1
|
Liu J, Xu L, Lu J, Shen X, Li D, Bai L, Li X, Yu Z, Li H. Roles of Adam8 in Neuroinflammation in experimental ischemic Stroke: Insights from single-cell and ribosome-bound mRNA sequencing. Exp Neurol 2025; 388:115207. [PMID: 40064361 DOI: 10.1016/j.expneurol.2025.115207] [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: 12/28/2024] [Revised: 02/27/2025] [Accepted: 03/05/2025] [Indexed: 03/21/2025]
Abstract
Stroke remains a leading cause of global mortality, with neuroinflammation significantly exacerbating clinical outcomes. Microglia serve as key mediators of post-stroke neuroinflammation, though the mechanisms driving their migration to injury sites remain poorly understood. In this study, using publicly available single-cell sequencing data (GSE234052), we identified a migration-associated microglial subtype in a murine model of distal middle cerebral artery occlusion (dMCAO). Additionally, ribosome-bound mRNA sequencing data (GSE225110) from microglia isolated from peri-infarct cortical tissue uncovered dMCAO-induced alterations in microglial mRNA translation. By integrating these datasets, we identified A Disintegrin And Metalloproteinase 8 (Adam8) as a key gene upregulated at both the transcriptional and translational levels post-dMCAO. Protein analysis revealed that both the precursor and active forms of Adam8 were predominantly expressed in microglia and significantly upregulated in peri-infarct regions following dMCAO. Notably, Adam8 inhibition with BK-1361 significantly reduced Adam8 cleavage, M1 microglial migration, inflammation, infarct size, and improved neurological outcomes. Bioinformatics analysis further identified Myo1e as a potential interacting partner of Adam8, a finding validated through immunofluorescence co-localization. These findings highlight Adam8 as a promising therapeutic target for mitigating post-stroke neuroinflammation and offer new insights into the mechanisms of microglial migration.
Collapse
Affiliation(s)
- Jiale Liu
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, Soochow University, Suzhou 215006, China; Institute of Stroke Research, Soochow University, Suzhou 215006, China
| | - Li Xu
- Intensive Care Unit of the Department of Anesthesiology, The First Affiliated Hospital of Soochow University, Soochow University, Suzhou 215006, China
| | - Jinxin Lu
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, Soochow University, Suzhou 215006, China; Institute of Stroke Research, Soochow University, Suzhou 215006, China
| | - Xi Shen
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, Soochow University, Suzhou 215006, China; Institute of Stroke Research, Soochow University, Suzhou 215006, China
| | - Di Li
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, Soochow University, Suzhou 215006, China; Institute of Stroke Research, Soochow University, Suzhou 215006, China
| | - Lei Bai
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, Soochow University, Suzhou 215006, China; Institute of Stroke Research, Soochow University, Suzhou 215006, China
| | - Xiang Li
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, Soochow University, Suzhou 215006, China; Institute of Stroke Research, Soochow University, Suzhou 215006, China.
| | - Zhengquan Yu
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, Soochow University, Suzhou 215006, China; Institute of Stroke Research, Soochow University, Suzhou 215006, China.
| | - Haiying Li
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, Soochow University, Suzhou 215006, China; Institute of Stroke Research, Soochow University, Suzhou 215006, China.
| |
Collapse
|
2
|
Sankowski R, Prinz M. A dynamic and multimodal framework to define microglial states. Nat Neurosci 2025:10.1038/s41593-025-01978-3. [PMID: 40394327 DOI: 10.1038/s41593-025-01978-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2024] [Accepted: 04/22/2025] [Indexed: 05/22/2025]
Abstract
The widespread use of single-cell RNA sequencing has generated numerous purportedly distinct and novel subsets of microglia. Here, we challenge this fragmented paradigm by proposing that microglia exist along a continuum rather than as discrete entities. We identify a methodological over-reliance on computational clustering algorithms as the fundamental issue, with arbitrary cluster numbers being interpreted as biological reality. Evidence suggests that the observed transcriptional diversity stems from a combination of microglial plasticity and technical noise, resulting in terminology describing largely overlapping cellular states. We introduce a continuous model of microglial states, where cell positioning along the continuum is determined by biological aging and cell-specific molecular contexts. The model accommodates the dynamic nature of microglia. We advocate for a parsimonious approach toward classification and terminology that acknowledges the continuous spectrum of microglial states, toward a robust framework for understanding these essential immune cells of the CNS.
Collapse
Affiliation(s)
- Roman Sankowski
- Institute of Neuropathology, Faculty of Medicine, University of Freiburg, Freiburg, Germany.
| | - Marco Prinz
- Institute of Neuropathology, Faculty of Medicine, University of Freiburg, Freiburg, Germany.
- Signalling Research Centres BIOSS and CIBSS, University of Freiburg, Freiburg, Germany.
| |
Collapse
|
3
|
Luo X, Deng H, Li Q, Zhao M, Zhang Y, Guo J, Wen Y, Chen G, Li J. Bulk transcriptome and single-nucleus RNA sequencing analyses highlight the role of recombination activating 1 in non-alcoholic fatty liver disease. Int J Biol Macromol 2025; 307:141919. [PMID: 40074128 DOI: 10.1016/j.ijbiomac.2025.141919] [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: 10/13/2024] [Revised: 03/07/2025] [Accepted: 03/08/2025] [Indexed: 03/14/2025]
Abstract
Non-alcoholic fatty liver disease (NAFLD) is a prevalent chronic condition with an incompletely understood pathogenesis. In this study, five candidate genes-RAG1, CKAP2, CENPK, TYMS, and BUB1-were identified as being associated with NAFLD progression through integrative bioinformatics analyses. A predictive model incorporating these genes demonstrated strong robustness and diagnostic accuracy. Single-nucleus RNA sequencing analysis further revealed that RAG1 plays a potential role in hepatocytes of NAFLD patients. Functional experiments using RNA interference to suppress RAG1 expression in HepG2 cells treated with oleic and palmitic acids showed reduced total glyceride and cholesterol levels, mitigated lipid accumulation, and alterations in pathways related to lipid metabolism, inflammation, and fibrosis. Furthermore, adeno-associated virus-specific knockdown of RAG1 in hepatocytes attenuated hepatic steatosis in high-fat diet-fed mice. These findings suggest that investigating the molecular mechanisms of hub genes like RAG1 may advance our understanding of NAFLD pathogenesis and inform therapeutic development.
Collapse
Affiliation(s)
- Xiaohua Luo
- Department of Liver Transplant, The Second Xiangya Hospital of Central South University, 410011 Changsha, China
| | - Hongbo Deng
- Department of Liver Transplant, The Second Xiangya Hospital of Central South University, 410011 Changsha, China
| | - Qiang Li
- Department of Liver Transplant, The Second Xiangya Hospital of Central South University, 410011 Changsha, China
| | - Miao Zhao
- Center for Medical Genetics and Hunan Key Laboratory of Medical Genetics, School of Life Science, Central South University, 410078 Changsha, China
| | - Yu Zhang
- Department of Liver Transplant, The Second Xiangya Hospital of Central South University, 410011 Changsha, China
| | - Junjie Guo
- Department of Liver Transplant, The Second Xiangya Hospital of Central South University, 410011 Changsha, China
| | - Yifan Wen
- Department of Liver Transplant, The Second Xiangya Hospital of Central South University, 410011 Changsha, China
| | - Guangshun Chen
- Department of Liver Transplant, The Second Xiangya Hospital of Central South University, 410011 Changsha, China.
| | - Jiequn Li
- Department of Liver Transplant, The Second Xiangya Hospital of Central South University, 410011 Changsha, China.
| |
Collapse
|
4
|
Guo B, Ling W, Kwon SH, Panwar P, Ghazanfar S, Martinowich K, Hicks SC. Integrating Spatially-Resolved Transcriptomics Data Across Tissues and Individuals: Challenges and Opportunities. SMALL METHODS 2025; 9:e2401194. [PMID: 39935130 DOI: 10.1002/smtd.202401194] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/01/2024] [Revised: 12/13/2024] [Indexed: 02/13/2025]
Abstract
Advances in spatially-resolved transcriptomics (SRT) technologies have propelled the development of new computational analysis methods to unlock biological insights. The lowering cost of SRT data generation presents an unprecedented opportunity to create large-scale spatial atlases and enable population-level investigation, integrating SRT data across multiple tissues, individuals, species, or phenotypes. Here, unique challenges are described in the SRT data integration, where the analytic impact of varying spatial and biological resolutions is characterized and explored. A succinct review of spatially-aware integration methods and computational strategies is provided. Exciting opportunities to advance computational algorithms amenable to atlas-scale datasets along with standardized preprocessing methods, leading to improved sensitivity and reproducibility in the future are further highlighted.
Collapse
Affiliation(s)
- Boyi Guo
- Department of Biostatistics, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, 21205, USA
| | - Wodan Ling
- Division of Biostatistics, Department of Population Health Sciences, Weill Cornell Medicine, New York, NY, 10065, USA
| | - Sang Ho Kwon
- Lieber Institute for Brain Development, Johns Hopkins Medical Campus, Baltimore, MD, 21205, USA
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins School of Medicine, Baltimore, MD, 21205, USA
- Biochemistry, Cellular, and Molecular Biology Graduate Program, Johns Hopkins School of Medicine, Baltimore, MD, 21205, USA
| | - Pratibha Panwar
- School of Mathematics and Statistics, The University of Sydney, Camperdown, NSW, 2006, Australia
- Sydney Precision Data Science Centre, University of Sydney, Camperdown, NSW, 2006, Australia
- Charles Perkins Centre, The University of Sydney, Camperdown, NSW, 2006, Australia
| | - Shila Ghazanfar
- School of Mathematics and Statistics, The University of Sydney, Camperdown, NSW, 2006, Australia
- Sydney Precision Data Science Centre, University of Sydney, Camperdown, NSW, 2006, Australia
- Charles Perkins Centre, The University of Sydney, Camperdown, NSW, 2006, Australia
| | - Keri Martinowich
- Lieber Institute for Brain Development, Johns Hopkins Medical Campus, Baltimore, MD, 21205, USA
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins School of Medicine, Baltimore, MD, 21205, USA
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins School of Medicine, Baltimore, MD, USA
- Johns Hopkins Kavli Neuroscience Discovery Institute, Johns Hopkins University, Baltimore, MD, 21218, USA
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD, 21218, USA
| | - Stephanie C Hicks
- Department of Biostatistics, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, 21205, USA
- Center for Computational Biology, Johns Hopkins University, Baltimore, MD, 21218, USA
- Malone Center for Engineering in Healthcare, Johns Hopkins University, Baltimore, MD, 21218, USA
| |
Collapse
|
5
|
Gui J, Fang M, Tu J, Chen X, Sun L. Exploring genetic and immune cell dynamics in systemic lupus erythematosus patients with Epstein-Barr virus infection via machine learning. Rheumatology (Oxford) 2025; 64:2656-2664. [PMID: 39361430 DOI: 10.1093/rheumatology/keae537] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2024] [Accepted: 08/30/2024] [Indexed: 10/05/2024] Open
Abstract
OBJECTIVES EBV is a widespread virus implicated in various diseases, including SLE. However, the specific genes and pathways altered in SLE patients with EBV infection remain unclear. We aimed to identify key genes and immune cells in SLE patients with EBV infection. METHODS The datasets of SLE (GSE50772 and GSE81622) and EBV infection (GSE85599 and GSE45918) were obtained from the Gene Expression Omnibus (GEO) database. Next, differential gene expression (DEGs) analyses were conducted to identify overlapping DEGs, and then enrichment analysis was performed. Machine learning was applied to identify key genes. Validation was conducted using receiver operating characteristic (ROC) curve analysis and expression level verification in test datasets and single-cell RNA sequencing. Immune cell infiltration patterns were analysed using CIBERSORTx, and clinical data were reviewed for SLE patients. RESULTS We identified 58 overlapping DEGs enriched in IFN-related pathways. Five overlapping DEGs (IFI27, TXK, RAPGEF6, PIK3IP1 and PSENEN) were selected as key genes by machine-learning algorithms, with IFI27 showing the highest diagnostic performance. The expression level of IFI27 was found to be higher in CD4 CTL, CD8-naïve and various B cell subsets of SLE patients with EBV infection. IFI27 showed significant correlation with B intermediate and CD4 CTL cells. Clinical data showed lower CD4 T cell proportions in SLE patients with EBV infection. CONCLUSION This study identified IFI27 as a key gene for SLE patients with EBV infection, influencing CD4 CTL and B cell subtypes. These findings enhance the understanding of the molecular mechanisms linking SLE and EBV infection, providing potential targets for diagnostic and therapeutic strategies.
Collapse
Affiliation(s)
- Jiajun Gui
- Department of Rheumatology and Immunology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Mengyuan Fang
- Department of Rheumatology and Immunology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Jianxin Tu
- Department of Rheumatology and Immunology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Xiaowei Chen
- Department of Rheumatology and Immunology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Li Sun
- Department of Rheumatology and Immunology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| |
Collapse
|
6
|
Zhu M, Hsu CW, Peralta Ogorek LL, Taylor IW, La Cavera S, Oliveira DM, Verma L, Mehra P, Mijar M, Sadanandom A, Perez-Cota F, Boerjan W, Nolan TM, Bennett MJ, Benfey PN, Pandey BK. Single-cell transcriptomics reveal how root tissues adapt to soil stress. Nature 2025:10.1038/s41586-025-08941-z. [PMID: 40307555 DOI: 10.1038/s41586-025-08941-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2024] [Accepted: 03/26/2025] [Indexed: 05/02/2025]
Abstract
Land plants thrive in soils showing vastly different properties and environmental stresses1. Root systems can adapt to contrasting soil conditions and stresses, yet how their responses are programmed at the individual cell scale remains unclear. Using single-cell RNA sequencing and spatial transcriptomic approaches, we showed major expression changes in outer root cell types when comparing the single-cell transcriptomes of rice roots grown in gel versus soil conditions. These tissue-specific transcriptional responses are related to nutrient homeostasis, cell wall integrity and defence in response to heterogeneous soil versus homogeneous gel growth conditions. We also demonstrate how the model soil stress, termed compaction, triggers expression changes in cell wall remodelling and barrier formation in outer and inner root tissues, regulated by abscisic acid released from phloem cells. Our study reveals how root tissues communicate and adapt to contrasting soil conditions at single-cell resolution.
Collapse
Affiliation(s)
- Mingyuan Zhu
- Department of Biology, Duke University, Durham, NC, USA
- Howard Hughes Medical Institute, Duke University, Durham, NC, USA
| | - Che-Wei Hsu
- Department of Biology, Duke University, Durham, NC, USA
- Howard Hughes Medical Institute, Duke University, Durham, NC, USA
| | - Lucas L Peralta Ogorek
- Plant and Crop Science Department, School of Biosciences, University of Nottingham, Nottingham, UK
| | - Isaiah W Taylor
- Department of Biology, Duke University, Durham, NC, USA
- Howard Hughes Medical Institute, Duke University, Durham, NC, USA
| | - Salvatore La Cavera
- Optics and Photonics Group, Faculty of Engineering, University of Nottingham, Nottingham, UK
| | - Dyoni M Oliveira
- Department of Plant Biotechnology and Bioinformatics, Ghent University, Ghent, Belgium
- VIB Center for Plant Systems Biology, Ghent, Belgium
| | - Lokesh Verma
- Plant and Crop Science Department, School of Biosciences, University of Nottingham, Nottingham, UK
| | - Poonam Mehra
- Plant and Crop Science Department, School of Biosciences, University of Nottingham, Nottingham, UK
| | - Medhavinee Mijar
- Department of Biology, Duke University, Durham, NC, USA
- Howard Hughes Medical Institute, Duke University, Durham, NC, USA
| | - Ari Sadanandom
- Department of Biosciences, University of Durham, Durham, UK
| | - Fernando Perez-Cota
- Optics and Photonics Group, Faculty of Engineering, University of Nottingham, Nottingham, UK
| | - Wout Boerjan
- Department of Plant Biotechnology and Bioinformatics, Ghent University, Ghent, Belgium
- VIB Center for Plant Systems Biology, Ghent, Belgium
| | - Trevor M Nolan
- Department of Biology, Duke University, Durham, NC, USA
- Howard Hughes Medical Institute, Duke University, Durham, NC, USA
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, USA
| | - Malcolm J Bennett
- Plant and Crop Science Department, School of Biosciences, University of Nottingham, Nottingham, UK.
| | - Philip N Benfey
- Department of Biology, Duke University, Durham, NC, USA.
- Howard Hughes Medical Institute, Duke University, Durham, NC, USA.
| | - Bipin K Pandey
- Plant and Crop Science Department, School of Biosciences, University of Nottingham, Nottingham, UK.
| |
Collapse
|
7
|
Fang C, Zhang X, Yang L, Sun L, Lu Y, Liu Y, Guo J, Wang M, Tan Y, Zhang J, Gao X, Zhu L, Liu G, Ren M, Xiao J, Zhang F, Ma S, Zhao R, Mei X, Qi D. Transcriptomic and morphologic vascular aberrations underlying FCDIIb etiology. Nat Commun 2025; 16:3320. [PMID: 40199880 PMCID: PMC11978774 DOI: 10.1038/s41467-025-58535-6] [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: 07/31/2023] [Accepted: 03/25/2025] [Indexed: 04/10/2025] Open
Abstract
Focal cortical dysplasia type II (FCDII) is a major cause of drug-resistant epilepsy, but genetic factors explain only some cases, suggesting other mechanisms. In this study, we conduct a molecular analysis of brain lesions and adjacent areas in FCDIIb patients. By analyzing over 217,506 single-nucleus transcriptional profiles from 15 individuals, we find significant changes in smooth muscle cells (SMCs) and astrocytes. We identify abnormal vascular malformations and a unique type of SMC that we call "Firework cells", which migrate from blood vessels into the brain parenchyma and associate with VIM+ cells. These abnormalities create localized ischemic-hypoxic (I/H) microenvironments, as confirmed by clinical data, further impairing astrocyte function, activating the HIF-1α/mTOR/S6 pathway, and causing neuronal loss. Using zebrafish models, we demonstrate that vascular abnormalities resulting in I/H environments promote seizures. Our results highlight vascular malformations as a factor in FCDIIb pathogenesis, suggesting potential therapeutic avenues.
Collapse
Affiliation(s)
- Chuantao Fang
- Center for Clinical Research and Translational Medicine, Yangpu Hospital, School of Medicine, Tongji University, Shanghai, China
- Institute of Pediatrics, Children's Hospital of Fudan University, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Obstetrics and Gynecology Hospital of Fudan University and Department of Neurology, Huashan Hospital of Fudan University, Fudan University, Shanghai, China
- Shanghai Tenth People's Hospital, Institute for Infectious Diseases and Vaccine Development, Tongji University School of Medicine, Shanghai, China
| | - Xiaodan Zhang
- Institute of Pediatrics, Children's Hospital of Fudan University, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Obstetrics and Gynecology Hospital of Fudan University and Department of Neurology, Huashan Hospital of Fudan University, Fudan University, Shanghai, China
| | - Lin Yang
- Institute of Pediatrics, Children's Hospital of Fudan University, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Obstetrics and Gynecology Hospital of Fudan University and Department of Neurology, Huashan Hospital of Fudan University, Fudan University, Shanghai, China
| | - Licheng Sun
- Center for Clinical Research and Translational Medicine, Yangpu Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Yujia Lu
- Center for Clinical Research and Translational Medicine, Yangpu Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Yi Liu
- Universidade de Vigo, Nutrition and Bromatology Group, Department of Analytical Chemistry and Food Science, Instituto de Agroecoloxía e Alimentación (IAA) - CITEXVI, Vigo, Spain
| | - Jingjing Guo
- Center for Clinical Research and Translational Medicine, Yangpu Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Min Wang
- Institute of Pediatrics, Children's Hospital of Fudan University, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Obstetrics and Gynecology Hospital of Fudan University and Department of Neurology, Huashan Hospital of Fudan University, Fudan University, Shanghai, China
| | - Yanfeng Tan
- Institute of Pediatrics, Children's Hospital of Fudan University, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Obstetrics and Gynecology Hospital of Fudan University and Department of Neurology, Huashan Hospital of Fudan University, Fudan University, Shanghai, China
| | - Jinsen Zhang
- Institute of Pediatrics, Children's Hospital of Fudan University, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Obstetrics and Gynecology Hospital of Fudan University and Department of Neurology, Huashan Hospital of Fudan University, Fudan University, Shanghai, China
| | - Xin Gao
- Shanghai Universal Medical Imaging Diagnostic Center, Shanghai, China
| | - Li Zhu
- Institute of Translational Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Guoping Liu
- Institute of Pediatrics, Children's Hospital of Fudan University, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Obstetrics and Gynecology Hospital of Fudan University and Department of Neurology, Huashan Hospital of Fudan University, Fudan University, Shanghai, China
| | - Maozhi Ren
- Institute of Urban Agriculture, Chinese Academy of Agricultural Sciences, Chengdu National Agricultural Science and Technology Center, Chengdu, China
| | - Jianbo Xiao
- Universidade de Vigo, Nutrition and Bromatology Group, Department of Analytical Chemistry and Food Science, Instituto de Agroecoloxía e Alimentación (IAA) - CITEXVI, Vigo, Spain
| | - Fayong Zhang
- Institute of Pediatrics, Children's Hospital of Fudan University, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Obstetrics and Gynecology Hospital of Fudan University and Department of Neurology, Huashan Hospital of Fudan University, Fudan University, Shanghai, China
| | - Shaojie Ma
- Institute of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Rui Zhao
- Institute of Pediatrics, Children's Hospital of Fudan University, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Obstetrics and Gynecology Hospital of Fudan University and Department of Neurology, Huashan Hospital of Fudan University, Fudan University, Shanghai, China.
- Department of Neurosurgery, Children's Hospital of Shanghai, Shanghai, China.
- Department of Neurosurgery, Hainan Women and Children's Medical Center, Haikou, China.
| | - Xinyu Mei
- Center for Clinical Research and Translational Medicine, Yangpu Hospital, School of Medicine, Tongji University, Shanghai, China.
| | - Dashi Qi
- Center for Clinical Research and Translational Medicine, Yangpu Hospital, School of Medicine, Tongji University, Shanghai, China.
