, CA, USA
- Department of Computational Medicine, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA
- Department of Human Genetics, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA
| | - A Sina Booeshaghi
- Department of Bioengineering, University of California, Berkeley, Berkeley, CA, USA.
| | - Páll Melsted
- deCODE Genetics/Amgen Inc., Reykjavik, Iceland.
- School of Engineering and Natural Sciences, University of Iceland, Reykjavik, Iceland.
| | - Lior Pachter
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, USA.
- Department of Computing and Mathematical Sciences, California Institute of Technology, Pasadena, CA, USA.
| |
Collapse
|
23
|
Zhao B, Song K, Wei DQ, Xiong Y, Ding J. scCobra allows contrastive cell embedding learning with domain adaptation for single cell data integration and harmonization. Commun Biol 2025; 8:233. [PMID: 39948393 PMCID: PMC11825689 DOI: 10.1038/s42003-025-07692-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2024] [Accepted: 02/06/2025] [Indexed: 02/16/2025] Open
Abstract
The rapid advancement of single-cell technologies has created an urgent need for effective methods to integrate and harmonize single-cell data. Technical and biological variations across studies complicate data integration, while conventional tools often struggle with reliance on gene expression distribution assumptions and over-correction. Here, we present scCobra, a deep generative neural network designed to overcome these challenges through contrastive learning with domain adaptation. scCobra effectively mitigates batch effects, minimizes over-correction, and ensures biologically meaningful data integration without assuming specific gene expression distributions. It enables online label transfer across datasets with batch effects, allowing continuous integration of new data without retraining. Additionally, scCobra supports batch effect simulation, advanced multi-omic integration, and scalable processing of large datasets. By integrating and harmonizing datasets from similar studies, scCobra expands the available data for investigating specific biological problems, improving cross-study comparability, and revealing insights that may be obscured in isolated datasets.
Collapse
Affiliation(s)
- Bowen Zhao
- State Key Laboratory of Microbial Metabolism, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai, China
- Meakins-Christie Laboratories, Department of Medicine, McGill University Health Centre, Montreal, QC, Canada
- Division of Experimental Medicine, Department of Medicine, McGill University, Montreal, QC, Canada
| | - Kailu Song
- Meakins-Christie Laboratories, Department of Medicine, McGill University Health Centre, Montreal, QC, Canada
- Quantitative Life Sciences, McGill University, Montreal, QC, Canada
| | - Dong-Qing Wei
- State Key Laboratory of Microbial Metabolism, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai, China
| | - Yi Xiong
- State Key Laboratory of Microbial Metabolism, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai, China.
| | - Jun Ding
- Meakins-Christie Laboratories, Department of Medicine, McGill University Health Centre, Montreal, QC, Canada.
- Division of Experimental Medicine, Department of Medicine, McGill University, Montreal, QC, Canada.
- Quantitative Life Sciences, McGill University, Montreal, QC, Canada.
- School of Computer Science, McGill University, Montreal, QC, Canada.
- Mila-Quebec AI Institute, Montreal, QC, Canada.
| |
Collapse
|
24
|
McManus RM, Komes MP, Griep A, Santarelli F, Schwartz S, Ramón Perea J, Schlachetzki JCM, Bouvier DS, Khalil MA, Lauterbach MA, Heinemann L, Schlüter T, Pour MS, Lovotti M, Stahl R, Duthie F, Rodríguez-Alcázar JF, Schmidt SV, Spitzer J, Noori P, Maillo A, Boettcher A, Herron B, McConville J, Gomez-Cabrero D, Tegnér J, Glass CK, Hiller K, Latz E, Heneka MT. NLRP3-mediated glutaminolysis controls microglial phagocytosis to promote Alzheimer's disease progression. Immunity 2025; 58:326-343.e11. [PMID: 39904338 DOI: 10.1016/j.immuni.2025.01.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2024] [Revised: 10/14/2024] [Accepted: 01/10/2025] [Indexed: 02/06/2025]
Abstract
Activation of the NLRP3 inflammasome has been implicated in the pathogenesis of Alzheimer's disease (AD) via the release of IL-1β and ASC specks. However, whether NLRP3 is involved in pathways beyond this remained unknown. Here, we found that Aβ deposition in vivo directly triggered NLRP3 activation in APP/PS1 mice, which model many features of AD. Loss of NLRP3 increased glutamine- and glutamate-related metabolism and increased expression of microglial Slc1a3, which was associated with enhanced mitochondrial and metabolic activity. The generation of α-ketoglutarate during this process impacted cellular function, including increased clearance of Aβ peptides as well as epigenetic and gene transcription changes. This pathway was conserved between murine and human cells. Critically, we could mimic this effect pharmacologically using NLRP3-specific inhibitors, but only with chronic NLRP3 inhibition. Together, these data demonstrate an additional role for NLRP3, where it can modulate mitochondrial and metabolic function, with important downstream consequences for the progression of AD.
Collapse
Affiliation(s)
- Róisín M McManus
- German Center for Neurodegenerative Diseases (DZNE), Venusberg Campus 1/99, 53127 Bonn, Germany; Institute of Innate Immunity, University Hospital Bonn, Bonn, Germany
| | - Max P Komes
- German Center for Neurodegenerative Diseases (DZNE), Venusberg Campus 1/99, 53127 Bonn, Germany; Institute of Innate Immunity, University Hospital Bonn, Bonn, Germany
| | - Angelika Griep
- German Center for Neurodegenerative Diseases (DZNE), Venusberg Campus 1/99, 53127 Bonn, Germany; Institute of Innate Immunity, University Hospital Bonn, Bonn, Germany
| | - Francesco Santarelli
- German Center for Neurodegenerative Diseases (DZNE), Venusberg Campus 1/99, 53127 Bonn, Germany; Institute of Innate Immunity, University Hospital Bonn, Bonn, Germany
| | | | - Juan Ramón Perea
- German Center for Neurodegenerative Diseases (DZNE), Venusberg Campus 1/99, 53127 Bonn, Germany; Department of Molecular Neuropathology, Centro de Biología Molecular "Severo Ochoa" (UAM-CSIC), Madrid 28049, Spain
| | - Johannes C M Schlachetzki
- Department of Neurosciences, University of California, San Diego, 9500 Gilman Drive, La Jolla, CA 92093, USA
| | - David S Bouvier
- Luxembourg Centre for Systems Biomedicine, University of Luxembourg, 4367 Belvaux, Luxembourg; Laboratoire National de Santé (LNS), National Center of Pathology (NCP), Dudelange, Luxembourg; Luxembourg Center of Neuropathology (LCNP), Dudelange, Luxembourg
| | - Michelle-Amirah Khalil
- Department of Bioinformatics and Biochemistry, Braunschweig Integrated Center of Systems Biology (BRICS), Technische Universität Braunschweig, Braunschweig, Germany
| | - Mario A Lauterbach
- German Center for Neurodegenerative Diseases (DZNE), Venusberg Campus 1/99, 53127 Bonn, Germany; Institute of Innate Immunity, University Hospital Bonn, Bonn, Germany
| | - Lea Heinemann
- German Center for Neurodegenerative Diseases (DZNE), Venusberg Campus 1/99, 53127 Bonn, Germany
| | - Titus Schlüter
- German Center for Neurodegenerative Diseases (DZNE), Venusberg Campus 1/99, 53127 Bonn, Germany; Institute of Innate Immunity, University Hospital Bonn, Bonn, Germany
| | - Mehran Shaban Pour
- German Center for Neurodegenerative Diseases (DZNE), Venusberg Campus 1/99, 53127 Bonn, Germany; Institute of Innate Immunity, University Hospital Bonn, Bonn, Germany
| | - Marta Lovotti
- Institute of Innate Immunity, University Hospital Bonn, Bonn, Germany
| | - Rainer Stahl
- Institute of Innate Immunity, University Hospital Bonn, Bonn, Germany
| | - Fraser Duthie
- Institute of Innate Immunity, University Hospital Bonn, Bonn, Germany
| | | | - Susanne V Schmidt
- Institute of Innate Immunity, University Hospital Bonn, Bonn, Germany
| | - Jasper Spitzer
- Institute of Innate Immunity, University Hospital Bonn, Bonn, Germany
| | - Peri Noori
- Unit of Computational Medicine, Center for Molecular Medicine, Department of Medicine Solna, Karolinska Institutet, Stockholm, Sweden
| | - Alberto Maillo
- Translational Bioinformatics Unit, Navarrabiomed, Universidad Pública de Navarra, Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Navarra, Spain
| | - Andreas Boettcher
- Novartis Institutes for BioMedical Research, Novartis Pharma AG, 4056 Basel, Switzerland
| | - Brian Herron
- Regional Neuropathology Service, Institute of Pathology, Royal Victoria Hospital, Belfast Health and Social Care Trust, Grosvenor Road, Belfast BT12 6BL, Northern Ireland
| | | | - David Gomez-Cabrero
- Translational Bioinformatics Unit, Navarrabiomed, Universidad Pública de Navarra, Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Navarra, Spain; Biological and Environmental Science and Engineering Division, King Abdullah University of Science and Technology (KAUST), Thuwal, Saudi Arabia
| | - Jesper Tegnér
- Unit of Computational Medicine, Center for Molecular Medicine, Department of Medicine Solna, Karolinska Institutet, Stockholm, Sweden; Biological and Environmental Science and Engineering Division, King Abdullah University of Science and Technology (KAUST), Thuwal, Saudi Arabia; Computer, Electrical and Mathematical Sciences and Engineering Division, King Abdullah University of Science and Technology (KAUST), Thuwal, Saudi Arabia; Science for Life Laboratory, Tomtebodavagen 23A, 17165, Solna, Sweden
| | - Christopher K Glass
- Department of Cellular and Molecular Medicine, University of California, San Diego, 9500 Gilman Drive, La Jolla, CA 92093, USA; Department of Medicine, University of California, San Diego, 9500 Gilman Drive, La Jolla, CA 92093, USA
| | - Karsten Hiller
- Department of Bioinformatics and Biochemistry, Braunschweig Integrated Center of Systems Biology (BRICS), Technische Universität Braunschweig, Braunschweig, Germany
| | - Eicke Latz
- German Center for Neurodegenerative Diseases (DZNE), Venusberg Campus 1/99, 53127 Bonn, Germany; Institute of Innate Immunity, University Hospital Bonn, Bonn, Germany; Centre of Molecular Inflammation Research, Norwegian University of Science and Technology, 7491 Trondheim, Norway; Division of Infectious Diseases and Immunology, University of Massachusetts Medical School, Worcester, MA 01605, USA; Deutsches Rheuma-Forschungszentrum (DRFZ), Charitéplatz 1, 10117 Berlin, Germany
| | - Michael T Heneka
- German Center for Neurodegenerative Diseases (DZNE), Venusberg Campus 1/99, 53127 Bonn, Germany; Institute of Innate Immunity, University Hospital Bonn, Bonn, Germany; Luxembourg Centre for Systems Biomedicine, University of Luxembourg, 4367 Belvaux, Luxembourg; Division of Infectious Diseases and Immunology, University of Massachusetts Medical School, Worcester, MA 01605, USA.
| |
Collapse
|
25
|
Mizoo T, Oka T, Sugahara O, Minato T, Higa T, Nakayama KI. GPLD1+ cancer stem cells contribute to chemotherapy resistance and tumour relapse in intestinal cancer. J Biochem 2025; 177:105-119. [PMID: 39743241 DOI: 10.1093/jb/mvae082] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2024] [Revised: 11/14/2024] [Accepted: 11/15/2024] [Indexed: 01/04/2025] Open
Abstract
Cancer stem cells (CSCs) play a central role in cancer progression, therapy resistance, and disease recurrence. With the use of a quadruple-mutant mouse intestinal cancer organoid model and single-cell RNA-sequencing analysis, we have now identified glycosylphosphatidylinositol-specific phospholipase D1 (GPLD1), an enzyme that catalyzes the cleavage of glycosylphosphatidylinositol (GPI) anchors of membrane proteins, as a marker of slowly cycling CSCs. Ablation of Gpld1+ cells in combination with 5-fluorouracil treatment greatly attenuated cell viability in and regrowth of the intestinal cancer organoids. In addition, we identified serine protease 8 (PRSS8) as a key substrate of GPLD1 in human colorectal cancer cells. GPLD1 cleaves the GPI anchor of PRSS8 and thereby mediates release of the protease from the plasma membrane, resulting in the activation of Wnt signalling and promotion of the epithelial-mesenchymal transition (EMT) in the cancer cells. Pharmacological inhibition of GPLD1 suppressed Wnt signalling activity and EMT in association with upregulation of the amount of functional PRSS8 at the plasma membrane. Our findings suggest that targeting of GPLD1 in colorectal cancer might contribute to a new therapeutic strategy that is based on suppression of Wnt signalling and EMT-related cancer progression driven by CSCs.
