1
|
Boakye AO, Obirikorang C, Afum-Adjei Awuah A, Adu EA, Winter D, Boham EE, Alani H, Newton SK, Almoustapha NST, Deke J, Dzadey WO, Adu-Amoah L, Kroduah SA, Grant MA, Asare G, Amoako-Adusei A, Loag W, Kettenbeil J, Sarkodie YA, Oduro-Mensah E, Yawson AE, Apanga S, Odotei Adjei R, Adobasom-Anane AG, Lorenz E, Souares A, Maiga-Ascofaré O, May J, Struck NS, Amuasi JH. Genetic association of ACE2 rs2285666 (C>T) and rs2106809 (A>G) and susceptibility to SARS-CoV-2 infection among the Ghanaian population. Front Genet 2025; 16:1555515. [PMID: 40491571 PMCID: PMC12146278 DOI: 10.3389/fgene.2025.1555515] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2025] [Accepted: 05/07/2025] [Indexed: 06/11/2025] Open
Abstract
Background Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), enters human cells using the angiotensin-converting enzyme 2 (ACE-2) receptor. ACE2 single nucleotide polymorphisms (SNPs) can influence susceptibility by affecting viral binding or gene expression. This study investigated the association between ACE2 SNPs, rs2285666 and rs2106809, and the SARS-CoV-2 infection susceptibility in a Ghanaian population. Methods Genomic DNA was extracted, using a magnetic bead-based method, from blood samples of a random-subset of 1,334 participants drawn from a two-stage cluster, population-based household cross-sectional SARS-CoV-2 IgG seroprevalence survey. Data collected included, socio-demographic characteristics, medical history, vaccination, and smoking status. Genotyping of the ACE2 SNPs was performed using Allele-Specific Oligonucleotide Polymerase Chain Reaction (ASO-PCR) combined with melting curve analysis. Logistic regression models were utilized to assess the association between the ACE2 SNPs and the susceptibility to SARS-CoV-2 infection. Results The median age of participants was 33 [Interquartile range (IQR) = 24-46] years. Females accounted for the majority of the sampled population, 64.3%. SARS-CoV-2-IgG seropositivity was (58.4%, 95%CI: 52.6%-64.2%) among the male population and (54.1%, 95%CI: 49.54%-58.61%) in the female population. There were no significant differences in overall allele or genotype frequencies of ACE2 SNPs between SARS-CoV-2 IgG seropositive and seronegative individuals for both females and males. Among females, those with the T allele of ACE2 rs2285666 had a 38% decreased susceptibility to SARS-CoV-2 infection under the dominant [adjusted odds ratio (aOR) = 0.62; 95%CI = 0.45-0.85, P = 0.003] and heterozygous advantage models (aOR = 0.62; 95%CI = 0.45-0.86, P = 0.004), after adjusting for confounders, but not thee recessive model (aOR = 0.41; 95%CI = 0.03-5.22, P = 0.490). No significant association was observed among males. Overall, the ACE2 rs2106809 was not associated with the susceptibility to SARS-CoV-2 infection in both males and females. Conclusion This study found no association between ACE2 rs2106809 genetic variant and susceptibility to SARS-CoV-2 infection, whilst the rs2285666 T-allele was associated with a decreased frequency for SARS-CoV-2 infection among Ghanaian females. These findings enhance our understanding of genetic factors influencing SARS-CoV-2 susceptibility, which could help identify at-risk populations and inform more targeted public health interventions in future outbreaks.
Collapse
Affiliation(s)
- Alexander Owusu Boakye
- Kumasi Centre for Collaborative Research in Tropical Medicine (KCCR), Kwame Nkrumah University of Science and Technology, Kumasi, Ghana
- Department of Molecular Medicine, School of Medicine and Dentistry, College of Health Sciences, Kwame Nkrumah University of Science and Technology (KNUST), Kumasi, Ghana
| | - Christian Obirikorang
- Kumasi Centre for Collaborative Research in Tropical Medicine (KCCR), Kwame Nkrumah University of Science and Technology, Kumasi, Ghana
- Department of Molecular Medicine, School of Medicine and Dentistry, College of Health Sciences, Kwame Nkrumah University of Science and Technology (KNUST), Kumasi, Ghana
| | - Anthony Afum-Adjei Awuah
- Kumasi Centre for Collaborative Research in Tropical Medicine (KCCR), Kwame Nkrumah University of Science and Technology, Kumasi, Ghana
- Department of Molecular Medicine, School of Medicine and Dentistry, College of Health Sciences, Kwame Nkrumah University of Science and Technology (KNUST), Kumasi, Ghana
| | - Evans Asamoah Adu
- Kumasi Centre for Collaborative Research in Tropical Medicine (KCCR), Kwame Nkrumah University of Science and Technology, Kumasi, Ghana
- Department of Molecular Medicine, School of Medicine and Dentistry, College of Health Sciences, Kwame Nkrumah University of Science and Technology (KNUST), Kumasi, Ghana
| | - Doris Winter
- Infectious Disease Epidemiology, Bernhard Nocht Institute for Tropical Medicine, Hamburg, Germany
| | - Eric Ebenezer Boham
- Kumasi Centre for Collaborative Research in Tropical Medicine (KCCR), Kwame Nkrumah University of Science and Technology, Kumasi, Ghana
- Department of Molecular Medicine, School of Medicine and Dentistry, College of Health Sciences, Kwame Nkrumah University of Science and Technology (KNUST), Kumasi, Ghana
| | - Hakim Alani
- Kumasi Centre for Collaborative Research in Tropical Medicine (KCCR), Kwame Nkrumah University of Science and Technology, Kumasi, Ghana
| | - Sylvester Kofi Newton
- Kumasi Centre for Collaborative Research in Tropical Medicine (KCCR), Kwame Nkrumah University of Science and Technology, Kumasi, Ghana
| | - Nana Safi Toure Almoustapha
- Kumasi Centre for Collaborative Research in Tropical Medicine (KCCR), Kwame Nkrumah University of Science and Technology, Kumasi, Ghana
| | - James Deke
- Kumasi Centre for Collaborative Research in Tropical Medicine (KCCR), Kwame Nkrumah University of Science and Technology, Kumasi, Ghana
- Department of Clinical Microbiology, School of Medicine and Dentistry, College of Health Sciences, Kwame Nkrumah University of Science and Technology (KNUST), Kumasi, Ghana
| | - Welbeck Odame Dzadey
- Kumasi Centre for Collaborative Research in Tropical Medicine (KCCR), Kwame Nkrumah University of Science and Technology, Kumasi, Ghana
| | - Louis Adu-Amoah
- Kumasi Centre for Collaborative Research in Tropical Medicine (KCCR), Kwame Nkrumah University of Science and Technology, Kumasi, Ghana
- Department of Molecular Medicine, School of Medicine and Dentistry, College of Health Sciences, Kwame Nkrumah University of Science and Technology (KNUST), Kumasi, Ghana
| | - Sally-Ann Kroduah
- Kumasi Centre for Collaborative Research in Tropical Medicine (KCCR), Kwame Nkrumah University of Science and Technology, Kumasi, Ghana
- Department of Global and International Health, School of Public Health, College of Health Sciences, Kwame Nkrumah University of Science and Technology (KNUST), Kumasi, Ghana
| | - Mary Ama Grant
- Kumasi Centre for Collaborative Research in Tropical Medicine (KCCR), Kwame Nkrumah University of Science and Technology, Kumasi, Ghana
| | - Gracelyn Asare
- Kumasi Centre for Collaborative Research in Tropical Medicine (KCCR), Kwame Nkrumah University of Science and Technology, Kumasi, Ghana
| | - Amos Amoako-Adusei
- Kumasi Centre for Collaborative Research in Tropical Medicine (KCCR), Kwame Nkrumah University of Science and Technology, Kumasi, Ghana
| | - Wibke Loag
- Infectious Disease Epidemiology, Bernhard Nocht Institute for Tropical Medicine, Hamburg, Germany
| | - Jenny Kettenbeil
- Infectious Disease Epidemiology, Bernhard Nocht Institute for Tropical Medicine, Hamburg, Germany
| | - Yaw Adu Sarkodie
- Department of Clinical Microbiology, School of Medicine and Dentistry, College of Health Sciences, Kwame Nkrumah University of Science and Technology (KNUST), Kumasi, Ghana
| | | | - Alfred Edwin Yawson
- Department of Community Health, Medical School, University of Ghana, Accra, Ghana
| | - Stephen Apanga
- Department of Community Health and Preventive Medicine, School of Medicine, University for Development Studies, Tamale, Ghana
| | - Rose Odotei Adjei
- Kumasi Centre for Collaborative Research in Tropical Medicine (KCCR), Kwame Nkrumah University of Science and Technology, Kumasi, Ghana
- Department of Global and International Health, School of Public Health, College of Health Sciences, Kwame Nkrumah University of Science and Technology (KNUST), Kumasi, Ghana
| | - Austin Gideon Adobasom-Anane
- Kumasi Centre for Collaborative Research in Tropical Medicine (KCCR), Kwame Nkrumah University of Science and Technology, Kumasi, Ghana
- Department of Global and International Health, School of Public Health, College of Health Sciences, Kwame Nkrumah University of Science and Technology (KNUST), Kumasi, Ghana
| | - Eva Lorenz
- Infectious Disease Epidemiology, Bernhard Nocht Institute for Tropical Medicine, Hamburg, Germany
- German Center for Infection Research (DZIF), Partner Site Hamburg-Borstel-Lübeck-Riems, Hamburg, Germany
| | - Aurélia Souares
- German Center for Infection Research (DZIF), Partner Site Heidelberg, Heidelberg, Germany
- Heidelberg Institute of Global Health (HIGH), Heidelberg University Hospital, Heidelberg, Germany
| | - Oumou Maiga-Ascofaré
- Kumasi Centre for Collaborative Research in Tropical Medicine (KCCR), Kwame Nkrumah University of Science and Technology, Kumasi, Ghana
- Infectious Disease Epidemiology, Bernhard Nocht Institute for Tropical Medicine, Hamburg, Germany
- German Center for Infection Research (DZIF), Partner Site Hamburg-Borstel-Lübeck-Riems, Hamburg, Germany
| | - Jürgen May
- Infectious Disease Epidemiology, Bernhard Nocht Institute for Tropical Medicine, Hamburg, Germany
- German Center for Infection Research (DZIF), Partner Site Hamburg-Borstel-Lübeck-Riems, Hamburg, Germany
- Department of Tropical Medicine I, University Medical Centre Hamburg Eppendorf (UKE), Hamburg, Germany
| | - Nicole S. Struck
- Infectious Disease Epidemiology, Bernhard Nocht Institute for Tropical Medicine, Hamburg, Germany
- German Center for Infection Research (DZIF), Partner Site Hamburg-Borstel-Lübeck-Riems, Hamburg, Germany
| | - John Humphery Amuasi
- Kumasi Centre for Collaborative Research in Tropical Medicine (KCCR), Kwame Nkrumah University of Science and Technology, Kumasi, Ghana
- Infectious Disease Epidemiology, Bernhard Nocht Institute for Tropical Medicine, Hamburg, Germany
- Department of Global and International Health, School of Public Health, College of Health Sciences, Kwame Nkrumah University of Science and Technology (KNUST), Kumasi, Ghana
- Department of Tropical Medicine I, University Medical Centre Hamburg Eppendorf (UKE), Hamburg, Germany
| |
Collapse
|
2
|
Yao Y, Xu Y, Zhang Y, Gui Y, Bai Q, Zhu Z, Peng H, Zhou Y, Chen ZJ, Sun J, Su J. Single Cell Inference of Cancer Drug Response Using Pathway-Based Transformer Network. SMALL METHODS 2025; 9:e2400991. [PMID: 39962810 DOI: 10.1002/smtd.202400991] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/01/2024] [Revised: 01/30/2025] [Indexed: 05/26/2025]
Abstract
Accurate prediction of cancer drug responses is crucial for personalized therapy. Single-cell RNA sequencing (scRNA-seq) captures cellular heterogeneity and rare resistant populations, offering valuable insights into treatment responses. However, the distinct distributions of bulk RNA-seq and scRNA-seq data hinder the transfer of drug response knowledge from large-scale cell line datasets. To address this, single-cell Pathway Drug Sensitivity (scPDS) model is developed, a Transformer-based deep learning method that predicts drug sensitivities from scRNA-seq data through pathway activation transformation. By integrating bulk RNA-seq data from extensive cell line datasets, scPDS improves accuracy and computational efficiency in scRNA-seq analysis. It is demonstrated that scPDS outperforms state-of-the-art methods in both time and memory consumption. When applied to breast cancer cells treated with bortezomib, scPDS showed that resistance increases initially but diminishes with prolonged exposure. The method also identifies drug-sensitive populations in bortezomib-resistant cells and predicts the efficacy of combination therapies, including docetaxel, gemcitabine, and irinotecan. Furthermore, scPDS successfully distinguishes between sensitive and resistant patients, predicting significantly different survival outcomes. In summary, scPDS offers a robust tool for predicting cellular responses, providing insights to optimize cancer treatment strategies.
Collapse
Affiliation(s)
- Yinghao Yao
- Oujiang Laboratory, Zhejiang Lab for Regenerative Medicine, Vision and Brain Health, Eye Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, 325101, China
| | - Yuandong Xu
- Oujiang Laboratory, Zhejiang Lab for Regenerative Medicine, Vision and Brain Health, Eye Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, 325101, China
| | - Yaru Zhang
- Oujiang Laboratory, Zhejiang Lab for Regenerative Medicine, Vision and Brain Health, Eye Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, 325101, China
| | - Yuanyuan Gui
- Oujiang Laboratory, Zhejiang Lab for Regenerative Medicine, Vision and Brain Health, Eye Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, 325101, China
| | - Qingshi Bai
- Oujiang Laboratory, Zhejiang Lab for Regenerative Medicine, Vision and Brain Health, Eye Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, 325101, China
| | - Zhengbiao Zhu
- School of Biomedical Engineering, Eye Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, 325027, China
| | - Hui Peng
- School of Biomedical Engineering, Eye Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, 325027, China
| | - Yijun Zhou
- School of Information and Communication Engineering, Hainan University, Haikou, 570228, China
- Hainan Institute of Real-World Data, Qionghai, Hainan, 571400, China
| | - Zhen Ji Chen
- Oujiang Laboratory, Zhejiang Lab for Regenerative Medicine, Vision and Brain Health, Eye Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, 325101, China
| | - Jie Sun
- School of Biomedical Engineering, Eye Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, 325027, China
| | - Jianzhong Su
- Oujiang Laboratory, Zhejiang Lab for Regenerative Medicine, Vision and Brain Health, Eye Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, 325101, China
- School of Information and Communication Engineering, Hainan University, Haikou, 570228, China
| |
Collapse
|
3
|
Li J, Ma Y, Cao Y, Zheng G, Ren Q, Chen C, Zhu Q, Zhou Y, Lu Y, Zhang Y, Deng C, Chen WH, Su J. Integrating microbial GWAS and single-cell transcriptomics reveals associations between host cell populations and the gut microbiome. Nat Microbiol 2025; 10:1210-1226. [PMID: 40195537 DOI: 10.1038/s41564-025-01978-w] [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: 03/13/2024] [Accepted: 03/05/2025] [Indexed: 04/09/2025]
Abstract
Microbial genome-wide association studies (GWAS) have uncovered numerous host genetic variants associated with gut microbiota. However, links between host genetics, the gut microbiome and specific cellular contexts remain unclear. Here we use a computational framework, scBPS (single-cell Bacteria Polygenic Score), to integrate existing microbial GWAS and single-cell RNA-sequencing profiles of 24 human organs, including the liver, pancreas, lung and intestine, to identify host tissues and cell types relevant to gut microbes. Analysing 207 microbial taxa and 254 host cell types, scBPS-inferred cellular enrichments confirmed known biology such as dominant communications between gut microbes and the digestive tissue module and liver epithelial cell compartment. scBPS also identified a robust association between Collinsella and the central-veinal hepatocyte subpopulation. We experimentally validated the causal effects of Collinsella on cholesterol metabolism in mice through single-nuclei RNA sequencing on liver tissue to identify relevant cell subpopulations. Mechanistically, oral gavage of Collinsella modulated cholesterol pathway gene expression in central-veinal hepatocytes. We further validated our approach using independent microbial GWAS data, alongside single-cell and bulk transcriptomic analyses, demonstrating its robustness and reproducibility. Together, scBPS enables a systematic mapping of the host-microbe crosstalk by linking cell populations to their interacting gut microbes.