- Institute of Pediatrics, Children's Hospital of Fudan University, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Obstetrics and Gynecology Hospital of Fudan University and Department of Neurology, Huashan Hospital of Fudan University, Fudan University, Shanghai, China.
| |
Collapse
|
8
|
Zhao W, Zhou L, Wang Y, Wang J, Sun YE, Wang Q. Single-cell transcriptome analyses of PBMCs reveal the immunological characteristics of individuals with phlegm-dampness constitution. Front Med 2025; 19:376-385. [PMID: 40126771 DOI: 10.1007/s11684-024-1113-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2024] [Accepted: 09/18/2024] [Indexed: 03/26/2025]
Abstract
Ancient traditional Chinese medicine (TCM) doctrine says "The superior doctor prevents illnesses," pointing out preventative medicine as the ultimate goal for medical care. TCM recognizes that genetic predisposition and environmental and lifestyle influences contribute to diseases. It divides people into eight constitutions in addition to one normal/healthy kind. People with one of the eight subhealth constitutions are prone to develop different kinds of corresponding illnesses. The goal for this type of categorization is to help people take preemptive measures to prevent or delay disease onset. As the peripheral immune system through surveying the body, it can capture information from essentially all organs and reflect anomalies occurring in each organ. Thus, the detailed profiling of the peripheral immune-system function can generally reflect a person's overall heath state. In this study, we performed the single-cell RNA sequencing (scRNA-seq) of peripheral blood mononuclear cells (PBMCs) from individuals with Tanshi (phlegm dampness) constitution. They were prone to develop metabolic disorders including diabetes. scRNA-seq revealed greatly reduced mucosal-associated invariable T cell content and heightened TNFα-NFκB, JAK-STAT, and interferon signaling. These findings indicated heightened chronic inflammation, as well as increased hypoxia/apoptosis responses, likely resulting from frequent sleep apnea that Tanshi individuals experienced. Altogether, this pilot study demonstrated effectiveness in using scRNA-seq to reveal molecular-immunological bases for constitution categorization, thereby substantiating that preventative medicine originated from TCM.
Collapse
Affiliation(s)
- Weibo Zhao
- National Institute of Traditional Chinese Medicine Constitution and Preventive Treatment of Diseases, Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Liqiang Zhou
- Faculty of Life and Health Sciences, Shenzhen University of Advanced Technology (SUAT), Shenzhen, 518107, China
| | - Yixing Wang
- Department of Internal Medicine of Traditional Chinese Medicine, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, 200120, China
| | - Ji Wang
- National Institute of Traditional Chinese Medicine Constitution and Preventive Treatment of Diseases, Beijing University of Chinese Medicine, Beijing, 100029, China.
| | - Yi Eve Sun
- Stem Cell Translational Research Center, Tongji Hospital, Tongji University School of Medicine, Shanghai, 200065, China.
| | - Qi Wang
- National Institute of Traditional Chinese Medicine Constitution and Preventive Treatment of Diseases, Beijing University of Chinese Medicine, Beijing, 100029, China.
| |
Collapse
|
9
|
Mwirigi JM, Sankaranarayanan I, Tavares-Ferreira D, Gabriel KA, Palomino S, Li Y, Uhelski ML, Shiers S, Franco-Enzástiga Ú, Wangzhou A, Lesnak JB, Bandaru S, Shrivastava A, Inturi N, Albrecht PJ, Dockum M, Cervantes AM, Horton P, Funk G, North RY, Tatsui CE, Corrales G, Yousuf MS, Curatolo M, Gereau RW, Patwardhan A, Dussor G, Dougherty PM, Rice FL, Price TJ. Expansion of OSMR expression and signaling in the human dorsal root ganglion links OSM to neuropathic pain. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.03.26.645611. [PMID: 40236060 PMCID: PMC11996445 DOI: 10.1101/2025.03.26.645611] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 04/17/2025]
Abstract
RNA sequencing studies on human dorsal root ganglion (hDRG) from patients suffering from neuropathic pain show upregulation of OSM, linking this IL-6 family cytokine to pain disorders. In mice, however, OSM signaling causes itch behaviors through a direct effect on its cognate receptor expressed uniquely by pruriceptive sensory neurons. We hypothesized that an expansion in function of OSM-OSM receptor (OSMR) in sensory disorders in humans could be explained by species differences in receptor expression and signaling. Our in situ hybridization and immunohistochemical findings demonstrate broad expression of OSMR in DRG nociceptors and afferent fibers innervating the superficial and deep skin of humans. In patch-clamp electrophysiology, OSM directly activates human sensory neurons engaging MAPK signaling to promote action potential firing. Using CRISPR editing we show that OSM activation of MAPK signaling is dependent on OSMR and not LIFR in hDRG. Bulk, single-nuclei, and single-cell RNA-seq of OSM-treated hDRG cultures reveal expansive similarities in the transcriptomic signature observed in pain DRGs from neuropathic patients, indicating that OSM alone can orchestrate transcriptomic signatures associated with pain. We conclude that OSM-OSMR signaling via MAPKs is a critical signaling factor for DRG plasticity that may underlie neuropathic pain in patients.
Collapse
|
10
|
Brinkmeier ML, Wang SQ, Pittman HA, Cheung LY, Prasov L. Myelin regulatory factor (MYRF) is a critical early regulator of retinal pigment epithelial development. PLoS Genet 2025; 21:e1011670. [PMID: 40233131 PMCID: PMC12052213 DOI: 10.1371/journal.pgen.1011670] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2024] [Revised: 05/05/2025] [Accepted: 04/01/2025] [Indexed: 04/17/2025] Open
Abstract
Myelin regulatory factor (Myrf) is a critical transcription factor in early retinal and retinal pigment epithelial development, and human variants in MYRF are a cause for nanophthalmos. Single cell RNA sequencing (scRNAseq) was performed on Myrf conditional knockout mice (Rx > Cre Myrffl/fl) at 3 developmental timepoints. Myrf was expressed specifically in the RPE, and expression was abrogated in Rx > Cre Myrffl/fl eyes. scRNAseq analysis revealed a loss of RPE cells at all timepoints resulting from cell death. GO-term analysis in the RPE revealed downregulation of melanogenesis and anatomic structure morphogenesis pathways, which were supported by electron microscopy and histologic analysis. Novel structural target genes including Ermn and Upk3b, along with macular degeneration and inherited retinal disease genes were identified as downregulated, and a strong upregulation of TGFß/BMP signaling and effectors was observed. Regulon analysis placed Myrf downstream or parallel to Pax6 and Mitf and upstream of Sox10 in RPE differentiation. Together, these results suggest a strong role for MYRF in the RPE maturation by regulating melanogenesis, cell survival, and cell structure, in part acting through suppression of TGFß signaling and activation of Sox10.
Collapse
Affiliation(s)
- Michelle L. Brinkmeier
- Department of Ophthalmology and Visual Sciences, University of Michigan, Ann Arbor, Michigan, United States of America
- Department of Human Genetics, University of Michigan, Ann Arbor, Michigan, United States of America
| | - Su Qing Wang
- Department of Ophthalmology and Visual Sciences, University of Michigan, Ann Arbor, Michigan, United States of America
| | - Hannah A. Pittman
- Department of Ophthalmology and Visual Sciences, University of Michigan, Ann Arbor, Michigan, United States of America
| | - Leonard Y. Cheung
- Department of Human Genetics, University of Michigan, Ann Arbor, Michigan, United States of America
- Department of Physiology and Biophysics, State University of New York at Stony Brook, Stony Brook, New York, United States of America
| | - Lev Prasov
- Department of Ophthalmology and Visual Sciences, University of Michigan, Ann Arbor, Michigan, United States of America
- Department of Human Genetics, University of Michigan, Ann Arbor, Michigan, United States of America
| |
Collapse
|
11
|
Llera-Oyola J, Pérez-Moraga R, Parras M, Rosón B. How to view the female reproductive tract through single-cell looking glasses. Am J Obstet Gynecol 2025; 232:S21-S43. [PMID: 40253081 DOI: 10.1016/j.ajog.2024.08.040] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2023] [Revised: 07/04/2024] [Accepted: 08/24/2024] [Indexed: 04/21/2025]
Abstract
Single-cell technologies have emerged as an unprecedented tool for biologists and clinicians, allowing them to assess organs and tissues at the level of individual cells. In the field of women's reproductive biology, single-cell studies have provided insights into the cellular and molecular processes that regulate reproductive and obstetrical functions in health and disease. The knowledge that these studies generate is helping clinicians to improve the understanding and diagnosis of infertility related issues or pregnancy complications and to find new avenues for their treatment. However, navigating the expansive landscape of this type of transcriptomic data analysis represents a pivotal challenge in current research. Single cell RNA sequencing involves isolating cells into droplets, reverse transcribing RNA to generate complementary DNA, with each droplet content uniquely labeled by a barcode. Upon sequencing the complementary DNAs, the barcodes enable the reassignment of sequencing reads to individual droplets, facilitating the reconstruction of the cellular landscape of the sample obtained from a tissue or organ and beyond. Researchers, equipped with the metaphorical 'single-cell glasses,' must adequately choose from a plethora of strategies to dissect and interpret cellular information. Sophisticated algorithms and the decision-making process are often underestimated, resulting in artefactual or cumbersome interpreted results. Computational biologists apply and innovate computational tools designed to process, model, and interpret expansive datasets. The ramifications of their work extend far beyond the realm of data processing; they give shape to the outcome of analyses, playing a pivotal role in drawing meaningful conclusions from the wealth of information garnered. In this review, we describe the wide variety of approaches and analytical steps available with enough detail to gain a concise picture of what a complete examination of a single-cell dataset would be. We commence with a discussion on key points in experimental design, highlighting crucial questions one should consider. Following this, we delve into the various preprocessing and quality control steps essential for any single-cell dataset. The subsequent section offers a detailed guide on constructing a single-cell atlas, exploring nuances such as differential characteristics in visualization and clustering techniques, as well as strategies for assigning identity to cell populations through gene marker annotations. Moving beyond the creation of an atlas, we explore methods for investigating pathological conditions. This involves conducting cell population comparison tests between conditions and analyzing specific cell-to-cell communications and cellular differentiation trajectories in both health and disease scenarios. This work aims to furnish a newcomer researcher and/or clinician with essential guidelines to embark on a single-cell adventure without succumbing to common pitfalls. By bridging the gap between theory and practice, it facilitates the translation of single-cell technologies into clinically relevant applications. Throughout the manuscript, practical examples of its usage in women's reproductive health studies are provided. Various sections delve into specific clinical scenarios, demonstrating how these guidelines can be instrumental in unraveling the molecular landscapes of diseases and physiological processes related to women's reproduction.
Collapse
Affiliation(s)
- Jaime Llera-Oyola
- Carlos Simon Foundation, INCLIVA Health Research Institute, Valencia, Spain
| | - Raúl Pérez-Moraga
- Carlos Simon Foundation, INCLIVA Health Research Institute, Valencia, Spain; R&D Department, Igenomix, Valencia, Spain
| | - Marcos Parras
- Carlos Simon Foundation, INCLIVA Health Research Institute, Valencia, Spain
| | - Beatriz Rosón
- Carlos Simon Foundation, INCLIVA Health Research Institute, Valencia, Spain.
| |
Collapse
|
12
|
Qiu Y, Wang Y, Liu J, Liu B, Sun K, Hou Q. Single-cell sequencing uncovers a high ESM1-expression endothelial cell subpopulation associated with bladder cancer progression and the immunosuppressive microenvironment. Sci Rep 2025; 15:10946. [PMID: 40159545 PMCID: PMC11955522 DOI: 10.1038/s41598-025-95731-2] [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: 01/11/2025] [Accepted: 03/24/2025] [Indexed: 04/02/2025] Open
Abstract
Despite remarkable advancements in therapeutic strategies, a considerable proportion of patients with bladder cancer (BC) still experience disease progression and unfavorable prognosis. The heterogeneity and biological functions of tumor endothelial cells (ECs) during BC progression remain poorly understood. We collected scRNA-seq data from BC samples and identified two EC subpopulations through hierarchical clustering analysis. The activity of signaling pathways in distinct EC subpopulations was assessed utilizing AUCell analysis. Gene regulatory networks (GRN) were constructed and analyzed for different EC subpopulations using the pySCENIC algorithm. Additionally, we investigated the association between the abundance of EC subpopulations and both clinical prognosis and immune cell infiltration. The biological effects of ESM1 protein on BC cells were further validated through EdU and Transwell assays. We analyzed 7,519 CD45-negative single cells from BC tissues and discerned two distinct EC subpopulations. The two subpopulations were characterized by high expression of ESM1 (S1 ECs) and CXCL2 (S2 ECs), respectively. In S1 ECs, we observed significant activation of signaling pathways involved in tumor promotion, including angiogenesis and cell proliferation. Additionally, our GRN analysis uncovered notable differences in transcription factor activity between S1 and S2 ECs. Moreover, ESM1 protein promoted proliferation and migration of BC cells. Patients with higher abundance of the S1 EC subpopulation exhibited more unfavorable clinical outcomes and increased infiltration of inhibitory immune cells. Our findings elucidate the transcriptional profiles and biological roles of the high ESM1-expression endothelial cell subpopulation in BC. This subpopulation is associated with poor prognosis and immunosuppressive tumor microenvironment. Accordingly, targeting endothelial cells with high ESM1 expression may offer a novel therapeutic strategy for patients with BC.
Collapse
Affiliation(s)
- Yifeng Qiu
- Guangdong Key Laboratory for Biomedical Measurements and Ultrasound Imaging, National-Regional Key Technology Engineering Laboratory for Medical Ultrasound, School of Biomedical Engineering, Shenzhen University Medical school, Shenzhen, 518060, China
- Department of Urology, Shenzhen University General Hospital, Shenzhen University, Shenzhen, Guangdong, China
- Shenzhen Key Laboratory for Systemic Aging and Intervention (SAI), Guangdong Key Laboratory of Genome Stability and Human Disease Prevention, School of Basic Medical Sciences, Marshall Laboratory of Biomedical Engineering, National Engineering Research Center for Biotechnology (Shenzhen), International Cancer Center, Shenzhen University, Shenzhen, Guangdong, China
- International Cancer Center, Shenzhen Key Laboratory, Hematology Institution of ShenzhenUniversity, Shenzhen, China
| | - Yuhan Wang
- Guangdong Key Laboratory for Biomedical Measurements and Ultrasound Imaging, National-Regional Key Technology Engineering Laboratory for Medical Ultrasound, School of Biomedical Engineering, Shenzhen University Medical school, Shenzhen, 518060, China
- Department of Urology, Shenzhen University General Hospital, Shenzhen University, Shenzhen, Guangdong, China
- Shenzhen Key Laboratory for Systemic Aging and Intervention (SAI), Guangdong Key Laboratory of Genome Stability and Human Disease Prevention, School of Basic Medical Sciences, Marshall Laboratory of Biomedical Engineering, National Engineering Research Center for Biotechnology (Shenzhen), International Cancer Center, Shenzhen University, Shenzhen, Guangdong, China
| | - Jiahe Liu
- Guangdong Key Laboratory for Biomedical Measurements and Ultrasound Imaging, National-Regional Key Technology Engineering Laboratory for Medical Ultrasound, School of Biomedical Engineering, Shenzhen University Medical school, Shenzhen, 518060, China
- Department of Urology, Shenzhen University General Hospital, Shenzhen University, Shenzhen, Guangdong, China
- Shenzhen Key Laboratory for Systemic Aging and Intervention (SAI), Guangdong Key Laboratory of Genome Stability and Human Disease Prevention, School of Basic Medical Sciences, Marshall Laboratory of Biomedical Engineering, National Engineering Research Center for Biotechnology (Shenzhen), International Cancer Center, Shenzhen University, Shenzhen, Guangdong, China
| | - Baohua Liu
- Shenzhen Key Laboratory for Systemic Aging and Intervention (SAI), Guangdong Key Laboratory of Genome Stability and Human Disease Prevention, School of Basic Medical Sciences, Marshall Laboratory of Biomedical Engineering, National Engineering Research Center for Biotechnology (Shenzhen), International Cancer Center, Shenzhen University, Shenzhen, Guangdong, China.
| | - Kai Sun
- Department of Radiology, the Third People's Hospital of Longgang District, Shenzhen Clinical Medical School, Guangzhou University of Chinese Medicine, Shenzhen, 518116, China.
| | - Qi Hou
- Department of Urology, Shenzhen University General Hospital, Shenzhen University, Shenzhen, Guangdong, China.
- Shenzhen Key Laboratory for Systemic Aging and Intervention (SAI), Guangdong Key Laboratory of Genome Stability and Human Disease Prevention, School of Basic Medical Sciences, Marshall Laboratory of Biomedical Engineering, National Engineering Research Center for Biotechnology (Shenzhen), International Cancer Center, Shenzhen University, Shenzhen, Guangdong, China.
- International Cancer Center, Shenzhen Key Laboratory, Hematology Institution of ShenzhenUniversity, Shenzhen, China.
| |
Collapse
|
13
|
Xu P, Cheng S, Yang X, Xu K, Hou W, Liu L, Peng K, Wen Y, Zhang F. Integrative single-cell analysis reveals transcriptional and epigenetic regulatory features of human developmental dysplasia of the hip. Osteoarthritis Cartilage 2025:S1063-4584(25)00866-0. [PMID: 40154730 DOI: 10.1016/j.joca.2025.02.788] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/14/2024] [Revised: 01/18/2025] [Accepted: 02/14/2025] [Indexed: 04/01/2025]
Abstract
OBJECTIVE Developmental dysplasia of the hip (DDH) is a developmental disorder that has long-term chronic pain and limited hip joint mobility. The aim of the current study is to understand the specific chondrocyte composition involved in DDH development, identify effective biomarkers for DDH prediction, and elucidate the gene regulatory elements driving DDH progression. METHOD In this study, we performed an integrated analysis combining single-cell RNA sequencing and single-cell assay for transposase-accessible chromatin sequencing to investigate the molecular programs and epigenetic changes governing human DDH pathogenesis. Validation of marker genes for distinct chondrocyte populations was performed via immunohistochemical assays, alongside characterization of regulatory elements specific to DDH. RESULTS Our analysis identified seven molecularly distinct chondrocyte populations in DDH cartilage, including a novel inflammatory chondrocyte population with unique molecular signatures. Furthermore, we reconstructed the differentiation trajectory of chondrocytes, shedding light on their roles in DDH pathogenesis. Integrative analyses of transcriptomic and chromatin accessibility profiles highlighted shared regulatory features and transcriptional programs among chondrocyte subtypes, with several regulatory elements linked to DDH progression. Immunohistochemical validation corroborated the presence of key marker genes in distinct chondrocyte subsets. CONCLUSION Our findings contribute to clarifying the cellular heterogeneity of DDH and offer insights into potential early diagnostic and therapeutic strategies for this condition.
Collapse
Affiliation(s)
- Peng Xu
- Department of Joint Surgery, HongHui Hospital, Xi'an Jiaotong University, Xi'an, Shaanxi 710061, PR China.
| | - Shiqiang Cheng
- Key Laboratory of Trace Elements and Endemic Diseases, Collaborative Innovation Center of Endemic Disease and Health Promotion for Silk Road Region, School of Public Health, Health Science Center, Xi'an Jiaotong University, Xi'an, Shaanxi 710061, PR China
| | - Xuena Yang
- Key Laboratory of Trace Elements and Endemic Diseases, Collaborative Innovation Center of Endemic Disease and Health Promotion for Silk Road Region, School of Public Health, Health Science Center, Xi'an Jiaotong University, Xi'an, Shaanxi 710061, PR China
| | - Ke Xu
- Department of Joint Surgery, HongHui Hospital, Xi'an Jiaotong University, Xi'an, Shaanxi 710061, PR China
| | - Weikun Hou
- Department of Joint Surgery, HongHui Hospital, Xi'an Jiaotong University, Xi'an, Shaanxi 710061, PR China
| | - Lin Liu
- Department of Joint Surgery, HongHui Hospital, Xi'an Jiaotong University, Xi'an, Shaanxi 710061, PR China
| | - Kan Peng
- Department of Joint Surgery, HongHui Hospital, Xi'an Jiaotong University, Xi'an, Shaanxi 710061, PR China
| | - Yan Wen
- Key Laboratory of Trace Elements and Endemic Diseases, Collaborative Innovation Center of Endemic Disease and Health Promotion for Silk Road Region, School of Public Health, Health Science Center, Xi'an Jiaotong University, Xi'an, Shaanxi 710061, PR China
| | - Feng Zhang
- Key Laboratory of Trace Elements and Endemic Diseases, Collaborative Innovation Center of Endemic Disease and Health Promotion for Silk Road Region, School of Public Health, Health Science Center, Xi'an Jiaotong University, Xi'an, Shaanxi 710061, PR China.
| |
Collapse
|
14
|
Hasel P, Cooper ML, Marchildon AE, Rufen-Blanchette U, Kim RD, Ma TC, Groh AMR, Hill EJ, Lewis EM, Januszewski M, Light SEW, Smith CJ, Stratton JA, Sloan SA, Kang UJ, Chao MV, Liddelow SA. Defining the molecular identity and morphology of glia limitans superficialis astrocytes in vertebrates. Cell Rep 2025; 44:115344. [PMID: 39982817 DOI: 10.1016/j.celrep.2025.115344] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2023] [Revised: 07/30/2024] [Accepted: 02/01/2025] [Indexed: 02/23/2025] Open
Abstract
Astrocytes are a highly abundant glial cell type and perform critical homeostatic functions in the central nervous system. Like neurons, astrocytes have many discrete heterogeneous subtypes. The subtype identity and functions are, at least in part, associated with their anatomical location and can be highly restricted to strategically important anatomical domains. Here, we report that astrocytes forming the glia limitans superficialis, the outermost border of the brain and spinal cord, are a highly specialized astrocyte subtype and can be identified by a single marker: myocilin (Myoc). We show that glia limitans superficialis astrocytes cover the entire brain and spinal cord surface, exhibit an atypical morphology, and are evolutionarily conserved from zebrafish, rodents, and non-human primates to humans. Identification of this highly specialized astrocyte subtype will advance our understanding of CNS homeostasis and potentially be targeted for therapeutic intervention to combat peripheral inflammatory effects on the CNS.