Collapse
Affiliation(s)
- Taisuke Mizoo
- Laboratory of Anticancer Strategies, Advanced Research Initiative, Institute of Science Tokyo, 1-5-45 Yushima, Bunkyo-ku, Tokyo 113-8510, Japan
- Department of Molecular and Cellular Biology, Medical Institute of Bioregulation, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka, Fukuoka 812-8582, Japan
| | - Takeru Oka
- Department of Molecular and Cellular Biology, Medical Institute of Bioregulation, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka, Fukuoka 812-8582, Japan
| | - Osamu Sugahara
- Laboratory of Anticancer Strategies, Advanced Research Initiative, Institute of Science Tokyo, 1-5-45 Yushima, Bunkyo-ku, Tokyo 113-8510, Japan
- Department of Molecular and Cellular Biology, Medical Institute of Bioregulation, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka, Fukuoka 812-8582, Japan
| | - Takafumi Minato
- Laboratory of Anticancer Strategies, Advanced Research Initiative, Institute of Science Tokyo, 1-5-45 Yushima, Bunkyo-ku, Tokyo 113-8510, Japan
- Department of Molecular and Cellular Biology, Medical Institute of Bioregulation, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka, Fukuoka 812-8582, Japan
| | - Tsunaki Higa
- Laboratory of Anticancer Strategies, Advanced Research Initiative, Institute of Science Tokyo, 1-5-45 Yushima, Bunkyo-ku, Tokyo 113-8510, Japan
- Department of Molecular and Cellular Biology, Medical Institute of Bioregulation, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka, Fukuoka 812-8582, Japan
| | - Keiichi I Nakayama
- Laboratory of Anticancer Strategies, Advanced Research Initiative, Institute of Science Tokyo, 1-5-45 Yushima, Bunkyo-ku, Tokyo 113-8510, Japan
- Department of Molecular and Cellular Biology, Medical Institute of Bioregulation, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka, Fukuoka 812-8582, Japan
| |
Collapse
|
26
|
Molla Desta G, Birhanu AG. Advancements in single-cell RNA sequencing and spatial transcriptomics: transforming biomedical research. Acta Biochim Pol 2025; 72:13922. [PMID: 39980637 PMCID: PMC11835515 DOI: 10.3389/abp.2025.13922] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2024] [Accepted: 01/20/2025] [Indexed: 02/22/2025]
Abstract
In recent years, significant advancements in biochemistry, materials science, engineering, and computer-aided testing have driven the development of high-throughput tools for profiling genetic information. Single-cell RNA sequencing (scRNA-seq) technologies have established themselves as key tools for dissecting genetic sequences at the level of single cells. These technologies reveal cellular diversity and allow for the exploration of cell states and transformations with exceptional resolution. Unlike bulk sequencing, which provides population-averaged data, scRNA-seq can detect cell subtypes or gene expression variations that would otherwise be overlooked. However, a key limitation of scRNA-seq is its inability to preserve spatial information about the RNA transcriptome, as the process requires tissue dissociation and cell isolation. Spatial transcriptomics is a pivotal advancement in medical biotechnology, facilitating the identification of molecules such as RNA in their original spatial context within tissue sections at the single-cell level. This capability offers a substantial advantage over traditional single-cell sequencing techniques. Spatial transcriptomics offers valuable insights into a wide range of biomedical fields, including neurology, embryology, cancer research, immunology, and histology. This review highlights single-cell sequencing approaches, recent technological developments, associated challenges, various techniques for expression data analysis, and their applications in disciplines such as cancer research, microbiology, neuroscience, reproductive biology, and immunology. It highlights the critical role of single-cell sequencing tools in characterizing the dynamic nature of individual cells.
Collapse
Affiliation(s)
- Getnet Molla Desta
- College of Veterinary Medicine, Jigjiga University, Jigjiga, Ethiopia
- Institute of Biotechnology, Addis Ababa University, Addis Ababa, Ethiopia
| | | |
Collapse
|
27
|
Zhang H, Li X, Song D, Yukselen O, Nanda S, Kucukural A, Li JJ, Garber M, Walhout AJ. Worm Perturb-Seq: massively parallel whole-animal RNAi and RNA-seq. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.02.02.636107. [PMID: 39975282 PMCID: PMC11838469 DOI: 10.1101/2025.02.02.636107] [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
The transcriptome provides a highly informative molecular phenotype to connect genotype to phenotype and is most frequently measured by RNA-sequencing (RNA-seq). Therefore, an ultimate goal is to perturb every gene and measure changes in the transcriptome. However, this remains challenging, especially in intact organisms due to different experimental and computational challenges. Here, we present 'Worm Perturb-Seq (WPS)', which provides high-resolution RNA-seq profiles for hundreds of replicate perturbations at a time in a living animal. WPS introduces multiple experimental advances that combine strengths of bulk and single cell RNA-seq, and that further provides an analytical framework, EmpirDE, that leverages the unique power of the large WPS datasets. EmpirDE identifies differentially expressed genes (DEGs) by using gene-specific empirical null distributions, rather than control conditions alone, thereby systematically removing technical biases and improving statistical rigor. We applied WPS to 103 Caenhorhabditis elegans nuclear hormone receptors (NHRs) to delineate a Gene Regulatory Network (GRN) and found that this GRN presents a striking 'pairwise modularity' where pairs of NHRs regulate shared target genes. We envision that the experimental and analytical advances of WPS should be useful not only for C. elegans, but will be broadly applicable to other models, including human cells.
Collapse
Affiliation(s)
- Hefei Zhang
- Department of Systems Biology, University of Massachusetts Chan Medical School, Worcester, MA, USA
| | - Xuhang Li
- Department of Systems Biology, University of Massachusetts Chan Medical School, Worcester, MA, USA
| | - Dongyuan Song
- Bioinformatics Interdepartmental Ph.D. Program, University of California, Los Angeles, CA, USA
| | | | - Shivani Nanda
- Department of Systems Biology, University of Massachusetts Chan Medical School, Worcester, MA, USA
| | - Alper Kucukural
- Via Scientific Inc. Cambridge, MA, USA
- Department of Genomics and Computational Biology, University of Massachusetts Chan Medical School, Worcester, MA, USA
| | - Jingyi Jessica Li
- Bioinformatics Interdepartmental Ph.D. Program, University of California, Los Angeles, CA, USA
- Department of Statistics and Data Science, Department of Biostatistics, Department of Computational Medicine, and Department of Human Genetics, University of California, Los Angeles, CA, USA
| | - Manuel Garber
- Department of Genomics and Computational Biology, University of Massachusetts Chan Medical School, Worcester, MA, USA
| | - Albertha J.M. Walhout
- Department of Systems Biology, University of Massachusetts Chan Medical School, Worcester, MA, USA
| |
Collapse
|
28
|
Lecornec N, Duchmann M, Itzykson R. Single-cell sequencing applications in acute myeloid leukemia. Leuk Lymphoma 2025; 66:175-189. [PMID: 39496597 DOI: 10.1080/10428194.2024.2422833] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2024] [Revised: 09/26/2024] [Accepted: 10/23/2024] [Indexed: 11/06/2024]
Abstract
Acute myeloid leukemia (AML) is a heterogeneous group of malignancies with poor prognosis. AML result from the proliferation of immature myeloid cells blocked at a variable stage of differentiation. Beyond inter-patient heterogeneity, AMLs are characterized by genetic and phenotypic intra-patient heterogeneity. Despite major advances in deciphering AML biology with bulk sequencing studies, pivotal questions remain unanswered. Analyses at the single-cell level could thus transform our understanding of these neoplasms. We review recent progresses in single-cell sequencing technologies from cell processing to bioinformatic pipelines. We next discuss how single-cell applications have helped understand the genetic and functional intra-leukemic heterogeneity, emphasizing aspects related to leukemic stem cells, clonal evolution and measurable residual disease (MRD) monitoring. We finally delineate how single-cell technologies could be implemented in routine clinical practice to improve patient management.
Collapse
MESH Headings
- Humans
- Leukemia, Myeloid, Acute/genetics
- Leukemia, Myeloid, Acute/diagnosis
- Leukemia, Myeloid, Acute/pathology
- Leukemia, Myeloid, Acute/therapy
- Single-Cell Analysis/methods
- Neoplasm, Residual/genetics
- Neoplasm, Residual/diagnosis
- Biomarkers, Tumor/genetics
- High-Throughput Nucleotide Sequencing/methods
- Clonal Evolution
- Neoplastic Stem Cells/pathology
- Neoplastic Stem Cells/metabolism
- Computational Biology/methods
- Prognosis
Collapse
Affiliation(s)
- Nicolas Lecornec
- Génomes, Biologie Cellulaire et Thérapeutique U944, INSERM, CNRS, Université Paris Cité, Paris, France
- Département d'Immuno-Hématologie Pédiatrique, Hôpital Robert-Debré, Assistance Publique Hôpitaux de Paris (AP-HP), Université Paris Cité, Paris, France
| | - Matthieu Duchmann
- Génomes, Biologie Cellulaire et Thérapeutique U944, INSERM, CNRS, Université Paris Cité, Paris, France
- Laboratoire d'Hématologie, Hôpital Saint-Louis, Assistance Publique-Hôpitaux de Paris (AP-HP), Université Paris Cité, Paris, France
| | - Raphael Itzykson
- Génomes, Biologie Cellulaire et Thérapeutique U944, INSERM, CNRS, Université Paris Cité, Paris, France
- Département Hématologie et Immunologie, Hôpital Saint-Louis, Assistance Publique-Hôpitaux de Paris, Paris, France
| |
Collapse
|
29
|
Marchant DB, Walbot V. The establishment of the anther somatic niche with single-cell sequencing. Dev Biol 2025; 518:37-47. [PMID: 39547468 DOI: 10.1016/j.ydbio.2024.11.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2023] [Revised: 10/25/2024] [Accepted: 11/12/2024] [Indexed: 11/17/2024]
Abstract
The anther is the developmental housing of pollen and therefore the male gametes of flowering plants. The meiotic cells from which pollen are derived must differentiate de novo from somatic anther cells and synchronously develop with the rest of the anther. Anthropogenic control over another development has become crucial for global agriculture so as to maintain inbred lines and generate hybrid seeds of many crops. Understanding the genes that underlie the proper differentiation, developmental landmarks, and functions of each anther cell type is thus fundamental to both basic and applied plant sciences. We investigated the development of the somatic niche of the maize (Zea mays) anther using single-cell RNA-seq (scRNA-seq). Extensive background knowledge on the birth then pace and pattern of cell division of the maize anther cell types and published examples of cell-type gene expression from in situ hybridization allowed us to identify the primary cell types within the anther lobe, as well as the connective cells between the four lobes. We established the developmental trajectories of somatic cell types from pre-meiosis to post-meiosis, identified putative marker genes for the somatic cell types that previously lacked any known specific functions, and addressed the possibility that tapetal cells sequentially differentiate. This comprehensive scRNA-seq dataset of the somatic niche of the maize anther will serve as a baseline for future analyses investigating male-sterile genotypes and the impact of environmental conditions on male fertility in flowering plants.
Collapse
Affiliation(s)
- D Blaine Marchant
- Department of Biology, University of Missouri - St. Louis, St. Louis, MO, 63121, USA; Department of Biology, Stanford University, Stanford, CA, 94305, USA.
| | - Virginia Walbot
- Department of Biology, Stanford University, Stanford, CA, 94305, USA.
| |
Collapse
|
30
|
Goss K, Horwitz EM. Single-cell multiomics to advance cell therapy. Cytotherapy 2025; 27:137-145. [PMID: 39530970 DOI: 10.1016/j.jcyt.2024.10.009] [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: 09/13/2024] [Revised: 10/21/2024] [Accepted: 10/21/2024] [Indexed: 11/16/2024]
Abstract
Single-cell RNA-sequencing (scRNAseq) was first introduced in 2009 and has evolved with many technological advancements over the last decade. Not only are there several scRNAseq platforms differing in many aspects, but there are also a large number of computational pipelines available for downstream analyses which are being developed at an exponential rate. Such computational data appear in many scientific publications in virtually every field of study; thus, investigators should be able to understand and interpret data in this rapidly evolving field. Here, we discuss key differences in scRNAseq platforms, crucial steps in scRNAseq experiments, standard downstream analyses and introduce newly developed multimodal approaches. We then discuss how single-cell omics has been applied to advance the field of cell therapy.