Collapse
Affiliation(s)
- Jingjing Li
- Oujiang Laboratory, Zhejiang Lab for Regenerative Medicine, Vision and Brain Health, Eye Hospital, Wenzhou Medical University, Wenzhou, China
| | - Yunlong Ma
- Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Yue Cao
- Key Laboratory of Molecular Biophysics of the Ministry of Education, Hubei Key Laboratory of Bioinformatics and Molecular-imaging, Center for Artificial Intelligence Biology, Department of Bioinformatics and Systems Biology, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, China
| | - Gongwei Zheng
- Oujiang Laboratory, Zhejiang Lab for Regenerative Medicine, Vision and Brain Health, Eye Hospital, Wenzhou Medical University, Wenzhou, China
| | - Qing Ren
- Oujiang Laboratory, Zhejiang Lab for Regenerative Medicine, Vision and Brain Health, Eye Hospital, Wenzhou Medical University, Wenzhou, China
| | - Cheng Chen
- Oujiang Laboratory, Zhejiang Lab for Regenerative Medicine, Vision and Brain Health, Eye Hospital, Wenzhou Medical University, Wenzhou, China
| | - Qunyan Zhu
- Oujiang Laboratory, Zhejiang Lab for Regenerative Medicine, Vision and Brain Health, Eye Hospital, Wenzhou Medical University, Wenzhou, China
| | - Yijun Zhou
- Oujiang Laboratory, Zhejiang Lab for Regenerative Medicine, Vision and Brain Health, Eye Hospital, Wenzhou Medical University, Wenzhou, China
| | - Yu Lu
- The Second School of Clinical Medicine, Institution of Medical Artificial Intelligence, Binzhou Medical University, Yantai, China
| | - Yaru Zhang
- Oujiang Laboratory, Zhejiang Lab for Regenerative Medicine, Vision and Brain Health, Eye Hospital, Wenzhou Medical University, Wenzhou, China
| | - Chunyu Deng
- Oujiang Laboratory, Zhejiang Lab for Regenerative Medicine, Vision and Brain Health, Eye Hospital, Wenzhou Medical University, Wenzhou, China
| | - Wei-Hua Chen
- Key Laboratory of Molecular Biophysics of the Ministry of Education, Hubei Key Laboratory of Bioinformatics and Molecular-imaging, Center for Artificial Intelligence Biology, Department of Bioinformatics and Systems Biology, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, China.
- The Second School of Clinical Medicine, Institution of Medical Artificial Intelligence, Binzhou Medical University, Yantai, China.
- School of Biological Science, Jining Medical University, Rizhao, China.
| | - Jianzhong Su
- Oujiang Laboratory, Zhejiang Lab for Regenerative Medicine, Vision and Brain Health, Eye Hospital, Wenzhou Medical University, Wenzhou, China.
| |
Collapse
|
4
|
Song L, Chen W, Hou J, Guo M, Yang J. Spatially resolved mapping of cells associated with human complex traits. Nature 2025; 641:932-941. [PMID: 40108460 PMCID: PMC12095064 DOI: 10.1038/s41586-025-08757-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2024] [Accepted: 02/07/2025] [Indexed: 03/22/2025]
Abstract
Depicting spatial distributions of disease-relevant cells is crucial for understanding disease pathology1,2. Here we present genetically informed spatial mapping of cells for complex traits (gsMap), a method that integrates spatial transcriptomics data with summary statistics from genome-wide association studies to map cells to human complex traits, including diseases, in a spatially resolved manner. Using embryonic spatial transcriptomics datasets covering 25 organs, we benchmarked gsMap through simulation and by corroborating known trait-associated cells or regions in various organs. Applying gsMap to brain spatial transcriptomics data, we reveal that the spatial distribution of glutamatergic neurons associated with schizophrenia more closely resembles that for cognitive traits than that for mood traits such as depression. The schizophrenia-associated glutamatergic neurons were distributed near the dorsal hippocampus, with upregulated expression of calcium signalling and regulation genes, whereas depression-associated glutamatergic neurons were distributed near the deep medial prefrontal cortex, with upregulated expression of neuroplasticity and psychiatric drug target genes. Our study provides a method for spatially resolved mapping of trait-associated cells and demonstrates the gain of biological insights (such as the spatial distribution of trait-relevant cells and related signature genes) through these maps.
Collapse
Affiliation(s)
- Liyang Song
- School of Life Sciences, Westlake University, Hangzhou, China
- Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, China
| | - Wenhao Chen
- School of Life Sciences, Westlake University, Hangzhou, China
- Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, China
| | - Junren Hou
- School of Life Sciences, Westlake University, Hangzhou, China
- Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, China
| | - Minmin Guo
- School of Life Sciences, Westlake University, Hangzhou, China
- Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, China
| | - Jian Yang
- School of Life Sciences, Westlake University, Hangzhou, China.
- Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, China.
| |
Collapse
|
5
|
Zhang F, Zhu M, Chen Y, Wang G, Yang H, Lu X, Li Y, Chang HM, Wu Y, Ma Y, Yuan S, Zhu W, Dong X, Zhao Y, Yu Y, Wang J, Mu L. Harnessing omics data for drug discovery and development in ovarian aging. Hum Reprod Update 2025; 31:240-268. [PMID: 39977580 DOI: 10.1093/humupd/dmaf002] [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: 09/29/2024] [Revised: 12/02/2024] [Indexed: 02/22/2025] Open
Abstract
BACKGROUND Ovarian aging occurs earlier than the aging of many other organs and has a lasting impact on women's overall health and well-being. However, effective interventions to slow ovarian aging remain limited, primarily due to an incomplete understanding of the underlying molecular mechanisms and drug targets. Recent advances in omics data resources, combined with innovative computational tools, are offering deeper insight into the molecular complexities of ovarian aging, paving the way for new opportunities in drug discovery and development. OBJECTIVE AND RATIONALE This review aims to synthesize the expanding multi-omics data, spanning genome, transcriptome, proteome, metabolome, and microbiome, related to ovarian aging, from both tissue-level and single-cell perspectives. We will specially explore how the analysis of these emerging omics datasets can be leveraged to identify novel drug targets and guide therapeutic strategies for slowing and reversing ovarian aging. SEARCH METHODS We conducted a comprehensive literature search in the PubMed database using a range of relevant keywords: ovarian aging, age at natural menopause, premature ovarian insufficiency (POI), diminished ovarian reserve (DOR), genomics, transcriptomics, epigenomics, DNA methylation, RNA modification, histone modification, proteomics, metabolomics, lipidomics, microbiome, single-cell, genome-wide association studies (GWAS), whole-exome sequencing, phenome-wide association studies (PheWAS), Mendelian randomization (MR), epigenetic target, drug target, machine learning, artificial intelligence (AI), deep learning, and multi-omics. The search was restricted to English-language articles published up to September 2024. OUTCOMES Multi-omics studies have uncovered key mechanisms driving ovarian aging, including DNA damage and repair deficiencies, inflammatory and immune responses, mitochondrial dysfunction, and cell death. By integrating multi-omics data, researchers can identify critical regulatory factors and mechanisms across various biological levels, leading to the discovery of potential drug targets. Notable examples include genetic targets such as BRCA2 and TERT, epigenetic targets like Tet and FTO, metabolic targets such as sirtuins and CD38+, protein targets like BIN2 and PDGF-BB, and transcription factors such as FOXP1. WIDER IMPLICATIONS The advent of cutting-edge omics technologies, especially single-cell technologies and spatial transcriptomics, has provided valuable insights for guiding treatment decisions and has become a powerful tool in drug discovery aimed at mitigating or reversing ovarian aging. As technology advances, the integration of single-cell multi-omics data with AI models holds the potential to more accurately predict candidate drug targets. This convergence offers promising new avenues for personalized medicine and precision therapies, paving the way for tailored interventions in ovarian aging. REGISTRATION NUMBER Not applicable.
Collapse
Affiliation(s)
- Fengyu Zhang
- Reproductive Medicine Center, Zhongshan Hospital, Fudan University, Shanghai, China
- The First School of Medicine, Wenzhou Medical University, Wenzhou, China
| | - Ming Zhu
- The First School of Medicine, Wenzhou Medical University, Wenzhou, China
| | - Yi Chen
- The First School of Medicine, Wenzhou Medical University, Wenzhou, China
| | - Guiquan Wang
- Xiamen Key Laboratory of Reproduction and Genetics, Department of Reproductive Medicine, Women and Children's Hospital, School of Medicine, Xiamen University, Xiamen, China
| | - Haiyan Yang
- Reproductive Medicine Center, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Xinmei Lu
- Reproductive Medicine Center, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Yan Li
- Reproductive Medicine Center, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Hsun-Ming Chang
- Department of Obstetrics and Gynecology, China Medical University Hospital, Taichung, Taiwan
| | - Yang Wu
- Institute of Rare Diseases, West China Hospital of Sichuan University, Chengdu, China
| | - Yunlong Ma
- Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Shuai Yuan
- Department of Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Unit of Cardiovascular and Nutritional Epidemiology, Institute of Environmental Medicine, Karolinska Institute, Stockholm, Sweden
| | - Wencheng Zhu
- Institute of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, China
| | - Xi Dong
- Reproductive Medicine Center, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Yue Zhao
- State Key Laboratory of Female Fertility Promotion, Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, China
| | - Yang Yu
- State Key Laboratory of Female Fertility Promotion, Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, China
- National Clinical Research Center for Obstetrics and Gynecology, Beijing, China
- Key Laboratory of Assisted Reproduction, Ministry of Education, Beijing, China
- Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive Technology, Beijing, China
| | - Jia Wang
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Liangshan Mu
- Reproductive Medicine Center, Zhongshan Hospital, Fudan University, Shanghai, China
| |
Collapse
|
6
|
Hou X, Li Z, Lin J, Lin W, Li L, Zheng X, Lai X, Zhu L, Guo P, Cai F, Zhang J, Li W, Yang C, Dai Y. Single-cell transcriptome integrated with genome-wide association study reveals heterogeneity of carotid and femoral plaques and its association with plaque stability. Sci Rep 2025; 15:11812. [PMID: 40189611 PMCID: PMC11973204 DOI: 10.1038/s41598-025-96434-4] [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: 01/28/2025] [Accepted: 03/28/2025] [Indexed: 04/09/2025] Open
Abstract
Carotid and femoral plaques exhibit varying degrees of stability; however, the relationships of different genes/cell types with plaque embolism are poorly understood. We evaluated differential gene/cell expression and investigated the cells/genes associated with carotid and femoral artery plaque embolism. sc-RNA-seq and bulk RNA data were obtained to identify differentially expressed genes (DEGs). Seven machine learning models were trained, and the top 10 DEGs across all models were selected. The most disturbed cells in carotid and femoral artery plaques were identified using Augur, while the genes and cells in the carotid plaque associated with embolism were analyzed through scPagwas. The differences in most disturbed cells and embolism-related cells were further analyzed. Compared with femoral plaques, carotid plaques had 80 downregulated and 90 upregulated genes. Machine learning identified the key DEGs between carotid and femoral plaques were predominantly from the HOX gene family. Natural Killer (NK) cells were the most significantly disturbed cells between carotid and femoral plaques, and they may be most strongly associated with plaque embolism. Among the differential genes in NK cells, CD2 was most associated with embolism. Our research may offer new insights into atherosclerosis at different locations.
Collapse
Affiliation(s)
- Xinhuang Hou
- Department of Vascular Surgery, The First Affiliated Hospital, Fujian Medical University, 20 Chazhong Road, Fuzhou, 350005, China.
- Department of Vascular Surgery, National Regional Medical Center, Binhai Campus of The First Affiliated Hospital, Fujian Medical University, Fuzhou, 350212, China.
| | - Zhipeng Li
- Department of Gastrointestinal Surgery, The First Affiliated Hospital, Fujian Medical University, Fuzhou, 350005, China
| | - Jun Lin
- Trauma Center and Emergency Surgery Department, The First Affiliated Hospital, Fujian Medical University, Fuzhou, 350005, China
| | - Wei Lin
- Department of Vascular Surgery, The First Affiliated Hospital, Fujian Medical University, 20 Chazhong Road, Fuzhou, 350005, China
| | - Luyao Li
- Department of Vascular Surgery, The First Affiliated Hospital, Fujian Medical University, 20 Chazhong Road, Fuzhou, 350005, China
| | - Xiaoqi Zheng
- Department of Vascular Surgery, The First Affiliated Hospital, Fujian Medical University, 20 Chazhong Road, Fuzhou, 350005, China
| | - Xiaoling Lai
- Department of Vascular Surgery, The First Affiliated Hospital, Fujian Medical University, 20 Chazhong Road, Fuzhou, 350005, China
| | - Lin Zhu
- Department of Nutrition and Food Safety, School of Public Health, Fujian Medical University, Fuzhou, 350122, China
| | - Pingfan Guo
- Department of Vascular Surgery, The First Affiliated Hospital, Fujian Medical University, 20 Chazhong Road, Fuzhou, 350005, China
- Department of Vascular Surgery, National Regional Medical Center, Binhai Campus of The First Affiliated Hospital, Fujian Medical University, Fuzhou, 350212, China
| | - Fanggang Cai
- Department of Vascular Surgery, The First Affiliated Hospital, Fujian Medical University, 20 Chazhong Road, Fuzhou, 350005, China
- Department of Vascular Surgery, National Regional Medical Center, Binhai Campus of The First Affiliated Hospital, Fujian Medical University, Fuzhou, 350212, China
| | - Jinchi Zhang
- Department of Vascular Surgery, The First Affiliated Hospital, Fujian Medical University, 20 Chazhong Road, Fuzhou, 350005, China
- Department of Vascular Surgery, National Regional Medical Center, Binhai Campus of The First Affiliated Hospital, Fujian Medical University, Fuzhou, 350212, China
| | - Wanglong Li
- Department of Vascular Surgery, The First Affiliated Hospital, Fujian Medical University, 20 Chazhong Road, Fuzhou, 350005, China
- Department of Vascular Surgery, National Regional Medical Center, Binhai Campus of The First Affiliated Hospital, Fujian Medical University, Fuzhou, 350212, China
| | - Changwei Yang
- Department of Nutrition and Food Safety, School of Public Health, Fujian Medical University, Fuzhou, 350122, China.
| | - Yiquan Dai
- Department of Vascular Surgery, The First Affiliated Hospital, Fujian Medical University, 20 Chazhong Road, Fuzhou, 350005, China.