Collapse
Affiliation(s)
- Philip Hasel
- Institute for Translational Neuroscience, NYU Grossman School of Medicine, New York, NY, USA; UK Dementia Research Institute at the University of Edinburgh, Edinburgh, UK; Centre for Discovery Brain Sciences, School of Biomedical Sciences, College of Medicine and Veterinary Medicine, The University of Edinburgh, Edinburgh, UK.
| | - Melissa L Cooper
- Institute for Translational Neuroscience, NYU Grossman School of Medicine, New York, NY, USA
| | - Anne E Marchildon
- Institute for Translational Neuroscience, NYU Grossman School of Medicine, New York, NY, USA
| | - Uriel Rufen-Blanchette
- Institute for Translational Neuroscience, NYU Grossman School of Medicine, New York, NY, USA
| | - Rachel D Kim
- Institute for Translational Neuroscience, NYU Grossman School of Medicine, New York, NY, USA
| | - Thong C Ma
- Fresco Institute for Parkinson's and Movement Disorders, Department of Neurology, NYU Grossman School of Medicine, New York, NY, USA; Parekh Center for Interdisciplinary Neurology, NYU Grossman School of Medicine, New York, NY, USA; Department of Neuroscience, NYU Grossman School of Medicine, New York, NY, USA
| | - Adam M R Groh
- Department of Neurology and Neurosurgery, Montreal Neurological Institute-Hospital, McGill University, Montréal, QC, Canada
| | - Emily J Hill
- Department of Human Genetics, Emory University School of Medicine, Atlanta, GA, USA
| | - Eleanor M Lewis
- UK Dementia Research Institute at the University of Edinburgh, Edinburgh, UK; Centre for Discovery Brain Sciences, School of Biomedical Sciences, College of Medicine and Veterinary Medicine, The University of Edinburgh, Edinburgh, UK
| | | | - Sarah E W Light
- Department of Biological Sciences, University of Notre Dame, Notre Dame, IN, USA; Center for Stem Cells and Regenerative Medicine, University of Notre Dame, Notre Dame, IN, USA
| | - Cody J Smith
- Department of Biological Sciences, University of Notre Dame, Notre Dame, IN, USA; Center for Stem Cells and Regenerative Medicine, University of Notre Dame, Notre Dame, IN, USA
| | - Jo Anne Stratton
- Department of Neurology and Neurosurgery, Montreal Neurological Institute-Hospital, McGill University, Montréal, QC, Canada
| | - Steven A Sloan
- Department of Human Genetics, Emory University School of Medicine, Atlanta, GA, USA
| | - Un Jung Kang
- Institute for Translational Neuroscience, NYU Grossman School of Medicine, New York, NY, USA; Fresco Institute for Parkinson's and Movement Disorders, Department of Neurology, NYU Grossman School of Medicine, New York, NY, USA; Parekh Center for Interdisciplinary Neurology, NYU Grossman School of Medicine, New York, NY, USA; Department of Neuroscience, NYU Grossman School of Medicine, New York, NY, USA
| | - Moses V Chao
- Institute for Translational Neuroscience, NYU Grossman School of Medicine, New York, NY, USA; Department of Neuroscience, NYU Grossman School of Medicine, New York, NY, USA; Department of Cell Biology, NYU Grossman School of Medicine, New York, NY, USA; Department of Psychiatry, NYU Grossman School of Medicine, New York, NY, USA
| | - Shane A Liddelow
- Institute for Translational Neuroscience, NYU Grossman School of Medicine, New York, NY, USA; Parekh Center for Interdisciplinary Neurology, NYU Grossman School of Medicine, New York, NY, USA; Department of Neuroscience, NYU Grossman School of Medicine, New York, NY, USA; Department of Ophthalmology, NYU Grossman School of Medicine, New York, NY, USA.
| |
Collapse
|
15
|
Zhao S, Yu H, Li Z, Chen W, Liu K, Dai H, Wang G, Zhang Z, Xie J, He Y, Li L. Single-cell RNA sequencing reveals a new mechanism of endothelial cell heterogeneity and healing in diabetic foot ulcers. Biol Direct 2025; 20:34. [PMID: 40121493 PMCID: PMC11929994 DOI: 10.1186/s13062-025-00628-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2025] [Accepted: 03/07/2025] [Indexed: 03/25/2025] Open
Abstract
Diabetic foot ulcers (DFU) are a common and severe complication among diabetic patients, posing a significant burden on patients' quality of life and healthcare systems due to their high incidence, amputation rates, and mortality. This study utilized single-cell RNA sequencing technology to deeply analyze the cellular heterogeneity of the skin on the feet ofDFU patients and the transcriptomic characteristics of endothelial cells, aiming to identify key cell populations and genes associated with the healing and progression of DFU. The study found that endothelial cells from DFU patients exhibited significant transcriptomic differences under various conditions, particularly in signaling pathways related to inflammatory responses and angiogenesis. Through trajectory analysis and cell communication research, we revealed the key role of endothelial cell subsets in the development of DFU and identified multiple important gene modules associated with the progression of DFU. Notably, the promoting effect of the SH3BGRL3 gene on endothelial cell proliferation, migration, and angiogenic capabilities under high glucose conditions was experimentally verified, providing a new potential target and theoretical basis for the treatment of DFU. This study not only enhances the understanding of the pathogenesis ofDFU but also provides a scientific basis for the development ofnew therapeutic strategies.
Collapse
Affiliation(s)
- Songyun Zhao
- Department of Plastic Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Hua Yu
- Department of Plastic Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Zihao Li
- Department of Plastic Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Wanying Chen
- Department of Plastic Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Kaibo Liu
- Department of Plastic Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Hao Dai
- Department of Plastic Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Gaoyi Wang
- Department of Plastic Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Zibing Zhang
- Department of Neurosurgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Jiaheng Xie
- Department of Plastic Surgery, Xiangya Hospital, Central South University, Changsha, China.
| | - Yucang He
- Department of Plastic Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China.
| | - Liqun Li
- Department of Plastic Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China.
- National Key Clinical Specialty (Wound Healing), The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China.
| |
Collapse
|
16
|
Jelcic I, Naghavian R, Fanaswala I, Macnair W, Esposito C, Calini D, Han Y, Marti Z, Raposo C, Sarabia Del Castillo J, Oldrati P, Erny D, Kana V, Zheleznyakova G, Al Nimer F, Tackenberg B, Reichen I, Khademi M, Piehl F, Robinson MD, Jelcic I, Sospedra M, Pelkmans L, Malhotra D, Reynolds R, Jagodic M, Martin R. T-bet+ CXCR3+ B cells drive hyperreactive B-T cell interactions in multiple sclerosis. Cell Rep Med 2025; 6:102027. [PMID: 40107244 PMCID: PMC11970401 DOI: 10.1016/j.xcrm.2025.102027] [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: 08/22/2023] [Revised: 05/16/2024] [Accepted: 02/20/2025] [Indexed: 03/22/2025]
Abstract
Multiple sclerosis (MS) is an autoimmune disease of the central nervous system (CNS). Self-peptide-dependent autoproliferation (AP) of B and T cells is a key mechanism in MS. Here, we show that pro-inflammatory B-T cell-enriched cell clusters (BTECs) form during AP and mirror features of a germinal center reaction. T-bet+CXCR3+ B cells are the main cell subset amplifying and sustaining their counterpart Th1 cells via interferon (IFN)-γ and are present in highly inflamed meningeal tissue. The underlying B cell activation signature is reflected by epigenetic modifications and receptor-ligand interactions with self-reactive T cells. AP+ CXCR3+ B cells show marked clonal evolution from memory to somatically hypermutated plasmablasts and upregulation of IFN-γ-related genes. Our data underscore a key role of T-bet+CXCR3+ B cells in the pathogenesis of MS in both the peripheral immune system and the CNS compartment, and thus they appear to be involved in both early relapsing-remitting disease and the chronic stage.
Collapse
Affiliation(s)
- Ivan Jelcic
- Neuroimmunology and MS Research Section (NIMS), Neurology Clinic, University of Zurich, University Hospital Zurich, 8091 Zurich, Switzerland; Roche Pharma Research and Early Development (pRED), Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd, Basel, Switzerland.
| | - Reza Naghavian
- Neuroimmunology and MS Research Section (NIMS), Neurology Clinic, University of Zurich, University Hospital Zurich, 8091 Zurich, Switzerland
| | - Imran Fanaswala
- SIB Swiss Institute of Bioinformatics, University of Zurich, Zurich, Switzerland; Department of Molecular Life Sciences, University of Zurich, Zurich, Switzerland
| | - Will Macnair
- Roche Pharma Research and Early Development (pRED), Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd, Basel, Switzerland
| | - Cinzia Esposito
- Department of Molecular Life Sciences, University of Zurich, Zurich, Switzerland
| | - Daniela Calini
- Roche Pharma Research and Early Development (pRED), Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd, Basel, Switzerland
| | - Yanan Han
- Department of Clinical Neuroscience, Karolinska Institutet, Center for Molecular Medicine, Karolinska University Hospital, Stockholm, Sweden
| | - Zoe Marti
- Neuroimmunology and MS Research Section (NIMS), Neurology Clinic, University of Zurich, University Hospital Zurich, 8091 Zurich, Switzerland; Cellerys AG, Schlieren, Switzerland
| | - Catarina Raposo
- Roche Pharma Research and Early Development (pRED), Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd, Basel, Switzerland
| | | | - Pietro Oldrati
- Neuroimmunology and MS Research Section (NIMS), Neurology Clinic, University of Zurich, University Hospital Zurich, 8091 Zurich, Switzerland; Cellerys AG, Schlieren, Switzerland
| | - Daniel Erny
- Neuroimmunology and MS Research Section (NIMS), Neurology Clinic, University of Zurich, University Hospital Zurich, 8091 Zurich, Switzerland; Institute of Neuropathology, University of Freiburg, Freiburg, Germany
| | - Veronika Kana
- Neuroimmunology and MS Research Section (NIMS), Neurology Clinic, University of Zurich, University Hospital Zurich, 8091 Zurich, Switzerland
| | - Galina Zheleznyakova
- Department of Clinical Neuroscience, Karolinska Institutet, Center for Molecular Medicine, Karolinska University Hospital, Stockholm, Sweden
| | - Faiez Al Nimer
- Neuroimmunology and MS Research Section (NIMS), Neurology Clinic, University of Zurich, University Hospital Zurich, 8091 Zurich, Switzerland; Department of Clinical Neuroscience, Karolinska Institutet, Center for Molecular Medicine, Karolinska University Hospital, Stockholm, Sweden
| | - Björn Tackenberg
- Product Development Medical Affairs, Neuroscience and Rare Disease, F. Hoffmann-La Roche Ltd, Basel, Switzerland
| | - Ina Reichen
- Neuroimmunology and MS Research Section (NIMS), Neurology Clinic, University of Zurich, University Hospital Zurich, 8091 Zurich, Switzerland
| | - Mohsen Khademi
- Department of Clinical Neuroscience, Karolinska Institutet, Center for Molecular Medicine, Karolinska University Hospital, Stockholm, Sweden
| | - Fredrik Piehl
- Department of Clinical Neuroscience, Karolinska Institutet, Center for Molecular Medicine, Karolinska University Hospital, Stockholm, Sweden
| | - Mark D Robinson
- SIB Swiss Institute of Bioinformatics, University of Zurich, Zurich, Switzerland; Department of Molecular Life Sciences, University of Zurich, Zurich, Switzerland
| | - Ilijas Jelcic
- Neuroimmunology and MS Research Section (NIMS), Neurology Clinic, University of Zurich, University Hospital Zurich, 8091 Zurich, Switzerland
| | - Mireia Sospedra
- Neuroimmunology and MS Research Section (NIMS), Neurology Clinic, University of Zurich, University Hospital Zurich, 8091 Zurich, Switzerland; Cellerys AG, Schlieren, Switzerland
| | - Lucas Pelkmans
- Department of Molecular Life Sciences, University of Zurich, Zurich, Switzerland
| | - Dheeraj Malhotra
- Roche Pharma Research and Early Development (pRED), Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd, Basel, Switzerland
| | | | - Maja Jagodic
- Department of Clinical Neuroscience, Karolinska Institutet, Center for Molecular Medicine, Karolinska University Hospital, Stockholm, Sweden
| | - Roland Martin
- Neuroimmunology and MS Research Section (NIMS), Neurology Clinic, University of Zurich, University Hospital Zurich, 8091 Zurich, Switzerland; Institute of Experimental Immunology, University of Zurich, Zurich, Switzerland; Therapeutic Design Unit, Center for Molecular Medicine, Department of Clinical Neurosciences, Karolinska Institutet, Stockholm, Sweden; Cellerys AG, Schlieren, Switzerland.
| |
Collapse
|
17
|
Wu CH, Zhou X, Chen M. Exploring and mitigating shortcomings in single-cell differential expression analysis with a new statistical paradigm. Genome Biol 2025; 26:58. [PMID: 40098192 PMCID: PMC11912664 DOI: 10.1186/s13059-025-03525-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2023] [Accepted: 03/05/2025] [Indexed: 03/19/2025] Open
Abstract
BACKGROUND Differential expression analysis is pivotal in single-cell transcriptomics for unraveling cell-type-specific responses to stimuli. While numerous methods are available to identify differentially expressed genes in single-cell data, recent evaluations of both single-cell-specific methods and methods adapted from bulk studies have revealed significant shortcomings in performance. In this paper, we dissect the four major challenges in single-cell differential expression analysis: excessive zeros, normalization, donor effects, and cumulative biases. These "curses" underscore the limitations and conceptual pitfalls in existing workflows. RESULTS To address the limitations of current single-cell differential expression analysis methods, we propose GLIMES, a statistical framework that leverages UMI counts and zero proportions within a generalized Poisson/Binomial mixed-effects model to account for batch effects and within-sample variation. We rigorously benchmarked GLIMES against six existing differential expression methods using three case studies and simulations across different experimental scenarios, including comparisons across cell types, tissue regions, and cell states. Our results demonstrate that GLIMES is more adaptable to diverse experimental designs in single-cell studies and effectively mitigates key shortcomings of current approaches, particularly those related to normalization procedures. By preserving biologically meaningful signals, GLIMES offers improved performance in detecting differentially expressed genes. CONCLUSIONS By using absolute RNA expression rather than relative abundance, GLIMES improves sensitivity, reduces false discoveries, and enhances biological interpretability. This paradigm shift challenges existing workflows and highlights the need for careful consideration of normalization strategies, ultimately paving the way for more accurate and robust single-cell transcriptomic analyses.
Collapse
Affiliation(s)
- Chih-Hsuan Wu
- Department of Statistics, University of Chicago, Chicago, USA
| | - Xiang Zhou
- Department of Biostatistics, University of Michigan, Ann Arbor, USA
| | - Mengjie Chen
- Department of Human Genetics and Department of Medicine, University of Chicago, Chicago, USA.
| |
Collapse
|
18
|
Wang M, Di Pietro-Torres A, Feregrino C, Luxey M, Moreau C, Fischer S, Fages A, Ritz D, Tschopp P. Distinct gene regulatory dynamics drive skeletogenic cell fate convergence during vertebrate embryogenesis. Nat Commun 2025; 16:2187. [PMID: 40038298 PMCID: PMC11880379 DOI: 10.1038/s41467-025-57480-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2024] [Accepted: 02/12/2025] [Indexed: 03/06/2025] Open
Abstract
Cell type repertoires have expanded extensively in metazoan animals, with some clade-specific cells being crucial to evolutionary success. A prime example are the skeletogenic cells of vertebrates. Depending on anatomical location, these cells originate from three different precursor lineages, yet they converge developmentally towards similar cellular phenotypes. Furthermore, their 'skeletogenic competency' arose at distinct evolutionary timepoints, thus questioning to what extent different skeletal body parts rely on truly homologous cell types. Here, we investigate how lineage-specific molecular properties are integrated at the gene regulatory level, to allow for skeletogenic cell fate convergence. Using single-cell functional genomics, we find that distinct transcription factor profiles are inherited from the three precursor states and incorporated at lineage-specific enhancer elements. This lineage-specific regulatory logic suggests that these regionalized skeletogenic cells are distinct cell types, rendering them amenable to individualized selection, to define adaptive morphologies and biomaterial properties in different parts of the vertebrate skeleton.
Collapse
Affiliation(s)
- Menghan Wang
- Zoology, Department of Environmental Sciences, University of Basel, Basel, Switzerland
- Department of Biomedicine, University Hospital Basel, University of Basel, Basel, Switzerland
| | - Ana Di Pietro-Torres
- Zoology, Department of Environmental Sciences, University of Basel, Basel, Switzerland
| | - Christian Feregrino
- Zoology, Department of Environmental Sciences, University of Basel, Basel, Switzerland
- Max Planck Institute for Molecular Genetics, Berlin, Germany
| | - Maëva Luxey
- Zoology, Department of Environmental Sciences, University of Basel, Basel, Switzerland
- MeLis, CNRS UMR 5284, INSERM U1314, Université Claude Bernard Lyon 1, Institut NeuroMyo Gène, Lyon, France
| | - Chloé Moreau
- Zoology, Department of Environmental Sciences, University of Basel, Basel, Switzerland
| | - Sabrina Fischer
- Zoology, Department of Environmental Sciences, University of Basel, Basel, Switzerland
| | - Antoine Fages
- Zoology, Department of Environmental Sciences, University of Basel, Basel, Switzerland
| | - Danilo Ritz
- Proteomics Core Facility, Biozentrum, University of Basel, Basel, Switzerland
| | - Patrick Tschopp
- Zoology, Department of Environmental Sciences, University of Basel, Basel, Switzerland.
| |
Collapse
|
19
|
Ahlmann-Eltze C, Huber W. Analysis of multi-condition single-cell data with latent embedding multivariate regression. Nat Genet 2025; 57:659-667. [PMID: 39753773 PMCID: PMC11906359 DOI: 10.1038/s41588-024-01996-0] [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: 03/20/2024] [Accepted: 10/18/2024] [Indexed: 03/15/2025]
Abstract
Identifying gene expression differences in heterogeneous tissues across conditions is a fundamental biological task, enabled by multi-condition single-cell RNA sequencing (RNA-seq). Current data analysis approaches divide the constituent cells into clusters meant to represent cell types, but such discrete categorization tends to be an unsatisfactory model of the underlying biology. Here, we introduce latent embedding multivariate regression (LEMUR), a model that operates without, or before, commitment to discrete categorization. LEMUR (1) integrates data from different conditions, (2) predicts each cell's gene expression changes as a function of the conditions and its position in latent space and (3) for each gene, identifies a compact neighborhood of cells with consistent differential expression. We apply LEMUR to cancer, zebrafish development and spatial gradients in Alzheimer's disease, demonstrating its broad applicability.
Collapse
Affiliation(s)
- Constantin Ahlmann-Eltze
- Genome Biology Unit, European Molecular Biology Laboratory (EMBL), Heidelberg, Germany.
- Faculty of Biosciences, Heidelberg University, Heidelberg, Germany.
| | - Wolfgang Huber
- Genome Biology Unit, European Molecular Biology Laboratory (EMBL), Heidelberg, Germany.
| |
Collapse
|
20
|
Dini A, Barker H, Piki E, Sharma S, Raivola J, Murumägi A, Ungureanu D. A multiplex single-cell RNA-Seq pharmacotranscriptomics pipeline for drug discovery. Nat Chem Biol 2025; 21:432-442. [PMID: 39482470 PMCID: PMC11867973 DOI: 10.1038/s41589-024-01761-8] [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: 08/23/2023] [Accepted: 09/22/2024] [Indexed: 11/03/2024]
Abstract
The gene-regulatory dynamics governing drug responses in cancer are yet to be fully understood. Here, we report a pipeline capable of producing high-throughput pharmacotranscriptomic profiling through live-cell barcoding using antibody-oligonucleotide conjugates. This pipeline combines drug screening with 96-plex single-cell RNA sequencing. We show the potential of this approach by exploring the heterogeneous transcriptional landscape of primary high-grade serous ovarian cancer (HGSOC) cells after treatment with 45 drugs, with 13 distinct classes of mechanisms of action. A subset of phosphatidylinositol 3-OH kinase (PI3K), protein kinase B (AKT) and mammalian target of rapamycin (mTOR) inhibitors induced the activation of receptor tyrosine kinases, such as the epithelial growth factor receptor (EGFR), and this was mediated by the upregulation of caveolin 1 (CAV1). This drug resistance feedback loop could be mitigated by the synergistic action of agents targeting PI3K-AKT-mTOR and EGFR for HGSOC with CAV1 and EGFR expression. Using this workflow could enable the personalized testing of patient-derived tumor samples at single-cell resolution.
Collapse
Affiliation(s)
- Alice Dini
- Disease Networks Unit, Faculty of Biochemistry and Molecular Medicine, University of Oulu, Oulu, Finland
| | - Harlan Barker
- Disease Networks Unit, Faculty of Biochemistry and Molecular Medicine, University of Oulu, Oulu, Finland
- Tampere University Hospital and Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| | - Emilia Piki
- Disease Networks Unit, Faculty of Biochemistry and Molecular Medicine, University of Oulu, Oulu, Finland
| | - Subodh Sharma
- Disease Networks Unit, Faculty of Biochemistry and Molecular Medicine, University of Oulu, Oulu, Finland
| | - Juuli Raivola
- Applied Tumor Genomics, Research Program Unit, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Astrid Murumägi
- Institute for Molecular Medicine Finland (FIMM), Helsinki Institute of Life Science (HiLIFE), University of Helsinki, Helsinki, Finland
| | - Daniela Ungureanu
- Disease Networks Unit, Faculty of Biochemistry and Molecular Medicine, University of Oulu, Oulu, Finland.
- Applied Tumor Genomics, Research Program Unit, Faculty of Medicine, University of Helsinki, Helsinki, Finland.
| |
Collapse
|
21
|
Lin Y, Zhang X, Sun D, Wang Q, Dou S, Zhou Q. Decoding the corneal immune microenvironment in healthy and diabetic mice during corneal wound healing. Ocul Surf 2025; 37:68-79. [PMID: 40023495 DOI: 10.1016/j.jtos.2025.02.010] [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: 02/24/2025] [Indexed: 03/04/2025]
Abstract
Diabetic keratopathy (DK) is an underdiagnosed ocular complication of diabetes mellitus. The changes of ocular immune microenvironment contribute to the pathogenesis of DK, while precise mechanisms remain inadequately understood. Here, we employed single-cell RNA sequencing (scRNA-seq) to elucidate the transcriptional alterations of immune cells from diabetic and healthy control mouse corneas during homeostasis and wound healing. Unbiased clustering analysis unveiled 3 major cell subsets and 11 subdivided cell clusters, including T cells, monocyte lineages, and neutrophil subpopulations. The further sub-clustering analysis demonstrated that T cells exhibited cytotoxicity characteristics in both homeostasis and wound healing of diabetic cornea. Moreover, dendritic cells preferred the migratory and maturation phenotype and may recruit and maintain cytotoxic T cells. Macrophages in diabetic cornea preferred the pro-inflammatory M1 phenotype. Under injury conditions, diabetic corneal neutrophils exhibited a more mature and functional possession of neutrophil extracellular traps (NETs). Furthermore, cell-cell communication revealed that the immune cells exhibited hyperactivation and pro-inflammatory responses, while the monocyte lineages exhibited the activating effect on T cells in diabetic cornea. This study represents the inaugural effort to establish a comprehensive scRNA-Seq transcriptomic profile of corneal immune cells during wound healing in healthy and diabetic mice, which offers a valuable reference for subsequent investigations into the pathological roles of immune cells in DK.