Collapse
Affiliation(s)
- Kyndal Goss
- Marcus Center for Advanced Cellular Therapy, Children's Healthcare of Atlanta, Atlanta, Georgia, USA; Aflac Cancer & Blood Disorders Center, Children's Healthcare of Atlanta, Atlanta, Georgia, USA; Graduate Division of Biology and Biomedical Sciences, Emory University Laney Graduate School, Atlanta, Georgia, USA
| | - Edwin M Horwitz
- Marcus Center for Advanced Cellular Therapy, Children's Healthcare of Atlanta, Atlanta, Georgia, USA; Aflac Cancer & Blood Disorders Center, Children's Healthcare of Atlanta, Atlanta, Georgia, USA; Department of Pediatrics, Emory University School of Medicine, Atlanta, Georgia, USA; Graduate Division of Biology and Biomedical Sciences, Emory University Laney Graduate School, Atlanta, Georgia, USA.
| |
Collapse
|
31
|
Tamminga SM, Van Der Wal MM, Saager ES, Van Der Gang LF, Boesjes CM, Hendriks A, Pannekoek Y, De Bruin MS, Van Wijk F, Van Sorge NM. Single-cell sequencing of human Langerhans cells identifies altered gene expression profiles in patients with atopic dermatitis. Immunohorizons 2025; 9:vlae009. [PMID: 39849992 PMCID: PMC11841975 DOI: 10.1093/immhor/vlae009] [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] [Accepted: 11/18/2024] [Indexed: 01/25/2025] Open
Abstract
Atopic dermatitis (AD) is characterized by dysregulated T cell immunity and skin microbiome dysbiosis with predominance of Staphylococcus aureus, which is associated with exacerbating AD skin inflammation. Specific glycosylation patterns of S. aureus cell wall structures amplify skin inflammation through interaction with Langerhans cells (LCs). Nevertheless, the role of LCs in AD remains poorly characterized. Here, we performed single cell RNA sequencing of primary epidermal LCs and dermal T cells, isolated from skin biopsies of AD patients and healthy control subjects, alongside specific glycoanalysis of S. aureus strains isolated from the AD lesions. Our findings revealed 4 LC subpopulations ie, 2 steady-state clusters [LC1 and LC1H] and 2 proinflammatory/matured subsets [LC2 and migratory LCs]. The latter 2 subsets were enriched in AD skin. AD LCs showed enhanced expression of C-type lectin receptors, the high-affinity IgE receptor, and activation of prostaglandin and leukotriene biosynthesis pathways, upregulated transcriptional signatures related to T cell activation pathways, and increased expression of CCL17 compared with healthy LCs. Correspondingly, T helper 2 and T regulatory cell populations were increased in AD lesions. Complementary, we performed bulk RNA sequencing of primary LCs stimulated with the S. aureus strains isolated from the AD lesions, which showed upregulation of T helper 2-related pathways. Our study provides proof-of-concept for a role of LCs in connecting the S. aureus-T cell axis in the AD inflammatory cycle.
Collapse
Affiliation(s)
- Sara M Tamminga
- Department of Medical Microbiology and Infection Prevention, Amsterdam UMC location University of Amsterdam, Amsterdam, the Netherlands
| | - M Marlot Van Der Wal
- Center for Translational Immunology, Wilhelmina Children’s Hospital, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands
| | - Elise S Saager
- Center for Translational Immunology, Wilhelmina Children’s Hospital, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands
| | - Lian F Van Der Gang
- National Expertise Center for Atopic Dermatitis, Department of Dermatology and Allergology, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Celeste M Boesjes
- National Expertise Center for Atopic Dermatitis, Department of Dermatology and Allergology, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Astrid Hendriks
- Department of Medical Microbiology and Infection Prevention, Amsterdam UMC location University of Amsterdam, Amsterdam, the Netherlands
| | - Yvonne Pannekoek
- Department of Medical Microbiology and Infection Prevention, Amsterdam UMC location University of Amsterdam, Amsterdam, the Netherlands
| | - Marjolein S De Bruin
- National Expertise Center for Atopic Dermatitis, Department of Dermatology and Allergology, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Femke Van Wijk
- Center for Translational Immunology, Wilhelmina Children’s Hospital, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands
| | - Nina M Van Sorge
- Department of Medical Microbiology and Infection Prevention, Amsterdam UMC location University of Amsterdam, Amsterdam, the Netherlands
- Netherlands Reference Laboratory for Bacterial Meningitis, Amsterdam UMC location AMC, Amsterdam, the Netherlands
| |
Collapse
|
32
|
Yang K, Islas N, Jewell S, Wu D, Jha A, Radens C, Pleiss J, Lynch K, Barash Y, Choi P. Machine learning-optimized targeted detection of alternative splicing. Nucleic Acids Res 2025; 53:gkae1260. [PMID: 39727154 PMCID: PMC11797022 DOI: 10.1093/nar/gkae1260] [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: 08/02/2024] [Revised: 10/31/2024] [Accepted: 12/10/2024] [Indexed: 12/28/2024] Open
Abstract
RNA sequencing (RNA-seq) is widely adopted for transcriptome analysis but has inherent biases that hinder the comprehensive detection and quantification of alternative splicing. To address this, we present an efficient targeted RNA-seq method that greatly enriches for splicing-informative junction-spanning reads. Local splicing variation sequencing (LSV-seq) utilizes multiplexed reverse transcription from highly scalable pools of primers anchored near splicing events of interest. Primers are designed using Optimal Prime, a novel machine learning algorithm trained on the performance of thousands of primer sequences. In experimental benchmarks, LSV-seq achieves high on-target capture rates and concordance with RNA-seq, while requiring significantly lower sequencing depth. Leveraging deep learning splicing code predictions, we used LSV-seq to target events with low coverage in GTEx RNA-seq data and newly discover hundreds of tissue-specific splicing events. Our results demonstrate the ability of LSV-seq to quantify splicing of events of interest at high-throughput and with exceptional sensitivity.
Collapse
Affiliation(s)
- Kevin Yang
- Department of Genetics, University of Pennsylvania, Philadelphia, PA 19104, USA
- Department of Pathology & Laboratory Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA
- Division of Cancer Pathobiology, Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Nathaniel Islas
- Department of Computer and Information Science, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - San Jewell
- Department of Genetics, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Di Wu
- Department of Computer and Information Science, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Anupama Jha
- Department of Genome Sciences, University of Washington, Seattle, WA 98195, USA
| | - Caleb M Radens
- Department of Genetics, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Jeffrey A Pleiss
- Department of Molecular Biology and Genetics, Cornell University, Ithaca, NY 14853, USA
| | - Kristen W Lynch
- Department of Biochemistry and Biophysics, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Yoseph Barash
- Department of Genetics, University of Pennsylvania, Philadelphia, PA 19104, USA
- Department of Computer and Information Science, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Peter S Choi
- Department of Pathology & Laboratory Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA
- Division of Cancer Pathobiology, Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA
| |
Collapse
|
33
|
Evangelista JE, Ali-Nasser T, Malek LE, Xie Z, Marino GB, Bester AC, Ma'ayan A. lncRNAlyzr: Enrichment Analysis for lncRNA Sets. J Mol Biol 2025:168938. [PMID: 40133794 DOI: 10.1016/j.jmb.2025.168938] [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: 11/14/2024] [Revised: 01/06/2025] [Accepted: 01/06/2025] [Indexed: 03/27/2025]
Abstract
lncRNAs make up a large portion of the human genome affecting many biological processes in normal physiology and diseases. However, human lncRNAs are understudied compared to protein-coding genes. While there are many tools for performing gene set enrichment analysis for coding genes, few tools exist for lncRNA enrichment analysis. lncRNAlyzr is a webserver application designed for lncRNAs enrichment analysis. lncRNAlyzr has a database containing 33 lncRNA set libraries created by computing correlations between lncRNAs and annotated coding gene sets. After users submit a set of lncRNAs to lncRNAlyzr, the enrichment analysis results are visualized as ball-and-stick subnetworks where nodes are lncRNAs connected to enrichment terms from across selected lncRNA set libraries. To demonstrate lncRNAlyzr, it was used to analyze the effects of knocking down the lncRNA CYTOR in K562 cells. Overall, lncRNAlyzr is an enrichment analysis tool for lncRNAs aiming to further our understanding of lncRNAs functional modules. lncRNAlyzr is available from: https://lncrnalyzr.maayanlab.cloud.
Collapse
Affiliation(s)
- John Erol Evangelista
- Mount Sinai Center for Bioinformatics, Department of Pharmacological Sciences, Department of Artificial Intelligence and Human Health, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, Box 1603, New York, NY 10029, USA.
| | - Tahleel Ali-Nasser
- Department of Biology, Technion-Israel Institute of Technology, 3200003 Haifa, Israel.
| | - Lauren E Malek
- Mount Sinai Center for Bioinformatics, Department of Pharmacological Sciences, Department of Artificial Intelligence and Human Health, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, Box 1603, New York, NY 10029, USA.
| | - Zhuorui Xie
- Mount Sinai Center for Bioinformatics, Department of Pharmacological Sciences, Department of Artificial Intelligence and Human Health, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, Box 1603, New York, NY 10029, USA.
| | - Giacomo B Marino
- Mount Sinai Center for Bioinformatics, Department of Pharmacological Sciences, Department of Artificial Intelligence and Human Health, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, Box 1603, New York, NY 10029, USA.
| | - Assaf C Bester
- Department of Biology, Technion-Israel Institute of Technology, 3200003 Haifa, Israel.
| | - Avi Ma'ayan
- Mount Sinai Center for Bioinformatics, Department of Pharmacological Sciences, Department of Artificial Intelligence and Human Health, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, Box 1603, New York, NY 10029, USA.
| |
Collapse
|
34
|
Argoetti A, Shalev D, Polyak G, Shima N, Biran H, Lahav T, Hashimshony T, Mandel-Gutfreund Y. lncRNA NORAD modulates STAT3/STAT1 balance and innate immune responses in human cells via interaction with STAT3. Nat Commun 2025; 16:571. [PMID: 39794357 PMCID: PMC11723954 DOI: 10.1038/s41467-025-55822-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2023] [Accepted: 12/30/2024] [Indexed: 01/13/2025] Open
Abstract
Long non-coding RNAs (lncRNAs) are pivotal regulators of cellular processes. Here we reveal an interaction between the lncRNA NORAD, noted for its role in DNA stability, and the immune related transcription factor STAT3 in embryonic and differentiated human cells. Results from NORAD knockdown experiments implicate NORAD in facilitating STAT3 nuclear localization and suppressing antiviral gene activation. In NORAD-deficient cells, STAT3 remains cytoplasmic, allowing STAT1 to enhance antiviral activity. Analysis of RNA expression data from in vitro experiments and clinical samples demonstrates reduced NORAD upon viral infection. Additionally, evolutionary conservation analysis suggests that this regulatory function of NORAD is restricted to humans, potentially owing to the introduction of an Alu element in hominoids. Our findings thus suggest that NORAD functions as a modulator of STAT3-mediated immune suppression, adding to the understanding of lncRNAs in immune regulation and evolutionary adaptation in host defense mechanisms.
Collapse
Affiliation(s)
- Amir Argoetti
- Technion-Israel Institute of Technology, Faculty of Biology, Emerson building, Haifa, Israel
| | - Dor Shalev
- Technion-Israel Institute of Technology, Faculty of Biology, Emerson building, Haifa, Israel
| | - Galia Polyak
- Technion-Israel Institute of Technology, Faculty of Biology, Emerson building, Haifa, Israel
| | - Noa Shima
- Technion-Israel Institute of Technology, Faculty of Biology, Emerson building, Haifa, Israel
| | - Hadas Biran
- Technion-Israel Institute of Technology, Faculty of Computer Science, Taub building, Haifa, Israel
| | - Tamar Lahav
- Technion-Israel Institute of Technology, Faculty of Biology, Emerson building, Haifa, Israel
| | - Tamar Hashimshony
- Technion-Israel Institute of Technology, Faculty of Biology, Emerson building, Haifa, Israel
| | - Yael Mandel-Gutfreund
- Technion-Israel Institute of Technology, Faculty of Biology, Emerson building, Haifa, Israel.
- Technion-Israel Institute of Technology, Faculty of Computer Science, Taub building, Haifa, Israel.
| |
Collapse
|
35
|
Triscott J, Lehner M, Benjak A, Reist M, Emerling BM, Ng CK, de Brot S, Rubin MA. Loss of PI5P4Kα Slows the Progression of a Pten Mutant Basal Cell Model of Prostate Cancer. Mol Cancer Res 2025; 23:33-45. [PMID: 39382632 PMCID: PMC7616865 DOI: 10.1158/1541-7786.mcr-24-0290] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2024] [Revised: 09/15/2024] [Accepted: 10/07/2024] [Indexed: 10/10/2024]
Abstract
Although early prostate cancer depends on the androgen receptor signaling pathway, which is predominant in luminal cells, there is much to be understood about the contribution of epithelial basal cells in cancer progression. Herein, we observe cell type-specific differences in the importance of the metabolic enzyme phosphatidylinositol 5-phosphate 4-kinase alpha (PI5P4Kα; gene name PIP4K2A) in the prostate epithelium. We report the development of a basal cell-specific genetically engineered mouse model targeting Pip4k2a alone or in combination with the tumor suppressor phosphatase and tensin homolog (Pten). PI5P4Kα is enriched in basal cells, and no major histopathologic changes were detectable following gene deletion. Notably, the combined loss of Pip4k2a slowed the development of Pten mutant mouse prostatic intraepithelial neoplasia. Through the inclusion of a lineage tracing reporter, we utilize single-cell RNA sequencing to evaluate changes resulting from in vivo downregulation of Pip4k2a and characterize cell populations influenced in the established Probasin-Cre- and cytokeratin 5-Cre-driven genetically engineered mouse model. Transcriptomic pathway analysis points toward the disruption of lipid metabolism as a mechanism for reduced tumor progression. This was functionally supported by shifts of carnitine lipids in LNCaP prostate cancer cells treated with siPIP4K2A. Overall, these data nominate PI5P4Kα as a target for PTEN mutant prostate cancer. Implications: PI5P4Kα is enriched in prostate basal cells, and its targeted loss slows the progression of a model of advanced prostate cancer.