- Department of Vascular Surgery, National Regional Medical Center, Binhai Campus of The First Affiliated Hospital, Fujian Medical University, Fuzhou, 350212, China.
| |
Collapse
|
7
|
Gao C, Wu J, Zhong F, Yang X, Liu H, Lai J, Cai J, Mao W, Xu H. Integrative analysis of genetic variability and functional traits in lung adenocarcinoma epithelial cells via single-cell RNA sequencing, GWAS, bayesian deconvolution, and machine learning. Genes Genomics 2025; 47:435-468. [PMID: 39992528 PMCID: PMC12000210 DOI: 10.1007/s13258-025-01621-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2024] [Accepted: 01/09/2025] [Indexed: 02/25/2025]
Abstract
BACKGROUND Lung adenocarcinoma remains a leading cause of cancer-related mortality worldwide, characterized by high genetic and cellular heterogeneity, especially within the tumor microenvironment. OBJECTIVE This study integrates single-cell RNA sequencing (scRNA-seq) with genome-wide association studies (GWAS) using Bayesian deconvolution and machine learning techniques to unravel the genetic and functional complexity of lung adenocarcinoma epithelial cells. METHODS We performed scRNA-seq and GWAS analysis to identify critical cell populations affected by genetic variations. Bayesian deconvolution and machine learning techniques were applied to investigate tumor progression, prognosis, and immune-epithelial cell interactions, particularly focusing on immune checkpoint markers such as PD-L1 and CTLA-4. RESULTS Our analysis highlights the importance of genes like SLC2A1, which regulates glucose metabolism and correlates with tumor invasiveness and poor prognosis. Immune-epithelial interactions suggest a suppressive tumor microenvironment, potentially hindering immune responses. Additionally, machine learning models identify core prognostic genes such as F12, GOLM1, and S100P, which are significantly associated with patient survival. CONCLUSIONS This comprehensive approach provides novel insights into lung adenocarcinoma biology, emphasizing the role of genetic and immune factors in tumor progression. The findings support the development of personalized therapeutic strategies targeting both tumor cells and the immune microenvironment.
Collapse
Affiliation(s)
- Chenggen Gao
- Jiangxi medical college, Nanchang university, Nanchang, China
| | - Jintao Wu
- Department of Thoracic Surgery, The Third Affiliated Hospital of Zhejiang Chinese Medicine University, Hangzhou, China
| | - Fangyan Zhong
- Jiangxi medical college, Nanchang university, Nanchang, China
- NHC Key Laboratory of Personalized Diagnosis and Treatment of Nasopharyngeal Carcinoma, Jiangxi Cancer Hospital, The Second Affiliated Hospital of Nanchang Medical College, Nanchang, China
| | - Xianxin Yang
- The fifth affiliated hospital of jinan university, Heyuan, Guangdong, China
| | - Hanwen Liu
- Department of general surgery, The Third Affiliated Hospital of Zhejiang Chinese Medical University, Nanchang, China
| | - Junming Lai
- Ganjiang New District Hospital of the First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Jing Cai
- Lung cancer center, The second affiliated hospital of Nanchang University, Nanchang, China
| | - Weimin Mao
- Department of Thoracic Surgery, Jiangxi Cancer HospitalJiangxi Province, Nanchang, China
| | - Huijuan Xu
- Department of Clinical Laboratory, Hangzhou TCM Hospital Affiliated to Zhejiang Chinese Medical University, Hangzhou, China.
| |
Collapse
|
8
|
Liu H, Ma Y, Gao N, Zhou Y, Li G, Zhu Q, Liu X, Li S, Deng C, Chen C, Yang Y, Ren Q, Hu H, Cai Y, Chen M, Xue Y, Zhang K, Qu J, Su J. Identification and characterization of human retinal stem cells capable of retinal regeneration. Sci Transl Med 2025; 17:eadp6864. [PMID: 40138453 DOI: 10.1126/scitranslmed.adp6864] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2024] [Revised: 09/12/2024] [Accepted: 03/04/2025] [Indexed: 03/29/2025]
Abstract
Human retinal stem cells hold great promise in regenerative medicine, yet their existence and characteristics remain elusive. Here, we performed single-cell multiomics and spatial transcriptomics of human fetal retinas and uncovered a cell subpopulation, human neural retinal stem-like cells (hNRSCs), distinct from retinal pigment epithelium stem-like cells and traditional retinal progenitor cells. We found that these hNRSCs reside in the peripheral retina in the ciliary marginal zone, exhibiting substantial self-renewal and differentiation potential. We conducted single-cell and spatial transcriptomic analyses of human retinal organoids (hROs) and revealed that hROs contain a population of hNRSCs with similar transcriptional profiles and developmental trajectories to hNRSCs in the fetal retina potentially capable of regenerating all retinal cells. Furthermore, we identified crucial transcription factors, such as MECOM, governing hNRSC commitment to neural retinogenesis and regulating repair processes in hROs. hRO-derived hNRSCs transplanted into the rd10 mouse model of retinitis pigmentosa differentiated and were integrated into the retina, alleviated retinal degeneration, and improved visual function. Overall, our work identifies and characterizes a distinct category of retinal stem cells from human retinas, underscoring their regenerative potential and promise for transplantation therapy.
Collapse
Affiliation(s)
- Hui Liu
- National Engineering Research Center of Ophthalmology and Optometry, Eye Hospital, Wenzhou Medical University, Wenzhou 325027, China
- Oujiang Laboratory, Zhejiang Lab for Regenerative Medicine, Vision and Brain Health, Wenzhou 325101, China
- State Key Laboratory of Eye Health, Eye Hospital, Wenzhou Medical University, Wenzhou 325027, China
| | - Yunlong Ma
- National Engineering Research Center of Ophthalmology and Optometry, Eye Hospital, Wenzhou Medical University, Wenzhou 325027, China
- Oujiang Laboratory, Zhejiang Lab for Regenerative Medicine, Vision and Brain Health, Wenzhou 325101, China
| | - Na Gao
- National Engineering Research Center of Ophthalmology and Optometry, Eye Hospital, Wenzhou Medical University, Wenzhou 325027, China
| | - Yijun Zhou
- National Engineering Research Center of Ophthalmology and Optometry, Eye Hospital, Wenzhou Medical University, Wenzhou 325027, China
| | - Gen Li
- Guangzhou National Laboratory, Guangzhou 510005, China
- Center for Biomedicine and Innovations, Faculty of Medicine, Macau University of Science and Technology, Taipa 999078, Macau, China
| | - Qunyan Zhu
- Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou 325011, China
| | - Xiaoyu Liu
- National Engineering Research Center of Ophthalmology and Optometry, Eye Hospital, Wenzhou Medical University, Wenzhou 325027, China
| | - Shasha Li
- Oujiang Laboratory, Zhejiang Lab for Regenerative Medicine, Vision and Brain Health, Wenzhou 325101, China
| | - Chunyu Deng
- National Engineering Research Center of Ophthalmology and Optometry, Eye Hospital, Wenzhou Medical University, Wenzhou 325027, China
- Oujiang Laboratory, Zhejiang Lab for Regenerative Medicine, Vision and Brain Health, Wenzhou 325101, China
| | - Cheng Chen
- National Engineering Research Center of Ophthalmology and Optometry, Eye Hospital, Wenzhou Medical University, Wenzhou 325027, China
- Oujiang Laboratory, Zhejiang Lab for Regenerative Medicine, Vision and Brain Health, Wenzhou 325101, China
| | - Yuhe Yang
- National Engineering Research Center of Ophthalmology and Optometry, Eye Hospital, Wenzhou Medical University, Wenzhou 325027, China
| | - Qing Ren
- National Engineering Research Center of Ophthalmology and Optometry, Eye Hospital, Wenzhou Medical University, Wenzhou 325027, China
| | - Huijuan Hu
- National Engineering Research Center of Ophthalmology and Optometry, Eye Hospital, Wenzhou Medical University, Wenzhou 325027, China
| | - Yaoyao Cai
- Department of Obstetrics, First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325000, China
| | - Ming Chen
- National Engineering Research Center of Ophthalmology and Optometry, Eye Hospital, Wenzhou Medical University, Wenzhou 325027, China
- State Key Laboratory of Eye Health, Eye Hospital, Wenzhou Medical University, Wenzhou 325027, China
| | - Yuanchao Xue
- National Engineering Research Center of Ophthalmology and Optometry, Eye Hospital, Wenzhou Medical University, Wenzhou 325027, China
- Key Laboratory of RNA Biology, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100190, China
| | - Kang Zhang
- Center for Biomedicine and Innovations, Faculty of Medicine, Macau University of Science and Technology, Taipa 999078, Macau, China
| | - Jia Qu
- National Engineering Research Center of Ophthalmology and Optometry, Eye Hospital, Wenzhou Medical University, Wenzhou 325027, China
- Oujiang Laboratory, Zhejiang Lab for Regenerative Medicine, Vision and Brain Health, Wenzhou 325101, China
| | - Jianzhong Su
- National Engineering Research Center of Ophthalmology and Optometry, Eye Hospital, Wenzhou Medical University, Wenzhou 325027, China
- Oujiang Laboratory, Zhejiang Lab for Regenerative Medicine, Vision and Brain Health, Wenzhou 325101, China
- State Key Laboratory of Eye Health, Eye Hospital, Wenzhou Medical University, Wenzhou 325027, China
- Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou 325011, China
| |
Collapse
|
9
|
Chi D, Zhang K, Zhang J, He Z, Zhou H, Huang W, Liu Y, Huang J, Zeng W, Bai X, Ou C, Ouyang H. Astrocytic pleiotrophin deficiency in the prefrontal cortex contributes to stress-induced depressive-like responses in male mice. Nat Commun 2025; 16:2528. [PMID: 40087317 PMCID: PMC11909280 DOI: 10.1038/s41467-025-57924-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2024] [Accepted: 03/03/2025] [Indexed: 03/17/2025] Open
Abstract
Astrocytes are closely linked to depression, and the prefrontal cortex (PFC) is an important brain region involved in major depressive disorder (MDD). However, the underlying mechanism by which astrocytes within PFC contribute to MDD remains unclear. Using single-nucleus RNA sequencing analyses, we show a significant reduction in astrocytes and attenuated pleiotrophin-protein tyrosine phosphatase receptor type Z1 (PTN-PTPRZ1) signaling in astrocyte-to-excitatory neuron communication in the PFC of male MDD patients. We find reduced astrocytes and PTN in the dorsomedial PFC of male mice with depression induced by chronic restraint and social defeat stress. Knockdown of astrocytic PTN induces depression-related responses, which is reversed by exogenous PTN supplementation or overexpression of astrocytic PTN. The antidepressant effects exerted by astrocytic PTN require interaction with PTPRZ1 in excitatory neurons, and PTN-PTPRZ1 activates the AKT signaling pathway to regulate depression-related responses. Our findings indicate the PTN-PTPRZ1-AKT pathway may be a potential therapeutic target for MDD.
Collapse
Affiliation(s)
- Dongmei Chi
- Department of Anesthesiology, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, P. R. China
- Department of Experimental Research, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Guangdong Esophageal Cancer Institute, Sun Yat-sen University Cancer Center, Guangzhou, P. R. China
| | - Kun Zhang
- Department of Anesthesiology, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, P. R. China
| | - Jianxing Zhang
- Department of Anesthesiology, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, P. R. China
| | - Zhaoli He
- Department of Anesthesiology, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, P. R. China
| | - Hongxia Zhou
- Department of Anesthesiology, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, P. R. China
| | - Wan Huang
- Department of Anesthesiology, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, P. R. China
| | - Yang Liu
- Department of Experimental Research, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Guangdong Esophageal Cancer Institute, Sun Yat-sen University Cancer Center, Guangzhou, P. R. China
| | - Jingxiu Huang
- Department of Anesthesiology, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, P. R. China
| | - Weian Zeng
- Department of Anesthesiology, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, P. R. China
| | - Xiaohui Bai
- Department of Anesthesiology, Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation; Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China.
| | - Chaopeng Ou
- Department of Anesthesiology, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, P. R. China.
| | - Handong Ouyang
- Department of Anesthesiology, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, P. R. China.
| |
Collapse
|
10
|
Yamamoto Y, Shirai Y, Edahiro R, Kumanogoh A, Okada Y. Large-scale cross-trait genetic analysis highlights shared genetic backgrounds of autoimmune diseases. Immunol Med 2025; 48:1-10. [PMID: 39171621 DOI: 10.1080/25785826.2024.2394258] [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: 06/25/2024] [Accepted: 08/15/2024] [Indexed: 08/23/2024] Open
Abstract
Disorders associated with the immune system burden multiple organs, although the shared biology exists across the diseases. Preceding family-based studies reveal that immune diseases are heritable to varying degrees, providing the basis for immunogenomics. The recent cost reduction in genetic analysis intensively promotes biobank-scale studies and the development of frameworks for statistical genetics. The accumulating multi-layer omics data, including genome-wide association studies (GWAS) and RNA-sequencing at single-cell resolution, enable us to dissect the genetic backgrounds of immune-related disorders. Although autoimmune and allergic diseases are generally categorized into different disease categories, epidemiological studies reveal the high incidence of autoimmune and allergic disease complications, suggesting the shared genetics and biology between the disease categories. Biobank resources and consortia cover multiple immune-related disorders to accumulate phenome-wide associations of genetic variants and enhance researchers to analyze the shared and heterogeneous genetic backgrounds. The emerging post-GWAS and integrative multi-omics analyses provide genetic and biological insights into the multicategorical disease associations.