Collapse
Affiliation(s)
- Yujing Lin
- State Key Laboratory Cultivation Base, Shandong Provincial Key Laboratory of Ophthalmology, Eye Institute of Shandong First Medical University, Qingdao, China; Qingdao Eye Hospital of Shandong First Medical University, Qingdao, China
| | - Xiaowen Zhang
- State Key Laboratory Cultivation Base, Shandong Provincial Key Laboratory of Ophthalmology, Eye Institute of Shandong First Medical University, Qingdao, China; Qingdao Eye Hospital of Shandong First Medical University, Qingdao, China
| | - Di Sun
- State Key Laboratory Cultivation Base, Shandong Provincial Key Laboratory of Ophthalmology, Eye Institute of Shandong First Medical University, Qingdao, China; Qingdao Eye Hospital of Shandong First Medical University, Qingdao, China
| | - Qun Wang
- State Key Laboratory Cultivation Base, Shandong Provincial Key Laboratory of Ophthalmology, Eye Institute of Shandong First Medical University, Qingdao, China; Qingdao Eye Hospital of Shandong First Medical University, Qingdao, China
| | - Shengqian Dou
- State Key Laboratory Cultivation Base, Shandong Provincial Key Laboratory of Ophthalmology, Eye Institute of Shandong First Medical University, Qingdao, China; Qingdao Eye Hospital of Shandong First Medical University, Qingdao, China.
| | - Qingjun Zhou
- State Key Laboratory Cultivation Base, Shandong Provincial Key Laboratory of Ophthalmology, Eye Institute of Shandong First Medical University, Qingdao, China; Qingdao Eye Hospital of Shandong First Medical University, Qingdao, China.
| |
Collapse
|
22
|
Grandke F, Fehlmann T, Kern F, Gate DM, Wolff TW, Leventhal O, Channappa D, Hirsch P, Wilson EN, Meese E, Liu C, Shi Q, Flotho M, Li Y, Chen C, Yu Y, Xu J, Junkin M, Wang Z, Wu T, Liu L, Hou Y, Andreasson KI, Gansen JS, Mass E, Poston K, Wyss-Coray T, Keller A. A single-cell atlas to map sex-specific gene-expression changes in blood upon neurodegeneration. Nat Commun 2025; 16:1965. [PMID: 40000636 PMCID: PMC11862118 DOI: 10.1038/s41467-025-56833-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2024] [Accepted: 01/29/2025] [Indexed: 02/27/2025] Open
Abstract
The clinical course and treatment of neurodegenerative disease are complicated by immune-system interference and chronic inflammatory processes, which remain incompletely understood. Mapping immune signatures in larger human cohorts through single-cell gene expression profiling supports our understanding of observed peripheral changes in neurodegeneration. Here, we employ single-cell gene expression profiling of over 909k peripheral blood mononuclear cells (PBMCs) from 121 healthy individuals, 48 patients with mild cognitive impairment (MCI), 46 with Parkinson's disease (PD), 27 with Alzheimer's disease (AD), and 15 with both PD and MCI. The dataset is interactively accessible through a freely available website ( https://www.ccb.uni-saarland.de/adrcsc ). In this work, we identify disease-associated changes in blood cell type composition and the gene expression in a sex-specific manner, offering insights into peripheral and solid tissue signatures in AD and PD.
Collapse
Affiliation(s)
- Friederike Grandke
- Clinical Bioinformatics, Saarland University, 66123, Saarbrücken, Germany
| | - Tobias Fehlmann
- Clinical Bioinformatics, Saarland University, 66123, Saarbrücken, Germany
| | - Fabian Kern
- Clinical Bioinformatics, Saarland University, 66123, Saarbrücken, Germany
- Helmholtz Institute for Pharmaceutical Research Saarland (HIPS)-Helmholtz Centre for Infection Re- search (HZI), Saarland University Campus, Saarbrücken, Germany
| | - David M Gate
- Department of Neurology and Neurological Sciences, Stanford University, Stanford, CA, 94305, USA
- Veterans Administration Palo Alto Healthcare System, Palo Alto, CA, USA
- Wu Tsai Neurosciences Institute, Stanford University, Stanford, CA, USA
- Chemistry, Engineering, and Medicine for Human Health, Stanford University, Stanford, CA, USA
| | | | - Olivia Leventhal
- Department of Neurology and Neurological Sciences, Stanford University, Stanford, CA, 94305, USA
| | - Divya Channappa
- Department of Neurology and Neurological Sciences, Stanford University, Stanford, CA, 94305, USA
| | - Pascal Hirsch
- Clinical Bioinformatics, Saarland University, 66123, Saarbrücken, Germany
| | - Edward N Wilson
- Department of Neurology and Neurological Sciences, Stanford University, Stanford, CA, 94305, USA
| | - Eckart Meese
- Department of Human Genetics, Saarland University, 66421, Homburg/Saar, Germany
| | | | | | - Matthias Flotho
- Clinical Bioinformatics, Saarland University, 66123, Saarbrücken, Germany
| | - Yongping Li
- Clinical Bioinformatics, Saarland University, 66123, Saarbrücken, Germany
- MGI Group, San Jose, CA, USA
| | | | - Yeya Yu
- MGI Group, San Jose, CA, USA
| | | | | | | | - Tao Wu
- MGI Group, San Jose, CA, USA
| | | | | | - Katrin I Andreasson
- Department of Neurology and Neurological Sciences, Stanford University, Stanford, CA, 94305, USA
- Wu Tsai Neurosciences Institute, Stanford University, Stanford, CA, USA
- Program in Immunology, Stanford University, Stanford, CA, USA
| | - Jenny S Gansen
- Clinical Bioinformatics, Saarland University, 66123, Saarbrücken, Germany
| | - Elvira Mass
- Life and Medical Sciences Institute, Developmental Biology of the Immune System, University of Bonn, Bonn, Germany
| | - Kathleen Poston
- Department of Neurology and Neurological Sciences, Stanford University, Stanford, CA, 94305, USA
| | - Tony Wyss-Coray
- Department of Neurology and Neurological Sciences, Stanford University, Stanford, CA, 94305, USA.
- Veterans Administration Palo Alto Healthcare System, Palo Alto, CA, USA.
- Wu Tsai Neurosciences Institute, Stanford University, Stanford, CA, USA.
- Chemistry, Engineering, and Medicine for Human Health, Stanford University, Stanford, CA, USA.
- The Phil and Penny Knight Initiative for Brain Resilience, Stanford University, Stanford, CA, USA.
| | - Andreas Keller
- Clinical Bioinformatics, Saarland University, 66123, Saarbrücken, Germany.
- Helmholtz Institute for Pharmaceutical Research Saarland (HIPS)-Helmholtz Centre for Infection Re- search (HZI), Saarland University Campus, Saarbrücken, Germany.
- Department of Neurology and Neurological Sciences, Stanford University, Stanford, CA, 94305, USA.
- PharmaScienceHub, Saarland University Campus, Saarbrücken, Germany.
| |
Collapse
|
23
|
Qian J, Shao X, Bao H, Fang Y, Guo W, Li C, Li A, Hua H, Fan X. Identification and characterization of cell niches in tissue from spatial omics data at single-cell resolution. Nat Commun 2025; 16:1693. [PMID: 39956823 PMCID: PMC11830827 DOI: 10.1038/s41467-025-57029-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2024] [Accepted: 02/03/2025] [Indexed: 02/18/2025] Open
Abstract
Deciphering the features, structure, and functions of the cell niche in tissues remains a major challenge. Here, we present scNiche, a computational framework to identify and characterize cell niches from spatial omics data at single-cell resolution. We benchmark scNiche with both simulated and biological datasets, and demonstrate that scNiche can effectively and robustly identify cell niches while outperforming other existing methods. In spatial proteomics data from human triple-negative breast cancer, scNiche reveals the influence of the microenvironment on cellular phenotypes, and further dissects patient-specific niches with distinct cellular compositions or phenotypic characteristics. By analyzing mouse liver spatial transcriptomics data across normal and early-onset liver failure donors, scNiche uncovers disease-specific liver injury niches, and further delineates the niche remodeling from normal liver to liver failure. Overall, scNiche enables decoding the cellular microenvironment in tissues from single-cell spatial omics data.
Collapse
Affiliation(s)
- Jingyang Qian
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China
- State Key Laboratory of Chinese Medicine Modernization, Innovation Center of Yangtze River Delta, Zhejiang University, Jiaxing, 314102, China
| | - Xin Shao
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China.
- State Key Laboratory of Chinese Medicine Modernization, Innovation Center of Yangtze River Delta, Zhejiang University, Jiaxing, 314102, China.
- Zhejiang Key Laboratory of Chinese Medicine Modernization, Innovation Center of Yangtze River Delta, Zhejiang University, Jiaxing, 314100, China.
| | - Hudong Bao
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China
| | - Yin Fang
- College of Computer Science and Technology, Zhejiang University, Hangzhou, 310013, China
| | - Wenbo Guo
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China
- State Key Laboratory of Chinese Medicine Modernization, Innovation Center of Yangtze River Delta, Zhejiang University, Jiaxing, 314102, China
- Zhejiang Key Laboratory of Chinese Medicine Modernization, Innovation Center of Yangtze River Delta, Zhejiang University, Jiaxing, 314100, China
| | - Chengyu Li
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China
- State Key Laboratory of Chinese Medicine Modernization, Innovation Center of Yangtze River Delta, Zhejiang University, Jiaxing, 314102, China
| | - Anyao Li
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China
- State Key Laboratory of Chinese Medicine Modernization, Innovation Center of Yangtze River Delta, Zhejiang University, Jiaxing, 314102, China
| | - Hua Hua
- Translational Chinese Medicine Key Laboratory of Sichuan Province, SiChuan Institute for Translational Chinese Medicine, Chengdu, 610041, China.
| | - Xiaohui Fan
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China.
- State Key Laboratory of Chinese Medicine Modernization, Innovation Center of Yangtze River Delta, Zhejiang University, Jiaxing, 314102, China.
- Zhejiang Key Laboratory of Chinese Medicine Modernization, Innovation Center of Yangtze River Delta, Zhejiang University, Jiaxing, 314100, China.
- Zhejiang Key Laboratory of Precision Diagnosis and Therapy for Major Gynecological Diseases, Women's Hospital, Zhejiang University School of Medicine, 310006, Hangzhou, China.
| |
Collapse
|
24
|
Yang J, Agrawal K, Stanley J, Li R, Jacobs N, Wang H, Lu C, Qu R, Clarke D, Chen Y, Jiang Y, Bai D, Zheng S, Fox H, Ho YC, Huttner A, Gerstein M, Kluger Y, Zhang L, Spudich S. Multi-omic Characterization of HIV Effects at Single Cell Level across Human Brain Regions. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.02.05.636707. [PMID: 39975288 PMCID: PMC11839123 DOI: 10.1101/2025.02.05.636707] [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
HIV infection exerts profound and long-lasting neurodegenerative effects on the central nervous system (CNS) that can persist despite antiretroviral therapy (ART). Here, we used single-nucleus multiome sequencing to map the transcriptomic and epigenetic landscapes of postmortem human brains from 13 healthy individuals and 20 individuals with HIV who have a history of treatment with ART. Our study spanned three distinct regions-the prefrontal cortex, insular cortex, and ventral striatum-enabling a comprehensive exploration of region-specific and cross-regional perturbations. We found widespread and persistent HIV-associated transcriptional and epigenetic alterations across multiple cell types. Detailed analyses of microglia revealed state changes marked by immune activation and metabolic dysregulation, while integrative multiomic profiling of astrocytes identified multiple subpopulations, including a reactive subpopulation unique to HIV-infected brains. These findings suggest that cells from people with HIV exhibit molecular shifts that may underlie ongoing neuroinflammation and CNS dysfunction. Furthermore, cell-cell communication analyses uncovered dysregulated and pro-inflammatory interactions among glial populations, underscoring the multifaceted and enduring impact of HIV on the brain milieu. Collectively, our comprehensive atlas of HIV-associated brain changes reveals distinct glial cell states with signatures of proinflammatory signaling and metabolic dysregulation, providing a framework for developing targeted therapies for HIV-associated neurological dysfunction.
Collapse
Affiliation(s)
- Junchen Yang
- Interdepartmental Program in Computational Biology and Bioinformatics, Yale University, New Haven, CT, USA
| | - Kriti Agrawal
- Interdepartmental Program in Computational Biology and Bioinformatics, Yale University, New Haven, CT, USA
| | - Jay Stanley
- Program in Applied Mathematics, Yale University, New Haven, CT, USA
| | - Ruiqi Li
- Interdepartmental Program in Computational Biology and Bioinformatics, Yale University, New Haven, CT, USA
| | - Nicholas Jacobs
- Department of Pathology, Yale University, New Haven, CT, USA
| | - Haowei Wang
- Department of Neurology, Yale University, New Haven, CT, USA
- Department of Neuroscience, Yale University, New Haven, CT, USA
| | - Chang Lu
- Interdepartmental Program in Computational Biology and Bioinformatics, Yale University, New Haven, CT, USA
| | - Rihao Qu
- Interdepartmental Program in Computational Biology and Bioinformatics, Yale University, New Haven, CT, USA
| | - Declan Clarke
- Department of Molecular Biophysics and Biochemistry, Yale University, New Haven, CT, USA
| | - Yuhang Chen
- Interdepartmental Program in Computational Biology and Bioinformatics, Yale University, New Haven, CT, USA
| | - Yunzhe Jiang
- Interdepartmental Program in Computational Biology and Bioinformatics, Yale University, New Haven, CT, USA
| | - Donglu Bai
- Interdepartmental Program in Computational Biology and Bioinformatics, Yale University, New Haven, CT, USA
| | - Suchen Zheng
- Interdepartmental Program in Computational Biology and Bioinformatics, Yale University, New Haven, CT, USA
| | - Howard Fox
- Department of Neurological Sciences, University of Nebraska School of Medicine, Omaha, NB, USA
| | - Ya-chi Ho
- Department of Microbial Pathogenesis, Yale University, New Haven, CT, USA
| | - Anita Huttner
- Department of Pathology, Yale University, New Haven, CT, USA
| | - Mark Gerstein
- Interdepartmental Program in Computational Biology and Bioinformatics, Yale University, New Haven, CT, USA
- Department of Molecular Biophysics and Biochemistry, Yale University, New Haven, CT, USA
- Department of Computer Science, Yale University, New Haven, CT, USA
- Department of Statistics and Data Science, Yale University, New Haven, CT, USA
- Department of Biomedical Informatics & Data Science, Yale University, New Haven, CT, USA
| | - Yuval Kluger
- Interdepartmental Program in Computational Biology and Bioinformatics, Yale University, New Haven, CT, USA
- Program in Applied Mathematics, Yale University, New Haven, CT, USA
- Department of Pathology, Yale University, New Haven, CT, USA
| | - Le Zhang
- Department of Neurology, Yale University, New Haven, CT, USA
- Department of Neuroscience, Yale University, New Haven, CT, USA
| | - Serena Spudich
- Department of Neurology, Yale University, New Haven, CT, USA
- Center for Brain and Mind Health, Yale University, New Haven, CT, USA
| |
Collapse
|
25
|
Glass MR, Matoba N, Beltran AA, Patel NK, Farah TM, Eswar K, Bhargava S, Huang K, Curtin I, Ahmed S, Srivastava M, Drake E, Davis LT, Yeturi M, Sun K, Love MI, Simon JM, St John T, Marrus N, Pandey J, Estes A, Dager S, Schultz RT, Botteron K, Evans A, Kim SH, Styner M, McKinstry RC, Collins DL, Volk H, Benke K, Zwaigenbaum L, Hazlett H, Beltran AS, Girault JB, Shen MD, Piven J, Stein JL. Early cell cycle genes in cortical organoid progenitors predict interindividual variability in infant brain growth trajectories. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.02.07.637106. [PMID: 39974968 PMCID: PMC11839139 DOI: 10.1101/2025.02.07.637106] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/21/2025]
Abstract
Human induced pluripotent stem cell (iPSC) derived cortical organoids (hCOs) model neurogenesis on an individual's genetic background. The degree to which hCO phenotypes recapitulate the brain growth of the participants from which they were derived is not well established. We generated up to 3 iPSC clones from each of 18 participants in the Infant Brain Imaging Study, who have undergone longitudinal brain imaging during infancy. We identified consistent hCO morphology and cortical cell types across clones from the same participant. hCO cross-sectional area and production of cortical hem cells were associated with in vivo cortical growth rates. Cell cycle associated genes expression in early progenitors at the crux of fate decision trajectories were correlated with cortical growth rate from 6-12 months of age, and were enriched in microcephaly and neurodevelopmental disorder genes. Our data suggest the hCOs capture inter-individual variation in cortical cell types influencing infant cortical surface area expansion.
Collapse
Affiliation(s)
- Madison R Glass
- Department of Genetics, University of North Carolina at Chapel Hill; Chapel Hill, NC, USA
- UNC Neuroscience Center, University of North Carolina at Chapel Hill; Chapel Hill, NC, USA
- These authors contributed equally
| | - Nana Matoba
- Department of Genetics, University of North Carolina at Chapel Hill; Chapel Hill, NC, USA
- UNC Neuroscience Center, University of North Carolina at Chapel Hill; Chapel Hill, NC, USA
- These authors contributed equally
| | - Alvaro A Beltran
- Department of Genetics, University of North Carolina at Chapel Hill; Chapel Hill, NC, USA
- UNC Neuroscience Center, University of North Carolina at Chapel Hill; Chapel Hill, NC, USA
| | - Niyanta K Patel
- Department of Genetics, University of North Carolina at Chapel Hill; Chapel Hill, NC, USA
- UNC Neuroscience Center, University of North Carolina at Chapel Hill; Chapel Hill, NC, USA
| | - Tala M Farah
- Department of Genetics, University of North Carolina at Chapel Hill; Chapel Hill, NC, USA
- UNC Neuroscience Center, University of North Carolina at Chapel Hill; Chapel Hill, NC, USA
| | - Karthik Eswar
- Department of Genetics, University of North Carolina at Chapel Hill; Chapel Hill, NC, USA
- UNC Neuroscience Center, University of North Carolina at Chapel Hill; Chapel Hill, NC, USA
| | - Shivam Bhargava
- Department of Genetics, University of North Carolina at Chapel Hill; Chapel Hill, NC, USA
- UNC Neuroscience Center, University of North Carolina at Chapel Hill; Chapel Hill, NC, USA
| | - Karen Huang
- Department of Genetics, University of North Carolina at Chapel Hill; Chapel Hill, NC, USA
- UNC Neuroscience Center, University of North Carolina at Chapel Hill; Chapel Hill, NC, USA
| | - Ian Curtin
- Department of Genetics, University of North Carolina at Chapel Hill; Chapel Hill, NC, USA
- UNC Neuroscience Center, University of North Carolina at Chapel Hill; Chapel Hill, NC, USA
| | - Sara Ahmed
- Department of Genetics, University of North Carolina at Chapel Hill; Chapel Hill, NC, USA
- UNC Neuroscience Center, University of North Carolina at Chapel Hill; Chapel Hill, NC, USA
| | - Mary Srivastava
- Department of Genetics, University of North Carolina at Chapel Hill; Chapel Hill, NC, USA
- UNC Neuroscience Center, University of North Carolina at Chapel Hill; Chapel Hill, NC, USA
| | - Emma Drake
- Department of Genetics, University of North Carolina at Chapel Hill; Chapel Hill, NC, USA
- UNC Neuroscience Center, University of North Carolina at Chapel Hill; Chapel Hill, NC, USA
| | - Liam T Davis
- Department of Genetics, University of North Carolina at Chapel Hill; Chapel Hill, NC, USA
- UNC Neuroscience Center, University of North Carolina at Chapel Hill; Chapel Hill, NC, USA
| | - Meghana Yeturi
- Department of Genetics, University of North Carolina at Chapel Hill; Chapel Hill, NC, USA
- UNC Neuroscience Center, University of North Carolina at Chapel Hill; Chapel Hill, NC, USA
| | - Kexin Sun
- Department of Genetics, University of North Carolina at Chapel Hill; Chapel Hill, NC, USA
- UNC Neuroscience Center, University of North Carolina at Chapel Hill; Chapel Hill, NC, USA
| | - Michael I Love
- Department of Genetics, University of North Carolina at Chapel Hill; Chapel Hill, NC, USA
- Department of Biostatistics, University of North Carolina at Chapel Hill; Chapel Hill, NC, USA
| | - Jeremy M Simon
- Department of Genetics, University of North Carolina at Chapel Hill; Chapel Hill, NC, USA
- UNC Neuroscience Center, University of North Carolina at Chapel Hill; Chapel Hill, NC, USA
- Present address: Department of Data Science, Dana-Farber Cancer Institute, Boston, MA, USA
- Present address: Department of Biostatistics, Harvard T.H. Chan School of Public Health, MA, USA
| | - Tanya St John
- Department of Speech and Hearing Sciences, University of Washington; Seattle, WA, USA
- University of Washington Autism Center, University of Washington; Seattle, WA, USA
| | - Natasha Marrus
- Department of Psychiatry, Washington University School of Medicine; St. Louis, MO, USA
| | - Juhi Pandey
- Department of Psychiatry, University of Pennsylvania; Philadelphia, PA, USA
- Center for Autism Research, Children's Hospital of Philadelphia; Philadelphia, PA, USA
- Department of Pediatrics, University of Pennsylvania; Philadelphia, USA
| | - Annette Estes
- Department of Speech and Hearing Sciences, University of Washington; Seattle, WA, USA
- University of Washington Autism Center, University of Washington; Seattle, WA, USA
| | - Stephen Dager
- Department of Radiology, University of Washington; Seattle, WA, USA
- Department of Bioengineering, University of Washington; Seattle, WA, USA
| | - Robert T Schultz
- Department of Psychiatry, University of Pennsylvania; Philadelphia, PA, USA
- Center for Autism Research, Children's Hospital of Philadelphia; Philadelphia, PA, USA
- Department of Pediatrics, University of Pennsylvania; Philadelphia, USA
| | - Kelly Botteron
- Department of Psychiatry, Washington University School of Medicine; St. Louis, MO, USA
| | - Alan Evans
- Department of Neurology and Neurosurgery, McGill University; Montreal, QC, CA
- Department of Psychiatry, McGill University; Montreal, QC, CA
- Department of Biomedical Engineering, McGill University; Montreal, QC, CA
| | - Sun Hyung Kim
- Carolina Institute for Developmental Disabilities, University of North Carolina at Chapel Hill; Chapel Hill, NC, USA
| | - Martin Styner
- Carolina Institute for Developmental Disabilities, University of North Carolina at Chapel Hill; Chapel Hill, NC, USA
- Department of Psychiatry, University of North Carolina at Chapel Hill; Chapel Hill, NC, USA
- Department of Computer Sciences, University of North Carolina at Chapel Hill; Chapel Hill, NC, USA
| | - Robert C McKinstry
- Department of Psychiatry, Washington University School of Medicine; St. Louis, MO, USA
| | - D Louis Collins
- Department of Neurology and Neurosurgery, McGill University; Montreal, QC, CA
- Department of Biomedical Engineering, McGill University; Montreal, QC, CA
| | - Heather Volk
- School of Public Health, Johns Hopkins; Baltimore, MD, USA
| | - Kelly Benke
- School of Public Health, Johns Hopkins; Baltimore, MD, USA
| | - Lonnie Zwaigenbaum
- Department of Developmental Pediatrics, University of Alberta; Edmonton, AB, CA
| | - Heather Hazlett
- Carolina Institute for Developmental Disabilities, University of North Carolina at Chapel Hill; Chapel Hill, NC, USA
- Department of Psychiatry, University of North Carolina at Chapel Hill; Chapel Hill, NC, USA
| | - Adriana S Beltran
- Department of Genetics, University of North Carolina at Chapel Hill; Chapel Hill, NC, USA
| | - Jessica B Girault
- Carolina Institute for Developmental Disabilities, University of North Carolina at Chapel Hill; Chapel Hill, NC, USA
- Department of Psychiatry, University of North Carolina at Chapel Hill; Chapel Hill, NC, USA
- These authors jointly supervised
| | - Mark D Shen
- UNC Neuroscience Center, University of North Carolina at Chapel Hill; Chapel Hill, NC, USA
- Carolina Institute for Developmental Disabilities, University of North Carolina at Chapel Hill; Chapel Hill, NC, USA
- Department of Psychiatry, University of North Carolina at Chapel Hill; Chapel Hill, NC, USA
- These authors jointly supervised
| | - Joseph Piven
- Carolina Institute for Developmental Disabilities, University of North Carolina at Chapel Hill; Chapel Hill, NC, USA
- Department of Psychiatry, University of North Carolina at Chapel Hill; Chapel Hill, NC, USA
- These authors jointly supervised
| | - Jason L Stein
- Department of Genetics, University of North Carolina at Chapel Hill; Chapel Hill, NC, USA
- UNC Neuroscience Center, University of North Carolina at Chapel Hill; Chapel Hill, NC, USA
- Carolina Institute for Developmental Disabilities, University of North Carolina at Chapel Hill; Chapel Hill, NC, USA
- These authors jointly supervised
- Lead contact
| |
Collapse
|
26
|
Macnair W, Calini D, Agirre E, Bryois J, Jäkel S, Smith RS, Kukanja P, Stokar-Regenscheit N, Ott V, Foo LC, Collin L, Schippling S, Urich E, Nutma E, Marzin M, Ansaloni F, Amor S, Magliozzi R, Heidari E, Robinson MD, Ffrench-Constant C, Castelo-Branco G, Williams A, Malhotra D. snRNA-seq stratifies multiple sclerosis patients into distinct white matter glial responses. Neuron 2025; 113:396-410.e9. [PMID: 39708806 DOI: 10.1016/j.neuron.2024.11.016] [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: 11/16/2023] [Revised: 09/11/2024] [Accepted: 11/25/2024] [Indexed: 12/23/2024]
Abstract
Poor understanding of the cellular and molecular basis of clinical and genetic heterogeneity in progressive multiple sclerosis (MS) has hindered the search for new effective therapies. To address this gap, we analyzed 632,000 single-nucleus RNA sequencing profiles from 156 brain tissue samples of MS and control donors to examine inter- and intra-donor heterogeneity. We found distinct cell type-specific gene expression changes between MS gray and white matter, highlighting clear pathology differences. MS lesion subtypes had different cellular compositions but surprisingly similar cell-type gene expression patterns both within and across patients, suggesting global changes. Most gene expression variability was instead explained by patient effects, allowing us to stratify patients and describe the different pathological processes occurring between patient subgroups. Future mapping of these brain molecular profiles with blood and/or CSF profiles from living MS patients will allow precision medicine approaches anchored in patient-specific pathological processes.