Collapse
Affiliation(s)
- Joanna Triscott
- Department for BioMedical Research, University of Bern, Bern, Switzerland
| | - Marika Lehner
- Department for BioMedical Research, University of Bern, Bern, Switzerland
| | - Andrej Benjak
- Department for BioMedical Research, University of Bern, Bern, Switzerland
| | - Matthias Reist
- Department for BioMedical Research, University of Bern, Bern, Switzerland
| | - Brooke M. Emerling
- Cancer Metabolism and Microenvironment Program, Sanford Burnham Prebys, La Jolla, California
| | - Charlotte K.Y. Ng
- Department for BioMedical Research, University of Bern, Bern, Switzerland
- Bern Center for Precision Medicine, University of Bern and Inselspital, Bern, Switzerland
- SIB Swiss Institute of Bioinformatics, Lausanne, Switzerland
| | - Simone de Brot
- Bern Center for Precision Medicine, University of Bern and Inselspital, Bern, Switzerland
- COMPATH, Institute of Animal Pathology, University of Bern, Bern, Switzerland
| | - Mark A. Rubin
- Department for BioMedical Research, University of Bern, Bern, Switzerland
- Bern Center for Precision Medicine, University of Bern and Inselspital, Bern, Switzerland
| |
Collapse
|
36
|
Sun F, Li H, Sun D, Fu S, Gu L, Shao X, Wang Q, Dong X, Duan B, Xing F, Wu J, Xiao M, Zhao F, Han JDJ, Liu Q, Fan X, Li C, Wang C, Shi T. Single-cell omics: experimental workflow, data analyses and applications. SCIENCE CHINA. LIFE SCIENCES 2025; 68:5-102. [PMID: 39060615 DOI: 10.1007/s11427-023-2561-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/07/2023] [Accepted: 04/18/2024] [Indexed: 07/28/2024]
Abstract
Cells are the fundamental units of biological systems and exhibit unique development trajectories and molecular features. Our exploration of how the genomes orchestrate the formation and maintenance of each cell, and control the cellular phenotypes of various organismsis, is both captivating and intricate. Since the inception of the first single-cell RNA technology, technologies related to single-cell sequencing have experienced rapid advancements in recent years. These technologies have expanded horizontally to include single-cell genome, epigenome, proteome, and metabolome, while vertically, they have progressed to integrate multiple omics data and incorporate additional information such as spatial scRNA-seq and CRISPR screening. Single-cell omics represent a groundbreaking advancement in the biomedical field, offering profound insights into the understanding of complex diseases, including cancers. Here, we comprehensively summarize recent advances in single-cell omics technologies, with a specific focus on the methodology section. This overview aims to guide researchers in selecting appropriate methods for single-cell sequencing and related data analysis.
Collapse
Affiliation(s)
- Fengying Sun
- Department of Clinical Laboratory, the Affiliated Wuhu Hospital of East China Normal University (The Second People's Hospital of Wuhu City), Wuhu, 241000, China
| | - Haoyan Li
- Pharmaceutical Informatics Institute, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China
| | - Dongqing Sun
- Key Laboratory of Spine and Spinal Cord Injury Repair and Regeneration (Tongji University), Ministry of Education, Orthopaedic Department, Tongji Hospital, Bioinformatics Department, School of Life Sciences and Technology, Tongji University, Shanghai, 200082, China
- Frontier Science Center for Stem Cells, School of Life Sciences and Technology, Tongji University, Shanghai, 200092, China
| | - Shaliu Fu
- Key Laboratory of Spine and Spinal Cord Injury Repair and Regeneration (Tongji University), Ministry of Education, Orthopaedic Department, Tongji Hospital, Bioinformatics Department, School of Life Sciences and Technology, Tongji University, Shanghai, 200082, China
- Translational Medical Center for Stem Cell Therapy and Institute for Regenerative Medicine, Shanghai East Hospital, Bioinformatics Department, School of Life Sciences and Technology, Tongji University, Shanghai, 200082, China
- Research Institute of Intelligent Computing, Zhejiang Lab, Hangzhou, 311121, China
- Shanghai Research Institute for Intelligent Autonomous Systems, Shanghai, 201210, China
| | - Lei Gu
- Center for Single-cell Omics, School of Public Health, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Xin Shao
- Pharmaceutical Informatics Institute, 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
| | - Qinqin Wang
- Center for Single-cell Omics, School of Public Health, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Xin Dong
- Key Laboratory of Spine and Spinal Cord Injury Repair and Regeneration (Tongji University), Ministry of Education, Orthopaedic Department, Tongji Hospital, Bioinformatics Department, School of Life Sciences and Technology, Tongji University, Shanghai, 200082, China
- Frontier Science Center for Stem Cells, School of Life Sciences and Technology, Tongji University, Shanghai, 200092, China
| | - Bin Duan
- Key Laboratory of Spine and Spinal Cord Injury Repair and Regeneration (Tongji University), Ministry of Education, Orthopaedic Department, Tongji Hospital, Bioinformatics Department, School of Life Sciences and Technology, Tongji University, Shanghai, 200082, China
- Translational Medical Center for Stem Cell Therapy and Institute for Regenerative Medicine, Shanghai East Hospital, Bioinformatics Department, School of Life Sciences and Technology, Tongji University, Shanghai, 200082, China
- Research Institute of Intelligent Computing, Zhejiang Lab, Hangzhou, 311121, China
- Shanghai Research Institute for Intelligent Autonomous Systems, Shanghai, 201210, China
| | - Feiyang Xing
- Key Laboratory of Spine and Spinal Cord Injury Repair and Regeneration (Tongji University), Ministry of Education, Orthopaedic Department, Tongji Hospital, Bioinformatics Department, School of Life Sciences and Technology, Tongji University, Shanghai, 200082, China
- Frontier Science Center for Stem Cells, School of Life Sciences and Technology, Tongji University, Shanghai, 200092, China
| | - Jun Wu
- Center for Bioinformatics and Computational Biology, Shanghai Key Laboratory of Regulatory Biology, the Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai, 200241, China
| | - Minmin Xiao
- Department of Clinical Laboratory, the Affiliated Wuhu Hospital of East China Normal University (The Second People's Hospital of Wuhu City), Wuhu, 241000, China.
| | - Fangqing Zhao
- Beijing Institutes of Life Science, Chinese Academy of Sciences, Beijing, 100101, China.
| | - Jing-Dong J Han
- Peking-Tsinghua Center for Life Sciences, Academy for Advanced Interdisciplinary Studies, Center for Quantitative Biology (CQB), Peking University, Beijing, 100871, China.
| | - Qi Liu
- Key Laboratory of Spine and Spinal Cord Injury Repair and Regeneration (Tongji University), Ministry of Education, Orthopaedic Department, Tongji Hospital, Bioinformatics Department, School of Life Sciences and Technology, Tongji University, Shanghai, 200082, China.
- Translational Medical Center for Stem Cell Therapy and Institute for Regenerative Medicine, Shanghai East Hospital, Bioinformatics Department, School of Life Sciences and Technology, Tongji University, Shanghai, 200082, China.
- Research Institute of Intelligent Computing, Zhejiang Lab, Hangzhou, 311121, China.
- Shanghai Research Institute for Intelligent Autonomous Systems, Shanghai, 201210, China.
| | - Xiaohui Fan
- Pharmaceutical Informatics Institute, 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.
- Zhejiang Key Laboratory of Precision Diagnosis and Therapy for Major Gynecological Diseases, Women's Hospital, Zhejiang University School of Medicine, Hangzhou, 310006, China.
| | - Chen Li
- Center for Single-cell Omics, School of Public Health, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China.
| | - Chenfei Wang
- Key Laboratory of Spine and Spinal Cord Injury Repair and Regeneration (Tongji University), Ministry of Education, Orthopaedic Department, Tongji Hospital, Bioinformatics Department, School of Life Sciences and Technology, Tongji University, Shanghai, 200082, China.
- Frontier Science Center for Stem Cells, School of Life Sciences and Technology, Tongji University, Shanghai, 200092, China.
| | - Tieliu Shi
- Department of Clinical Laboratory, the Affiliated Wuhu Hospital of East China Normal University (The Second People's Hospital of Wuhu City), Wuhu, 241000, China.
- Center for Bioinformatics and Computational Biology, Shanghai Key Laboratory of Regulatory Biology, the Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai, 200241, China.
- Key Laboratory of Advanced Theory and Application in Statistics and Data Science-MOE, School of Statistics, East China Normal University, Shanghai, 200062, China.
| |
Collapse
|
37
|
Bouwman M, de Bakker DEM, Honkoop H, Giovou AE, Versteeg D, Boender AR, Nguyen PD, Slotboom M, Colquhoun D, Vigil-Garcia M, Kooijman L, Janssen R, Hooijkaas IB, Günthel M, Visser KJ, Klerk M, Zentilin L, Giacca M, Kaslin J, Boink GJJ, van Rooij E, Christoffels VM, Bakkers J. Cross-species comparison reveals that Hmga1 reduces H3K27me3 levels to promote cardiomyocyte proliferation and cardiac regeneration. NATURE CARDIOVASCULAR RESEARCH 2025; 4:64-82. [PMID: 39747457 PMCID: PMC11738996 DOI: 10.1038/s44161-024-00588-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/15/2023] [Accepted: 11/26/2024] [Indexed: 01/04/2025]
Abstract
In contrast to adult mammalian hearts, the adult zebrafish heart efficiently replaces cardiomyocytes lost after injury. Here we reveal shared and species-specific injury response pathways and a correlation between Hmga1, an architectural non-histone protein, and regenerative capacity, as Hmga1 is required and sufficient to induce cardiomyocyte proliferation and required for heart regeneration. In addition, Hmga1 was shown to reactivate developmentally silenced genes, likely through modulation of H3K27me3 levels, poising them for a pro-regenerative gene program. Furthermore, AAV-mediated Hmga1 expression in injured adult mouse hearts led to controlled cardiomyocyte proliferation in the border zone and enhanced heart function, without cardiomegaly and adverse remodeling. Histone modification mapping in mouse border zone cardiomyocytes revealed a similar modulation of H3K27me3 marks, consistent with findings in zebrafish. Our study demonstrates that Hmga1 mediates chromatin remodeling and drives a regenerative program, positioning it as a promising therapeutic target to enhance cardiac regeneration after injury.
Collapse
Affiliation(s)
- Mara Bouwman
- Hubrecht Institute, Royal Netherlands Academy of Arts and Sciences (KNAW) and University Medical Center Utrecht, Utrecht, The Netherlands
| | - Dennis E M de Bakker
- Hubrecht Institute, Royal Netherlands Academy of Arts and Sciences (KNAW) and University Medical Center Utrecht, Utrecht, The Netherlands
- Leibniz Institute on Aging, Fritz Lipmann Institute (FLI), Jena, Germany
| | - Hessel Honkoop
- Hubrecht Institute, Royal Netherlands Academy of Arts and Sciences (KNAW) and University Medical Center Utrecht, Utrecht, The Netherlands
| | - Alexandra E Giovou
- Department of Medical Biology, Amsterdam Cardiovascular Sciences, Amsterdam University Medical Centers, Amsterdam, The Netherlands
| | - Danielle Versteeg
- Hubrecht Institute, Royal Netherlands Academy of Arts and Sciences (KNAW) and University Medical Center Utrecht, Utrecht, The Netherlands
| | - Arie R Boender
- Department of Medical Biology, Amsterdam Cardiovascular Sciences, Amsterdam University Medical Centers, Amsterdam, The Netherlands
- PacingCure BV, Amsterdam, The Netherlands
| | - Phong D Nguyen
- Hubrecht Institute, Royal Netherlands Academy of Arts and Sciences (KNAW) and University Medical Center Utrecht, Utrecht, The Netherlands
- Institut Curie, Université PSL, CNRS UMR3215, INSERM U934, Paris, France
| | - Merel Slotboom
- Hubrecht Institute, Royal Netherlands Academy of Arts and Sciences (KNAW) and University Medical Center Utrecht, Utrecht, The Netherlands
| | - Daniel Colquhoun
- Australian Regenerative Medicine Institute, Monash University, Melbourne, VIC, Australia
| | - Marta Vigil-Garcia
- Hubrecht Institute, Royal Netherlands Academy of Arts and Sciences (KNAW) and University Medical Center Utrecht, Utrecht, The Netherlands
| | - Lieneke Kooijman
- Hubrecht Institute, Royal Netherlands Academy of Arts and Sciences (KNAW) and University Medical Center Utrecht, Utrecht, The Netherlands
| | - Rob Janssen
- Department of Medical Biology, Amsterdam Cardiovascular Sciences, Amsterdam University Medical Centers, Amsterdam, The Netherlands
| | - Ingeborg B Hooijkaas
- Department of Medical Biology, Amsterdam Cardiovascular Sciences, Amsterdam University Medical Centers, Amsterdam, The Netherlands
| | - Marie Günthel
- Department of Medical Biology, Amsterdam Cardiovascular Sciences, Amsterdam University Medical Centers, Amsterdam, The Netherlands
| | - Kimberly J Visser
- Hubrecht Institute, Royal Netherlands Academy of Arts and Sciences (KNAW) and University Medical Center Utrecht, Utrecht, The Netherlands
| | - Mischa Klerk
- Department of Medical Biology, Amsterdam Cardiovascular Sciences, Amsterdam University Medical Centers, Amsterdam, The Netherlands
| | - Lorena Zentilin
- International Centre for Genetic Engineering and Biotechnology (ICGEB), University of Trieste, Trieste, Italy
| | - Mauro Giacca
- International Centre for Genetic Engineering and Biotechnology (ICGEB), University of Trieste, Trieste, Italy
- School of Cardiovascular and Metabolic Medicine and Sciences, King's College London, London, UK
| | - Jan Kaslin
- Australian Regenerative Medicine Institute, Monash University, Melbourne, VIC, Australia
| | - Gerard J J Boink
- Department of Medical Biology, Amsterdam Cardiovascular Sciences, Amsterdam University Medical Centers, Amsterdam, The Netherlands
- PacingCure BV, Amsterdam, The Netherlands
- Department of Cardiology, Amsterdam Cardiovascular Sciences, Amsterdam University Medical Centers, Amsterdam, The Netherlands
| | - Eva van Rooij
- Hubrecht Institute, Royal Netherlands Academy of Arts and Sciences (KNAW) and University Medical Center Utrecht, Utrecht, The Netherlands
- Department of Cardiology, Division of Heart and Lungs, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Vincent M Christoffels
- Department of Medical Biology, Amsterdam Cardiovascular Sciences, Amsterdam University Medical Centers, Amsterdam, The Netherlands
| | - Jeroen Bakkers
- Hubrecht Institute, Royal Netherlands Academy of Arts and Sciences (KNAW) and University Medical Center Utrecht, Utrecht, The Netherlands.