Collapse
Affiliation(s)
- Yuji Yamamoto
- Department of Statistical Genetics, Osaka University Graduate School of Medicine, Suita, Japan
- Department of Respiratory Medicine and Clinical Immunology, Osaka University Graduate School of Medicine, Suita, Japan
| | - Yuya Shirai
- Department of Statistical Genetics, Osaka University Graduate School of Medicine, Suita, Japan
- Department of Respiratory Medicine and Clinical Immunology, Osaka University Graduate School of Medicine, Suita, Japan
- Laboratory of Statistical Immunology, Immunology Frontier Research Center (WPI-IFReC), Osaka University, Suita, Japan
- RIKEN Center for Integrative Medical Sciences, Laboratory for Systems Genetics, Yokohama, Japan
| | - Ryuya Edahiro
- Department of Statistical Genetics, Osaka University Graduate School of Medicine, Suita, Japan
- Department of Respiratory Medicine and Clinical Immunology, Osaka University Graduate School of Medicine, Suita, Japan
- RIKEN Center for Integrative Medical Sciences, Laboratory for Systems Genetics, Yokohama, Japan
| | - Atsushi Kumanogoh
- Department of Respiratory Medicine and Clinical Immunology, Osaka University Graduate School of Medicine, Suita, Japan
- Department of Immunopathology, Immunology Frontier Research Center (WPI-IFReC), Osaka University, Suita, Japan
- Integrated Frontier Research for Medical Science Division, Institute for Open and Transdisciplinary Research Initiatives (OTRI), Osaka University, Suita, Japan
- Center for Infectious Diseases for Education and Research (CiDER), Osaka University, Suita, Japan
- Japan Agency for Medical Research and Development, Core Research for Evolutional Science and Technology (AMED-CREST), Tokyo, Japan
- Center for Advanced Modalities and DDS (CAMaD), Osaka University, Suita, Japan
| | - Yukinori Okada
- Department of Statistical Genetics, Osaka University Graduate School of Medicine, Suita, Japan
- Laboratory of Statistical Immunology, Immunology Frontier Research Center (WPI-IFReC), Osaka University, Suita, Japan
- RIKEN Center for Integrative Medical Sciences, Laboratory for Systems Genetics, Yokohama, Japan
- Center for Infectious Diseases for Education and Research (CiDER), Osaka University, Suita, Japan
- Center for Advanced Modalities and DDS (CAMaD), Osaka University, Suita, Japan
- Department of Genome Informatics, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
- Premium Research Institute for Human Metaverse Medicine (WPI-PRIMe), Osaka University, Suita, Japan
| |
Collapse
|
11
|
Dand N, Stuart PE, Bowes J, Ellinghaus D, Nititham J, Saklatvala JR, Teder-Laving M, Thomas LF, Traks T, Uebe S, Assmann G, Baudry D, Behrens F, Billi AC, Brown MA, Burkhardt H, Capon F, Chung R, Curtis CJ, Duckworth M, Ellinghaus E, FitzGerald O, Gerdes S, Griffiths CEM, Gulliver S, Helliwell PS, Ho P, Hoffmann P, Holmen OL, Huang ZM, Hveem K, Jadon D, Köhm M, Kraus C, Lamacchia C, Lee SH, Ma F, Mahil SK, McHugh N, McManus R, Modalsli EH, Nissen MJ, Nöthen M, Oji V, Oksenberg JR, Patrick MT, Perez White BE, Ramming A, Rech J, Rosen C, Sarkar MK, Schett G, Schmidt B, Tejasvi T, Traupe H, Voorhees JJ, Wacker EM, Warren RB, Wasikowski R, Weidinger S, Wen X, Zhang Z, Barton A, Chandran V, Esko T, Foerster J, Franke A, Gladman DD, Gudjonsson JE, Gulliver W, Hüffmeier U, Kingo K, Kõks S, Liao W, Løset M, Mägi R, Nair RP, Rahman P, Reis A, Smith CH, Di Meglio P, Barker JN, Tsoi LC, Simpson MA, Elder JT. GWAS meta-analysis of psoriasis identifies new susceptibility alleles impacting disease mechanisms and therapeutic targets. Nat Commun 2025; 16:2051. [PMID: 40021644 PMCID: PMC11871359 DOI: 10.1038/s41467-025-56719-8] [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: 10/04/2023] [Accepted: 01/28/2025] [Indexed: 03/03/2025] Open
Abstract
Psoriasis is a common, debilitating immune-mediated skin disease. Genetic studies have identified biological mechanisms of psoriasis risk, including those targeted by effective therapies. However, the genetic liability to psoriasis is not fully explained by variation at robustly identified risk loci. To refine the genetic map of psoriasis susceptibility we meta-analysed 18 GWAS comprising 36,466 cases and 458,078 controls and identified 109 distinct psoriasis susceptibility loci, including 46 that have not been previously reported. These include susceptibility variants at loci in which the therapeutic targets IL17RA and AHR are encoded, and deleterious coding variants supporting potential new drug targets (including in STAP2, CPVL and POU2F3). We conducted a transcriptome-wide association study to identify regulatory effects of psoriasis susceptibility variants and cross-referenced these against single cell expression profiles in psoriasis-affected skin, highlighting roles for the transcriptional regulation of haematopoietic cell development and epigenetic modulation of interferon signalling in psoriasis pathobiology.
Collapse
Grants
- R01 ES033634 NIEHS NIH HHS
- R01AR050511, R01AR054966, R01AR063611, R01AR065183 U.S. Department of Health & Human Services | National Institutes of Health (NIH)
- BRC_1215_20006, NIHR302258, NIHR203308, BRC-1215-20014 DH | National Institute for Health Research (NIHR)
- 980 Maudsley Charity
- RG2/10, ST1/19, ST3/20 Psoriasis Association
- EXC 2167-390884018, CRC1181-2/project A05 Deutsche Forschungsgemeinschaft (German Research Foundation)
- STR130505 Guy's and St Thomas' Charity
- K01 AR072129, P30 AR075043, UC2 AR081033, R01AR042742 U.S. Department of Health & Human Services | National Institutes of Health (NIH)
- K08 AR078251 NIAMS NIH HHS
- P30 AR075043 NIAMS NIH HHS
- K01 AR072129 NIAMS NIH HHS
- 814364 National Psoriasis Foundation (NPF)
- R01 AR042742 NIAMS NIH HHS
- PUT1465, PRG1189, PRG1911, PRG1291 Eesti Teadusagentuur (Estonian Research Council)
- 2014-2020.4.01.15-0012 EC | European Regional Development Fund (Europski Fond za Regionalni Razvoj)
- U01AI119125 U.S. Department of Health & Human Services | National Institutes of Health (NIH)
- LF-OC-22-001033 LEO Pharma Research Foundation
- 821511 Innovative Medicines Initiative (IMI)
- RG-1611-26299 National Multiple Sclerosis Society (National MS Society)
- MR/S003126/1 RCUK | Medical Research Council (MRC)
- U01 AI119125 NIAID NIH HHS
- R01ES033634, R35GM138121, K08 AR078251, R01AR065174 U.S. Department of Health & Human Services | National Institutes of Health (NIH)
- R01 AR054966 NIAMS NIH HHS
- R01 AR050511 NIAMS NIH HHS
- R01 AR065174 NIAMS NIH HHS
- R35 GM138121 NIGMS NIH HHS
- 01EC1407A, 01EC1401C Bundesministerium für Bildung und Forschung (Federal Ministry of Education and Research)
- SI 236/8-1, SI236/9-1, ER 155/6-1 Deutsche Forschungsgemeinschaft (German Research Foundation)
- UC2 AR081033 NIAMS NIH HHS
- R01 AR065183 NIAMS NIH HHS
- R01 AR063611 NIAMS NIH HHS
- U.S. Department of Health & Human Services | National Institutes of Health (NIH)
- Versus Arthritis - grant reference number 21754 Additional funding support from the following bodies are also acknowledged, as detailed in the funding section of the manuscript: Ann Arbor Veterans Hospital; Babcock Memorial Trust; Cambridge Arthritis Research Endeavour (CARE); Dermatology Foundation; Faculty of Medicine and Health Sciences, NTNU; German Centre for Neurodegenerative Disorders (DZNE), Bonn; German Ministry of Education and Science; Heinz Nixdorf Foundation (Germany); Joint Research Committee between St Olav’s Hospital and the Faculty of Medicine and Health Sciences, NTNU; Krembil Foundation; Liaison Committee for Education, Research, and Innovation in Central Norway; The Michael J. Fox Foundation; MSWA; National Institutes of Health; Perron Institute for Neurological and Translational Science; Pfizer Chair Research Award in Rheumatology; Research Council of Norway; Shake It Up Australia; Stiftelsen Kristian Gerhard Jebsen; Taubman Medical Research Institute; University of Michigan
Collapse
Affiliation(s)
- Nick Dand
- Department of Medical & Molecular Genetics, School of Basic & Medical Biosciences, Faculty of Life Sciences & Medicine, King's College London, London, UK
- Health Data Research UK, London, UK
| | - Philip E Stuart
- Department of Dermatology, University of Michigan Medical School, Ann Arbor, MI, USA
| | - John Bowes
- Centre for Genetics and Genomics Versus Arthritis, The University of Manchester, Manchester, UK
- National Institute for Health and Care Research (NIHR) Manchester Biomedical Research Centre, The University of Manchester, Manchester, UK
| | - David Ellinghaus
- Institute of Clinical Molecular Biology, Christian-Albrechts-University of Kiel, Kiel, Germany
| | - Joanne Nititham
- Deparment of Dermatology, University of California San Francisco, San Francisco, CA, USA
| | - Jake R Saklatvala
- Department of Medical & Molecular Genetics, School of Basic & Medical Biosciences, Faculty of Life Sciences & Medicine, King's College London, London, UK
| | | | - Laurent F Thomas
- Department of Clinical and Molecular Medicine, NTNU - Norwegian University of Science and Technology, Trondheim, Norway
- K.G. Jebsen Center for Genetic Epidemiology, Department of Public Health and Nursing, NTNU - Norwegian University of Science and Technology, Trondheim, Norway
- BioCore - Bioinformatics Core Facility, NTNU - Norwegian University of Science and Technology, Trondheim, Norway
- Clinic of Laboratory Medicine, St. Olavs Hospital, Trondheim University Hospital, Trondheim, Norway
| | - Tanel Traks
- Department of Dermatology and Venereology, Institute of Clinical Medicine, University of Tartu, Tartu, Estonia
| | - Steffen Uebe
- Institute of Human Genetics, Universitätsklinikum Erlangen, FAU Erlangen-Nürnberg, Erlangen, Germany
| | - Gunter Assmann
- RUB University Hospital JWK Minden, Department of Rheumatology, Minden, Germany
- Jose-Carreras Centrum for Immuno- and Gene Therapy, University of Saarland Medical School, Homburg, Germany
| | - David Baudry
- St John's Institute of Dermatology, School of Basic & Medical Biosciences, Faculty of Life Sciences & Medicine, King's College London, London, UK
| | - Frank Behrens
- Division of Translational Rheumatology, Immunology - Inflammation Medicine, University Hospital, Goethe University, Frankfurt am Main, Germany
- Fraunhofer Institute for Translational Medicine and Pharmacology ITMP, Frankfurt am Main, Germany
- Fraunhofer Cluster of Excellence Immune-mediated Diseases CIMD, Frankfurt am Main, Germany
- Division of Rheumatology, University Hospital, Goethe University, Frankfurt am Main, Germany
| | - Allison C Billi
- Department of Dermatology, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Matthew A Brown
- Department of Medical & Molecular Genetics, School of Basic & Medical Biosciences, Faculty of Life Sciences & Medicine, King's College London, London, UK
- Genomics England, Canary Wharf, London, UK
| | - Harald Burkhardt
- Fraunhofer Institute for Translational Medicine and Pharmacology ITMP, Frankfurt am Main, Germany
- Fraunhofer Cluster of Excellence Immune-mediated Diseases CIMD, Frankfurt am Main, Germany
- Division of Rheumatology, University Hospital, Goethe University, Frankfurt am Main, Germany
| | - Francesca Capon
- Department of Medical & Molecular Genetics, School of Basic & Medical Biosciences, Faculty of Life Sciences & Medicine, King's College London, London, UK
| | - Raymond Chung
- Institute of Psychiatry, Psychology and Neuroscience, King's College London, Denmark Hill, Camberwell, London, UK
- National Institute for Health and Care Research (NIHR) Biomedical Research Centre, South London and Maudsley Hospital, London, UK
| | - Charles J Curtis
- Institute of Psychiatry, Psychology and Neuroscience, King's College London, Denmark Hill, Camberwell, London, UK
- National Institute for Health and Care Research (NIHR) Biomedical Research Centre, South London and Maudsley Hospital, London, UK
| | - Michael Duckworth
- St John's Institute of Dermatology, School of Basic & Medical Biosciences, Faculty of Life Sciences & Medicine, King's College London, London, UK
| | - Eva Ellinghaus
- Institute of Clinical Molecular Biology, Christian-Albrechts-University of Kiel, Kiel, Germany
| | - Oliver FitzGerald
- UCD School of Medicine and Medical Sciences and Conway Institute of Biomolecular and Biomedical Research, University College Dublin, Dublin, Ireland
| | - Sascha Gerdes
- Department of Dermatology, Venereology and Allergy, University Hospital Schleswig-Holstein, Campus Kiel, Kiel, Germany
| | - Christopher E M Griffiths
- St John's Institute of Dermatology, School of Basic & Medical Biosciences, Faculty of Life Sciences & Medicine, King's College London, London, UK
- Centre for Dermatology Research, University of Manchester, NIHR Manchester Biomedical Research Centre, Manchester, UK
- Department of Dermatology, King's College Hospital NHS Foundation Trust, London, UK
| | | | - Philip S Helliwell
- Leeds Institute of Rheumatic and Musculoskeletal Medicine, University of Leeds, Leeds, UK
| | - Pauline Ho
- Centre for Genetics and Genomics Versus Arthritis, The University of Manchester, Manchester, UK
- National Institute for Health and Care Research (NIHR) Manchester Biomedical Research Centre, The University of Manchester, Manchester, UK
- The Kellgren Centre for Rheumatology, Manchester University NHS Foundation Trust, Manchester, UK
| | - Per Hoffmann
- Institute of Human Genetics, University of Bonn, School of Medicine & University Hospital Bonn, Bonn, Germany
| | - Oddgeir L Holmen
- HUNT Research Centre, Department of Public Health and Nursing, NTNU - Norwegian University of Science and Technology, Levanger, Norway
- Levanger Hospital, Nord-Trøndelag Hospital Trust, Levanger, Norway
| | - Zhi-Ming Huang
- Deparment of Dermatology, University of California San Francisco, San Francisco, CA, USA
| | - Kristian Hveem
- K.G. Jebsen Center for Genetic Epidemiology, Department of Public Health and Nursing, NTNU - Norwegian University of Science and Technology, Trondheim, Norway
- HUNT Research Centre, Department of Public Health and Nursing, NTNU - Norwegian University of Science and Technology, Levanger, Norway
- Levanger Hospital, Nord-Trøndelag Hospital Trust, Levanger, Norway
| | - Deepak Jadon
- Department of Medicine, University of Cambridge, Cambridge, UK
| | - Michaela Köhm
- Division of Translational Rheumatology, Immunology - Inflammation Medicine, University Hospital, Goethe University, Frankfurt am Main, Germany
- Fraunhofer Institute for Translational Medicine and Pharmacology ITMP, Frankfurt am Main, Germany
- Fraunhofer Cluster of Excellence Immune-mediated Diseases CIMD, Frankfurt am Main, Germany
- Division of Rheumatology, University Hospital, Goethe University, Frankfurt am Main, Germany
| | - Cornelia Kraus
- Institute of Human Genetics, Universitätsklinikum Erlangen, FAU Erlangen-Nürnberg, Erlangen, Germany
| | - Céline Lamacchia
- Division of Rheumatology, Geneva University Hospital, Geneva, Switzerland
| | - Sang Hyuck Lee
- Institute of Psychiatry, Psychology and Neuroscience, King's College London, Denmark Hill, Camberwell, London, UK
- National Institute for Health and Care Research (NIHR) Biomedical Research Centre, South London and Maudsley Hospital, London, UK
| | - Feiyang Ma
- Department of Dermatology, University of Michigan Medical School, Ann Arbor, MI, USA
- Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, MI, USA
| | - Satveer K Mahil
- St John's Institute of Dermatology, School of Basic & Medical Biosciences, Faculty of Life Sciences & Medicine, King's College London, London, UK
- St John's Institute of Dermatology, Guy's and St Thomas' National Health Service (NHS) Foundation Trust, London, UK
| | - Neil McHugh
- Department of Life Sciences, University of Bath, Bath, UK
| | - Ross McManus
- Department of Clinical Medicine, Trinity Translational Medicine Institute, Trinity College Dublin, Dublin, Ireland
| | - Ellen H Modalsli
- Department of Clinical and Molecular Medicine, NTNU - Norwegian University of Science and Technology, Trondheim, Norway
- Department of Dermatology, Clinic of Orthopedy, Rheumatology and Dermatology, St. Olavs Hospital, Trondheim University Hospital, Trondheim, Norway
| | - Michael J Nissen
- Division of Rheumatology, Geneva University Hospital, Geneva, Switzerland