Collapse
Affiliation(s)
- Will Macnair
- Roche Pharma Research and Early Development, Neuroscience and Rare Diseases, Roche Innovation Center, Basel, Switzerland.
| | - Daniela Calini
- Roche Pharma Research and Early Development, Neuroscience and Rare Diseases, Roche Innovation Center, Basel, Switzerland
| | - Eneritz Agirre
- Laboratory of Molecular Neurobiology, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, 17177 Stockholm, Sweden
| | - Julien Bryois
- Roche Pharma Research and Early Development, Neuroscience and Rare Diseases, Roche Innovation Center, Basel, Switzerland
| | - Sarah Jäkel
- Institute for Stroke and Dementia Research (ISD), Klinikum der Universität München, Ludwig-Maximilians Universität, Munich, Germany; Munich Cluster of Systems Neurology (SyNergy), Munich, Germany
| | - Rebecca Sherrard Smith
- Centre for Regenerative Medicine, Institute for Regeneration and Repair, MS Society Edinburgh Centre for MS Research, The University of Edinburgh, 5 Little France Drive, Edinburgh EH16 4UU, UK
| | - Petra Kukanja
- Laboratory of Molecular Neurobiology, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, 17177 Stockholm, Sweden
| | - Nadine Stokar-Regenscheit
- Roche Pharma Research and Early Development (pRED), Pharmaceutical Sciences, Pathology and Applied Safety Sciences, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd., Grenzacherstrasse 124, 4070 Basel, Switzerland
| | - Virginie Ott
- Roche Pharma Research and Early Development (pRED), Pharmaceutical Sciences, Pathology and Applied Safety Sciences, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd., Grenzacherstrasse 124, 4070 Basel, Switzerland
| | - Lynette C Foo
- Roche Pharma Research and Early Development, Neuroscience and Rare Diseases, Roche Innovation Center, Basel, Switzerland
| | - Ludovic Collin
- Roche Pharma Research and Early Development, Neuroscience and Rare Diseases, Roche Innovation Center, Basel, Switzerland
| | - Sven Schippling
- Roche Pharma Research and Early Development, Neuroscience and Rare Diseases, Roche Innovation Center, Basel, Switzerland
| | - Eduard Urich
- Roche Pharma Research and Early Development, Neuroscience and Rare Diseases, Roche Innovation Center, Basel, Switzerland
| | - Erik Nutma
- Department of Neurobiology and Aging, Biomedical Primate Research Centre, Rijswijk, the Netherlands
| | - Manuel Marzin
- Department of Neurobiology and Aging, Biomedical Primate Research Centre, Rijswijk, the Netherlands
| | - Federico Ansaloni
- Laboratory of Molecular Neurobiology, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, 17177 Stockholm, Sweden
| | - Sandra Amor
- Department of Neurobiology and Aging, Biomedical Primate Research Centre, Rijswijk, the Netherlands
| | - Roberta Magliozzi
- Department of Neurosciences, Biomedicine and Movement Sciences, University of Verona, Verona, Italy
| | - Elyas Heidari
- Department of Molecular Life Sciences and SIB Swiss Institute of Bioinformatics, University of Zurich, Winterthurerstrasse 190, 8057 Zurich, Switzerland
| | - Mark D Robinson
- Department of Molecular Life Sciences and SIB Swiss Institute of Bioinformatics, University of Zurich, Winterthurerstrasse 190, 8057 Zurich, Switzerland
| | - Charles Ffrench-Constant
- Faculty of Medicine and Health Sciences, University of East Anglia, Norwich Research Park, Norwich NR4 7TJ, UK.
| | - Gonçalo Castelo-Branco
- Laboratory of Molecular Neurobiology, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, 17177 Stockholm, Sweden.
| | - Anna Williams
- Centre for Regenerative Medicine, Institute for Regeneration and Repair, MS Society Edinburgh Centre for MS Research, The University of Edinburgh, 5 Little France Drive, Edinburgh EH16 4UU, UK.
| | - Dheeraj Malhotra
- Roche Pharma Research and Early Development, Neuroscience and Rare Diseases, Roche Innovation Center, Basel, Switzerland.
| |
Collapse
|
27
|
Nakatsuka N, Adler D, Jiang L, Hartman A, Cheng E, Klann E, Satija R. A Reproducibility Focused Meta-Analysis Method for Single-Cell Transcriptomic Case-Control Studies Uncovers Robust Differentially Expressed Genes. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2024.10.15.618577. [PMID: 39463993 PMCID: PMC11507907 DOI: 10.1101/2024.10.15.618577] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 10/29/2024]
Abstract
We assessed the reproducibility of differentially expressed genes (DEGs) in previously published Alzheimer's (AD), Parkinson's (PD), Schizophrenia (SCZ), and COVID-19 scRNA-seq studies. While transcriptional scores from DEGs of individual PD and COVID-19 datasets had moderate predictive power for case-control status of other datasets (AUC=0.77 and 0.75), genes from individual AD and SCZ datasets had poor predictive power (AUC=0.68 and 0.55). We developed a non-parametric meta-analysis method, SumRank, based on reproducibility of relative differential expression ranks across datasets, and found DEGs with improved predictive power (AUC=0.88, 0.91, 0.78, and 0.62). By multiple other metrics, specificity and sensitivity of these genes were substantially higher than those discovered by dataset merging and inverse variance weighted p-value aggregation methods. The DEGs revealed known and novel biological pathways, and we validate BCAT1 as down-regulated in AD mouse oligodendrocytes. Lastly, we evaluate factors influencing reproducibility of individual studies as a prospective guide for experimental design.
Collapse
|
28
|
Wu Y, Fan Y, Miao Y, Li Y, Du G, Chen Z, Diao J, Chen YA, Ye M, You R, Chen A, Chen Y, Li W, Guo W, Dong J, Zhang X, Wang Y, Gu J. uniLIVER: a human liver cell atlas for data-driven cellular state mapping. J Genet Genomics 2025:S1673-8527(25)00032-3. [PMID: 39892777 DOI: 10.1016/j.jgg.2025.01.017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2025] [Accepted: 01/22/2025] [Indexed: 02/04/2025]
Abstract
The liver performs several vital functions such as metabolism, toxin removal, and glucose storage through the coordination of various cell types. With the recent breakthrough of the single-cell/single-nucleus RNA-seq (sc/snRNA-seq) techniques, there is a great opportunity to establish a reference cell map of the liver at single-cell resolution with transcriptome-wise features. In this study, we build a unified liver cell atlas uniLIVER (http://lifeome.net/database/uniliver) by integrative analysis of a large-scale sc/snRNA-seq data collection of normal human liver with 331,125 cells and 79 samples from 6 datasets. Moreover, we introduce LiverCT, a novel machine learning based method for mapping any query dataset to the liver reference map by introducing the definition of "variant" cellular states analogy to the sequence variants in genomic analysis. Applying LiverCT on liver cancer datasets, we find that the "deviated" states of T cells are highly correlated with the stress pathway activities in hepatocellular carcinoma, and the enrichments of tumor cells with the hepatocyte-cholangiocyte "intermediate" states significantly indicate poor prognosis. Besides, we find that the tumor cells of different patients have different zonation tendencies and this zonation tendency is also significantly associated with the prognosis. This reference atlas mapping framework can also be extended to any other tissues.
Collapse
Affiliation(s)
- Yanhong Wu
- MOE Key Lab of Bioinformatics, BNRIST Bioinformatics Division, Department of Automation, Tsinghua University, Beijing 100084, China
| | - Yuhan Fan
- MOE Key Lab of Bioinformatics, BNRIST Bioinformatics Division, Department of Automation, Tsinghua University, Beijing 100084, China
| | - Yuxin Miao
- MOE Key Lab of Bioinformatics, BNRIST Bioinformatics Division, Department of Automation, Tsinghua University, Beijing 100084, China
| | - Yuman Li
- MOE Key Lab of Bioinformatics, BNRIST Bioinformatics Division, Department of Automation, Tsinghua University, Beijing 100084, China
| | - Guifang Du
- Hepato-Pancreato-Biliary Center, Beijing Tsinghua Changgung Hospital, Tsinghua University, Beijing 102218, China; Clinical Translational Science Center, Beijing Tsinghua Changgung Hospital, Tsinghua University, Beijing 102218, China
| | - Zeyu Chen
- MOE Key Lab of Bioinformatics, BNRIST Bioinformatics Division, Department of Automation, Tsinghua University, Beijing 100084, China
| | - Jinmei Diao
- Hepato-Pancreato-Biliary Center, Beijing Tsinghua Changgung Hospital, Tsinghua University, Beijing 102218, China; Clinical Translational Science Center, Beijing Tsinghua Changgung Hospital, Tsinghua University, Beijing 102218, China
| | - Yu-Ann Chen
- Hepato-Pancreato-Biliary Center, Beijing Tsinghua Changgung Hospital, Tsinghua University, Beijing 102218, China; Clinical Translational Science Center, Beijing Tsinghua Changgung Hospital, Tsinghua University, Beijing 102218, China
| | - Mingli Ye
- Fuzhou Institute of Data Technology, Fuzhou, Fujian 350207, China
| | - Renke You
- Fuzhou Institute of Data Technology, Fuzhou, Fujian 350207, China
| | - Amin Chen
- Fuzhou Institute of Data Technology, Fuzhou, Fujian 350207, China
| | - Yixin Chen
- MOE Key Lab of Bioinformatics, BNRIST Bioinformatics Division, Department of Automation, Tsinghua University, Beijing 100084, China
| | - Wenrui Li
- MOE Key Lab of Bioinformatics, BNRIST Bioinformatics Division, Department of Automation, Tsinghua University, Beijing 100084, China
| | - Wenbo Guo
- MOE Key Lab of Bioinformatics, BNRIST Bioinformatics Division, Department of Automation, Tsinghua University, Beijing 100084, China
| | - Jiahong Dong
- Hepato-Pancreato-Biliary Center, Beijing Tsinghua Changgung Hospital, Tsinghua University, Beijing 102218, China; Clinical Translational Science Center, Beijing Tsinghua Changgung Hospital, Tsinghua University, Beijing 102218, China
| | - Xuegong Zhang
- MOE Key Lab of Bioinformatics, BNRIST Bioinformatics Division, Department of Automation, Tsinghua University, Beijing 100084, China; Center for Synthetic and Systems Biology, School of Life Sciences and School of Medicine, Tsinghua University, Beijing 100084, China
| | - Yunfang Wang
- Hepato-Pancreato-Biliary Center, Beijing Tsinghua Changgung Hospital, Tsinghua University, Beijing 102218, China; Clinical Translational Science Center, Beijing Tsinghua Changgung Hospital, Tsinghua University, Beijing 102218, China.
| | - Jin Gu
- MOE Key Lab of Bioinformatics, BNRIST Bioinformatics Division, Department of Automation, Tsinghua University, Beijing 100084, China.
| |
Collapse
|
29
|
Shanmugam V, Tokcan N, Chafamo D, Sullivan S, Borji M, Martin H, Newton G, Nadaf N, Hanbury S, Barrera I, Cable D, Weir J, Ashenberg O, Pinkus G, Rodig S, Uhler C, Macosko E, Shipp M, Louissaint A, Chen F, Golub T. Genome-scale spatial mapping of the Hodgkin lymphoma microenvironment identifies tumor cell survival factors. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.01.24.631210. [PMID: 39896575 PMCID: PMC11785141 DOI: 10.1101/2025.01.24.631210] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 02/04/2025]
Abstract
A key challenge in cancer research is to identify the secreted factors that contribute to tumor cell survival. Nowhere is this more evident than in Hodgkin lymphoma, where malignant Hodgkin Reed Sternberg (HRS) cells comprise only 1-5% of the tumor mass, the remainder being infiltrating immune cells that presumably are required for the survival of the HRS cells. Until now, there has been no way to characterize the complex Hodgkin lymphoma tumor microenvironment at genome scale. Here, we performed genome-wide transcriptional profiling with spatial and single-cell resolution. We show that the neighborhood surrounding HRS cells forms a distinct niche involving 31 immune and stromal cell types and is enriched in CD4+ T cells, myeloid and follicular dendritic cells, while being depleted of plasma cells. Moreover, we used machine learning to nominate ligand-receptor pairs enriched in the HRS cell niche. Specifically, we identified IL13 as a candidate survival factor. In support of this hypothesis, recombinant IL13 augmented the proliferation of HRS cells in vitro. In addition, genome-wide CRISPR/Cas9 loss-of-function studies across more than 1,000 human cancer cell lines showed that IL4R and IL13RA1, the heterodimeric partners that constitute the IL13 receptor, were uniquely required for the survival of HRS cells. Moreover, monoclonal antibodies targeting either IL4R or IL13R phenocopied the genetic loss of function studies. IL13-targeting antibodies are already FDA-approved for atopic dermatitis, suggesting that clinical trials testing such agents should be explored in patients with Hodgkin lymphoma.
Collapse
Affiliation(s)
- Vignesh Shanmugam
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Department of Pathology, Brigham and Women's Hospital, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Neriman Tokcan
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Department of Mathematics, University of Massachusetts Boston, Boston, MA, USA
| | - Daniel Chafamo
- Klarman Cell Observatory, Broad Institute, Cambridge, MA, USA
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Sean Sullivan
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Department of Microbiology, University of Chicago, Chicago, IL, USA
| | - Mehdi Borji
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Haley Martin
- Department of Pathology, Massachusetts General Hospital, Boston, MA, USA
| | - Gail Newton
- Department of Pathology, Massachusetts General Hospital, Boston, MA, USA
| | - Naeem Nadaf
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | | | | | - Dylan Cable
- Department of Biostatistics, University of Michigan, Ann Arbor, MI, USA
| | - Jackson Weir
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Biological and Biomedical Sciences Program, Harvard University, Cambridge, MA, USA
| | - Orr Ashenberg
- Klarman Cell Observatory, Broad Institute, Cambridge, MA, USA
| | - Geraldine Pinkus
- Department of Pathology, Brigham and Women's Hospital, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Scott Rodig
- Department of Pathology, Brigham and Women's Hospital, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Caroline Uhler
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Laboratory for Information & Decision Systems, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Evan Macosko
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Department of Neurobiology, Harvard Medical School, Boston, MA, USA
- Department of Psychiatry, Massachusetts General Hospital, Boston, MA, USA
| | - Margaret Shipp
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Harvard Medical School, Boston, MA, USA
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Abner Louissaint
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Department of Pathology, Massachusetts General Hospital, Boston, MA, USA
| | - Fei Chen
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA, USA
| | - Todd Golub
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Harvard Medical School, Boston, MA, USA
- Department of Pediatric Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Division of Pediatric Hematology/Oncology, Boston Children's Hospital, Boston, MA, USA
| |
Collapse
|
30
|
Cheng J, Jin X, Smyth GK, Chen Y. Benchmarking cell type annotation methods for 10x Xenium spatial transcriptomics data. BMC Bioinformatics 2025; 26:22. [PMID: 39833693 PMCID: PMC11744978 DOI: 10.1186/s12859-025-06044-0] [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: 11/11/2024] [Accepted: 01/09/2025] [Indexed: 01/22/2025] Open
Abstract
BACKGROUND Imaging-based spatial transcriptomics technologies allow us to explore spatial gene expression profiles at the cellular level. Cell type annotation of imaging-based spatial data is challenging due to the small gene panel, but it is a crucial step for downstream analyses. Many good reference-based cell type annotation tools have been developed for single-cell RNA sequencing and sequencing-based spatial transcriptomics data. However, the performance of the reference-based cell type annotation tools on imaging-based spatial transcriptomics data has not been well studied yet. RESULTS We compared performance of five reference-based methods (SingleR, Azimuth, RCTD, scPred and scmapCell) with the marker-gene-based manual annotation method on an imaging-based Xenium data of human breast cancer. A practical workflow has been demonstrated for preparing a high-quality single-cell RNA reference, evaluating the accuracy, and estimating the running time for reference-based cell type annotation tools. CONCLUSIONS SingleR was the best performing reference-based cell type annotation tool for the Xenium platform, being fast, accurate and easy to use, with results closely matching those of manual annotation.
Collapse
Affiliation(s)
- Jinming Cheng
- Bioinformatics Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, 3052, Australia
- Department of Medical Biology, The University of Melbourne, Parkville, VIC, 3010, Australia
| | - Xinyi Jin
- Bioinformatics Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, 3052, Australia
- Department of Medical Biology, The University of Melbourne, Parkville, VIC, 3010, Australia
| | - Gordon K Smyth
- Bioinformatics Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, 3052, Australia.
- School of Mathematics and Statistics, The University of Melbourne, Parkville, VIC, 3010, Australia.
| | - Yunshun Chen
- Bioinformatics Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, 3052, Australia.
- Department of Medical Biology, The University of Melbourne, Parkville, VIC, 3010, Australia.
- ACRF Cancer Biology and Stem Cells Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, 3052, Australia.
| |
Collapse
|
31
|
Simmons SK, Adiconis X, Haywood N, Parker J, Lin Z, Liao Z, Tuncali I, Al’Khafaji AM, Shin A, Jagadeesh K, Gosik K, Gatzen M, Smith JT, El Kodsi DN, Kuras Y, Baecher-Allan C, Serrano GE, Beach TG, Garimella K, Rozenblatt-Rosen O, Regev A, Dong X, Scherzer CR, Levin JZ. Experimental and Computational Methods for Allelic Imbalance Analysis from Single-Nucleus RNA-seq Data. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2024.08.13.607784. [PMID: 39185246 PMCID: PMC11343128 DOI: 10.1101/2024.08.13.607784] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/27/2024]
Abstract
Single-cell RNA-seq (scRNA-seq) is emerging as a powerful tool for understanding gene function across diverse cells. Recently, this has included the use of allele-specific expression (ASE) analysis to better understand how variation in the human genome affects RNA expression at the single-cell level. We reasoned that because intronic reads are more prevalent in single-nucleus RNA-Seq (snRNA-Seq), and introns are under lower purifying selection and thus enriched for genetic variants, that snRNA-seq should facilitate single-cell analysis of ASE. Here we demonstrate how experimental and computational choices can improve the results of allelic imbalance analysis. We explore how experimental choices, such as RNA source, read length, sequencing depth, genotyping, etc., impact the power of ASE-based methods. We developed a new suite of computational tools to process and analyze scRNA-seq and snRNA-seq for ASE. As hypothesized, we extracted more ASE information from reads in intronic regions than those in exonic regions and show how read length can be set to increase power. Additionally, hybrid selection improved our power to detect allelic imbalance in genes of interest. We also explored methods to recover allele-specific isoform expression levels from both long- and short-read snRNA-seq. To further investigate ASE in the context of human disease, we applied our methods to a Parkinson's disease cohort of 94 individuals and show that ASE analysis had more power than eQTL analysis to identify significant SNP/gene pairs in our direct comparison of the two methods. Overall, we provide an end-to-end experimental and computational approach for future studies.