- Department of Pediatric Cardiology, Division of Pediatrics, University Medical Center Utrecht, Utrecht, The Netherlands.
| |
Collapse
|
38
|
Zhao Y, Shang J, Qin B, Zhang L, He X, Ge D, Ren Q, Liu JX. pscAdapt: Pre-Trained Domain Adaptation Network Based on Structural Similarity for Cell Type Annotation in Single Cell RNA-seq Data. IEEE J Biomed Health Inform 2025; 29:724-732. [PMID: 39325614 DOI: 10.1109/jbhi.2024.3468310] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/28/2024]
Abstract
Cell type annotation refers to the process of categorizing and labeling cells to identify their specific cell types, which is crucial for understanding cell functions and biological processes. Although many methods have been developed for automated cell type annotation, they often encounter challenges such as batch effects due to variations in data distribution across platforms and species, thereby compromising their performance. To address batch effects, in this study, a pre-trained domain adaptation model based on structural similarity, named pscAdapt, is proposed for cell type annotation. Specifically, a pre-trained strategy is employed to initialize model parameters to learn the data distribution of source domain. This strategy is also combined with an adversarial learning strategy to train the domain adaptation network for achieving domain level alignment and reducing domain discrepancy. Furthermore, to better distinguish different types of cells, a structural similarity loss is designed, aiming to shorten distances between cells of the same type and increase distances between cells of different types in feature space, thus achieving cell level alignment and enhancing the discriminability of cell types. Comprehensive experiments were conducted on simulated datasets, cross-platforms datasets and cross-species datasets to validate the effectiveness of pscAdapt, results of which demonstrate that pscAdapt outperforms several popular cell type annotation methods.
Collapse
|
39
|
Lahaie SC, Brezner N, Murai KK. Single-cell omics and heterogeneity of neuroglial cells. HANDBOOK OF CLINICAL NEUROLOGY 2025; 209:265-275. [PMID: 40122628 DOI: 10.1016/b978-0-443-19104-6.00013-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/25/2025]
Abstract
Our bodies contain a rich diversity of cell types with unique physiologic properties. Interestingly, cells within our bodies contain the same DNA content, yet they can vary dramatically with respect to their molecular, structural, and functional properties. The need to better understand cellular complexity and diversity in biologic systems has led to a technical revolution in the field through the development of sophisticated single-cell "omic" approaches. This allows the investigation of the genome, epigenome, transcriptome, and proteome of individual cells derived from complex samples or tissues, such as nervous system tissue. These methods are allowing scientists to detect distinct cell populations and cellular states in different species (including rodent and human) and molecular transitions of cell populations across the lifespan. Recent studies have revealed that astrocytes, oligodendrocytes, and microglia exhibit greater molecular and functional heterogeneity than originally thought and innovative single-cell technologies have allowed a more comprehensive and less biased view of this cellular diversity. The chapter begins by providing a primer of single-cell transcriptomic and spatial transcriptomic approaches that have been particularly influential in uncovering single-cell diversity of neuroglial cells in the brain. It then takes a closer look at how these technologies have been pivotal in defining neuroglial cell subtypes and for determining their spatial relationships within the CNS. Then, it concludes with discussion of how the recent technical advances and discoveries have provoked new questions about the origin, organization, and functional purpose of diverse neuroglial cell subtypes.
Collapse
Affiliation(s)
- Sylvie C Lahaie
- Centre for Research in Neuroscience, Department of Neurology & Neurosurgery, Brain Repair and Integrative Neuroscience Program, Research Institute of the McGill University Health Centre, Montreal General Hospital, Montreal, QC, Canada
| | - Naama Brezner
- Centre for Research in Neuroscience, Department of Neurology & Neurosurgery, Brain Repair and Integrative Neuroscience Program, Research Institute of the McGill University Health Centre, Montreal General Hospital, Montreal, QC, Canada
| | - Keith K Murai
- Centre for Research in Neuroscience, Department of Neurology & Neurosurgery, Brain Repair and Integrative Neuroscience Program, Research Institute of the McGill University Health Centre, Montreal General Hospital, Montreal, QC, Canada; Quantitative Life Sciences Graduate Program, McGill University, Montreal, QC, Canada.
| |
Collapse
|
40
|
Wu H, Yang ASP, Stelloo S, Roos FJM, te Morsche RHM, Verkerk AH, Luna-Velez MV, Wingens L, de Wilt JHW, Sauerwein RW, Mulder KW, van Heeringen SJ, Verstegen MMA, van der Laan LJW, Marks H, Bártfai R. Multi-omics analysis reveals distinct gene regulatory mechanisms between primary and organoid-derived human hepatocytes. Dis Model Mech 2025; 18:dmm050883. [PMID: 39878507 PMCID: PMC11810045 DOI: 10.1242/dmm.050883] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2024] [Accepted: 11/25/2024] [Indexed: 01/31/2025] Open
Abstract
Hepatic organoid cultures are a powerful model to study liver development and diseases in vitro. However, hepatocyte-like cells differentiated from these organoids remain immature compared to primary human hepatocytes (PHHs), which are the benchmark in the field. Here, we applied integrative single-cell transcriptome and chromatin accessibility analysis to reveal gene regulatory mechanisms underlying these differences. We found that, in mature human hepatocytes, activator protein 1 (AP-1) factors co-occupy regulatory regions with hepatocyte-specific transcription factors, including HNF4A, suggesting their potential cooperation in governing hepatic gene expression. Comparative analysis identified distinct transcription factor sets that are specifically active in either PHHs or intrahepatic cholangiocyte organoid (ICO)-derived human hepatocytes. ELF3 was one of the factors uniquely expressed in ICO-derived hepatocytes, and its expression negatively correlated with hepatic marker gene expression. Functional analysis further revealed that ELF3 depletion increased the expression of key hepatic markers in ICO-derived hepatocytes. Our integrative analysis provides insights into the transcriptional regulatory networks of PHHs and hepatic organoids, thereby informing future strategies for developing improved hepatic models.
Collapse
Affiliation(s)
- Haoyu Wu
- Department of Molecular Biology, Faculty of Science, Radboud Institute for Molecular Life Science, Radboud University, Nijmegen 6525GA, The Netherlands
| | - Annie S. P. Yang
- Center for Infectious Diseases, Department of Medical Microbiology, Radboud University Medical Center, Nijmegen 6500HB, The Netherlands
| | - Suzan Stelloo
- Department of Molecular Biology, Faculty of Science, Radboud Institute for Molecular Life Science, Radboud University, Nijmegen 6525GA, The Netherlands
- Department of Molecular Biology, Faculty of Science, Radboud Institute for Molecular Life Science, Oncode Institute, Radboud University, Nijmegen 6525GA, The Netherlands
| | - Floris J. M. Roos
- Department of Surgery, Erasmus University Medical Center Transplant Institute, University Medical Center Rotterdam,Rotterdam 3000CA, TheNetherlands
| | - René H. M. te Morsche
- Department of Gastroenterology and Hepatology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen 6500HB, The Netherlands
| | - Anne H. Verkerk
- Department of Molecular Biology, Faculty of Science, Radboud Institute for Molecular Life Science, Radboud University, Nijmegen 6525GA, The Netherlands
| | - Maria V. Luna-Velez
- Department of Molecular Biology, Faculty of Science, Radboud Institute for Molecular Life Science, Radboud University, Nijmegen 6525GA, The Netherlands
- Department of Molecular Biology, Faculty of Science, Radboud Institute for Molecular Life Science, Oncode Institute, Radboud University, Nijmegen 6525GA, The Netherlands
| | - Laura Wingens
- Department of Molecular Developmental Biology, Faculty of Science, Radboud Institute for Molecular Life Sciences, Radboud University, Nijmegen 6525GA, The Netherlands
| | - Johannes H. W. de Wilt
- Department of Surgery, Radboud University Medical Center, Nijmegen 6500HB, The Netherlands
| | - Robert W. Sauerwein
- Center for Infectious Diseases, Department of Medical Microbiology, Radboud University Medical Center, Nijmegen 6500HB, The Netherlands
| | - Klaas W. Mulder
- Department of Molecular Developmental Biology, Faculty of Science, Radboud Institute for Molecular Life Sciences, Radboud University, Nijmegen 6525GA, The Netherlands
| | - Simon J. van Heeringen
- Department of Molecular Developmental Biology, Faculty of Science, Radboud Institute for Molecular Life Sciences, Radboud University, Nijmegen 6525GA, The Netherlands
| | - Monique M. A. Verstegen
- Department of Surgery, Erasmus University Medical Center Transplant Institute, University Medical Center Rotterdam,Rotterdam 3000CA, TheNetherlands
| | - Luc J. W. van der Laan
- Department of Surgery, Erasmus University Medical Center Transplant Institute, University Medical Center Rotterdam,Rotterdam 3000CA, TheNetherlands
| | - Hendrik Marks
- Department of Molecular Biology, Faculty of Science, Radboud Institute for Molecular Life Science, Radboud University, Nijmegen 6525GA, The Netherlands
| | - Richárd Bártfai
- Department of Molecular Biology, Faculty of Science, Radboud Institute for Molecular Life Science, Radboud University, Nijmegen 6525GA, The Netherlands
| |
Collapse
|
41
|
Matsuoka R, Kitajima K, Nii T, Zou Z, Tanaka K, Joo K, Ohkawa Y, Ohga S, Meno C. Hyperglycaemia induces diet-dependent defects of the left-right axis by lowering intracellular pH. Biochim Biophys Acta Mol Basis Dis 2025; 1871:167550. [PMID: 39442590 DOI: 10.1016/j.bbadis.2024.167550] [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: 08/06/2024] [Revised: 10/02/2024] [Accepted: 10/17/2024] [Indexed: 10/25/2024]
Abstract
Pregestational diabetes is a risk factor for congenital anomalies, including heterotaxy syndrome, a rare birth defect characterized by the abnormal arrangement of organs relative to the left-right (L-R) body axis. To provide insight into the underlying mechanism by which diabetes induces heterotaxy, we here analyzed the L-R axis of mouse embryos of diabetic dams. Various Pitx2 expression patterns indicative of disruption of L-R axis formation were apparent in such embryos. Expression of Nodal at the node, which triggers a Nodal-Pitx2 expression cascade in lateral plate mesoderm, showed marked regression associated with L-R axis malformation. This regression was similar to that apparent in Wnt3a-/- embryos, and canonical Wnt signalling was indeed found to be downregulated in embryos of diabetic dams. RNA sequencing revealed dysregulation of glycolysis in embryos of diabetic dams, and high glucose lowered intracellular pH in the primitive streak, leading to the suppression of Wnt signalling and the regression of Nodal expression. Of note, maternal vitamin A intake increased the incidence of L-R axis defects in embryos of diabetic dams, with dysregulation of retinoic acid metabolism being apparent in these embryos and in Wnt3a-/- embryos. Our results shed light on the mechanisms underlying embryopathies associated with maternal diabetes and suggest the importance of diet for prevention of heterotaxy.