| | - Markus Nöthen
- Institute of Human Genetics, University of Bonn, School of Medicine & University Hospital Bonn, Bonn, Germany
| | - Vinzenz Oji
- Department of Dermatology, University of Münster, Münster, Germany
| | - Jorge R Oksenberg
- Weill Institute for Neurosciences, Department of Neurology, University of California, San Francisco, CA, USA
| | - Matthew T Patrick
- Department of Dermatology, University of Michigan Medical School, Ann Arbor, MI, USA
| | | | - Andreas Ramming
- Department of Internal Medicine 3, Friedrich-Alexander-University Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen, Ulmenweg 18, 91054, Erlangen, Germany
- Deutsches Zentrum Immuntherapie (DZI), Friedrich-Alexander-University Erlangen-Nürnberg and Universitätsklinikum Erlangen, Erlangen, Germany
| | - Jürgen Rech
- Department of Internal Medicine 3, Friedrich-Alexander-University Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen, Ulmenweg 18, 91054, Erlangen, Germany
- Deutsches Zentrum Immuntherapie (DZI), Friedrich-Alexander-University Erlangen-Nürnberg and Universitätsklinikum Erlangen, Erlangen, Germany
| | - Cheryl Rosen
- Division of Dermatology, Toronto Western Hospital, University of Toronto, Toronto, Ontario, Canada
| | - Mrinal K Sarkar
- Department of Dermatology, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Georg Schett
- Department of Internal Medicine 3, Friedrich-Alexander-University Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen, Ulmenweg 18, 91054, Erlangen, Germany
- Deutsches Zentrum Immuntherapie (DZI), Friedrich-Alexander-University Erlangen-Nürnberg and Universitätsklinikum Erlangen, Erlangen, Germany
| | - Börge Schmidt
- Institute of Medical Informatics, Biometry and Epidemiology, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Trilokraj Tejasvi
- Department of Dermatology, University of Michigan Medical School, Ann Arbor, MI, USA
- Ann Arbor Veterans Affairs Hospital, Ann Arbor, MI, USA
| | - Heiko Traupe
- Department of Dermatology, University of Münster, Münster, Germany
| | - John J Voorhees
- Department of Dermatology, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Eike Matthias Wacker
- Institute of Clinical Molecular Biology, Christian-Albrechts-University of Kiel, Kiel, Germany
| | - Richard B Warren
- Division of Musculoskeletal and Dermatological Sciences, School of Biological Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, UK
- Centre for Dermatology Research, Salford Royal Hospital, Northern Care Alliance NHS Foundation Trust, Manchester Academic Health Science Centre, Manchester, M6 8HD, UK
| | - Rachael Wasikowski
- Department of Dermatology, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Stephan Weidinger
- Department of Dermatology, Venereology and Allergy, University Hospital Schleswig-Holstein, Campus Kiel, Kiel, Germany
| | - Xiaoquan Wen
- Department of Biostatistics, Center for Statistical Genetics, University of Michigan, Ann Arbor, MI, USA
| | - Zhaolin Zhang
- Department of Dermatology, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Anne Barton
- Centre for Genetics and Genomics Versus Arthritis, The University of Manchester, Manchester, UK
- National Institute for Health and Care Research (NIHR) Manchester Biomedical Research Centre, The University of Manchester, Manchester, UK
- The Kellgren Centre for Rheumatology, Manchester University NHS Foundation Trust, Manchester, UK
| | - Vinod Chandran
- Schroeder Arthritis Institute, Krembil Research Institute and Toronto Western Hospital, University Health Network and University of Toronto, Toronto, Ontario, Canada
| | - Tõnu Esko
- Institute of Genomics, University of Tartu, Tartu, Estonia
| | - John Foerster
- College of Medicine, Dentistry, and Nursing, University of Dundee, Dundee, UK
| | - Andre Franke
- Institute of Clinical Molecular Biology, Christian-Albrechts-University of Kiel, Kiel, Germany
| | - Dafna D Gladman
- Schroeder Arthritis Institute, Krembil Research Institute and Toronto Western Hospital, University Health Network and University of Toronto, Toronto, Ontario, Canada
| | - Johann E Gudjonsson
- Department of Dermatology, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Wayne Gulliver
- Newlab Clinical Research Inc, St. John's, NL, Canada
- Department of Dermatology, Discipline of Medicine, Faculty of Medicine, Memorial University of Newfoundland, St. John's, NL, Canada
| | - Ulrike Hüffmeier
- Institute of Human Genetics, Universitätsklinikum Erlangen, FAU Erlangen-Nürnberg, Erlangen, Germany
| | - Külli Kingo
- Department of Dermatology and Venereology, Institute of Clinical Medicine, University of Tartu, Tartu, Estonia
- Dermatology Clinic, Tartu University Hospital, Tartu, Estonia
| | - Sulev Kõks
- Perron Institute for Neurological and Translational Science, Nedlands, WA, 6009, Australia
- Centre for Molecular Medicine and Innovative Therapeutics, Health Futures Institute, Murdoch University, Perth, WA, 6150, Australia
| | - Wilson Liao
- Deparment of Dermatology, University of California San Francisco, San Francisco, CA, USA
| | - Mari Løset
- K.G. Jebsen Center for Genetic Epidemiology, Department of Public Health and Nursing, NTNU - Norwegian University of Science and Technology, Trondheim, Norway
- Department of Dermatology, Clinic of Orthopedy, Rheumatology and Dermatology, St. Olavs Hospital, Trondheim University Hospital, Trondheim, Norway
| | - Reedik Mägi
- Estonian Genome Centre, Institute of Genomics, University of Tartu, Tartu, Estonia
| | - Rajan P Nair
- Department of Dermatology, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Proton Rahman
- Memorial University of Newfoundland, St. John's, NL, Canada
| | - André Reis
- Institute of Human Genetics, Universitätsklinikum Erlangen, FAU Erlangen-Nürnberg, Erlangen, Germany
| | - Catherine H Smith
- St John's Institute of Dermatology, School of Basic & Medical Biosciences, Faculty of Life Sciences & Medicine, King's College London, London, UK
- St John's Institute of Dermatology, Guy's and St Thomas' National Health Service (NHS) Foundation Trust, London, UK
| | - Paola Di Meglio
- St John's Institute of Dermatology, School of Basic & Medical Biosciences, Faculty of Life Sciences & Medicine, King's College London, London, UK
| | - Jonathan N Barker
- St John's Institute of Dermatology, School of Basic & Medical Biosciences, Faculty of Life Sciences & Medicine, King's College London, London, UK
- St John's Institute of Dermatology, Guy's and St Thomas' National Health Service (NHS) Foundation Trust, London, UK
| | - Lam C Tsoi
- Department of Dermatology, University of Michigan Medical School, Ann Arbor, MI, USA
- Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, MI, USA
- Department of Biostatistics, Center for Statistical Genetics, University of Michigan, Ann Arbor, MI, USA
| | - Michael A Simpson
- Department of Medical & Molecular Genetics, School of Basic & Medical Biosciences, Faculty of Life Sciences & Medicine, King's College London, London, UK.
| | - James T Elder
- Department of Dermatology, University of Michigan Medical School, Ann Arbor, MI, USA.
- Ann Arbor Veterans Affairs Hospital, Ann Arbor, MI, USA.
| |
Collapse
|
12
|
Feng R, Li X, Li B, Luan T, He J, Liu G, Yue J. Integrating transcriptomics and scPagwas analysis predicts naïve CD4 T cell-related gene DRAM2 as a potential biomarker and therapeutic target for colorectal cancer. BMC Cancer 2025; 25:317. [PMID: 39984869 PMCID: PMC11843817 DOI: 10.1186/s12885-025-13731-x] [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: 10/09/2024] [Accepted: 02/13/2025] [Indexed: 02/23/2025] Open
Abstract
OBJECTIVE The interaction between T cells, particularly naïve CD4 T cells (CD4Tn), and colorectal cancer (CRC) is highly complex. CD4Tn play a crucial role in modulating immune responses within the tumor microenvironment, yet the precise mechanisms by which they influence tumor progression remain elusive. This study aims to explore the relationship between CRC and CD4Tn, identify biomarkers and therapeutic targets, and focus on the role of CD4Tn in shaping the immune environment of CRC. METHODS Single-cell transcriptomics, alongside the scPagwas algorithm, were employed to identify pivotal T cell subsets involved in CRC progression. Bulk transcriptomic data were further analyzed using deconvolution algorithms to elucidate the roles of these key T cell subsets. The abundance of naïve CD4 T cells (CD4Tn) was specifically assessed to gauge patient responses to immunotherapy, alterations in the immune microenvironment, and correlations with genetic mutations. Key genes linked to CD4Tn were identified using weighted gene co-expression network analysis and Pearson correlation scores. The SMR algorithm was subsequently used for validation, with experimental verification following. RESULTS Through single-cell transcriptomics and the scPagwas algorithm, CD4Tn was confirmed as a critical cell type in CRC progression. High infiltration of CD4Tn cells in CRC patients was correlated with poorer prognosis and suboptimal responses to immunotherapy. SMR analysis suggested a potential causal link between DRAM2 gene expression and CRC progression. Experimental knockdown of DRAM2 in colorectal cancer cells significantly inhibited tumor growth. CONCLUSION The DRAM2 gene, associated with CD4Tn cells, appears to play a pivotal role in the advancement of CRC and may represent a promising therapeutic target for treatment.
Collapse
Affiliation(s)
- Rui Feng
- Chongqing Key Laboratory of Molecular Oncology and Epigenetics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Xiaofang Li
- Department of Pharmacy, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Benhua Li
- The Second People's Hospital of Liangshan Yi Autonomous Prefecture, Xichang, China
| | - Tiankuo Luan
- Department of Human Anatomy, Basic Medical School, Chongqing Medical University, Chongqing, China
| | - Jiaming He
- Department of Human Anatomy, Basic Medical School, Chongqing Medical University, Chongqing, China
| | - Guojing Liu
- Department of Neurosurgery, The University-Town Hospital of Chongqing Medical University, NO.55 of university-town middle Road, Shapingba District, Chongqing, 400000, China.
| | - Jian Yue
- Department of Breast Surgery, Gaozhou People's Hospital, No.89 Xiguan Road, Gaozhou, Guangdong, 525200, China.
| |
Collapse
|
13
|
Meng Z, Zhang C, Liu S, Li W, Wang Y, Zhang Q, Peng B, Ye W, Jiang Y, Song Y, Guo M, Chang X, Shao L. Exploring genetic loci linked to COVID-19 severity and immune response through multi-trait GWAS analyses. Front Genet 2025; 16:1502839. [PMID: 40034745 PMCID: PMC11873281 DOI: 10.3389/fgene.2025.1502839] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2024] [Accepted: 01/27/2025] [Indexed: 03/05/2025] Open
Abstract
Introduction COVID-19 severity has been linked to immune factors, with excessive immune responses like cytokine storms contributing to mortality. However, the genetic basis of these immune responses is not well understood. This study aimed to explore the genetic connection between COVID-19 severity and blood cell traits, given their close relationship with immunity. Materials and methods GWAS summary statistics for COVID-19 and blood cell counts were analyzed using Linkage Disequilibrium Score Regression (LDSC) to estimate genetic correlations and heritabilities. For traits with significant correlations, a Multi-Trait GWAS Analysis (MTAG) was performed to identify pleiotropic loci shared between COVID-19 and blood cell counts. Results Our MTAG analysis identified four pleiotropic loci associated with COVID-19 severity, five loci linked to hospitalized cases, and one locus related to general patients. Among these, two novel loci were identified in the high-risk population, with rs55779981 located near RAVER1 and rs73009538 near CARM1. In hospitalized patients, two previously unrecognized loci were detected, namely, rs115545251 near GFI1 and rs3181049 near RAVER1, while in general patients, rs11065822 near CUX2 emerged as a newly identified locus. We also identified potential target genes, including those involved in inflammation signaling (CARM1), endothelial dysfunction (INTS12), and antiviral immune response (RAVER1), which may require further investigation. Conclusion Our study offers insights into the genetic overlap between COVID-19 and immune factors, suggesting potential directions for future research and clinical exploration.
Collapse
Affiliation(s)
- Ziang Meng
- Department of Infectious Disease, Central Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
| | - Chumeng Zhang
- The Second School of Clinical Medicine of Shandong First Medical University, Tai’an, Shandong, China
| | - Shuai Liu
- Agricultural Products Quality and Safety Center of Jinan, Jinan, Shandong, China
| | - Wen Li
- College of Medical Information and Artificial Intelligence, Shandong First Medical University, Jinan, Shandong, China
| | - Yue Wang
- College of Medical Information and Artificial Intelligence, Shandong First Medical University, Jinan, Shandong, China
| | - Qingyi Zhang
- College of Medical Information and Artificial Intelligence, Shandong First Medical University, Jinan, Shandong, China
| | - Bichen Peng
- College of Medical Information and Artificial Intelligence, Shandong First Medical University, Jinan, Shandong, China
| | - Weiyi Ye
- College of Medical Information and Artificial Intelligence, Shandong First Medical University, Jinan, Shandong, China
| | - Yue Jiang
- College of Medical Information and Artificial Intelligence, Shandong First Medical University, Jinan, Shandong, China
| | - Yingchao Song
- College of Medical Information and Artificial Intelligence, Shandong First Medical University, Jinan, Shandong, China
| | - Miao Guo
- School of Life Sciences, Shandong First Medical University, Shandong, China
| | - Xiao Chang
- College of Medical Information and Artificial Intelligence, Shandong First Medical University, Jinan, Shandong, China
| | - Lei Shao
- Department of Infectious Disease, Central Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
| |
Collapse
|
14
|
Huang S, Yin H. A Multi-Omics-Based Exploration of the Predictive Role of MSMB in Prostate Cancer Recurrence: A Study Using Bayesian Inverse Convolution and 10 Machine Learning Combinations. Biomedicines 2025; 13:487. [PMID: 40002900 PMCID: PMC11853722 DOI: 10.3390/biomedicines13020487] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2024] [Revised: 02/02/2025] [Accepted: 02/05/2025] [Indexed: 02/27/2025] Open
Abstract
Background: Prostate cancer (PCa) is a prevalent malignancy among elderly men. Biochemical recurrence (BCR), which typically occurs after radical treatments such as radical prostatectomy or radiation therapy, serves as a critical indicator of potential disease progression. However, reliable and effective methods for predicting BCR in PCa patients remain limited. Methods: In this study, we used Bayesian deconvolution combined with 10 machine learning algorithms to build a five-gene model for predicting PCa progression. The model and the five selected genes were externally validated. Various analyses such as prognosis, clinical subgroups, tumor microenvironment, immunity, genetic variants, and drug sensitivity were performed on MSMB/Epithelial_cells subgroups. Results: Our model outperformed 102 previously published prognostic features. Notably, PCa patients with a high proportion of MSMB/epithelial cells were characterized by a greater progression-free Interval (PFI), a higher proportion of early-stage tumors, a lower stromal component, and a reduced presence of tumor-associated fibroblasts (CAF). The high proportion of MSMB/epithelial cells was also associated with higher frequencies of SPOP and TP53 mutations. Drug sensitivity analysis revealed that patients with a poorer prognosis and lower MSMB/epithelial cell ratio showed increased sensitivity to cyclophosphamide, cisplatin, and dasatinib. Conclusions: The model developed in this study provides a robust and accurate tool for predicting PCa progression. It offers significant potential for enhancing risk stratification and informing personalized treatment strategies for PCa patients.