Collapse
Affiliation(s)
- Sean K. Simmons
- Aligning Science Across Parkinson’s (ASAP) Collaborative Research Network, Chevy Chase, MD 20815, USA
- Klarman Cell Observatory, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Xian Adiconis
- Aligning Science Across Parkinson’s (ASAP) Collaborative Research Network, Chevy Chase, MD 20815, USA
- Klarman Cell Observatory, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Nathan Haywood
- Aligning Science Across Parkinson’s (ASAP) Collaborative Research Network, Chevy Chase, MD 20815, USA
- Klarman Cell Observatory, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Jacob Parker
- Aligning Science Across Parkinson’s (ASAP) Collaborative Research Network, Chevy Chase, MD 20815, USA
- Stephen & Denise Adams Center for Parkinson’s Disease Research of Yale School of Medicine, New Haven, CT 06510, USA
| | - Zechuan Lin
- Aligning Science Across Parkinson’s (ASAP) Collaborative Research Network, Chevy Chase, MD 20815, USA
- Stephen & Denise Adams Center for Parkinson’s Disease Research of Yale School of Medicine, New Haven, CT 06510, USA
| | - Zhixiang Liao
- Aligning Science Across Parkinson’s (ASAP) Collaborative Research Network, Chevy Chase, MD 20815, USA
- Precision Neurology Program of Brigham & Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Idil Tuncali
- Aligning Science Across Parkinson’s (ASAP) Collaborative Research Network, Chevy Chase, MD 20815, USA
- Precision Neurology Program of Brigham & Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Aziz M. Al’Khafaji
- Broad Clinical Labs, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Asa Shin
- Broad Clinical Labs, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Karthik Jagadeesh
- Klarman Cell Observatory, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Kirk Gosik
- Klarman Cell Observatory, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Michael Gatzen
- Data Sciences Platform, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Jonathan T. Smith
- Data Sciences Platform, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Daniel N. El Kodsi
- Aligning Science Across Parkinson’s (ASAP) Collaborative Research Network, Chevy Chase, MD 20815, USA
- Precision Neurology Program of Brigham & Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Yuliya Kuras
- Aligning Science Across Parkinson’s (ASAP) Collaborative Research Network, Chevy Chase, MD 20815, USA
- Precision Neurology Program of Brigham & Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Clare Baecher-Allan
- Dept. of Neurology, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA 02115, USA
| | - Geidy E. Serrano
- Aligning Science Across Parkinson’s (ASAP) Collaborative Research Network, Chevy Chase, MD 20815, USA
- Banner Sun Health Research Institute, Sun City, AZ 85351, USA
| | - Thomas G. Beach
- Aligning Science Across Parkinson’s (ASAP) Collaborative Research Network, Chevy Chase, MD 20815, USA
- Banner Sun Health Research Institute, Sun City, AZ 85351, USA
| | - Kiran Garimella
- Data Sciences Platform, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Orit Rozenblatt-Rosen
- Klarman Cell Observatory, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
- Present address: Genentech, South San Francisco, CA 94080, USA
| | - Aviv Regev
- Klarman Cell Observatory, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
- Present address: Genentech, South San Francisco, CA 94080, USA
| | - Xianjun Dong
- Aligning Science Across Parkinson’s (ASAP) Collaborative Research Network, Chevy Chase, MD 20815, USA
- Stephen & Denise Adams Center for Parkinson’s Disease Research of Yale School of Medicine, New Haven, CT 06510, USA
| | - Clemens R. Scherzer
- Aligning Science Across Parkinson’s (ASAP) Collaborative Research Network, Chevy Chase, MD 20815, USA
- Stephen & Denise Adams Center for Parkinson’s Disease Research of Yale School of Medicine, New Haven, CT 06510, USA
| | - Joshua Z. Levin
- Aligning Science Across Parkinson’s (ASAP) Collaborative Research Network, Chevy Chase, MD 20815, USA
- Klarman Cell Observatory, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| |
Collapse
|
32
|
Chen Y, Chen L, Lun AL, Baldoni P, Smyth G. edgeR v4: powerful differential analysis of sequencing data with expanded functionality and improved support for small counts and larger datasets. Nucleic Acids Res 2025; 53:gkaf018. [PMID: 39844453 PMCID: PMC11754124 DOI: 10.1093/nar/gkaf018] [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: 01/17/2024] [Revised: 11/22/2024] [Accepted: 01/08/2025] [Indexed: 01/24/2025] Open
Abstract
edgeR is an R/Bioconductor software package for differential analyses of sequencing data in the form of read counts for genes or genomic features. Over the past 15 years, edgeR has been a popular choice for statistical analysis of data from sequencing technologies such as RNA-seq or ChIP-seq. edgeR pioneered the use of the negative binomial distribution to model read count data with replicates and the use of generalized linear models to analyze complex experimental designs. edgeR implements empirical Bayes moderation methods to allow reliable inference when the number of replicates is small. This article announces edgeR version 4, which includes new developments across a range of application areas. Infrastructure improvements include support for fractional counts, implementation of model fitting in C and a new statistical treatment of the quasi-likelihood pipeline that improves accuracy for small counts. The revised package has new functionality for differential methylation analysis, differential transcript expression, differential transcript and exon usage, testing relative to a fold-change threshold and pathway analysis. This article reviews the statistical framework and computational implementation of edgeR, briefly summarizing all the existing features and functionalities but with special attention to new features and those that have not been described previously.
Collapse
Affiliation(s)
- Yunshun Chen
- Bioinformatics Division, WEHI, Parkville, VIC 3052, Australia
- ACRF Cancer Biology and Stem Cells Division, WEHI, Parkville, VIC 3052, Australia
| | - Lizhong Chen
- Bioinformatics Division, WEHI, Parkville, VIC 3052, Australia
- Department of Medical Biology, The University of Melbourne, Parkville, VIC 3010, Australia
| | - Aaron T L Lun
- Computational Sciences, Genentech Inc, 1 DNA Way, South San Francisco, CA 94080, United States
| | - Pedro L Baldoni
- Bioinformatics Division, WEHI, Parkville, VIC 3052, Australia
- Department of Medical Biology, The University of Melbourne, Parkville, VIC 3010, Australia
| | - Gordon K Smyth
- Bioinformatics Division, WEHI, Parkville, VIC 3052, Australia
- School of Mathematics and Statistics, The University of Melbourne, Parkville, VIC 3010, Australia
| |
Collapse
|
33
|
Calarco JA, Taylor SR, Miller DM. Detecting gene expression in Caenorhabditis elegans. Genetics 2025; 229:1-108. [PMID: 39693264 PMCID: PMC11979774 DOI: 10.1093/genetics/iyae167] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2024] [Accepted: 09/30/2024] [Indexed: 12/20/2024] Open
Abstract
Reliable methods for detecting and analyzing gene expression are necessary tools for understanding development and investigating biological responses to genetic and environmental perturbation. With its fully sequenced genome, invariant cell lineage, transparent body, wiring diagram, detailed anatomy, and wide array of genetic tools, Caenorhabditis elegans is an exceptionally useful model organism for linking gene expression to cellular phenotypes. The development of new techniques in recent years has greatly expanded our ability to detect gene expression at high resolution. Here, we provide an overview of gene expression methods for C. elegans, including techniques for detecting transcripts and proteins in situ, bulk RNA sequencing of whole worms and specific tissues and cells, single-cell RNA sequencing, and high-throughput proteomics. We discuss important considerations for choosing among these techniques and provide an overview of publicly available online resources for gene expression data.
Collapse
Affiliation(s)
- John A Calarco
- Department of Cell and Systems Biology, University of Toronto, Toronto, ON, Canada, M5S 3G5
| | - Seth R Taylor
- Department of Cell Biology and Physiology, Brigham Young University, Provo, UT 84602, USA
| | - David M Miller
- Department of Cell and Developmental Biology, Vanderbilt University, Nashville, TN 37240, USA
- Neuroscience Program, Vanderbilt University, Nashville, TN 37240, USA
| |
Collapse
|
34
|
Gulati GS, D'Silva JP, Liu Y, Wang L, Newman AM. Profiling cell identity and tissue architecture with single-cell and spatial transcriptomics. Nat Rev Mol Cell Biol 2025; 26:11-31. [PMID: 39169166 DOI: 10.1038/s41580-024-00768-2] [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] [Accepted: 07/16/2024] [Indexed: 08/23/2024]
Abstract
Single-cell transcriptomics has broadened our understanding of cellular diversity and gene expression dynamics in healthy and diseased tissues. Recently, spatial transcriptomics has emerged as a tool to contextualize single cells in multicellular neighbourhoods and to identify spatially recurrent phenotypes, or ecotypes. These technologies have generated vast datasets with targeted-transcriptome and whole-transcriptome profiles of hundreds to millions of cells. Such data have provided new insights into developmental hierarchies, cellular plasticity and diverse tissue microenvironments, and spurred a burst of innovation in computational methods for single-cell analysis. In this Review, we discuss recent advancements, ongoing challenges and prospects in identifying and characterizing cell states and multicellular neighbourhoods. We discuss recent progress in sample processing, data integration, identification of subtle cell states, trajectory modelling, deconvolution and spatial analysis. Furthermore, we discuss the increasing application of deep learning, including foundation models, in analysing single-cell and spatial transcriptomics data. Finally, we discuss recent applications of these tools in the fields of stem cell biology, immunology, and tumour biology, and the future of single-cell and spatial transcriptomics in biological research and its translation to the clinic.
Collapse
Affiliation(s)
- Gunsagar S Gulati
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | | | - Yunhe Liu
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Linghua Wang
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
- The University of Texas MD Anderson Cancer Center UTHealth Houston Graduate School of Biomedical Sciences, Houston, TX, USA
| | - Aaron M Newman
- Department of Biomedical Data Science, Stanford University, Stanford, CA, USA.
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University, Stanford, CA, USA.
- Stanford Cancer Institute, Stanford University, Stanford, CA, USA.
- Chan Zuckerberg Biohub - San Francisco, San Francisco, CA, USA.
| |
Collapse
|
35
|
King EM, Zhao Y, Moore CM, Steinhart B, Anderson KC, Vestal B, Moore PK, McManus SA, Evans CM, Mould KJ, Redente EF, McCubbrey AL, Janssen WJ. Gpnmb and Spp1 mark a conserved macrophage injury response masking fibrosis-specific programming in the lung. JCI Insight 2024; 9:e182700. [PMID: 39509324 PMCID: PMC11665561 DOI: 10.1172/jci.insight.182700] [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: 05/31/2024] [Accepted: 10/30/2024] [Indexed: 11/15/2024] Open
Abstract
Macrophages are required for healthy repair of the lungs following injury, but they are also implicated in driving dysregulated repair with fibrosis. How these 2 distinct outcomes of lung injury are mediated by different macrophage subsets is unknown. To assess this, single-cell RNA-Seq was performed on lung macrophages isolated from mice treated with LPS or bleomycin. Macrophages were categorized based on anatomic location (airspace versus interstitium), developmental origin (embryonic versus recruited monocyte derived), time after inflammatory challenge, and injury model. Analysis of the integrated dataset revealed that macrophage subset clustering was driven by macrophage origin and tissue compartment rather than injury model. Gpnmb-expressing recruited macrophages that were enriched for genes typically associated with fibrosis were present in both injury models. Analogous GPNMB-expressing macrophages were identified in datasets from both fibrotic and nonfibrotic lung disease in humans. We conclude that this subset represents a conserved response to tissue injury and is not sufficient to drive fibrosis. Beyond this conserved response, we identified that recruited macrophages failed to gain resident-like programming during fibrotic repair. Overall, fibrotic versus nonfibrotic tissue repair is dictated by dynamic shifts in macrophage subset programming and persistence of recruited macrophages.
Collapse
Affiliation(s)
- Emily M. King
- Medical Scientist Training Program, University of Colorado School of Medicine, Aurora, Colorado, USA
| | - Yifan Zhao
- Center for Genes, Environment, and Health, and
| | | | | | | | | | - Peter K. Moore
- Department of Medicine, National Jewish Health, Denver, Colorado, USA
- Department of Medicine, University of Colorado School of Medicine, Aurora, Colorado, USA
| | | | - Christopher M. Evans
- Department of Medicine, University of Colorado School of Medicine, Aurora, Colorado, USA
| | - Kara J. Mould
- Department of Medicine, National Jewish Health, Denver, Colorado, USA
- Department of Medicine, University of Colorado School of Medicine, Aurora, Colorado, USA
| | - Elizabeth F. Redente
- Department of Medicine, University of Colorado School of Medicine, Aurora, Colorado, USA
- Department of Pediatrics, National Jewish Health, Denver, Colorado, USA
| | - Alexandra L. McCubbrey
- Department of Medicine, National Jewish Health, Denver, Colorado, USA
- Department of Medicine, University of Colorado School of Medicine, Aurora, Colorado, USA
| | - William J. Janssen
- Department of Medicine, National Jewish Health, Denver, Colorado, USA
- Department of Medicine, University of Colorado School of Medicine, Aurora, Colorado, USA
| |
Collapse
|
36
|
Frouard J, Telwatte S, Luo X, Elphick N, Thomas R, Arneson D, Roychoudhury P, Butte AJ, Wong JK, Hoh R, Deeks SG, Lee SA, Roan NR, Yukl S. HIV-SEQ REVEALS GLOBAL HOST GENE EXPRESSION DIFFERENCES BETWEEN HIV-TRANSCRIBING CELLS FROM VIREMIC AND SUPPRESSED PEOPLE WITH HIV. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.12.17.629023. [PMID: 39763963 PMCID: PMC11702770 DOI: 10.1101/2024.12.17.629023] [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: 01/18/2025]
Abstract
"Active" reservoir cells transcribing HIV can perpetuate chronic inflammation in virally suppressed people with HIV (PWH) and likely contribute to viral rebound after antiretroviral therapy (ART) interruption, so they represent an important target for new therapies. These cells, however, are difficult to study using single-cell RNA-seq (scRNA-seq) due to their low frequency and low levels of HIV transcripts, which are usually not polyadenylated. Here, we developed "HIV-seq" to enable more efficient capture of HIV transcripts - including non-polyadenylated ones - for scRNA-seq analysis of cells from PWH. By spiking in a set of custom-designed capture sequences targeting conserved regions of the HIV genome during scRNA-seq, we increased our ability to find HIV RNA+ cells from PWH by up to 44%. Implementing HIV-seq in conjunction with surface phenotyping by CITE-seq on paired blood specimens from PWH before vs. after ART suppression, we found that HIV RNA+ cells were enriched among T effector memory (Tem) cells during both viremia and ART suppression, but exhibited a cytotoxic signature during viremia only. By contrast, HIV RNA+ cells from the ART-suppressed timepoints exhibited a distinct anti-inflammatory signature involving elevated TGF-β and diminished IFN signaling. Overall, these findings demonstrate that active reservoir cells exhibit transcriptional features distinct from HIV RNA+ cells during viremia, and underscore HIV-seq as a useful tool to better understand the mechanisms by which HIV-transcribing cells can persist during ART.
Collapse
Affiliation(s)
- Julie Frouard
- Gladstone Institutes, San Francisco, CA 94158, USA
- Department of Urology, University of California, San Francisco, CA 94158, USA
| | - Sushama Telwatte
- San Francisco Veterans Affairs (VA) Medical Center and University of California, San Francisco, CA, USA
- Department of Infectious Diseases, The University of Melbourne at the Peter Doherty Institute of Infection and Immunity, Melbourne, Australia
| | - Xiaoyu Luo
- Gladstone Institutes, San Francisco, CA 94158, USA
- Department of Urology, University of California, San Francisco, CA 94158, USA
| | | | | | - Douglas Arneson
- Bakar Computational Health Sciences Institute, University of California, San Francisco, San Francisco, CA, USA
| | - Pavitra Roychoudhury
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, Washington, USA; Viral and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, USA
| | - Atul J Butte
- Bakar Computational Health Sciences Institute, University of California, San Francisco, San Francisco, CA, USA
| | - Joseph K Wong
- San Francisco Veterans Affairs (VA) Medical Center and University of California, San Francisco, CA, USA
| | - Rebecca Hoh
- Division of HIV, Infectious Diseases and Global Medicine, University of California, San Francisco, USA
| | - Steven G Deeks
- Division of HIV, Infectious Diseases and Global Medicine, University of California, San Francisco, USA
| | - Sulggi A Lee
- Zuckerberg San Francisco General Hospital and the University of California, San Francisco, USA
| | - Nadia R Roan
- Gladstone Institutes, San Francisco, CA 94158, USA
- Department of Urology, University of California, San Francisco, CA 94158, USA
| | - Steven Yukl
- San Francisco Veterans Affairs (VA) Medical Center and University of California, San Francisco, CA, USA
| |
Collapse
|
37
|
Lee D, Vicari JM, Porras C, Spencer C, Pjanic M, Wang X, Kinrot S, Weiler P, Kosoy R, Bendl J, Prashant NM, Psychogyiou K, Malakates P, Hennigan E, Monteiro Fortes J, Zheng S, Therrien K, Mathur D, Kleopoulos SP, Shao Z, Argyriou S, Alvia M, Casey C, Hong A, Beaumont KG, Sebra R, Kellner CP, Bennett DA, Yuan GC, Voloudakis G, Theis FJ, Haroutunian V, Hoffman GE, Fullard JF, Roussos P. Plasticity of Human Microglia and Brain Perivascular Macrophages in Aging and Alzheimer's Disease. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2023.10.25.23297558. [PMID: 39677435 PMCID: PMC11643149 DOI: 10.1101/2023.10.25.23297558] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 12/17/2024]
Abstract
The complex roles of myeloid cells, including microglia and perivascular macrophages, are central to the neurobiology of Alzheimer's disease (AD), yet they remain incompletely understood. Here, we profiled 832,505 human myeloid cells from the prefrontal cortex of 1,607 unique donors covering the human lifespan and varying degrees of AD neuropathology. We delineated 13 transcriptionally distinct myeloid subtypes organized into 6 subclasses and identified AD-associated adaptive changes in myeloid cells over aging and disease progression. The GPNMB subtype, linked to phagocytosis, increased significantly with AD burden and correlated with polygenic AD risk scores. By organizing AD-risk genes into a regulatory hierarchy, we identified and validated MITF as an upstream transcriptional activator of GPNMB, critical for maintaining phagocytosis. Through cell-to-cell interaction networks, we prioritized APOE-SORL1 and APOE-TREM2 ligand-receptor pairs, associated with AD progression. In both human and mouse models, TREM2 deficiency disrupted GPNMB expansion and reduced phagocytic function, suggesting that GPNMB's role in neuroprotection was TREM2-dependent. Our findings clarify myeloid subtypes implicated in aging and AD, advancing the mechanistic understanding of their role in AD and aiding therapeutic discovery.
Collapse
Affiliation(s)
- Donghoon Lee
- Center for Disease Neurogenomics, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - James M. Vicari
- Center for Disease Neurogenomics, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Christian Porras
- Center for Disease Neurogenomics, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Collin Spencer
- Center for Disease Neurogenomics, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Milos Pjanic
- Center for Disease Neurogenomics, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Xinyi Wang
- Center for Disease Neurogenomics, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Seon Kinrot
- Center for Disease Neurogenomics, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Philipp Weiler
- Institute of Computational Biology, Helmholtz Center Munich, Munich, Germany
- Department of Mathematics, Technical University of Munich, Munich, Germany
| | - Roman Kosoy
- Center for Disease Neurogenomics, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Jaroslav Bendl
- Center for Disease Neurogenomics, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - N M Prashant
- Center for Disease Neurogenomics, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Konstantina Psychogyiou
- Center for Disease Neurogenomics, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Periklis Malakates
- Center for Disease Neurogenomics, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Evelyn Hennigan
- Center for Disease Neurogenomics, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Jennifer Monteiro Fortes
- Center for Disease Neurogenomics, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Shiwei Zheng
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Black Family Stem Cell Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Karen Therrien
- Center for Disease Neurogenomics, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Deepika Mathur
- Center for Disease Neurogenomics, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Steven P. Kleopoulos
- Center for Disease Neurogenomics, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Zhiping Shao
- Center for Disease Neurogenomics, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Stathis Argyriou
- Center for Disease Neurogenomics, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Marcela Alvia
- Center for Disease Neurogenomics, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Clara Casey
- Center for Disease Neurogenomics, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Aram Hong
- Center for Disease Neurogenomics, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Kristin G. Beaumont
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Robert Sebra
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | | | - David A. Bennett
- Rush Alzheimer's Disease Center, Rush University Medical Center, Chicago, IL, USA
| | - Guo-Cheng Yuan
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Black Family Stem Cell Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - George Voloudakis
- Center for Disease Neurogenomics, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Fabian J. Theis
- Institute of Computational Biology, Helmholtz Center Munich, Munich, Germany
- Department of Mathematics, Technical University of Munich, Munich, Germany
- TUM School of Life Sciences Weihenstephan, Technical University of Munich, Munich, Germany
| | - Vahram Haroutunian
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Mental Illness Research, Education and Clinical Centers, James J. Peters VA Medical Center, Bronx, New York
| | - Gabriel E. Hoffman
- Center for Disease Neurogenomics, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Mental Illness Research, Education and Clinical Centers, James J. Peters VA Medical Center, Bronx, New York
| | - John F. Fullard
- Center for Disease Neurogenomics, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Panos Roussos
- Center for Disease Neurogenomics, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Mental Illness Research, Education and Clinical Centers, James J. Peters VA Medical Center, Bronx, New York
| |
Collapse
|
38
|
Andrieu T, Duo A, Duempelmann L, Patzak M, Saner FAM, Skrabalova J, Donato C, Nestorov P, Mueller MD. Single-Cell RNA Sequencing of PBMCs Identified Junction Plakoglobin (JUP) as Stratification Biomarker for Endometriosis. Int J Mol Sci 2024; 25:13071. [PMID: 39684780 DOI: 10.3390/ijms252313071] [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: 10/15/2024] [Revised: 11/29/2024] [Accepted: 11/29/2024] [Indexed: 12/18/2024] Open
Abstract
This study aimed to identify unique characteristics in the peripheral blood mononuclear cells (PBMCs) of endometriosis patients and develop a non-invasive early diagnostic tool. Using single-cell RNA sequencing (scRNA-seq), we constructed the first single-cell atlas of PBMCs from endometriosis patients based on 107,964 cells and 25,847 genes. Within CD16+ monocytes, we discovered JUP as a dysregulated gene. To assess its diagnostic potential, we measured peritoneal fluid (PF) and serum JUP levels in a large cohort of 199 patients including 20 women with ovarian cancer (OC). JUP was barely detectable in PF but was significantly elevated in the serum of patients with endometriosis and OC, with levels 1.33 and 2.34 times higher than controls, respectively. Additionally, JUP was found in conditioned culture media of CD14+/CD16+ monocytes aligning with our scRNA-seq data. Serum JUP levels correlated with endometriosis severity and endometrioma presence but were unaffected by dysmenorrhea, menstrual cycle, or adenomyosis. When combined with CA125 (cancer antigen 125) JUP enhanced the specificity of endometriosis diagnosis from 89.13% (CA125 measured alone) to 100%. While sensitivity remains a challenge at 19%, our results suggest that JUP's potential to enhance diagnostic accuracy warrants additional investigation. Furthermore, employing serum JUP as a stratification marker unlocked the potential to identify additional endometriosis-related genes, offering novel insights into disease pathogenesis.