Collapse
Affiliation(s)
- Ryohei Matsuoka
- Department of Developmental Biology, Graduate School of Medical Sciences, Kyushu University, Fukuoka 812-8582, Japan; Department of Pediatrics, Graduate School of Medical Sciences, Kyushu University, Fukuoka 812-8582, Japan
| | - Keiko Kitajima
- Department of Developmental Biology, Graduate School of Medical Sciences, Kyushu University, Fukuoka 812-8582, Japan
| | - Takenobu Nii
- Department of Developmental Biology, Graduate School of Medical Sciences, Kyushu University, Fukuoka 812-8582, Japan
| | - Zhaonan Zou
- Department of Developmental Biology, Graduate School of Medical Sciences, Kyushu University, Fukuoka 812-8582, Japan
| | - Kaori Tanaka
- Division of Transcriptomics, Medical Institute of Bioregulation, Kyushu University, Fukuoka 812-8582, Japan
| | - Kunihiko Joo
- Department of Cardiovascular Surgery, Kyushu University Hospital, Fukuoka 812-8582, Japan
| | - Yasuyuki Ohkawa
- Division of Transcriptomics, Medical Institute of Bioregulation, Kyushu University, Fukuoka 812-8582, Japan
| | - Shouichi Ohga
- Department of Pediatrics, Graduate School of Medical Sciences, Kyushu University, Fukuoka 812-8582, Japan
| | - Chikara Meno
- Department of Developmental Biology, Graduate School of Medical Sciences, Kyushu University, Fukuoka 812-8582, Japan.
| |
Collapse
|
42
|
Li W, He H, Wang H, Wen W. Dynamics of liver cancer cellular taxa revealed through single-cell RNA sequencing: Advances and challenges. Cancer Lett 2024; 611:217394. [PMID: 39689824 DOI: 10.1016/j.canlet.2024.217394] [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: 08/20/2024] [Revised: 10/13/2024] [Accepted: 12/14/2024] [Indexed: 12/19/2024]
Abstract
Liver cancer is a leading cause of death worldwide, representing a substantial public health challenge. The advent of single-cell RNA sequencing has significantly advanced our understanding of cellular dynamics from the onset of liver cancer to therapeutic intervention. This technology has unveiled profound insights into cancer heterogeneity and the tumor microenvironment (TME), enabling the identification of key molecular drivers and phenotypic landscapes of liver cancer at a single-cell resolution. This review highlights recent advancements in mapping functional cell subsets, phenotypic alterations, and the diversity of the TME. These insights are pivotal for advancing targeted therapies and developing prognostic tools. Moreover, this review covers the ongoing challenges and advances from tumor initiation to progression, offering a detailed perspective on advancing personalized treatment.
Collapse
Affiliation(s)
- Wenxin Li
- Third Affiliated Hospital of Naval Medical University (Second Military Medical University), National Center for Liver Cancer, Shanghai, 200438, China; Department of Clinical Laboratory Medicine, Third Affiliated Hospital of Naval Medical University (Second Military Medical University), Shanghai, 200438, China
| | - Huisi He
- Third Affiliated Hospital of Naval Medical University (Second Military Medical University), National Center for Liver Cancer, Shanghai, 200438, China; Department of Oncology, Third Affiliated Hospital of Naval Medical University (Second Military Medical University), Shanghai, 200438, China
| | - Hongyang Wang
- Third Affiliated Hospital of Naval Medical University (Second Military Medical University), National Center for Liver Cancer, Shanghai, 200438, China; The Ministry of Education Key Laboratory on Signaling Regulation and Targeting Therapy of Liver Cancer, Shanghai, 200438, China.
| | - Wen Wen
- Third Affiliated Hospital of Naval Medical University (Second Military Medical University), National Center for Liver Cancer, Shanghai, 200438, China; Department of Clinical Laboratory Medicine, Third Affiliated Hospital of Naval Medical University (Second Military Medical University), Shanghai, 200438, China; The Ministry of Education Key Laboratory on Signaling Regulation and Targeting Therapy of Liver Cancer, Shanghai, 200438, China.
| |
Collapse
|
43
|
De Jonghe J, Opzoomer JW, Vilas-Zornoza A, Nilges BS, Crane P, Vicari M, Lee H, Lara-Astiaso D, Gross T, Morf J, Schneider K, Cudini J, Ramos-Mucci L, Mooijman D, Tiklová K, Salas SM, Langseth CM, Kashikar ND, Schapiro D, Lundeberg J, Nilsson M, Shalek AK, Cribbs AP, Taylor-King JP. scTrends: A living review of commercial single-cell and spatial 'omic technologies. CELL GENOMICS 2024; 4:100723. [PMID: 39667347 PMCID: PMC11701258 DOI: 10.1016/j.xgen.2024.100723] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/05/2024] [Revised: 08/05/2024] [Accepted: 11/15/2024] [Indexed: 12/14/2024]
Abstract
Understanding the rapidly evolving landscape of single-cell and spatial omic technologies is crucial for advancing biomedical research and drug development. We provide a living review of both mature and emerging commercial platforms, highlighting key methodologies and trends shaping the field. This review spans from foundational single-cell technologies such as microfluidics and plate-based methods to newer approaches like combinatorial indexing; on the spatial side, we consider next-generation sequencing and imaging-based spatial transcriptomics. Finally, we highlight emerging methodologies that may fundamentally expand the scope for data generation within pharmaceutical research, creating opportunities to discover and validate novel drug mechanisms. Overall, this review serves as a critical resource for navigating the commercialization and application of single-cell and spatial omic technologies in pharmaceutical and academic research.
Collapse
Affiliation(s)
| | - James W Opzoomer
- Cell Communication Lab, Department of Oncology, University College London Cancer Institute, London, UK; Relation Therapeutics, London, UK
| | | | | | | | - Marco Vicari
- Science for Life Laboratory, Department of Gene Technology, KTH Royal Institute of Technology, Solna, Sweden
| | - Hower Lee
- spatialist AB, Stockholm, Sweden; Science for Life Laboratory, Department of Biochemistry and Biophysics, Stockholm University, 171 65 Solna, Sweden
| | - David Lara-Astiaso
- Department of Hematology, University of Cambridge, Cambridge, UK; Wellcome-MRC Cambridge Stem Cell Institute, Cambridge, UK
| | | | - Jörg Morf
- Skyhawk Therapeutics, Basel, Switzerland
| | | | | | | | | | - Katarína Tiklová
- Science for Life Laboratory, Department of Biochemistry and Biophysics, Stockholm University, 171 65 Solna, Sweden
| | - Sergio Marco Salas
- spatialist AB, Stockholm, Sweden; Science for Life Laboratory, Department of Biochemistry and Biophysics, Stockholm University, 171 65 Solna, Sweden
| | - Christoffer Mattsson Langseth
- spatialist AB, Stockholm, Sweden; Science for Life Laboratory, Department of Biochemistry and Biophysics, Stockholm University, 171 65 Solna, Sweden
| | | | - Denis Schapiro
- Institute for Computational Biomedicine, Heidelberg University, Faculty of Medicine, Heidelberg University Hospital, Heidelberg, Germany; Institute of Pathology, Heidelberg University Hospital, Heidelberg, Germany; Translational Spatial Profiling Center (TSPC), Heidelberg, Germany
| | - Joakim Lundeberg
- Science for Life Laboratory, Department of Gene Technology, KTH Royal Institute of Technology, Solna, Sweden
| | - Mats Nilsson
- Science for Life Laboratory, Department of Biochemistry and Biophysics, Stockholm University, 171 65 Solna, Sweden
| | - Alex K Shalek
- Relation Therapeutics, London, UK; Institute for Medical Engineering and Science, Department of Chemistry and Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA; Broad Institute of MIT and Harvard, Cambridge, MA, USA; Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA, USA
| | - Adam P Cribbs
- Caeruleus Genomics, Oxford, UK; Botnar Research Centre, Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, National Institute of Health Research Oxford Biomedical Research Unit (BRU), University of Oxford, Oxford, UK; Oxford Centre for Translational Myeloma Research University of Oxford, Oxford, UK.
| | | |
Collapse
|
44
|
Shi R, Chen H, Zhang W, Leak RK, Lou D, Chen K, Chen J. Single-cell RNA sequencing in stroke and traumatic brain injury: Current achievements, challenges, and future perspectives on transcriptomic profiling. J Cereb Blood Flow Metab 2024:271678X241305914. [PMID: 39648853 PMCID: PMC11626557 DOI: 10.1177/0271678x241305914] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/31/2024] [Revised: 10/19/2024] [Accepted: 11/06/2024] [Indexed: 12/10/2024]
Abstract
Single-cell RNA sequencing (scRNA-seq) is a high-throughput transcriptomic approach with the power to identify rare cells, discover new cellular subclusters, and describe novel genes. scRNA-seq can simultaneously reveal dynamic shifts in cellular phenotypes and heterogeneities in cellular subtypes. Since the publication of the first protocol on scRNA-seq in 2009, this evolving technology has continued to improve, through the use of cell-specific barcodes, adoption of droplet-based systems, and development of advanced computational methods. Despite induction of the cellular stress response during the tissue dissociation process, scRNA-seq remains a popular technology, and commercially available scRNA-seq methods have been applied to the brain. Recent advances in spatial transcriptomics now allow the researcher to capture the positional context of transcriptional activity, strengthening our knowledge of cellular organization and cell-cell interactions in spatially intact tissues. A combination of spatial transcriptomic data with proteomic, metabolomic, or chromatin accessibility data is a promising direction for future research. Herein, we provide an overview of the workflow, data analyses methods, and pros and cons of scRNA-seq technology. We also summarize the latest achievements of scRNA-seq in stroke and acute traumatic brain injury, and describe future applications of scRNA-seq and spatial transcriptomics.
Collapse
Affiliation(s)
- Ruyu Shi
- Department of Human Genetics, School of Public Health, University of Pittsburgh, USA
| | - Huaijun Chen
- Pittsburgh Institute of Brain Disorders & Recovery and Department of Neurology, University of Pittsburgh, Pittsburgh, PA, USA
- Geriatric Research, Education and Clinical Center, Veterans Affairs Pittsburgh Health Care System, Pittsburgh, PA, USA
| | - Wenting Zhang
- Pittsburgh Institute of Brain Disorders & Recovery and Department of Neurology, University of Pittsburgh, Pittsburgh, PA, USA
- Geriatric Research, Education and Clinical Center, Veterans Affairs Pittsburgh Health Care System, Pittsburgh, PA, USA
| | - Rehana K Leak
- Graduate School of Pharmaceutical Sciences, School of Pharmacy, Duquesne University, Pittsburgh, PA, USA
| | - Dequan Lou
- Department of Medicine, School of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Kong Chen
- Department of Medicine, School of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Jun Chen
- Pittsburgh Institute of Brain Disorders & Recovery and Department of Neurology, University of Pittsburgh, Pittsburgh, PA, USA
- Geriatric Research, Education and Clinical Center, Veterans Affairs Pittsburgh Health Care System, Pittsburgh, PA, USA
| |
Collapse
|
45
|
Mayeur H, Leyhr J, Mulley J, Leurs N, Michel L, Sharma K, Lagadec R, Aury JM, Osborne OG, Mulhair P, Poulain J, Mangenot S, Mead D, Smith M, Corton C, Oliver K, Skelton J, Betteridge E, Dolucan J, Dudchenko O, Omer AD, Weisz D, Aiden EL, McCarthy SA, Sims Y, Torrance J, Tracey A, Howe K, Baril T, Hayward A, Martinand-Mari C, Sanchez S, Haitina T, Martin K, Korsching SI, Mazan S, Debiais-Thibaud M. The Sensory Shark: High-quality Morphological, Genomic and Transcriptomic Data for the Small-spotted Catshark Scyliorhinus Canicula Reveal the Molecular Bases of Sensory Organ Evolution in Jawed Vertebrates. Mol Biol Evol 2024; 41:msae246. [PMID: 39657112 PMCID: PMC11979771 DOI: 10.1093/molbev/msae246] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2024] [Revised: 11/21/2024] [Accepted: 11/21/2024] [Indexed: 12/16/2024] Open
Abstract
Cartilaginous fishes (chondrichthyans: chimeras and elasmobranchs -sharks, skates, and rays) hold a key phylogenetic position to explore the origin and diversifications of jawed vertebrates. Here, we report and integrate reference genomic, transcriptomic, and morphological data in the small-spotted catshark Scyliorhinus canicula to shed light on the evolution of sensory organs. We first characterize general aspects of the catshark genome, confirming the high conservation of genome organization across cartilaginous fishes, and investigate population genomic signatures. Taking advantage of a dense sampling of transcriptomic data, we also identify gene signatures for all major organs, including chondrichthyan specializations, and evaluate expression diversifications between paralogs within major gene families involved in sensory functions. Finally, we combine these data with 3D synchrotron imaging and in situ gene expression analyses to explore chondrichthyan-specific traits and more general evolutionary trends of sensory systems. This approach brings to light, among others, novel markers of the ampullae of Lorenzini electrosensory cells, a duplication hotspot for crystallin genes conserved in jawed vertebrates, and a new metazoan clade of the transient-receptor potential (TRP) family. These resources and results, obtained in an experimentally tractable chondrichthyan model, open new avenues to integrate multiomics analyses for the study of elasmobranchs and jawed vertebrates.