Collapse
Affiliation(s)
| | - Hang Yin
- Department of Urology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing 100020, China;
| |
Collapse
|
15
|
Zhang Y, Xu H, Han X, Yu Q, Zheng L, Xiao H. PMAIP1-mediated glucose metabolism and its impact on the tumor microenvironment in breast cancer: Integration of multi-omics analysis and experimental validation. Transl Oncol 2025; 52:102267. [PMID: 39740516 PMCID: PMC11750568 DOI: 10.1016/j.tranon.2024.102267] [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: 11/13/2024] [Revised: 12/19/2024] [Accepted: 12/24/2024] [Indexed: 01/02/2025] Open
Abstract
BACKGROUND Glucose metabolism in breast cancer has a potential effect on tumor progression and is related to the immune microenvironment. Thus, this study aimed to develop a glucose metabolism-tumor microenvironment score to provide new perspectives on breast cancer treatment. METHOD Data were acquired from the Gene Expression Omnibus and UCSC Xena databases, and glucose-metabolism-related genes were acquired from the Gene Set Enrichment Analysis database. Genes with significant prognostic value were identified, and immune infiltration analysis was conducted, and a prognostic model was constructed based on the results of these analyses. The results were validated by in vitro experiments with MCF-7 and MCF-10A cell lines, including expression validation, functional experiments, and bulk sequencing. Single-cell analysis was also conducted to explore the role of specific cell clusters in breast cancer, and Bayes deconvolution was used to further investigate the associations between cell clusters and tumor phenotypes of breast cancer. RESULTS Four significant prognostic genes (PMAIP1, PGK1, SIRT7, and SORBS1) were identified, and, through immune infiltration analysis, a combined prognostic model based on glucose metabolism and immune infiltration was established. The model was used to classify clinical subtypes of breast cancer, and PMAIP1 was identified as a potential critical gene related to glucose metabolism in breast cancer. Single-cell analysis and Bayes deconvolution jointly confirmed the protective role of the PMAIP1+ luminal cell cluster.
Collapse
Affiliation(s)
- Yidong Zhang
- School of Pharmacy, Faculty of Medicine, Macau University of Science and Technology, Macau SAR, China; School of Pharmacy, Queen's University of Belfast, Belfast, Northern Ireland, United Kingdom
| | - Hang Xu
- School of Pharmacy, Faculty of Medicine, Macau University of Science and Technology, Macau SAR, China; Department of Pharmacy, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, China
| | - Xuedan Han
- School of Life Science and Technology, China Pharmaceutical University, 639 Longmian Road, Nanjing 211198, China
| | - Qiyi Yu
- Mellon College of Science, Carnage Mellon University, Pittsburgh, Pennsylvania, USA
| | - Lufeng Zheng
- School of Life Science and Technology, China Pharmaceutical University, 639 Longmian Road, Nanjing 211198, China.
| | - Hua Xiao
- Department of General Surgery, Nanjing First Hospital, Nanjing Medical University, Nanjing, Jiangsu, China.
| |
Collapse
|
16
|
Ma Y, Jiang D, Li J, Zheng G, Deng Y, Gou X, Gao S, Chen C, Zhou Y, Zhang Y, Deng C, Yao Y, Han H, Su J. Systematic dissection of pleiotropic loci and critical regulons in excitatory neurons and microglia relevant to neuropsychiatric and ocular diseases. Transl Psychiatry 2025; 15:24. [PMID: 39856056 PMCID: PMC11760387 DOI: 10.1038/s41398-025-03243-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/18/2024] [Revised: 12/08/2024] [Accepted: 01/14/2025] [Indexed: 01/27/2025] Open
Abstract
Advancements in single-cell multimodal techniques have greatly enhanced our understanding of disease-relevant loci identified through genome-wide association studies (GWASs). To investigate the biological connections between the eye and brain, we integrated bulk and single-cell multiomic profiles with GWAS summary statistics for eight neuropsychiatric and five ocular diseases. Our analysis uncovered five latent factors explaining 61.7% of the genetic variance across these 13 diseases, revealing diverse correlational patterns among them. We identified 45 pleiotropic loci with 91 candidate genes that contribute to disease risk. By integrating GWAS and single-cell profiles, we implicated excitatory neurons and microglia as key contributors in the eye-brain connections. Polygenic enrichment analysis further identified 15 pleiotropic regulons in excitatory neurons and 16 in microglia that were linked to comorbid conditions. Functionally, excitatory neuron-specific regulons were involved in axon guidance and synaptic activity, while microglia-specific regulons were associated with immune response and cell activation. In sum, these findings underscore the genetic link between psychiatric disorders and ocular diseases.
Collapse
Affiliation(s)
- Yunlong Ma
- Oujiang Laboratory, Zhejiang Lab for Regenerative Medicine, Vision and Brain Health, Eye Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, China.
- Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
- Lifespan Brain Institute at Penn Med and the Children's Hospital of Philadelphia, Philadelphia, PA, USA.
| | - Dingping Jiang
- Oujiang Laboratory, Zhejiang Lab for Regenerative Medicine, Vision and Brain Health, Eye Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, China
- National Engineering Research Center of Ophthalmology and Optometry, Eye Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Jingjing Li
- Oujiang Laboratory, Zhejiang Lab for Regenerative Medicine, Vision and Brain Health, Eye Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, China
- National Engineering Research Center of Ophthalmology and Optometry, Eye Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Gongwei Zheng
- Oujiang Laboratory, Zhejiang Lab for Regenerative Medicine, Vision and Brain Health, Eye Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, China
- National Engineering Research Center of Ophthalmology and Optometry, Eye Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Yao Deng
- Oujiang Laboratory, Zhejiang Lab for Regenerative Medicine, Vision and Brain Health, Eye Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, China
- National Engineering Research Center of Ophthalmology and Optometry, Eye Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Xuanxuan Gou
- Oujiang Laboratory, Zhejiang Lab for Regenerative Medicine, Vision and Brain Health, Eye Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, China
- National Engineering Research Center of Ophthalmology and Optometry, Eye Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Shuaishuai Gao
- Oujiang Laboratory, Zhejiang Lab for Regenerative Medicine, Vision and Brain Health, Eye Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, China
- National Engineering Research Center of Ophthalmology and Optometry, Eye Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Cheng Chen
- Oujiang Laboratory, Zhejiang Lab for Regenerative Medicine, Vision and Brain Health, Eye Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, China
- Institute of Genomic Medicine, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Yijun Zhou
- Oujiang Laboratory, Zhejiang Lab for Regenerative Medicine, Vision and Brain Health, Eye Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, China
- Institute of Genomic Medicine, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Yaru Zhang
- Oujiang Laboratory, Zhejiang Lab for Regenerative Medicine, Vision and Brain Health, Eye Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, China
- Institute of Genomic Medicine, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Chunyu Deng
- Oujiang Laboratory, Zhejiang Lab for Regenerative Medicine, Vision and Brain Health, Eye Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, China
- National Engineering Research Center of Ophthalmology and Optometry, Eye Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Yinghao Yao
- Institute of Genomic Medicine, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Haijun Han
- School of Medicine, Hangzhou City University, Hangzhou, China
| | - Jianzhong Su
- Oujiang Laboratory, Zhejiang Lab for Regenerative Medicine, Vision and Brain Health, Eye Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, China.
- National Engineering Research Center of Ophthalmology and Optometry, Eye Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, China.
- Institute of Genomic Medicine, Wenzhou Medical University, Wenzhou, Zhejiang, China.
| |
Collapse
|
17
|
Chen Z, Mei K, Tan F, Zhou Y, Du H, Wang M, Gu R, Huang Y. Integrative multi-omics analysis for identifying novel therapeutic targets and predicting immunotherapy efficacy in lung adenocarcinoma. CANCER DRUG RESISTANCE (ALHAMBRA, CALIF.) 2025; 8:3. [PMID: 39935429 PMCID: PMC11810459 DOI: 10.20517/cdr.2024.91] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/18/2024] [Revised: 12/18/2024] [Accepted: 12/31/2024] [Indexed: 02/13/2025]
Abstract
Aim: Lung adenocarcinoma (LUAD), the most prevalent subtype of non-small cell lung cancer (NSCLC), presents significant clinical challenges due to its high mortality and limited therapeutic options. The molecular heterogeneity and the development of therapeutic resistance further complicate treatment, underscoring the need for a more comprehensive understanding of its cellular and molecular characteristics. This study sought to delineate novel cellular subpopulations and molecular subtypes of LUAD, identify critical biomarkers, and explore potential therapeutic targets to enhance treatment efficacy and patient prognosis. Methods: An integrative multi-omics approach was employed to incorporate single-cell RNA sequencing (scRNA-seq), bulk transcriptomic analysis, and genome-wide association study (GWAS) data from multiple LUAD patient cohorts. Advanced computational approaches, including Bayesian deconvolution and machine learning algorithms, were used to comprehensively characterize the tumor microenvironment, classify LUAD subtypes, and develop a robust prognostic model. Results: Our analysis identified eleven distinct cellular subpopulations within LUAD, with epithelial cells predominating and exhibiting high mutation frequencies in Tumor Protein 53 (TP53) and Titin (TTN) genes. Two molecular subtypes of LUAD [consensus subtype (CS)1 and CS2] were identified, each showing distinct immune landscapes and clinical outcomes. The CS2 subtype, characterized by increased immune cell infiltration, demonstrated a more favorable prognosis and higher sensitivity to immunotherapy. Furthermore, a multi-omics-driven machine learning signature (MOMLS) identified ribonucleotide reductase M1 (RRM1) as a critical biomarker associated with chemotherapy response. Based on this model, several potential therapeutic agents targeting different subtypes were proposed. Conclusion: This study presents a comprehensive multi-omics framework for understanding the molecular complexity of LUAD, providing insights into cellular heterogeneity, molecular subtypes, and potential therapeutic targets. Differential sensitivity to immunotherapy among various cellular subpopulations was identified, paving the way for future immunotherapy-focused research.
Collapse
Affiliation(s)
- Zilu Chen
- Nanjing University of Chinese Medicine, Nanjing 210023, Jiangsu, China
- Authors contributed equally
| | - Kun Mei
- Nanjing University of Chinese Medicine, Nanjing 210023, Jiangsu, China
- Department of Cardiothoracic Surgery, The Third Affiliated Hospital of Soochow University, Changzhou 213003, Jiangsu, China
- Authors contributed equally
| | - Foxing Tan
- Nanjing University of Chinese Medicine, Nanjing 210023, Jiangsu, China
| | - Yuheng Zhou
- Nanjing University of Chinese Medicine, Nanjing 210023, Jiangsu, China
| | - Haolin Du
- Nanjing University of Chinese Medicine, Nanjing 210023, Jiangsu, China
| | - Min Wang
- Department of Cardiothoracic Surgery, The Third Affiliated Hospital of Soochow University, Changzhou 213003, Jiangsu, China
| | - Renjun Gu
- School of Chinese Medicine and School of Integrated Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing 210023, Jiangsu, China
- Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing 210046, Jiangsu, China
| | - Yan Huang
- Department of Ultrasound, Nanjing Hospital of Chinese Medicine Affiliated with Nanjing University of Chinese Medicine, Nanjing 210022, Jiangsu, China
| |
Collapse
|
18
|
Yin M, Feng C, Yu Z, Zhang Y, Li Y, Wang X, Song C, Guo M, Li C. sc2GWAS: a comprehensive platform linking single cell and GWAS traits of human. Nucleic Acids Res 2025; 53:D1151-D1161. [PMID: 39565208 PMCID: PMC11701642 DOI: 10.1093/nar/gkae1008] [Citation(s) in RCA: 19] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2024] [Revised: 10/01/2024] [Accepted: 10/23/2024] [Indexed: 11/21/2024] Open
Abstract
Identifying cell populations associated with risk variants is essential for uncovering cell-specific mechanisms that drive disease development and progression. Integrating genome-wide association studies (GWAS) with single-cell RNA sequencing (scRNA-seq) has become an effective strategy for detecting trait-cell relationships. The accumulation of trait-related single cell data has led to an urgent need for its comprehensively processing. To address this, we developed sc2GWAS (https://bio.liclab.net/sc2GWAS/), which aims to document large-scale GWAS trait-cell regulatory pairs at single-cell resolution and provide comprehensive annotations and enrichment analyses for these related pairs. The current version of sc2GWAS curates a total of 15 078 310 candidate trait-cell pairs from > 6 300 000 individual cells, offering a valuable resource for exploring complex regulatory relationships between traits and cells. We applied strict quality control measures on both scRNA-seq data and GWAS data, ensuring the reliability and accuracy of the datasets for the identification of trait-relevant cells and genes. In addition, sc2GWAS provides ranked lists of trait-relevant genes and extensive (epi) genetic annotations, making it a valuable resource for downstream analyses. We demonstrate the utility of the platform by investigating Alzheimer's disease, where we identified significant associations between the disease and microglial cells, with the APOE gene emerging as particularly significant. This platform facilitates detailed research into complex trait-cell and trait-gene interactions, we anticipate that sc2GWAS will become a comprehensive and valuable platform for exploring GWAS trait-cell regulatory mechanisms.