Collapse
Affiliation(s)
- Thomas Andrieu
- Endometriosis & Gynaecological Oncology Laboratory (EndoGO), Department for Biomedical Research (DBMR), University of Bern, Murtenstrasse 35, 3008 Bern, Switzerland
- Inselspital Universitätsspital Bern, Women's Hospital-Universitätsklinik für Frauenheilkunde, Friedbühlstrasse 19, 3010 Bern, Switzerland
| | - Angelo Duo
- Scailyte AG, True Precision Medicine Through Single-Cell Science, Lichtstrasse 35, 4056 Basel, Switzerland
| | - Lea Duempelmann
- Endometriosis & Gynaecological Oncology Laboratory (EndoGO), Department for Biomedical Research (DBMR), University of Bern, Murtenstrasse 35, 3008 Bern, Switzerland
- Inselspital Universitätsspital Bern, Women's Hospital-Universitätsklinik für Frauenheilkunde, Friedbühlstrasse 19, 3010 Bern, Switzerland
| | - Magdalena Patzak
- Endometriosis & Gynaecological Oncology Laboratory (EndoGO), Department for Biomedical Research (DBMR), University of Bern, Murtenstrasse 35, 3008 Bern, Switzerland
- Inselspital Universitätsspital Bern, Women's Hospital-Universitätsklinik für Frauenheilkunde, Friedbühlstrasse 19, 3010 Bern, Switzerland
| | - Flurina Annacarina Maria Saner
- Endometriosis & Gynaecological Oncology Laboratory (EndoGO), Department for Biomedical Research (DBMR), University of Bern, Murtenstrasse 35, 3008 Bern, Switzerland
- Inselspital Universitätsspital Bern, Women's Hospital-Universitätsklinik für Frauenheilkunde, Friedbühlstrasse 19, 3010 Bern, Switzerland
| | - Jitka Skrabalova
- Endometriosis & Gynaecological Oncology Laboratory (EndoGO), Department for Biomedical Research (DBMR), University of Bern, Murtenstrasse 35, 3008 Bern, Switzerland
- Inselspital Universitätsspital Bern, Women's Hospital-Universitätsklinik für Frauenheilkunde, Friedbühlstrasse 19, 3010 Bern, Switzerland
| | - Cinzia Donato
- Scailyte AG, True Precision Medicine Through Single-Cell Science, Lichtstrasse 35, 4056 Basel, Switzerland
| | - Peter Nestorov
- Scailyte AG, True Precision Medicine Through Single-Cell Science, Lichtstrasse 35, 4056 Basel, Switzerland
| | - Michael D Mueller
- Endometriosis & Gynaecological Oncology Laboratory (EndoGO), Department for Biomedical Research (DBMR), University of Bern, Murtenstrasse 35, 3008 Bern, Switzerland
- Inselspital Universitätsspital Bern, Women's Hospital-Universitätsklinik für Frauenheilkunde, Friedbühlstrasse 19, 3010 Bern, Switzerland
| |
Collapse
|
39
|
Huo J, Wang Z, Zhao W, Chen M, Li H, He F, Tian X, Ma Y, Husanova F, Ma L, Ni Y, Ding H, Li W, Xu H. Investigating intra-tumoural heterogeneity and microenvironment diversity in primary cardiac angiosarcoma through single-cell RNA sequencing. Clin Transl Med 2024; 14:e70113. [PMID: 39658531 PMCID: PMC11631565 DOI: 10.1002/ctm2.70113] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2024] [Revised: 11/04/2024] [Accepted: 11/16/2024] [Indexed: 12/12/2024] Open
Abstract
BACKGROUND Primary cardiac angiosarcoma (PCAS) is a rare and aggressive heart tumour with limited treatment options and a poor prognosis. Understanding cellular heterogeneity and tumour microenvironment (TME) is crucial for the development of effective therapies. Here, we investigated the intratumoural heterogeneity and TME diversity of PCAS using single-cell RNA sequencing (scRNA-seq). METHODS We performed scRNA-seq analysis on tumour samples from four patients with PCAS, supplemented with multicolour immunohistochemistry for identification. We used scRNA-seq data from five normal cardiac tissue samples downloaded from public databases for comparative analyses. Bioinformatic analyses, including Cell Ranger, Seurat, Monocle2, hdWGCNA, SCENIC and NicheNet, were utilized to identify distinct cell populations, transcriptional patterns, and co-regulating gene modules. RESULTS Our analysis revealed significant intratumoural heterogeneity in PCAS driven by diverse biological processes such as protein synthesis, degradation, and RIG-I signalling inhibition. The SCENIC analysis identified three primary transcription factors' clusters (CEBPB, MYC and TAL1). T-cell subset analysis showed exhausted antigen-specific T-cells, complicating the efficacy of immune checkpoint blockade. Furthermore, we observed suppressive macrophages (SPP1+ and OLR1+) and reduced mitochondrial gene MT-RNR2 (MTRNR2L12) expression in TME-infiltrating cells, indicating impaired mitochondrial function. CONCLUSION This study elucidates the complex cellular landscape and immune microenvironment of PCAS, highlighting potential molecular targets for the development of novel therapies. These findings underscore the importance of a multifaceted therapeutic approach for addressing the challenges posed by PCAS's heterogeneity and immune evasion. KEY POINTS Insights into the heterogeneity and transcriptional patterns of sarcoma cells may explain the challenges in treating primary cardiac angiosarcoma (PCAS) using the current therapeutic modalities. Characterization of the immune microenvironment revealed significant immunosuppression mediated by specific myeloid cell populations (SPP1+ and OLR1+ macrophages). Identification of mitochondrial dysfunction in immune cells within the PCAS microenvironment, particularly the notable downregulation of the MTRNR2L12 protein, suggests a new avenue for therapeutic targeting.
Collapse
Affiliation(s)
- Jingyuan Huo
- Department of Cardiovascular SurgerySchool of Medicinethe First Affiliated Hospital of Zhejiang UniversityHangzhouChina
| | - Zhen Wang
- Department of Cardiovascular SurgerySchool of Medicinethe First Affiliated Hospital of Zhejiang UniversityHangzhouChina
| | - Wenting Zhao
- Department of CardiologySchool of Medicinethe First Affiliated Hospital of Zhejiang UniversityHangzhouChina
| | - Miao Chen
- Department of Cardiovascular SurgerySchool of Medicinethe First Affiliated Hospital of Zhejiang UniversityHangzhouChina
| | - Haoyang Li
- Department of Cardiovascular SurgerySchool of Medicinethe First Affiliated Hospital of Zhejiang UniversityHangzhouChina
| | - Fengpu He
- Department of Cardiovascular SurgerySchool of Medicinethe First Affiliated Hospital of Zhejiang UniversityHangzhouChina
| | - Xiao Tian
- Department of Cardiovascular SurgerySchool of Medicinethe First Affiliated Hospital of Zhejiang UniversityHangzhouChina
| | - Yaqi Ma
- Department of PathologySchool of Medicinethe First Affiliated Hospital of Zhejiang UniversityHangzhouChina
| | - Firyuza Husanova
- Department of Cardiovascular SurgerySchool of Medicinethe First Affiliated Hospital of Zhejiang UniversityHangzhouChina
| | - Liang Ma
- Department of Cardiovascular SurgerySchool of Medicinethe First Affiliated Hospital of Zhejiang UniversityHangzhouChina
| | - Yiming Ni
- Department of Cardiovascular SurgerySchool of Medicinethe First Affiliated Hospital of Zhejiang UniversityHangzhouChina
| | - Hongda Ding
- Department of General SurgeryShengjing Hospital of China Medical UniversityShenyangChina
| | - Weidong Li
- Department of Cardiovascular SurgerySchool of Medicinethe First Affiliated Hospital of Zhejiang UniversityHangzhouChina
| | - Hongfei Xu
- Department of Cardiovascular SurgerySchool of Medicinethe First Affiliated Hospital of Zhejiang UniversityHangzhouChina
| |
Collapse
|
40
|
Yermalovich AV, Mohsenin Z, Cowdin M, Giotti B, Gupta A, Feng A, Golomb L, Wheeler DB, Xu K, Tsankov A, Cleaver O, Meyerson M. An essential role for Cmtr2 in mammalian embryonic development. Dev Biol 2024; 516:47-58. [PMID: 39094818 DOI: 10.1016/j.ydbio.2024.07.019] [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: 07/14/2023] [Revised: 07/26/2024] [Accepted: 07/30/2024] [Indexed: 08/04/2024]
Abstract
CMTR2 is an mRNA cap methyltransferase with poorly understood physiological functions. It catalyzes 2'-O-ribose methylation of the second transcribed nucleotide of mRNAs, potentially serving to mark RNAs as "self" to evade the cellular innate immune response. Here we analyze the consequences of Cmtr2 deficiency in mice. We discover that constitutive deletion of Cmtr2 results in mouse embryos that die during mid-gestation, exhibiting defects in embryo size, placental malformation and yolk sac vascularization. Endothelial cell deletion of Cmtr2 in mice results in vascular and hematopoietic defects, and perinatal lethality. Detailed characterization of the constitutive Cmtr2 KO phenotype shows an activation of the p53 pathway and decreased proliferation, but no evidence of interferon pathway activation. In summary, our study reveals the essential roles of Cmtr2 in mammalian cells beyond its immunoregulatory function.
Collapse
Affiliation(s)
- Alena V Yermalovich
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, 02215, USA; Broad Institute of Harvard and MIT, Cambridge, MA, 02142, USA
| | - Zarin Mohsenin
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, 02215, USA; Broad Institute of Harvard and MIT, Cambridge, MA, 02142, USA
| | - Mitzy Cowdin
- Department of Molecular Biology, Center for Regenerative Science and Medicine, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Bruno Giotti
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Akansha Gupta
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, 02215, USA; Broad Institute of Harvard and MIT, Cambridge, MA, 02142, USA
| | - Alice Feng
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, 02215, USA; Broad Institute of Harvard and MIT, Cambridge, MA, 02142, USA
| | - Lior Golomb
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, 02215, USA; Broad Institute of Harvard and MIT, Cambridge, MA, 02142, USA
| | - Douglas B Wheeler
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, 02215, USA; Broad Institute of Harvard and MIT, Cambridge, MA, 02142, USA
| | - Kelly Xu
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, 02215, USA; Broad Institute of Harvard and MIT, Cambridge, MA, 02142, USA
| | - Alexander Tsankov
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Ondine Cleaver
- Department of Molecular Biology, Center for Regenerative Science and Medicine, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Matthew Meyerson
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, 02215, USA; Broad Institute of Harvard and MIT, Cambridge, MA, 02142, USA; Center for Cancer Genomics, Dana-Farber Cancer Institute, Boston, MA, 02215, USA; Departments of Genetics and Medicine, Harvard Medical School, Boston, MA, 02115, USA.
| |
Collapse
|
41
|
Zhang H, Fan K, Zhang Z, Guo Y, Mo X. Genome-wide identification of cell type-specific susceptibility genes for Juvenile dermatomyositis through the analysis of N 6-methyladenosine-associated SNPs. Autoimmunity 2024; 57:2419117. [PMID: 39447013 DOI: 10.1080/08916934.2024.2419117] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2024] [Revised: 08/12/2024] [Accepted: 10/15/2024] [Indexed: 10/26/2024]
Abstract
Genome-wide association studies (GWASs) have pinpointed genetic loci associated with juvenile dermatomyositis (JDM). Functional genes within the GWAS loci may be cell type-specific, but their identity remains largely unknown. N6-methyladenosine (m6A) plays a pivotal role in regulating various cellular processes and is linked to autoimmune diseases. This study aimed to underscore the potential functional genes within the GWAS loci through the analysis of m6A-associated SNPs (m6A-SNPs), specifically within relevant cell types. JDM-associated m6A-SNPs were identified from the GWAS summary dataset. The correlation between m6A-SNPs and gene expression was assessed through bulk tissue and single-cell eQTL analyses. To further investigate the relationship between gene expression and JDM, Mendelian randomization analysis was employed. Additionally, differential expression analyses were conducted on bulk tissues, as well as single-cell transcriptomic data comprising 6 JDM patients and 11 juvenile controls (99,396 cells). Seven m6A-SNPs associated with JDM were identified. Bulk tissue analysis revealed differential expression of HLA-DPA1, HLA-DPB1, MICB, HLA-A, HLA-F, HLA-DQB2, HLA-DRB5, TAP2, PSMB9, MICA, AIF1, and DDX39B influenced by m6A-SNPs, all showing associations with JDM in both differential expression and Mendelian randomization analyses. In single-cell analysis, the six m6A-SNPs within the HLA locus acted as cell-type-specific eQTLs, correlating with the expression of HLA-A, HLA-B, HLA-C, HLA-DPB1, HLA-DQA1, HLA-DQB1 and HLA-DRB1 in myeloid, T or B cells. Notably, these genes displayed abnormal expression in T, B, and myeloid cells of JDM patients. The present study identified m6A-SNPs within JDM susceptibility genes, shedding light on the intricate interplay between m6A-SNPs, gene expression, and JDM.
Collapse
Affiliation(s)
- Huan Zhang
- Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, Department of Epidemiology, School of Public Health, Suzhou Medical College of Soochow University, China
| | - Kedi Fan
- Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, Department of Epidemiology, School of Public Health, Suzhou Medical College of Soochow University, China
- Center for Genetic Epidemiology and Genomics, School of Public Health, Suzhou Medical College of Soochow University, China
| | - Zhentao Zhang
- Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, Department of Epidemiology, School of Public Health, Suzhou Medical College of Soochow University, China
- Center for Genetic Epidemiology and Genomics, School of Public Health, Suzhou Medical College of Soochow University, China
| | - Yufan Guo
- Department of Rheumatology, the First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, People's Republic of China
| | - Xingbo Mo
- Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, Department of Epidemiology, School of Public Health, Suzhou Medical College of Soochow University, China
- Center for Genetic Epidemiology and Genomics, School of Public Health, Suzhou Medical College of Soochow University, China
| |
Collapse
|
42
|
Nahman O, Few-Cooper TJ, Shen-Orr SS. Cell-specific priors rescue differential gene expression in spatial spot-based technologies. Brief Bioinform 2024; 26:bbae621. [PMID: 39679437 DOI: 10.1093/bib/bbae621] [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: 04/17/2024] [Revised: 10/18/2024] [Accepted: 11/14/2024] [Indexed: 12/17/2024] Open
Abstract
Spatial transcriptomics (ST), a breakthrough technology, captures the complex structure and state of tissues through the spatial profiling of gene expression. A variety of ST technologies have now emerged, most prominently spot-based platforms such as Visium. Despite the widespread use of ST and its distinct data characteristics, the vast majority of studies continue to analyze ST data using algorithms originally designed for older technologies such as single-cell (SC) and bulk RNA-seq-particularly when identifying differentially expressed genes (DEGs). However, it remains unclear whether these algorithms are still valid or appropriate for ST data. Therefore, here, we sought to characterize the performance of these methods by constructing an in silico simulator of ST data with a controllable and known DEG ground truth. Surprisingly, our findings reveal little variation in the performance of classic DEG algorithms-all of which fail to accurately recapture known DEGs to significant levels. We further demonstrate that cellular heterogeneity within spots is a primary cause of this poor performance and propose a simple gene-selection scheme, based on prior knowledge of cell-type specificity, to overcome this. Notably, our approach outperforms existing data-driven methods designed specifically for ST data and offers improved DEG recovery and reliability rates. In summary, our work details a conceptual framework that can be used upstream, agnostically, of any DEG algorithm to improve the accuracy of ST analysis and any downstream findings.
Collapse
Affiliation(s)
- Ornit Nahman
- Department of Immunology, Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, 1 Efron St., Haifa, 3525433, Israel
| | - Timothy J Few-Cooper
- Department of Immunology, Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, 1 Efron St., Haifa, 3525433, Israel
| | - Shai S Shen-Orr
- Department of Immunology, Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, 1 Efron St., Haifa, 3525433, Israel
| |
Collapse
|
43
|
Hoffman GE, Lee D, Bendl J, Prashant N, Hong A, Casey C, Alvia M, Shao Z, Argyriou S, Therrien K, Venkatesh S, Voloudakis G, Haroutunian V, Fullard JF, Roussos P. Efficient differential expression analysis of large-scale single cell transcriptomics data using dreamlet. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.03.17.533005. [PMID: 36993704 PMCID: PMC10055252 DOI: 10.1101/2023.03.17.533005] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 04/13/2023]
Abstract
Advances in single-cell and -nucleus transcriptomics have enabled generation of increasingly large-scale datasets from hundreds of subjects and millions of cells. These studies promise to give unprecedented insight into the cell type specific biology of human disease. Yet performing differential expression analyses across subjects remains difficult due to challenges in statistical modeling of these complex studies and scaling analyses to large datasets. Our open-source R package dreamlet (DiseaseNeurogenomics.github.io/dreamlet) uses a pseudobulk approach based on precision-weighted linear mixed models to identify genes differentially expressed with traits across subjects for each cell cluster. Designed for data from large cohorts, dreamlet is substantially faster and uses less memory than existing workflows, while supporting complex statistical models and controlling the false positive rate. We demonstrate computational and statistical performance on published datasets, and a novel dataset of 1.4M single nuclei from postmortem brains of 150 Alzheimer's disease cases and 149 controls.
Collapse
Affiliation(s)
- Gabriel E. Hoffman
- Center for Disease Neurogenomics
- Department of Psychiatry
- Department of Genetics and Genomic Sciences
- Center for Precision Medicine and Translational Therapeutics, Mental Illness Research, Education and Clinical Centers, James J. Peters VA Medical Center, Bronx, New York
| | - Donghoon Lee
- Center for Disease Neurogenomics
- Department of Psychiatry
- Department of Genetics and Genomic Sciences
- Friedman Brain Institute
| | - Jaroslav Bendl
- Center for Disease Neurogenomics
- Department of Psychiatry
- Department of Genetics and Genomic Sciences
- Friedman Brain Institute
| | - N.M. Prashant
- Center for Disease Neurogenomics
- Department of Psychiatry
- Department of Genetics and Genomic Sciences
- Friedman Brain Institute
| | - Aram Hong
- Center for Disease Neurogenomics
- Department of Psychiatry
- Department of Genetics and Genomic Sciences
- Friedman Brain Institute
| | - Clara Casey
- Center for Disease Neurogenomics
- Department of Psychiatry
- Department of Genetics and Genomic Sciences
- Friedman Brain Institute
| | - Marcela Alvia
- Center for Disease Neurogenomics
- Department of Psychiatry
- Department of Genetics and Genomic Sciences
- Friedman Brain Institute
| | - Zhiping Shao
- Center for Disease Neurogenomics
- Department of Psychiatry
- Department of Genetics and Genomic Sciences
- Friedman Brain Institute
| | - Stathis Argyriou
- Center for Disease Neurogenomics
- Department of Psychiatry
- Department of Genetics and Genomic Sciences
- Friedman Brain Institute
| | - Karen Therrien
- Center for Disease Neurogenomics
- Department of Psychiatry
- Department of Genetics and Genomic Sciences
- Friedman Brain Institute
| | - Sanan Venkatesh
- Center for Disease Neurogenomics
- Department of Psychiatry
- Department of Genetics and Genomic Sciences
- Friedman Brain Institute
| | - Georgios Voloudakis
- Center for Disease Neurogenomics
- Department of Psychiatry
- Department of Genetics and Genomic Sciences
- Friedman Brain Institute
- Center for Precision Medicine and Translational Therapeutics, Mental Illness Research, Education and Clinical Centers, James J. Peters VA Medical Center, Bronx, New York
| | - Vahram Haroutunian
- Department of Psychiatry
- Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Center for Precision Medicine and Translational Therapeutics, Mental Illness Research, Education and Clinical Centers, James J. Peters VA Medical Center, Bronx, New York
| | - John F. Fullard
- Center for Disease Neurogenomics
- Department of Psychiatry
- Department of Genetics and Genomic Sciences
- Friedman Brain Institute
| | - Panos Roussos
- Center for Disease Neurogenomics
- Department of Psychiatry
- Department of Genetics and Genomic Sciences
- Friedman Brain Institute
- Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Center for Precision Medicine and Translational Therapeutics, Mental Illness Research, Education and Clinical Centers, James J. Peters VA Medical Center, Bronx, New York
| |
Collapse
|
44
|
Shan X, Zhao H. Inferring Cell-Type-Specific Co-Expressed Genes from Single Cell Data. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.11.08.622700. [PMID: 39605403 PMCID: PMC11601408 DOI: 10.1101/2024.11.08.622700] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/29/2024]
Abstract
Background Cell-type-specific gene co-expression networks are widely used to characterize gene relationships. Although many methods have been developed to infer such co-expression networks from single-cell data, the lack of consideration of false positive control in many evaluations may lead to incorrect conclusions because higher reproducibility, higher functional coherence, and a larger overlap with known biological networks may not imply better performance if the false positives are not well controlled. Results In this study, we have developed an efficient and effective simulation tool to derive empirical p-values in co-expression inference to appropriately control false positives in assessing method performance. We studied the power of the p-value-based approach in inferring cell-type-specific co-expressions from single-cell data using both simulated and real data. We also highlight the need to adjust for random overlaps between the inferred and known networks when the number of selected correlated gene pairs varies substantially across different methods. We further illustrate the expression level bias in known biological networks and the impact of such bias in method assessment. Conclusion Our study indicates the importance of controlling false positives in the inference of co-expressed genes to achieve more reliable results and proposes a simulation-based p-value method to achieve this.
Collapse
|
45
|
Ross JL, Puigdelloses-Vallcorba M, Piñero G, Soni N, Thomason W, DeSisto J, Angione A, Tsankova NM, Castro MG, Schniederjan M, Wadhwani NR, Raju GP, Morgenstern P, Becher OJ, Green AL, Tsankov AM, Hambardzumyan D. Microglia and monocyte-derived macrophages drive progression of pediatric high-grade gliomas and are transcriptionally shaped by histone mutations. Immunity 2024; 57:2669-2687.e6. [PMID: 39395421 PMCID: PMC11578068 DOI: 10.1016/j.immuni.2024.09.007] [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: 10/06/2023] [Revised: 06/04/2024] [Accepted: 09/11/2024] [Indexed: 10/14/2024]
Abstract
Pediatric high-grade gliomas (pHGGs), including hemispheric pHGGs and diffuse midline gliomas (DMGs), harbor mutually exclusive tumor location-specific histone mutations. Using immunocompetent de novo mouse models of pHGGs, we demonstrated that myeloid cells were the predominant infiltrating non-neoplastic cell population. Single-cell RNA sequencing (scRNA-seq), flow cytometry, and immunohistochemistry illustrated the presence of heterogeneous myeloid cell populations shaped by histone mutations and tumor location. Disease-associated myeloid (DAM) cell phenotypes demonstrating immune permissive characteristics were identified in murine and human pHGG samples. H3.3K27M DMGs, the most aggressive DMG, demonstrated enrichment of DAMs. Genetic ablation of chemokines Ccl8 and Ccl12 resulted in a reduction of DAMs and an increase in lymphocyte infiltration, leading to increased survival of tumor-bearing mice. Pharmacologic inhibition of chemokine receptors CCR1 and CCR5 resulted in extended survival and decreased myeloid cell infiltration. This work establishes the tumor-promoting role of myeloid cells in DMG and the potential therapeutic opportunities for targeting them.