Collapse
Affiliation(s)
- Hélène Mayeur
- Sorbonne Université, CNRS, Biologie Intégrative des Organismes Marins, BIOM, Banyuls-sur-mer, France
| | - Jake Leyhr
- Department of Organismal Biology, Uppsala University, Uppsala, Sweden
| | - John Mulley
- School of Environmental and Natural Sciences, Bangor University, Bangor, Gwynedd LL57 2UW, UK
| | - Nicolas Leurs
- Institut des Sciences de l'Evolution de Montpellier, ISEM, University of Montpellier, CNRS, IRD, EPHE, Montpellier, France
| | - Léo Michel
- Sorbonne Université, CNRS, Biologie Intégrative des Organismes Marins, BIOM, Banyuls-sur-mer, France
| | - Kanika Sharma
- Institute of Genetics, Faculty of Mathematics and Natural Sciences of the University at Cologne, Cologne 50674, Germany
| | - Ronan Lagadec
- Sorbonne Université, CNRS, Biologie Intégrative des Organismes Marins, BIOM, Banyuls-sur-mer, France
| | - Jean-Marc Aury
- Génomique Métabolique, Génoscope, Institut François Jacob, CEA, CNRS, Univ Evry, Université Paris-Saclay, Evry 91057, France
| | - Owen G Osborne
- School of Environmental and Natural Sciences, Bangor University, Bangor, Gwynedd LL57 2UW, UK
| | - Peter Mulhair
- Department of Biology, University of Oxford, Oxford OX1 3SZ, UK
| | - Julie Poulain
- Génomique Métabolique, Génoscope, Institut François Jacob, CEA, CNRS, Univ Evry, Université Paris-Saclay, Evry 91057, France
| | - Sophie Mangenot
- Génomique Métabolique, Génoscope, Institut François Jacob, CEA, CNRS, Univ Evry, Université Paris-Saclay, Evry 91057, France
| | - Daniel Mead
- Sequencing Department, Wellcome Sanger Institute, Cambridge CB10 1SA, UK
| | - Michelle Smith
- Sequencing Department, Wellcome Sanger Institute, Cambridge CB10 1SA, UK
| | - Craig Corton
- Sequencing Department, Wellcome Sanger Institute, Cambridge CB10 1SA, UK
| | - Karen Oliver
- Sequencing Department, Wellcome Sanger Institute, Cambridge CB10 1SA, UK
| | - Jason Skelton
- Sequencing Department, Wellcome Sanger Institute, Cambridge CB10 1SA, UK
| | - Emma Betteridge
- Sequencing Department, Wellcome Sanger Institute, Cambridge CB10 1SA, UK
| | - Jale Dolucan
- The Center for Genome Architecture and Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA
- The Center for Theoretical Biological Physics, Rice University, Houston, TX 77030, USA
| | - Olga Dudchenko
- The Center for Genome Architecture and Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA
- The Center for Theoretical Biological Physics, Rice University, Houston, TX 77030, USA
| | - Arina D Omer
- The Center for Genome Architecture and Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA
- The Center for Theoretical Biological Physics, Rice University, Houston, TX 77030, USA
| | - David Weisz
- The Center for Genome Architecture and Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA
- The Center for Theoretical Biological Physics, Rice University, Houston, TX 77030, USA
| | - Erez L Aiden
- The Center for Genome Architecture and Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA
- The Center for Theoretical Biological Physics, Rice University, Houston, TX 77030, USA
- Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Shane A McCarthy
- Sequencing Department, Wellcome Sanger Institute, Cambridge CB10 1SA, UK
| | - Ying Sims
- Sequencing Department, Wellcome Sanger Institute, Cambridge CB10 1SA, UK
| | - James Torrance
- Sequencing Department, Wellcome Sanger Institute, Cambridge CB10 1SA, UK
| | - Alan Tracey
- Sequencing Department, Wellcome Sanger Institute, Cambridge CB10 1SA, UK
| | - Kerstin Howe
- Sequencing Department, Wellcome Sanger Institute, Cambridge CB10 1SA, UK
| | - Tobias Baril
- Centre for Ecology and Conservation, University of Exeter, Cornwall TR10 9FE, UK
| | - Alexander Hayward
- Centre for Ecology and Conservation, University of Exeter, Cornwall TR10 9FE, UK
| | - Camille Martinand-Mari
- Institut des Sciences de l'Evolution de Montpellier, ISEM, University of Montpellier, CNRS, IRD, EPHE, Montpellier, France
| | - Sophie Sanchez
- Department of Organismal Biology, Uppsala University, Uppsala, Sweden
- European Synchrotron Radiation Facility, Grenoble, France
| | - Tatjana Haitina
- Department of Organismal Biology, Uppsala University, Uppsala, Sweden
| | - Kyle Martin
- Department of Earth Sciences, Natural History Museum, London SW7 5BD, UK
| | - Sigrun I Korsching
- Institute of Genetics, Faculty of Mathematics and Natural Sciences of the University at Cologne, Cologne 50674, Germany
| | - Sylvie Mazan
- Sorbonne Université, CNRS, Biologie Intégrative des Organismes Marins, BIOM, Banyuls-sur-mer, France
| | - Mélanie Debiais-Thibaud
- Institut des Sciences de l'Evolution de Montpellier, ISEM, University of Montpellier, CNRS, IRD, EPHE, Montpellier, France
| |
Collapse
|
46
|
Neugebauer J, Raulien N, Arndt L, Akkermann D, Hobusch C, Lindhorst A, Fröba J, Gericke M. The Impact of Resident Adipose Tissue Macrophages on Adipocyte Homeostasis and Dedifferentiation. Int J Mol Sci 2024; 25:13019. [PMID: 39684730 DOI: 10.3390/ijms252313019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2024] [Revised: 11/29/2024] [Accepted: 11/29/2024] [Indexed: 12/18/2024] Open
Abstract
Obesity is concurrent with immunological dysregulation, resulting in chronic low-grade inflammation and cellular dysfunction. In pancreatic islets, this loss of function has been correlated with mature β-cells dedifferentiating into a precursor-like state through constant exposure to inflammatory stressors. As mature adipocytes likewise have the capability to dedifferentiate in vitro and in vivo, we wanted to analyze this cellular change in relation to adipose tissue (AT) inflammation and adipose tissue macrophage (ATM) activity. Using our organotypic AT explant culture method combined with a double-reporter mouse model for labeling ATMs and mature adipocytes, we were able to visualize and quantify dedifferentiated fat (DFAT) cells in AT explants. Preliminary testing showed increased dedifferentiation after tamoxifen (TAM) stimulation, making TAM-dependent lineage-tracing models unsuitable for quantification of naturally occurring DFAT cells. The regulatory role of ATMs in adipocyte dedifferentiation was shown through macrophage depletion using Plexxicon 5622 or clodronate liposomes, which significantly increased DFAT cell levels. Subsequent bulk RNA sequencing of macrophage-depleted explants revealed enrichment of the tumor necrosis factor α (TNFα) signaling pathway as well as downregulation of associated genes. Direct stimulation with TNFα decreased adipocyte dedifferentiation, while application of a TNFα-neutralizing antibody did not significantly alter DFAT cell levels. Our findings suggest a regulatory role of resident ATMs in maintaining the mature adipocyte phenotype and preventing excessive adipocyte dedifferentiation. The specific regulatory pathways as well as the impact that DFAT cells might have on ATMs, and vice versa, are subject to further investigation.
Collapse
Affiliation(s)
- Julia Neugebauer
- Institute of Anatomy, Leipzig University, 04103 Leipzig, Germany
| | - Nora Raulien
- Institute of Anatomy, Leipzig University, 04103 Leipzig, Germany
| | - Lilli Arndt
- Institute of Anatomy, Leipzig University, 04103 Leipzig, Germany
| | - Dagmar Akkermann
- Paul-Flechsig-Institute, Leipzig University, 04103 Leipzig, Germany
| | | | | | - Janine Fröba
- Institute of Anatomy, Leipzig University, 04103 Leipzig, Germany
| | - Martin Gericke
- Institute of Anatomy, Leipzig University, 04103 Leipzig, Germany
| |
Collapse
|
47
|
Xu P, Yuan Z, Lu X, Zhou P, Qiu D, Qiao Z, Zhou Z, Guan L, Jia Y, He X, Sun L, Wan Y, Wang M, Yu Y. RAG-seq: NSR-primed and Transposase Tagmentation-mediated Strand-specific Total RNA Sequencing in Single Cells. GENOMICS, PROTEOMICS & BIOINFORMATICS 2024; 22:qzae072. [PMID: 39388199 DOI: 10.1093/gpbjnl/qzae072] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/27/2024] [Revised: 09/24/2024] [Accepted: 09/30/2024] [Indexed: 10/15/2024]
Abstract
Single-cell RNA sequencing (scRNA-seq) has transformed our understanding of cellular diversity with unprecedented resolution. However, many current methods are limited in capturing full-length transcripts and discerning strand orientation. Here, we present RAG-seq, an innovative strand-specific total RNA sequencing technique that combines not-so-random (NSR) primers with Tn5 transposase-mediated tagmentation. RAG-seq overcomes previous limitations by delivering comprehensive transcript coverage and maintaining strand orientation, which are essential for accurate quantification of overlapping genes and detection of antisense transcripts. Through optimized reverse transcription with oligo-dT primers, rRNA depletion via Depletion of Abundant Sequences by Hybridization (DASH), and linear amplification, RAG-seq enhances sensitivity and reproducibility, especially for low-input samples and single cells. Application to mouse oocytes and early embryos highlights RAG-seq's superior performance in identifying stage-specific antisense transcripts, shedding light on their regulatory roles during early development. This advancement represents a significant leap in transcriptome analysis within complex biological contexts.
Collapse
Affiliation(s)
- Ping Xu
- China-Japan Union Hospital of Jilin University, Jilin University, Changchun 130033, China
- School of Life Sciences, Jilin University, Changchun 130012, China
- Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou 510623, China
| | - Zhiheng Yuan
- Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou 510623, China
| | - Xiaohua Lu
- Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China
| | - Peng Zhou
- Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou 510623, China
| | - Ding Qiu
- Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou 510623, China
| | - Zhenghao Qiao
- Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou 510623, China
| | - Zhongcheng Zhou
- Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou 510623, China
| | - Li Guan
- Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou 510623, China
| | - Yongkang Jia
- Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou 510623, China
| | - Xuan He
- Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou 510623, China
| | - Ling Sun
- Center for Reproductive Medicine, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou 510623, China
| | - Youzhong Wan
- China-Japan Union Hospital of Jilin University, Jilin University, Changchun 130033, China
| | - Ming Wang
- Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou 510623, China
| | - Yang Yu
- Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou 510623, China
- Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China
| |
Collapse
|
48
|
Drougard A, Ma EH, Wegert V, Sheldon R, Panzeri I, Vatsa N, Apostle S, Fagnocchi L, Schaf J, Gossens K, Völker J, Pang S, Bremser A, Dror E, Giacona F, Sagar S, Henderson MX, Prinz M, Jones RG, Pospisilik JA. An acute microglial metabolic response controls metabolism and improves memory. eLife 2024; 12:RP87120. [PMID: 39625057 PMCID: PMC11614388 DOI: 10.7554/elife.87120] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/06/2024] Open
Abstract
Chronic high-fat feeding triggers metabolic dysfunction including obesity, insulin resistance, and diabetes. How high-fat intake first triggers these pathophysiological states remains unknown. Here, we identify an acute microglial metabolic response that rapidly translates intake of high-fat diet (HFD) to a surprisingly beneficial effect on metabolism and spatial/learning memory. High-fat intake rapidly increases palmitate levels in cerebrospinal fluid and triggers a wave of microglial metabolic activation characterized by mitochondrial membrane activation and fission as well as metabolic skewing toward aerobic glycolysis. These effects are detectable throughout the brain and can be detected within as little as 12 hr of HFD exposure. In vivo, microglial ablation and conditional DRP1 deletion show that the microglial metabolic response is necessary for the acute effects of HFD. 13C-tracing experiments reveal that in addition to processing via β-oxidation, microglia shunt a substantial fraction of palmitate toward anaplerosis and re-release of bioenergetic carbons into the extracellular milieu in the form of lactate, glutamate, succinate, and intriguingly, the neuroprotective metabolite itaconate. Together, these data identify microglia as a critical nutrient regulatory node in the brain, metabolizing away harmful fatty acids and liberating the same carbons as alternate bioenergetic and protective substrates for surrounding cells. The data identify a surprisingly beneficial effect of short-term HFD on learning and memory.