Collapse
Affiliation(s)
- Mingxue Yin
- The First Affiliated Hospital & National Health Commission Key Laboratory of Birth Defect Research and Prevention, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, China
- Insititute of Biochemistry and Molecular Biology, Hengyang Medical College, University of South China, Hengyang, Hunan 421001, China
- The First Affiliated Hospital, Cardiovascular Lab of Big Data and Imaging Artificial Intelligence, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, China
- Hunan Provincial Key Laboratory of Multi-omics And Artificial Intelligence of Cardiovascular Diseases, University of South China, Hengyang, Hunan 421001, China
| | - Chenchen Feng
- School of Computer, University of South China, Hengyang, Hunan 421001, China
| | - Zhengmin Yu
- The First Affiliated Hospital & National Health Commission Key Laboratory of Birth Defect Research and Prevention, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, China
- Insititute of Biochemistry and Molecular Biology, Hengyang Medical College, University of South China, Hengyang, Hunan 421001, China
- The First Affiliated Hospital, Cardiovascular Lab of Big Data and Imaging Artificial Intelligence, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, China
- Hunan Provincial Key Laboratory of Multi-omics And Artificial Intelligence of Cardiovascular Diseases, University of South China, Hengyang, Hunan 421001, China
- School of Computer, University of South China, Hengyang, Hunan 421001, China
| | - Yuexin Zhang
- The First Affiliated Hospital & National Health Commission Key Laboratory of Birth Defect Research and Prevention, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, China
- Insititute of Biochemistry and Molecular Biology, Hengyang Medical College, University of South China, Hengyang, Hunan 421001, China
- The First Affiliated Hospital, Cardiovascular Lab of Big Data and Imaging Artificial Intelligence, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, China
- Hunan Provincial Key Laboratory of Multi-omics And Artificial Intelligence of Cardiovascular Diseases, University of South China, Hengyang, Hunan 421001, China
| | - Ye Li
- The First Affiliated Hospital & National Health Commission Key Laboratory of Birth Defect Research and Prevention, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, China
- Insititute of Biochemistry and Molecular Biology, Hengyang Medical College, University of South China, Hengyang, Hunan 421001, China
- The First Affiliated Hospital, Cardiovascular Lab of Big Data and Imaging Artificial Intelligence, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, China
- Hunan Provincial Key Laboratory of Multi-omics And Artificial Intelligence of Cardiovascular Diseases, University of South China, Hengyang, Hunan 421001, China
| | - Xuan Wang
- The First Affiliated Hospital & National Health Commission Key Laboratory of Birth Defect Research and Prevention, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, China
- The First Affiliated Hospital, Cardiovascular Lab of Big Data and Imaging Artificial Intelligence, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, China
- Hunan Provincial Key Laboratory of Multi-omics And Artificial Intelligence of Cardiovascular Diseases, University of South China, Hengyang, Hunan 421001, China
| | - Chao Song
- The First Affiliated Hospital & National Health Commission Key Laboratory of Birth Defect Research and Prevention, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, China
- The First Affiliated Hospital, Cardiovascular Lab of Big Data and Imaging Artificial Intelligence, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, China
- Hunan Provincial Key Laboratory of Multi-omics And Artificial Intelligence of Cardiovascular Diseases, University of South China, Hengyang, Hunan 421001, China
| | - Maozu Guo
- School of Electrical and Information Engineering, Beijing University of Civil Engineering and Architecture, Beijing 100044, China
| | - Chunquan Li
- The First Affiliated Hospital & National Health Commission Key Laboratory of Birth Defect Research and Prevention, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, China
- Insititute of Biochemistry and Molecular Biology, Hengyang Medical College, University of South China, Hengyang, Hunan 421001, China
- The First Affiliated Hospital, Cardiovascular Lab of Big Data and Imaging Artificial Intelligence, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, China
- Hunan Provincial Key Laboratory of Multi-omics And Artificial Intelligence of Cardiovascular Diseases, University of South China, Hengyang, Hunan 421001, China
- School of Computer, University of South China, Hengyang, Hunan 421001, China
- The First Affiliated Hospital, Institute of Cardiovascular Disease, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, China
- The First Affiliated Hospital, Department of Cardiology, Hengyang Medical School, University of South China, Hengyang, China
- Department of Cell Biology and Genetics, School of Basic Medical Sciences, Hengyang Medical School, University of South China, Hengyang, Hunan421001, China
- Key Laboratory of Rare Pediatric Diseases, Ministry of Education, University of South China, Hengyang, Hunan 421001, China
| |
Collapse
|
19
|
Yu YF, Gong HF, Li WJ, Wu M, Hu G. Exploring the causal relationship of gut microbiota in nonunion: a Mendelian randomization analysis mediated by immune cell. Front Microbiol 2024; 15:1447877. [PMID: 39736989 PMCID: PMC11683590 DOI: 10.3389/fmicb.2024.1447877] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2024] [Accepted: 11/25/2024] [Indexed: 01/01/2025] Open
Abstract
Background Emerging research indicates that gut microbiota (GM) are pivotal in the regulation of immune-mediated bone diseases. Nonunion, a bone metabolic disorder, has an unclear causal relationship with GM and immune cells. This study aims to elucidate the causal relationship between GM and nonunion using Mendelian Randomization (MR) and to explore the mediating role of immune cells. Methods Using a two-step, two-sample Mendelian randomization approach, this study explores the causal link between GM and nonunion, as well as the mediating role of immune cells in this relationship. Data were sourced from multiple cohorts and consortiums, including the MiBioGen consortium. GM data were derived from a recently published dataset of 473 gut microbiota, and nonunion data were obtained from genome-wide association studies (GWAS). Results MR analysis identified 12 bacterial genera with protective effects against nonunion and seven bacterial genera associated with a higher risk of nonunion, including Agathobacter sp000434275, Aureimonas, Clostridium M, Lachnospirales, Megamonas funiformis, and Peptoccia. Reverse MR analysis indicated that nonunion does not influence GM. Additionally, MR analysis identified 12 immune cell types positively associated with nonunion and 14 immune cell types negatively associated with nonunion. Building on these findings, we conducted mediation MR analysis to identify 24 crucial GM and immune cell-mediated relationships affecting nonunion. Notably, Campylobacter D, Megamonas funiformis, Agathobacter sp000434275, Lachnospirales, Clostridium E sporosphaeroides, and Clostridium M significantly regulated nonunion through multiple immune cell characteristics. Conclusions To our knowledge, our research results are the first to emphasize a causal relationship between the gut microbiome and nonunion, potentially mediated by immune cells. The correlations and mediation effects identified in our study provide valuable insights into potential therapeutic strategies targeting the gut microbiome, informing global action plans.
Collapse
Affiliation(s)
- Yun-fei Yu
- Department of Orthopedics, Wuxi Hospital of Traditional Chinese Medicine, Wuxi, Jiangsu, China
| | - Hai-Feng Gong
- Department of Trauma Surgery, Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| | - Wei-Ju Li
- Department of Orthopedics, Guangdong Provincial Second Hospital of Traditional Chinese Medicine, Guangzhou, Guangdong, China
| | - Mao Wu
- Department of Orthopedics, Wuxi Hospital of Traditional Chinese Medicine, Wuxi, Jiangsu, China
| | - Gang Hu
- Department of Orthopedics, Wuxi Hospital of Traditional Chinese Medicine, Wuxi, Jiangsu, China
| |
Collapse
|
20
|
Townsend HA, Rosenberger KJ, Vanderlinden LA, Inamo J, Zhang F. Evaluating methods for integrating single-cell data and genetics to understand inflammatory disease complexity. Front Immunol 2024; 15:1454263. [PMID: 39703500 PMCID: PMC11655331 DOI: 10.3389/fimmu.2024.1454263] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2024] [Accepted: 11/07/2024] [Indexed: 12/21/2024] Open
Abstract
Background Understanding genetic underpinnings of immune-mediated inflammatory diseases is crucial to improve treatments. Single-cell RNA sequencing (scRNA-seq) identifies cell states expanded in disease, but often overlooks genetic causality due to cost and small genotyping cohorts. Conversely, large genome-wide association studies (GWAS) are commonly accessible. Methods We present a 3-step robust benchmarking analysis of integrating GWAS and scRNA-seq to identify genetically relevant cell states and genes in inflammatory diseases. First, we applied and compared the results of three recent algorithms, based on pathways (scGWAS), single-cell disease scores (scDRS), or both (scPagwas), according to accuracy/sensitivity and interpretability. While previous studies focused on coarse cell types, we used disease-specific, fine-grained single-cell atlases (183,742 and 228,211 cells) and GWAS data (Ns of 97,173 and 45,975) for rheumatoid arthritis (RA) and ulcerative colitis (UC). Second, given the lack of scRNA-seq for many diseases with GWAS, we further tested the tools' resolution limits by differentiating between similar diseases with only one fine-grained scRNA-seq atlas. Lastly, we provide a novel evaluation of noncoding SNP incorporation methods by testing which enabled the highest sensitivity/accuracy of known cell-state calls. Results We first found that single-cell based tools scDRS and scPagwas called superior numbers of supported cell states that were overlooked by scGWAS. While scGWAS and scPagwas were advantageous for gene exploration, scDRS effectively accounted for batch effect and captured cellular heterogeneity of disease-relevance without single-cell genotyping. For noncoding SNP integration, we found a key trade-off between statistical power and confidence with positional (e.g. MAGMA) and non-positional approaches (e.g. chromatin-interaction, eQTL). Even when directly incorporating noncoding SNPs through 5' scRNA-seq measures of regulatory elements, non disease-specific atlases gave misleading results by not containing disease-tissue specific transcriptomic patterns. Despite this criticality of tissue-specific scRNA-seq, we showed that scDRS enabled deconvolution of two similar diseases with a single fine-grained scRNA-seq atlas and separate GWAS. Indeed, we identified supported and novel genetic-phenotype linkages separating RA and ankylosing spondylitis, and UC and crohn's disease. Overall, while noting evolving single-cell technologies, our study provides key findings for integrating expanding fine-grained scRNA-seq, GWAS, and noncoding SNP resources to unravel the complexities of inflammatory diseases.
Collapse
Affiliation(s)
- Hope A. Townsend
- Biofrontiers Institute, University of Colorado Boulder, Boulder, CO, United States
- Department of Molecular, Cellular, Developmental Biology, University of Colorado Boulder, Boulder, CO, United States
| | - Kaylee J. Rosenberger
- Biofrontiers Institute, University of Colorado Boulder, Boulder, CO, United States
- Department of Ecology and Evolutionary Biology, University of Colorado Boulder, Boulder, CO, United States
| | - Lauren A. Vanderlinden
- Department of Medicine, Division of Rheumatology, University of Colorado Anschutz Medical Campus, Denver, CO, United States
- Department of Biomedical Informatics, Center for Health AI, University of Colorado Anschutz Medical Campus, Denver, CO, United States
| | - Jun Inamo
- Department of Medicine, Division of Rheumatology, University of Colorado Anschutz Medical Campus, Denver, CO, United States
- Department of Biomedical Informatics, Center for Health AI, University of Colorado Anschutz Medical Campus, Denver, CO, United States
| | - Fan Zhang
- Biofrontiers Institute, University of Colorado Boulder, Boulder, CO, United States
- Department of Medicine, Division of Rheumatology, University of Colorado Anschutz Medical Campus, Denver, CO, United States
- Department of Biomedical Informatics, Center for Health AI, University of Colorado Anschutz Medical Campus, Denver, CO, United States
| |
Collapse
|
21
|
Huang H, Zheng H. Mendelian randomization study of the relationship between blood and urine biomarkers and lung cancer. Front Oncol 2024; 14:1453246. [PMID: 39687887 PMCID: PMC11646849 DOI: 10.3389/fonc.2024.1453246] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2024] [Accepted: 11/11/2024] [Indexed: 12/18/2024] Open
Abstract
Introduction Identifying suitable biomarkers is crucial for exploring the pathogenesis, early screening, and therapeutic monitoring of lung cancer. This study aims to analyze comprehensively the associations between lung cancer and biomarkers in blood and urine. Methods Bidirectional two-sample Mendelian randomization (MR) was used to evaluate the potential causal relationships between blood and urine biomarkers and lung cancer. We obtained Single nucleotide polymorphisms (SNPs) related to lung cancer from the 2021 Finnish database of genome-wide association studies, including small cell lung cancer (SCLC), total non-small cell lung cancer (NSCLC), lung adenocarcinoma (LAC), and lung squamous cell carcinoma (LSCC).Data on blood and urine biomarkers were derived from the UK Biobank cohort, comprising 376,807 participants. Results We found a potential inverse causal relationship between total bilirubin and SCLC (β=-0.285, P=0.015, FDR=0.12). Urate was inversely associated with NSCLC (β=-0.158, P=0.004, FDR=0.036*). Serum calcium showed a possible inverse relationship with lung squamous cell carcinoma (β=-0.256, P=0.046, FDR=0.138), while urinary creatinine was positively associated (β=1.233, P=0.024, FDR=0.216). Non-albumin proteins (β=-0.272, P=0.020, FDR=0.180) and total protein (β=-0.402, P=0.009, FDR=0.072) were inversely related to lung squamous cell carcinoma. The AST/ALT ratio was positively associated with lung adenocarcinoma (β=0.293, P=0.009, FDR=0.072). Our reverse Mendelian randomization study found a positive causal association between small cell lung cancer and serum creatinine (β=0.022, P=0.002, FDR=0.018*), while it was inversely associated with the estimated glomerular filtration rate(eGFR)(β=-0.022, P=0.003, FDR=0.027*). A positive causal relationship was also observed with cystatin C (β=0.026, P=0.005, FDR=0.045*) and glycated hemoglobin HbA1c (β=0.013, P=0.014, FDR=0.028*). A negative causal relationship was observed with Gamma_glutamyltransferase (β=-0.013, P=0.019, FDR=0.152). For non-small cell lung cancer, a negative causal relationship was found with albumin (β=-0.024, P=0.002, FDR=0.016*), while a potentially positive causal relationship was observed with cystatin C (β=0.022, P=0.006, FDR=0.054). Possible negative causal relationships were also observed with phosphate (β=-0.013, P=0.008, FDR=0.072) and urinary potassium (β=-0.011, P=0.012, FDR=0.108), while a potential positive causal relationship was observed with C-reactive protein (β=0.013, P=0.040, FDR=0.280).Regarding lung squamous cell carcinoma, an inverse causal relationship was found with eGFR (β=-0.022, P=9.58e-06, FDR=8.62×10-5*), while a positive causal relationship was observed with serum creatinine (β=0.021, P=1.16e-4, FDR=1.05×10-3*). Potential positive causal relationships were observed with Urate (β=0.012, P=0.020, FDR=0.180), urea (β=0.010, P=0.046, FDR=0.141), and glycated hemoglobin HbA1c (β=0.020, P=0.049, FDR P=0.098), whereas a potential negative causal relationship was observed with sex hormone-binding globulin(SHBG) (β=-0.020, P=0.036, FDR=0.108).Lastly, adenocarcinoma was found to have a positive causal association with alkaline phosphatase (β=0.015, P=0.006, FDR=0.033*). Conclusion Our study provides a robust theoretical basis for the early screening and therapeutic monitoring of lung cancer and contributes to understanding the pathogenesis of the disease.