Collapse
Affiliation(s)
- James L Ross
- Department of Microbiology and Immunology, Emory Vaccine Center, Emory University School of Medicine, Atlanta, GA, USA
| | - Montserrat Puigdelloses-Vallcorba
- Department of Oncological Sciences, The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Department of Neurosurgery, Mount Sinai Icahn School of Medicine, New York, NY, USA
| | - Gonzalo Piñero
- Department of Oncological Sciences, The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Department of Neurosurgery, Mount Sinai Icahn School of Medicine, New York, NY, USA
| | - Nishant Soni
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Wes Thomason
- Department of Oncological Sciences, The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Department of Neurosurgery, Mount Sinai Icahn School of Medicine, New York, NY, USA
| | - John DeSisto
- Morgan Adams Foundation Pediatric Brain Tumor Research Program, University of Colorado Anschutz Medical Campus, Aurora, CO, USA; Cell Biology, Stem Cells and Development Graduate Program, Aurora, CO, USA; Center for Cancer and Blood Disorders, Children's Hospital Colorado, Aurora, CO, USA
| | - Angelo Angione
- Department of Oncological Sciences, The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Department of Neurosurgery, Mount Sinai Icahn School of Medicine, New York, NY, USA
| | - Nadejda M Tsankova
- Department of Pathology and Molecular and Cell-Based Medicine, Mount Sinai Icahn School of Medicine, New York, NY 10029, USA
| | - Maria G Castro
- Departments of Neurosurgery and Cell & Developmental Biology, The University of Michigan School of Medicine, Ann Arbor, MI, USA
| | - Matthew Schniederjan
- Department of Pathology and Laboratory Medicine, Emory University, Atlanta, GA, USA
| | - Nitin R Wadhwani
- Department of Pathology and Laboratory Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - G Praveen Raju
- Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Department of Pediatrics, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Peter Morgenstern
- Department of Neurosurgery, Mount Sinai Icahn School of Medicine, New York, NY, USA
| | - Oren J Becher
- Department of Oncological Sciences, The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Department of Pediatrics, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Adam L Green
- Morgan Adams Foundation Pediatric Brain Tumor Research Program, University of Colorado Anschutz Medical Campus, Aurora, CO, USA; Cell Biology, Stem Cells and Development Graduate Program, Aurora, CO, USA; Center for Cancer and Blood Disorders, Children's Hospital Colorado, Aurora, CO, USA
| | - Alexander M Tsankov
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Dolores Hambardzumyan
- Department of Oncological Sciences, The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Department of Neurosurgery, Mount Sinai Icahn School of Medicine, New York, NY, USA.
| |
Collapse
|
46
|
Zhang H, Zhang Z, Fan K, Chen Y, Xu P, Guo Y, Mo X. Deciphering cell-specific genetic insights: Unraveling the immunogenetic landscape of systemic lupus erythematosus. Mol Immunol 2024; 175:165-175. [PMID: 39476438 DOI: 10.1016/j.molimm.2024.10.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2024] [Revised: 10/13/2024] [Accepted: 10/24/2024] [Indexed: 11/11/2024]
Abstract
Functional genes within genomic loci associated with systemic lupus erythematosus (SLE), as identified by genome-wide association studies, exhibit cell-specific characteristics. This study delves into the impact of genetic variants within SLE loci on gene expression in different types of immune cells, unraveling the complex interplay between genetics and immunopathogenesis. Through the integration of genetic association and single-cell transcriptomic sequencing data, we identified potential cell-specific susceptibility genes for SLE across diverse immune cell subsets. The single-cell eQTL analysis revealed 30,409 associations involving 3583 SLE-associated SNPs. These SNPs exhibited associations with expression levels of 147 genes across 14 distinct cell types. The single-cell summary data-based Mendelian randomization (SMR) analysis identified 119 significant associations between the expression levels of 44 genes and SLE. Notably, myeloid cells exhibited associations solely within the MHC region, while T, B, and natural killer cells showed associations with both MHC and non-MHC genes in relation to SLE. Analysis of single-cell transcriptomic data from 33 children SLE cases and 11 match controls (227,303 cells), as well as 7 adult SLE cases and 5 match controls (78,414 cells) highlights differential expression of key genes. Notably, genetic variants within HLA-DRB1, HLA-DRB5, HLA-DQA1, HLA-DQB1, IRF7, IRF5, BLK and HLA-DPA1 play a pivotal role in mediating immune dysregulation in specific immune cell types. Our study contributes to a comprehensive understanding of the intricate relationships between genetics, gene expression and SLE susceptibility. The findings shed light on the cell-specific impacts of genetic variants within SLE-associated genomic loci.
Collapse
Affiliation(s)
- Huan Zhang
- Department of Epidemiology, School of Public Health, Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, MOE Key Laboratory of Geriatric Diseases and Immunology, Suzhou Medical College of Soochow University, China
| | - Zhentao Zhang
- Department of Epidemiology, School of Public Health, Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, MOE Key Laboratory of Geriatric Diseases and Immunology, Suzhou Medical College of Soochow University, China; Center for Genetic Epidemiology and Genomics, School of Public Health, Suzhou Medical College of Soochow University, China
| | - Kedi Fan
- Department of Epidemiology, School of Public Health, Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, MOE Key Laboratory of Geriatric Diseases and Immunology, Suzhou Medical College of Soochow University, China; Center for Genetic Epidemiology and Genomics, School of Public Health, Suzhou Medical College of Soochow University, China
| | - Yuxi Chen
- Department of Epidemiology, School of Public Health, Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, MOE Key Laboratory of Geriatric Diseases and Immunology, Suzhou Medical College of Soochow University, China
| | - Peng Xu
- Department of Epidemiology, School of Public Health, Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, MOE Key Laboratory of Geriatric Diseases and Immunology, Suzhou Medical College of Soochow University, China
| | - Yufan Guo
- Department of Rheumatology, the First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215000, China.
| | - Xingbo Mo
- Department of Epidemiology, School of Public Health, Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, MOE Key Laboratory of Geriatric Diseases and Immunology, Suzhou Medical College of Soochow University, China; Center for Genetic Epidemiology and Genomics, School of Public Health, Suzhou Medical College of Soochow University, China.
| |
Collapse
|
47
|
Yang Y, Lu X, Liu N, Ma S, Zhang H, Zhang Z, Yang K, Jiang M, Zheng Z, Qiao Y, Hu Q, Huang Y, Zhang Y, Xiong M, Liu L, Jiang X, Reddy P, Dong X, Xu F, Wang Q, Zhao Q, Lei J, Sun S, Jing Y, Li J, Cai Y, Fan Y, Yan K, Jing Y, Haghani A, Xing M, Zhang X, Zhu G, Song W, Horvath S, Rodriguez Esteban C, Song M, Wang S, Zhao G, Li W, Izpisua Belmonte JC, Qu J, Zhang W, Liu GH. Metformin decelerates aging clock in male monkeys. Cell 2024; 187:6358-6378.e29. [PMID: 39270656 DOI: 10.1016/j.cell.2024.08.021] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2024] [Revised: 07/10/2024] [Accepted: 08/12/2024] [Indexed: 09/15/2024]
Abstract
In a rigorous 40-month study, we evaluated the geroprotective effects of metformin on adult male cynomolgus monkeys, addressing a gap in primate aging research. The study encompassed a comprehensive suite of physiological, imaging, histological, and molecular evaluations, substantiating metformin's influence on delaying age-related phenotypes at the organismal level. Specifically, we leveraged pan-tissue transcriptomics, DNA methylomics, plasma proteomics, and metabolomics to develop innovative monkey aging clocks and applied these to gauge metformin's effects on aging. The results highlighted a significant slowing of aging indicators, notably a roughly 6-year regression in brain aging. Metformin exerts a substantial neuroprotective effect, preserving brain structure and enhancing cognitive ability. The geroprotective effects on primate neurons were partially mediated by the activation of Nrf2, a transcription factor with anti-oxidative capabilities. Our research pioneers the systemic reduction of multi-dimensional biological age in primates through metformin, paving the way for advancing pharmaceutical strategies against human aging.
Collapse
Affiliation(s)
- Yuanhan Yang
- Key Laboratory of Organ Regeneration and Reconstruction, State Key Laboratory of Membrane Biology, State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Xiaoyong Lu
- China National Center for Bioinformation, Beijing, China; Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Ning Liu
- State Key Laboratory of Brain and Cognitive Science, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China; College of Life Sciences, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Shuai Ma
- Key Laboratory of Organ Regeneration and Reconstruction, State Key Laboratory of Membrane Biology, State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China; Institute for Stem Cell and Regeneration, CAS, Beijing 100101, China; Beijing Institute for Stem Cell and Regenerative Medicine, Beijing 100101, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Hui Zhang
- Key Laboratory of Organ Regeneration and Reconstruction, State Key Laboratory of Membrane Biology, State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China; University of Chinese Academy of Sciences, Beijing 100049, China; Beijing Institute of Heart, Lung and Blood Vessel Diseases, Beijing Anzhen Hospital, Capital Medical University, Beijing 100029, China
| | - Zhiyi Zhang
- State Key Laboratory of Brain and Cognitive Science, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China
| | - Kuan Yang
- China National Center for Bioinformation, Beijing, China; Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Mengmeng Jiang
- Key Laboratory of Organ Regeneration and Reconstruction, State Key Laboratory of Membrane Biology, State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China; Institute for Stem Cell and Regeneration, CAS, Beijing 100101, China; Beijing Institute for Stem Cell and Regenerative Medicine, Beijing 100101, China
| | - Zikai Zheng
- China National Center for Bioinformation, Beijing, China; Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Yicheng Qiao
- State Key Laboratory of Brain and Cognitive Science, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China; College of Life Sciences, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Qinchao Hu
- Key Laboratory of Organ Regeneration and Reconstruction, State Key Laboratory of Membrane Biology, State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China; Hospital of Stomatology, Guanghua School of Stomatology, Guangdong Provincial Key Laboratory of Stomatology, Sun Yat-Sen University, Guangzhou 510060, China
| | - Ying Huang
- Chongqing Fifth People's Hospital, Chongqing 400060, China
| | - Yiyuan Zhang
- Key Laboratory of Organ Regeneration and Reconstruction, State Key Laboratory of Membrane Biology, State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China; Institute for Stem Cell and Regeneration, CAS, Beijing 100101, China; Beijing Institute for Stem Cell and Regenerative Medicine, Beijing 100101, China
| | - Muzhao Xiong
- China National Center for Bioinformation, Beijing, China; Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Lixiao Liu
- China National Center for Bioinformation, Beijing, China; Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Xiaoyu Jiang
- Key Laboratory of Organ Regeneration and Reconstruction, State Key Laboratory of Membrane Biology, State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Pradeep Reddy
- Altos Labs San Diego Institute of Science, San Diego, CA, USA
| | - Xueda Dong
- State Key Laboratory of Brain and Cognitive Science, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China; Sino-Danish College, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Fanshu Xu
- State Key Laboratory of Brain and Cognitive Science, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China; College of Life Sciences, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Qiaoran Wang
- China National Center for Bioinformation, Beijing, China; Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Qian Zhao
- National Clinical Research Center for Geriatric Disorders, Aging Translational Medicine Center, International Center for Aging and Cancer, Xuanwu Hospital Capital Medical University, Beijing 100053, China
| | - Jinghui Lei
- National Clinical Research Center for Geriatric Disorders, Aging Translational Medicine Center, International Center for Aging and Cancer, Xuanwu Hospital Capital Medical University, Beijing 100053, China
| | - Shuhui Sun
- Beijing Institute of Heart, Lung and Blood Vessel Diseases, Beijing Anzhen Hospital, Capital Medical University, Beijing 100029, China
| | - Ying Jing
- National Clinical Research Center for Geriatric Disorders, Aging Translational Medicine Center, International Center for Aging and Cancer, Xuanwu Hospital Capital Medical University, Beijing 100053, China
| | - Jingyi Li
- Key Laboratory of Organ Regeneration and Reconstruction, State Key Laboratory of Membrane Biology, State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China; Institute for Stem Cell and Regeneration, CAS, Beijing 100101, China; Beijing Institute for Stem Cell and Regenerative Medicine, Beijing 100101, China; Aging Biomarker Consortium (ABC), Beijing 100101, China
| | - Yusheng Cai
- Key Laboratory of Organ Regeneration and Reconstruction, State Key Laboratory of Membrane Biology, State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China; Institute for Stem Cell and Regeneration, CAS, Beijing 100101, China; Beijing Institute for Stem Cell and Regenerative Medicine, Beijing 100101, China
| | - Yanling Fan
- China National Center for Bioinformation, Beijing, China; Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing, China
| | - Kaowen Yan
- Key Laboratory of Organ Regeneration and Reconstruction, State Key Laboratory of Membrane Biology, State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China; Institute for Stem Cell and Regeneration, CAS, Beijing 100101, China; Beijing Institute for Stem Cell and Regenerative Medicine, Beijing 100101, China
| | - Yaobin Jing
- Key Laboratory of Organ Regeneration and Reconstruction, State Key Laboratory of Membrane Biology, State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China; Institute for Stem Cell and Regeneration, CAS, Beijing 100101, China; Beijing Institute for Stem Cell and Regenerative Medicine, Beijing 100101, China; International Center for Aging and Cancer, Hainan Medical University, Haikou 571199, China
| | - Amin Haghani
- Altos Labs San Diego Institute of Science, San Diego, CA, USA
| | - Mengen Xing
- Oujiang Laboratory, Center for Geriatric Medicine and Institute of Aging, Key Laboratory of Alzheimer's Disease of Zhejiang Province, Zhejiang Provincial Clinical Research for Mental Disorders, The First-Affiliated Hospital, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Xuan Zhang
- Department of Rheumatology, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Clinical Immunology Center, Chinese Academy of Medical Sciences, Beijing 100730, China
| | - Guodong Zhu
- Institute of Gerontology, Guangzhou Geriatric Hospital, Guangzhou Medical University, Guangzhou, China
| | - Weihong Song
- Oujiang Laboratory, Center for Geriatric Medicine and Institute of Aging, Key Laboratory of Alzheimer's Disease of Zhejiang Province, Zhejiang Provincial Clinical Research for Mental Disorders, The First-Affiliated Hospital, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China
| | - Steve Horvath
- Altos Labs San Diego Institute of Science, San Diego, CA, USA
| | | | - Moshi Song
- Key Laboratory of Organ Regeneration and Reconstruction, State Key Laboratory of Membrane Biology, State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China; Beijing Institute for Stem Cell and Regenerative Medicine, Beijing 100101, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Si Wang
- National Clinical Research Center for Geriatric Disorders, Aging Translational Medicine Center, International Center for Aging and Cancer, Xuanwu Hospital Capital Medical University, Beijing 100053, China; Aging Biomarker Consortium (ABC), Beijing 100101, China
| | - Guoguang Zhao
- Department of Neurosurgery, Xuanwu Hospital Capital Medical University, Beijing 100053, China; National Medical Center for Neurological Diseases, Beijing 100053, China; Beijing Municipal Geriatric Medical Research Center, Beijing 100053, China
| | - Wei Li
- Key Laboratory of Organ Regeneration and Reconstruction, State Key Laboratory of Membrane Biology, State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China; Institute for Stem Cell and Regeneration, CAS, Beijing 100101, China; Beijing Institute for Stem Cell and Regenerative Medicine, Beijing 100101, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | | | - Jing Qu
- Key Laboratory of Organ Regeneration and Reconstruction, State Key Laboratory of Membrane Biology, State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China; Institute for Stem Cell and Regeneration, CAS, Beijing 100101, China; Beijing Institute for Stem Cell and Regenerative Medicine, Beijing 100101, China; University of Chinese Academy of Sciences, Beijing 100049, China; Beijing Institute of Heart, Lung and Blood Vessel Diseases, Beijing Anzhen Hospital, Capital Medical University, Beijing 100029, China; Aging Biomarker Consortium (ABC), Beijing 100101, China.
| | - Weiqi Zhang
- China National Center for Bioinformation, Beijing, China; Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing, China; Institute for Stem Cell and Regeneration, CAS, Beijing 100101, China; Beijing Institute for Stem Cell and Regenerative Medicine, Beijing 100101, China; University of Chinese Academy of Sciences, Beijing 100049, China; Aging Biomarker Consortium (ABC), Beijing 100101, China.
| | - Guang-Hui Liu
- Key Laboratory of Organ Regeneration and Reconstruction, State Key Laboratory of Membrane Biology, State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China; Institute for Stem Cell and Regeneration, CAS, Beijing 100101, China; Beijing Institute for Stem Cell and Regenerative Medicine, Beijing 100101, China; National Clinical Research Center for Geriatric Disorders, Aging Translational Medicine Center, International Center for Aging and Cancer, Xuanwu Hospital Capital Medical University, Beijing 100053, China; University of Chinese Academy of Sciences, Beijing 100049, China; Aging Biomarker Consortium (ABC), Beijing 100101, China.
| |
Collapse
|
48
|
Zhang C, Wu Q, Yang H, Zhang H, Liu C, Yang B, Hu Q. Ferroptosis-related gene signature for predicting prognosis and identifying potential therapeutic drug in EGFR wild-type lung adenocarcinoma. Commun Biol 2024; 7:1416. [PMID: 39478024 PMCID: PMC11525656 DOI: 10.1038/s42003-024-07117-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2024] [Accepted: 10/22/2024] [Indexed: 11/02/2024] Open
Abstract
Epidermal growth factor receptor wild type lung adenocarcinoma (EGFRWT LUAD) still has limited treatment options and unsatisfactory clinical outcomes. Ferroptosis, as a form of cell death, has been reported to play a dual role in regulating tumor cell survival. In this study, we constructed a 3-ferroptosis-gene signature, FeSig, and verified its accuracy and efficacy in predicting EGFRWT LUAD prognosis at both the RNA and protein levels. Patients with higher FeSig scores were found to have worse clinical outcomes. Additionally, we explored the relationship between FeSig and tumor microenvironment, revealing that enhanced interactions between fibroblasts and tumor cells in FeSighigh patients causing tumor resistance to ferroptosis. To address this challenge, we screened potential drugs from NCI-60 (The US National Cancer Institute 60 human tumour cell line anticancer drug screen) and Connectivity map database, ultimately identifying 6-mercatopurine (6-MP) as a promising candidate. Both in vitro and in vivo experiments demonstrated its efficacy in treating FeSighigh EGFRWT LUAD tumor models. In summary, we develop a novel FeSig for predicting prognosis and guiding drug application.
Collapse
Affiliation(s)
- Chuankai Zhang
- Department of Thoracic Surgery, The First Affiliated Hospital of University of Science and Technology of China (USTC), Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China.
| | - Qi Wu
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of USTC, Center for Advanced Interdisciplinary Science and Biomedicine of IHM, Division of Life Sciences and Medicine, University of Science and Technology of China, 230001, Hefei, China
| | - Hongwei Yang
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of USTC, Center for Advanced Interdisciplinary Science and Biomedicine of IHM, Division of Life Sciences and Medicine, University of Science and Technology of China, 230001, Hefei, China
| | - Hui Zhang
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of USTC, Center for Advanced Interdisciplinary Science and Biomedicine of IHM, Division of Life Sciences and Medicine, University of Science and Technology of China, 230001, Hefei, China
| | - Changqing Liu
- Department of Thoracic Surgery, The First Affiliated Hospital of University of Science and Technology of China (USTC), Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Bo Yang
- The First Affiliated Hospital of University of Science and Technology of China (USTC), Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China.
| | - Qingsong Hu
- Department of Thoracic Surgery, The First Affiliated Hospital of University of Science and Technology of China (USTC), Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China.
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of USTC, Center for Advanced Interdisciplinary Science and Biomedicine of IHM, Division of Life Sciences and Medicine, University of Science and Technology of China, 230001, Hefei, China.
| |
Collapse
|
49
|
Zhong T, Li X, Lei K, Tang R, Deng Q, Love PE, Zhou Z, Zhao B, Li X. TGF-β-mediated crosstalk between TIGIT + Tregs and CD226 +CD8 + T cells in the progression and remission of type 1 diabetes. Nat Commun 2024; 15:8894. [PMID: 39406740 PMCID: PMC11480485 DOI: 10.1038/s41467-024-53264-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2023] [Accepted: 10/08/2024] [Indexed: 10/19/2024] Open
Abstract
Type 1 diabetes (T1D) is a chronic autoimmune condition characterized by hyperglycemia resulting from the destruction of insulin-producing β-cells that is traditionally deemed irreversible, but partial remission (PR) with temporary reversal of hyperglycemia is sometimes observed. Here we use single-cell RNA sequencing to delineate the immune cell landscape across patients in different T1D stages. Together with cohort validation and functional assays, we observe dynamic changes in TIGIT+CCR7- Tregs and CD226+CCR7-CD8+ cytotoxic T cells during the peri-remission phase. Machine learning algorithms further identify TIGIT+CCR7- Tregs and CD226+CD8+ T cells as biomarkers for β-cell function decline in a predictive model, while cell communication analysis and in vitro assays suggest that TIGIT+CCR7- Tregs may inhibit CD226+CCR7-CD8+ T cells via TGF-β signaling. Lastly, in both cyclophosphamide-induced and streptozotocin (STZ)-induced mouse diabetes models, CD226 inhibition postpones insulitis onset and reduces hyperglycemia severity. Our results thus identify two interrelated immune cell subsets that may serve as biomarkers for monitoring disease progression and targets for therapeutic intervention of T1D.
Collapse
MESH Headings
- Diabetes Mellitus, Type 1/immunology
- Diabetes Mellitus, Type 1/metabolism
- Animals
- T-Lymphocytes, Regulatory/immunology
- T-Lymphocytes, Regulatory/metabolism
- CD8-Positive T-Lymphocytes/immunology
- CD8-Positive T-Lymphocytes/metabolism
- Antigens, Differentiation, T-Lymphocyte/metabolism
- Antigens, Differentiation, T-Lymphocyte/immunology
- Mice
- Humans
- Transforming Growth Factor beta/metabolism
- Receptors, Immunologic/metabolism
- Receptors, Immunologic/genetics
- Male
- Disease Progression
- Female
- Diabetes Mellitus, Experimental/immunology
- Adult
- Mice, Inbred NOD
- Receptors, CCR7/metabolism
- Receptors, CCR7/genetics
- Insulin-Secreting Cells/metabolism
- Insulin-Secreting Cells/immunology
- Adolescent
- Young Adult
- Cell Communication/immunology
Collapse
Affiliation(s)
- Ting Zhong
- National Clinical Research Center for Metabolic Diseases, Key Laboratory of Diabetes Immunology, Ministry of Education, and Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Xinyu Li
- National Clinical Research Center for Metabolic Diseases, Key Laboratory of Diabetes Immunology, Ministry of Education, and Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Kang Lei
- National Clinical Research Center for Metabolic Diseases, Key Laboratory of Diabetes Immunology, Ministry of Education, and Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Rong Tang
- National Clinical Research Center for Metabolic Diseases, Key Laboratory of Diabetes Immunology, Ministry of Education, and Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Qiaolin Deng
- Department of Physiology and Pharmacology, Karolinska Institute, 17177, Solna, Sweden
| | - Paul E Love
- Section on Hematopoiesis and Lymphocyte Biology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, USA
| | - Zhiguang Zhou
- National Clinical Research Center for Metabolic Diseases, Key Laboratory of Diabetes Immunology, Ministry of Education, and Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China.
| | - Bin Zhao
- National Clinical Research Center for Metabolic Diseases, Key Laboratory of Diabetes Immunology, Ministry of Education, and Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China.
- CSU-Sinocare Research Center for Nutrition and Metabolic Health, Xiangya School of Public Health, Central South University, Changsha, Hunan, China.
- Furong Laboratory, Changsha, Hunan, China.
| | - Xia Li
- National Clinical Research Center for Metabolic Diseases, Key Laboratory of Diabetes Immunology, Ministry of Education, and Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China.
| |
Collapse
|
50
|
Wang H, Torous W, Gong B, Purdom E. Visualizing scRNA-Seq data at population scale with GloScope. Genome Biol 2024; 25:259. [PMID: 39380041 PMCID: PMC11463121 DOI: 10.1186/s13059-024-03398-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2023] [Accepted: 09/20/2024] [Indexed: 10/10/2024] Open
Abstract
Increasingly, scRNA-Seq studies explore cell populations across different samples and the effect of sample heterogeneity on organism's phenotype. However, relatively few bioinformatic methods have been developed which adequately address the variation between samples for such population-level analyses. We propose a framework for representing the entire single-cell profile of a sample, which we call a GloScope representation. We implement GloScope on scRNA-Seq datasets from study designs ranging from 12 to over 300 samples and demonstrate how GloScope allows researchers to perform essential bioinformatic tasks at the sample-level, in particular visualization and quality control assessment.
Collapse
Affiliation(s)
- Hao Wang
- Division of Biostatistics, University of California, Berkeley, CA, USA
| | - William Torous
- Department of Statistics, University of California, Berkeley, CA, USA
| | - Boying Gong
- Division of Biostatistics, University of California, Berkeley, CA, USA
| | - Elizabeth Purdom
- Department of Statistics, University of California, Berkeley, CA, USA.
- Center for Computational Biology, University of California, Berkeley, CA, USA.
| |
Collapse
|