Collapse
Affiliation(s)
- Anne Drougard
- Department of Epigenetics, Van Andel Research InstituteGrand RapidsUnited States
- Max Planck Institute of Immunobiology and EpigeneticsFreiburgGermany
| | - Eric H Ma
- Department of Metabolism and Nutritional Programming, Van Andel Research InstituteGrand RapidsUnited States
| | - Vanessa Wegert
- Department of Epigenetics, Van Andel Research InstituteGrand RapidsUnited States
- Max Planck Institute of Immunobiology and EpigeneticsFreiburgGermany
| | - Ryan Sheldon
- Metabolomics and Bioenergetics Core, Van Andel InstituteGrand RapidsUnited States
| | - Ilaria Panzeri
- Department of Epigenetics, Van Andel Research InstituteGrand RapidsUnited States
- Max Planck Institute of Immunobiology and EpigeneticsFreiburgGermany
| | - Naman Vatsa
- Department of Neurodegenerative Sciences, Van Andel Research InstituteGrand RapidsUnited States
| | - Stefanos Apostle
- Department of Epigenetics, Van Andel Research InstituteGrand RapidsUnited States
| | - Luca Fagnocchi
- Department of Epigenetics, Van Andel Research InstituteGrand RapidsUnited States
| | - Judith Schaf
- Max Planck Institute of Immunobiology and EpigeneticsFreiburgGermany
| | - Klaus Gossens
- Max Planck Institute of Immunobiology and EpigeneticsFreiburgGermany
| | - Josephine Völker
- Max Planck Institute of Immunobiology and EpigeneticsFreiburgGermany
| | - Shengru Pang
- Institute of Neuropathology, Medical Faculty, University of FreiburgFreiburgGermany
| | - Anna Bremser
- Max Planck Institute of Immunobiology and EpigeneticsFreiburgGermany
| | - Erez Dror
- Max Planck Institute of Immunobiology and EpigeneticsFreiburgGermany
| | - Francesca Giacona
- Department of Epigenetics, Van Andel Research InstituteGrand RapidsUnited States
| | - Sagar Sagar
- Max Planck Institute of Immunobiology and EpigeneticsFreiburgGermany
- Department of Medicine II, University Hospital FreiburgFreiburgGermany
| | - Michael X Henderson
- Department of Neurodegenerative Sciences, Van Andel Research InstituteGrand RapidsUnited States
| | - Marco Prinz
- Institute of Neuropathology, Medical Faculty, University of FreiburgFreiburgGermany
- Centre for NeuroModulation (NeuroModBasics), University of FreiburgFreiburgGermany
- Signaling Research Centers BIOSS and CIBSS, University of FreiburgFreiburgGermany
| | - Russell G Jones
- Department of Metabolism and Nutritional Programming, Van Andel Research InstituteGrand RapidsUnited States
| | - John Andrew Pospisilik
- Department of Epigenetics, Van Andel Research InstituteGrand RapidsUnited States
- Max Planck Institute of Immunobiology and EpigeneticsFreiburgGermany
| |
Collapse
|
49
|
Tian C, Zhang Y, Tong Y, Kock KH, Sim DY, Liu F, Dong J, Jing Z, Wang W, Gao J, Tan LM, Han KY, Tomofuji Y, Nakano M, Buyamin EV, Sonthalia R, Ando Y, Hatano H, Sonehara K, Jin X, Loh M, Chambers J, Hon CC, Choi M, Park JE, Ishigaki K, Okamura T, Fujio K, Okada Y, Park WY, Shin JW, Roca X, Prabhakar S, Liu B. Single-cell RNA sequencing of peripheral blood links cell-type-specific regulation of splicing to autoimmune and inflammatory diseases. Nat Genet 2024; 56:2739-2752. [PMID: 39627432 PMCID: PMC11631754 DOI: 10.1038/s41588-024-02019-8] [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: 12/07/2023] [Accepted: 10/30/2024] [Indexed: 12/12/2024]
Abstract
Alternative splicing contributes to complex traits, but whether this differs in trait-relevant cell types across diverse genetic ancestries is unclear. Here we describe cell-type-specific, sex-biased and ancestry-biased alternative splicing in ~1 M peripheral blood mononuclear cells from 474 healthy donors from the Asian Immune Diversity Atlas. We identify widespread sex-biased and ancestry-biased differential splicing, most of which is cell-type-specific. We identify 11,577 independent cis-splicing quantitative trait loci (sQTLs), 607 trans-sGenes and 107 dynamic sQTLs. Colocalization between cis-eQTLs and trans-sQTLs revealed a cell-type-specific regulatory relationship between HNRNPLL and PTPRC. We observed an enrichment of cis-sQTL effects in autoimmune and inflammatory disease heritability. Specifically, we functionally validated an Asian-specific sQTL disrupting the 5' splice site of TCHP exon 4 that putatively modulates the risk of Graves' disease in East Asian populations. Our work highlights the impact of ancestral diversity on splicing and provides a roadmap to dissect its role in complex diseases at single-cell resolution.
Collapse
Affiliation(s)
- Chi Tian
- Department of Pharmacy and Pharmaceutical Sciences, Faculty of Science, National University of Singapore, Singapore, Singapore
| | - Yuntian Zhang
- Department of Biomedical Informatics, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Yihan Tong
- Department of Pharmacy and Pharmaceutical Sciences, Faculty of Science, National University of Singapore, Singapore, Singapore
| | - Kian Hong Kock
- Genome Institute of Singapore (GIS), Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
| | - Donald Yuhui Sim
- School of Biological Sciences, Nanyang Technological University, Singapore, Singapore
| | - Fei Liu
- Department of Pharmacy and Pharmaceutical Sciences, Faculty of Science, National University of Singapore, Singapore, Singapore
| | - Jiaqi Dong
- Department of Biomedical Informatics, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Zhixuan Jing
- Department of Biomedical Informatics, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Wenjing Wang
- Department of Pharmacy and Pharmaceutical Sciences, Faculty of Science, National University of Singapore, Singapore, Singapore
- Department of Biomedical Informatics, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Junbin Gao
- Department of Pharmacy and Pharmaceutical Sciences, Faculty of Science, National University of Singapore, Singapore, Singapore
| | - Le Min Tan
- Genome Institute of Singapore (GIS), Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
| | - Kyung Yeon Han
- Samsung Genome Institute, Samsung Medical Center, Seoul, South Korea
| | - Yoshihiko Tomofuji
- Laboratory for Systems Genetics, RIKEN Center for Integrative Medical Sciences, Yokohama City, Japan
- Department of Statistical Genetics, Osaka University Graduate School of Medicine, Suita, Japan
- Department of Genome Informatics, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Masahiro Nakano
- Laboratory for Autoimmune Diseases, RIKEN Center for Integrative Medical Sciences, Yokohama City, Japan
- Laboratory for Human Immunogenetics, RIKEN Center for Integrative Medical Sciences, Yokohama City, Japan
| | - Eliora Violain Buyamin
- Genome Institute of Singapore (GIS), Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
| | - Radhika Sonthalia
- Genome Institute of Singapore (GIS), Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
| | - Yoshinari Ando
- Laboratory for Advanced Genomics Circuit, RIKEN Center for Integrative Medical Sciences, Yokohama City, Japan
- Laboratory for Transcriptome Technology, RIKEN Center for Integrative Medical Sciences, Yokohama City, Japan
| | - Hiroaki Hatano
- Laboratory for Human Immunogenetics, RIKEN Center for Integrative Medical Sciences, Yokohama City, Japan
| | - Kyuto Sonehara
- Laboratory for Systems Genetics, RIKEN Center for Integrative Medical Sciences, Yokohama City, Japan
- Department of Statistical Genetics, Osaka University Graduate School of Medicine, Suita, Japan
- Department of Genome Informatics, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Xin Jin
- BGI Research, Shenzhen, China
- The Innovation Centre of Ministry of Education for Development and Diseases, School of Medicine, South China University of Technology, Guangzhou, China
- Shanxi Medical University-BGI Collaborative Center for Future Medicine, Shanxi Medical University, Taiyuan, China
- Shenzhen Key Laboratory of Transomics Biotechnologies, BGI Research, Shenzhen, China
| | - Marie Loh
- Genome Institute of Singapore (GIS), Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, Singapore
- Department of Epidemiology and Biostatistics, School of Public Health, Imperial College London, London, UK
| | - John Chambers
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, Singapore
| | - Chung-Chau Hon
- Laboratory for Genome Information Analysis, RIKEN Center for Integrative Medical Sciences, Graduate School of Integrated Sciences for Life, Hiroshima University, Higashi-Hiroshima, Japan
| | - Murim Choi
- Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul, South Korea
| | - Jong-Eun Park
- Graduate School of Medical Science and Engineering, KAIST, Daejeon, South Korea
| | - Kazuyoshi Ishigaki
- Laboratory for Human Immunogenetics, RIKEN Center for Integrative Medical Sciences, Yokohama City, Japan
| | - Tomohisa Okamura
- Department of Allergy and Rheumatology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Keishi Fujio
- Department of Allergy and Rheumatology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Yukinori Okada
- Laboratory for Systems Genetics, RIKEN Center for Integrative Medical Sciences, Yokohama City, Japan
- Department of Statistical Genetics, Osaka University Graduate School of Medicine, Suita, Japan
- Department of Genome Informatics, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
- Laboratory of Statistical Immunology, Immunology Frontier Research Center (WPI-IFReC), Osaka University, Suita, Japan
- Premium Research Institute for Human Metaverse Medicine (WPI-PRIMe), Osaka University, Suita, Japan
| | - Woong-Yang Park
- Samsung Genome Institute, Samsung Medical Center, Seoul, South Korea
| | - Jay W Shin
- Genome Institute of Singapore (GIS), Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
- Laboratory for Advanced Genomics Circuit, RIKEN Center for Integrative Medical Sciences, Yokohama City, Japan
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Xavier Roca
- School of Biological Sciences, Nanyang Technological University, Singapore, Singapore
| | - Shyam Prabhakar
- Genome Institute of Singapore (GIS), Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, Singapore
| | - Boxiang Liu
- Department of Pharmacy and Pharmaceutical Sciences, Faculty of Science, National University of Singapore, Singapore, Singapore.
- Department of Biomedical Informatics, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore.
- Genome Institute of Singapore (GIS), Agency for Science, Technology and Research (A*STAR), Singapore, Singapore.
- Precision Medicine Translational Research Programme, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore.
- NUS Centre for Cancer Research, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore.
- Cardiovascular-Metabolic Disease Translational Research Programme, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore.
| |
Collapse
|
50
|
Yanginlar C, Rother N, Post TGJM, Jacobs M, Jonkman I, Brouns M, Rinzema S, Martens JHA, Vermeulen M, Joosten LAB, Netea MG, Hilbrands LB, Choudhry ZA, van der Vlag J, Duivenvoorden R. Trained innate immunity in response to nuclear antigens in systemic lupus erythematosus. J Autoimmun 2024; 149:103335. [PMID: 39549487 DOI: 10.1016/j.jaut.2024.103335] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2023] [Revised: 10/10/2024] [Accepted: 11/03/2024] [Indexed: 11/18/2024]
Abstract
Systemic lupus erythematosus (SLE) is an autoimmune disease directed against nuclear antigens, including those derived from apoptotic microparticles (MPs) and neutrophil extracellular traps (NETs). Here we investigated whether nuclear autoantigens can induce trained immunity in SLE patients. Trained immunity is a de facto innate immune memory elicited by an initial stimulus that induces a more vigorous long-term inflammatory response to subsequent stimuli. Isolated monocytes were stimulated with SLE-typical nuclear antigens, neutrophil extracellular traps (NETs), and apoptotic microparticles (MPs) or plasma from SLE patients. After five days of rest, cells were restimulated with Toll-like receptor (TLR) agonists, and cytokine production was measured using ELISA. Functional, transcriptomic and epigenetic changes in monocytes from SLE patients were evaluated by ex vivo stimulations, flow cytometric analysis, RNA sequencing, and chromatin immunoprecipitation (ChIP) sequencing for histone 3 lysine 4 trimethylation. We found that in vitro, both MPs and NETs, as well as plasma from SLE patients, can induce trained immunity. Furthermore, circulating monocytes from SLE patients produce increased levels of pro-inflammatory cytokines after stimulation with TLR ligands, indicating trained immunity. This is accompanied by deregulation in histone 3 lysine 4 trimethylation and increased expression of metabolism and inflammation-related genes. Our findings demonstrate that trained immunity can develop against nuclear antigens and that trained immunity is involved in the immunological dysregulation in SLE patients.
Collapse
Affiliation(s)
- Cansu Yanginlar
- Department of Nephrology, Radboud Research Institute for Medical Innovation, Radboud university medical center, Nijmegen, the Netherlands
| | - Nils Rother
- Department of Nephrology, Radboud Research Institute for Medical Innovation, Radboud university medical center, Nijmegen, the Netherlands
| | - Tomas G J M Post
- Department of Nephrology, Radboud Research Institute for Medical Innovation, Radboud university medical center, Nijmegen, the Netherlands
| | - Maaike Jacobs
- Department of Nephrology, Radboud Research Institute for Medical Innovation, Radboud university medical center, Nijmegen, the Netherlands
| | - Inge Jonkman
- Department of Nephrology, Radboud Research Institute for Medical Innovation, Radboud university medical center, Nijmegen, the Netherlands
| | - Montsy Brouns
- Department of Internal Medicine, Dr. Horacio Oduber Hospital, Oranjestad, Aruba
| | - Sybren Rinzema
- Department of Molecular Biology, Faculty of Science, Oncode Institute, Radboud University Nijmegen, Nijmegen, the Netherlands
| | - Joost H A Martens
- Department of Molecular Biology, Faculty of Science, Oncode Institute, Radboud University Nijmegen, Nijmegen, the Netherlands
| | - Michiel Vermeulen
- Department of Molecular Biology, Faculty of Science, Oncode Institute, Radboud University Nijmegen, Nijmegen, the Netherlands
| | - Leo A B Joosten
- Department of Internal Medicine, Radboud Research Institute for Medical Innovation, Radboud university medical center, Nijmegen, the Netherlands; Department of Medical Genetics, University of Medicine and Pharmacy, Iuliu Haţieganu, Cluj-Napoca, Romania
| | - Mihai G Netea
- Department of Internal Medicine, Radboud Research Institute for Medical Innovation, Radboud university medical center, Nijmegen, the Netherlands; Department of Immunology and Metabolism, Life and Medical Sciences Institute, University of Bonn, Bonn, Germany
| | - Luuk B Hilbrands
- Department of Nephrology, Radboud Research Institute for Medical Innovation, Radboud university medical center, Nijmegen, the Netherlands
| | - Zaheeb A Choudhry
- Department of Internal Medicine, Dr. Horacio Oduber Hospital, Oranjestad, Aruba
| | - Johan van der Vlag
- Department of Nephrology, Radboud Research Institute for Medical Innovation, Radboud university medical center, Nijmegen, the Netherlands
| | - Raphaël Duivenvoorden
- Department of Nephrology, Radboud Research Institute for Medical Innovation, Radboud university medical center, Nijmegen, the Netherlands; Biomolecular Engineering and Imaging Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
| |
Collapse
|