Collapse
Affiliation(s)
| | - Haijun Zheng
- The First People's Hospital of Chenzhou, Chenzhou, China
| |
Collapse
|
22
|
Ye L, Ye C, Li P, Wang Y, Ma W. Inferring the genetic relationships between unsupervised deep learning-derived imaging phenotypes and glioblastoma through multi-omics approaches. Brief Bioinform 2024; 26:bbaf037. [PMID: 39879386 PMCID: PMC11775472 DOI: 10.1093/bib/bbaf037] [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: 10/23/2024] [Revised: 12/20/2024] [Accepted: 01/15/2025] [Indexed: 01/31/2025] Open
Abstract
This study aimed to investigate the genetic association between glioblastoma (GBM) and unsupervised deep learning-derived imaging phenotypes (UDIPs). We employed a combination of genome-wide association study (GWAS) data, single-nucleus RNA sequencing (snRNA-seq), and scPagwas (pathway-based polygenic regression framework) methods to explore the genetic links between UDIPs and GBM. Two-sample Mendelian randomization analyses were conducted to identify causal relationships between UDIPs and GBM. Colocalization analysis was performed to validate genetic associations, while scPagwas analysis was used to evaluate the relevance of key UDIPs to GBM at the cellular level. Among 512 UDIPs tested, 23 were found to have significant causal associations with GBM. Notably, UDIPs such as T1-33 (OR = 1.007, 95% CI = 1.001 to 1.012, P = .022), T1-34 (OR = 1.012, 95% CI = 1.001-1.023, P = .028), and T1-96 (OR = 1.009, 95% CI = 1.001-1.019, P = .046) were found to have a genetic association with GBM. Furthermore, T1-34 and T1-96 were significantly associated with GBM recurrence, with P-values < .0001 and P < .001, respectively. In addition, scPagwas analysis revealed that T1-33, T1-34, and T1-96 are distinctively linked to different GBM subtypes, with T1-33 showing strong associations with the neural progenitor-like subtype (NPC2), T1-34 with mesenchymal (MES2) and neural progenitor (NPC1) cells, and T1-96 with the NPC2 subtype. T1-33, T1-34, and T1-96 hold significant potential for predicting tumor recurrence and aiding in the development of personalized GBM treatment strategies.
Collapse
Affiliation(s)
- Liguo Ye
- Department of Neurosurgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100730, China
| | - Cheng Ye
- Department of Neurosurgery, Xuanwu Hospital, Capital Medical University, Beijing, People's Republic of China
| | - Pengtao Li
- Department of Neurosurgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100730, China
| | - Yu Wang
- Department of Neurosurgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100730, China
| | - Wenbin Ma
- Department of Neurosurgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100730, China
| |
Collapse
|
23
|
Zaied RE, Gokuladhas S, Walker C, O’Sullivan JM. Unspecified asthma, childhood-onset, and adult-onset asthma have different causal genes: a Mendelian randomization analysis. Front Immunol 2024; 15:1412032. [PMID: 39628479 PMCID: PMC11611866 DOI: 10.3389/fimmu.2024.1412032] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2024] [Accepted: 10/28/2024] [Indexed: 12/06/2024] Open
Abstract
Introduction Asthma is a heterogeneous condition that is characterized by reversible airway obstruction. Childhood-onset asthma (COA) and adult-onset asthma (AOA) are two prominent asthma subtypes, each with unique etiological factors and prognosis, which suggests the existence of both shared and distinct risk factors. Methods Here, we employed a two-sample Mendelian randomization analysis to elucidate the causal association between genes within lung and whole-blood-specific gene regulatory networks (GRNs) and the development of unspecified asthma, COA, and AOA using the Wald ratio method. Lung and whole blood-specific GRNs, encompassing spatial eQTLs (instrumental variables) and their target genes (exposures), were utilized as exposure data. Genome-wide association studies for unspecified asthma, COA, and AOA were used as outcome data in this investigation. Results We identified 101 genes that were causally linked to unspecified asthma, 39 genes causally associated with COA, and ten genes causally associated with AOA. Among the identified genes, 29 were shared across some, or all of the asthma subtypes. Of the identified causal genes, ORMDL3 had the strongest causal association with both unspecified asthma (OR: 1.49; 95% CI:1.42-1.57; p=7.30x10-51) and COA (OR: 3.37; 95% CI: 3.02-3.76; p=1.95x10-102), whereas PEBP1P3 had the strongest causal association with AOA (OR: 1.28; 95% CI: 1.16-1.41; p=0.007). Discussion This study identified shared and unique genetic factors causally associated with different asthma subtypes. In so doing, our study emphasizes the need to move beyond perceiving asthma as a singular condition to enable the development of therapeutic interventions that target sub-type specific causal genes.
Collapse
Affiliation(s)
- Roan E. Zaied
- The Liggins Institute, The University of Auckland, Auckland, New Zealand
| | - Sreemol Gokuladhas
- The Liggins Institute, The University of Auckland, Auckland, New Zealand
| | - Caroline Walker
- Faculty of Medical and Health Sciences, The University of Auckland, Auckland, New Zealand
| | - Justin M. O’Sullivan
- The Liggins Institute, The University of Auckland, Auckland, New Zealand
- The Maurice Wilkins Centre, The University of Auckland, Auckland, New Zealand
- Australian Parkinsons Mission, Garvan Institute of Medical Research, Sydney, NSW, Australia
- MRC Lifecourse Epidemiology Unit, University of Southampton, Southampton, United Kingdom
- Singapore Institute for Clinical Sciences, Agency for Science Technology and Research, Singapore, Singapore
| |
Collapse
|
24
|
Huang Z, Zheng Y, Wang W, Zhou W, Zhang Y, Wei C, Zhang X, Jin X, Yin J. Uncovering disease-related multicellular pathway modules on large-scale single-cell transcriptomes with scPAFA. Commun Biol 2024; 7:1523. [PMID: 39550507 PMCID: PMC11569158 DOI: 10.1038/s42003-024-07238-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2024] [Accepted: 11/08/2024] [Indexed: 11/18/2024] Open
Abstract
Pathway analysis is a crucial analytical phase in disease research on single-cell RNA sequencing (scRNA-seq) data, offering biological interpretations based on prior knowledge. However, currently available tools for generating cell-level pathway activity scores (PAS) exhibit computational inefficacy in large-scale scRNA-seq datasets. Additionally, disease-related pathways are often identified through cross-condition comparisons within specific cell types, overlooking potential patterns that involve multiple cell types. Here, we present single-cell pathway activity factor analysis (scPAFA), a Python library designed for large-scale single-cell datasets allowing rapid PAS computation and uncovering biologically interpretable disease-related multicellular pathway modules, which are low-dimensional representations of disease-related PAS alterations in multiple cell types. Application on colorectal cancer (CRC) datasets and large-scale lupus atlas over 1.2 million cells demonstrated that scPAFA can achieve over 40-fold reductions in the runtime of PAS computation and further identified reliable and interpretable multicellular pathway modules that capture the heterogeneity of CRC and transcriptional abnormalities in lupus patients, respectively. Overall, scPAFA presents a valuable addition to existing research tools in disease research, with the potential to reveal complex disease mechanisms and support biomarker discovery at the pathway level.
Collapse
Affiliation(s)
- Zhuoli Huang
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, 100049, China
- BGI Research, Shenzhen, 518083, China
| | - Yuhui Zheng
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, 100049, China
- BGI Research, Shenzhen, 518083, China
| | - Weikai Wang
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, 100049, China
- BGI Research, Shenzhen, 518083, China
| | - Wenwen Zhou
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, 100049, China
- BGI Research, Shenzhen, 518083, China
| | - Yanbo Zhang
- Shanxi Medical University-BGI Collaborative Center for Future Medicine, Shanxi Medical University, Taiyuan, 030001, China
- Department of Health Statistics, School of Public Health, Shanxi Medical University, Taiyuan, 030001, China
| | - Chen Wei
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, 100049, China
- BGI Research, Shenzhen, 518083, China
| | - Xiuqing Zhang
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, 100049, China
- BGI Research, Shenzhen, 518083, China
| | - Xin Jin
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, 100049, China.
- BGI Research, Shenzhen, 518083, China.
- Shanxi Medical University-BGI Collaborative Center for Future Medicine, Shanxi Medical University, Taiyuan, 030001, China.
| | - Jianhua Yin
- BGI Research, Shenzhen, 518083, China.
- Shanxi Medical University-BGI Collaborative Center for Future Medicine, Shanxi Medical University, Taiyuan, 030001, China.
| |
Collapse
|
25
|
Yao X, Li Z, Lei Y, Liu Q, Chen S, Zhang H, Dong X, He K, Guo J, Li MJ, Wang X, Yan H. Single-Cell Multiomics Profiling Reveals Heterogeneity of Müller Cells in the Oxygen-Induced Retinopathy Model. Invest Ophthalmol Vis Sci 2024; 65:8. [PMID: 39504047 PMCID: PMC11547256 DOI: 10.1167/iovs.65.13.8] [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: 01/16/2024] [Accepted: 09/28/2024] [Indexed: 11/10/2024] Open
Abstract
Purpose Retinal neovascularization poses heightened risks of vision loss and blindness. Despite its clinical significance, the molecular mechanisms underlying the pathogenesis of retinal neovascularization remain elusive. This study utilized single-cell multiomics profiling in an oxygen-induced retinopathy (OIR) model to comprehensively investigate the intricate molecular landscape of retinal neovascularization. Methods Mice were exposed to hyperoxia to induce the OIR model, and retinas were isolated for nucleus isolation. The cellular landscape of the single-nucleus suspensions was extensively characterized through single-cell multiomics sequencing. Single-cell data were integrated with genome-wide association study (GWAS) data to identify correlations between ocular cell types and diabetic retinopathy. Cell communication analysis among cells was conducted to unravel crucial ligand-receptor signals. Trajectory analysis and dynamic characterization of Müller cells were performed, followed by integration with human retinal data for pathway analysis. Results The multiomics dataset revealed six major ocular cell classes, with Müller cells/astrocytes showing significant associations with proliferative diabetic retinopathy (PDR). Cell communication analysis highlighted pathways that are associated with vascular proliferation and neurodevelopment, such as Vegfa-Vegfr2, Igf1-Igf1r, Nrxn3-Nlgn1, and Efna5-Epha4. Trajectory analysis identified a subset of Müller cells expressing genes linked to photoreceptor degeneration. Multiomics data integration further unveiled positively regulated genes in OIR Müller cells/astrocytes associated with axon development and neurotransmitter transmission. Conclusions This study significantly advances our understanding of the intricate cellular and molecular mechanisms underlying retinal neovascularization, emphasizing the pivotal role of Müller cells. The identified pathways provide valuable insights into potential therapeutic targets for PDR, offering promising directions for further research and clinical interventions.
Collapse
Affiliation(s)
- Xueming Yao
- Department of Ophthalmology, Tianjin Medical University General Hospital, Tianjin, China
- School of Medicine, Nankai University, Tianjin, China
| | - Ziqi Li
- Department of Ophthalmology, Tianjin Medical University General Hospital, Tianjin, China
- Laboratory of Molecular Ophthalmology, Tianjin Medical University, Tianjin, China
| | - Yi Lei
- Department of Pharmacology, Tianjin Key Laboratory of Inflammation Biology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
- Laboratory of Molecular Ophthalmology, Tianjin Medical University, Tianjin, China
| | - Qiangyun Liu
- Department of Ophthalmology, Tianjin Medical University General Hospital, Tianjin, China
- Laboratory of Molecular Ophthalmology, Tianjin Medical University, Tianjin, China
| | - Siyue Chen
- Department of Ophthalmology, Tianjin Medical University General Hospital, Tianjin, China
- Laboratory of Molecular Ophthalmology, Tianjin Medical University, Tianjin, China
| | - Haokun Zhang
- Laboratory of Molecular Ophthalmology, Tianjin Medical University, Tianjin, China
| | - Xue Dong
- Department of Pharmacology, Tianjin Key Laboratory of Inflammation Biology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
- Laboratory of Molecular Ophthalmology, Tianjin Medical University, Tianjin, China
| | - Kai He
- Department of Ophthalmology, Tianjin Medical University General Hospital, Tianjin, China
- Laboratory of Molecular Ophthalmology, Tianjin Medical University, Tianjin, China
| | - Ju Guo
- Department of Ophthalmology, Tianjin Medical University General Hospital, Tianjin, China
| | - Mulin Jun Li
- Department of Bioinformatics, The Province and Ministry Co-Sponsored Collaborative Innovation Center for Medical Epigenetics, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
| | - Xiaohong Wang
- Department of Pharmacology, Tianjin Key Laboratory of Inflammation Biology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
- Laboratory of Molecular Ophthalmology, Tianjin Medical University, Tianjin, China
| | - Hua Yan
- Department of Ophthalmology, Tianjin Medical University General Hospital, Tianjin, China
- Laboratory of Molecular Ophthalmology, Tianjin Medical University, Tianjin, China
- School of Medicine, Nankai University, Tianjin, China
| |
Collapse
|
26
|
Huang Z, Peng S, Cen T, Wang X, Ma L, Cao Z. Association between biological ageing and periodontitis: Evidence from a cross-sectional survey and multi-omics Mendelian randomization analysis. J Clin Periodontol 2024; 51:1369-1383. [PMID: 38956929 DOI: 10.1111/jcpe.14040] [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: 03/04/2024] [Revised: 06/07/2024] [Accepted: 06/21/2024] [Indexed: 07/04/2024]
Abstract
AIM To investigate the relationship and potential causality between biological ageing and periodontitis. MATERIALS AND METHODS We obtained the National Health and Nutrition Examination Survey (NHANES) and genome-wide association study (GWAS) summary statistics as well as single-cell sequencing data. Multivariate regression analysis based on cross-sectional data, Mendelian randomization (MR) and multi-omics integration analysis were employed to explore the causal association and potential molecular mechanisms between biological ageing and periodontitis. Additionally, two-step MR mediation analysis explored the risk factors in biological ageing-mediated periodontitis. RESULTS We analysed data from 3189 participants in the NHANES data and found that higher biological age was associated with increased risk of periodontitis. MR analyses revealed causal associations between biological age measures and periodontitis risk. Frailty (odds ratio [OR] = 2.08, 95% confidence interval [CI]: 1.04-4.18, p = .039) and GrimAge acceleration (OR = 1.16, 95% CI: 1.01-1.32, p = .033) were causally associated with periodontitis risk, and these results were validated in a large-scale meta-periodontitis GWAS dataset. Additionally, the risk effects of body mass index, waist circumference and lifetime smoking on periodontitis were partially mediated by frailty and GrimAge acceleration. CONCLUSIONS Evidence from cross-sectional survey and MR analysis suggests that biological ageing increases the risk of periodontitis. Additionally, improving the associated risk factors can help prevent both ageing and periodontitis.
Collapse
Affiliation(s)
- Zhendong Huang
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Simin Peng
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Ting Cen
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Xiaoxuan Wang
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, China
- Department of Periodontology, School & Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Li Ma
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, China
- Department of Periodontology, School & Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Zhengguo Cao
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, China
- Department of Periodontology, School & Hospital of Stomatology, Wuhan University, Wuhan, China
| |
Collapse
|
27
|
Kong Y, Jiang J, Kong W, Qin S. DRCTdb: disease-related cell type analysis to decode cell type effect and underlying regulatory mechanisms. Commun Biol 2024; 7:1205. [PMID: 39341994 PMCID: PMC11439014 DOI: 10.1038/s42003-024-06833-y] [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: 04/10/2024] [Accepted: 09/03/2024] [Indexed: 10/01/2024] Open
Abstract
Understanding the molecular mechanisms underlying genetic diseases is challenging due to environmental and genetic factors. Genome-wide association studies (GWAS) have identified numerous genetic loci, but their functional implications are largely unknown. Single-cell multiomics sequencing has emerged as a powerful tool to study disease-specific cell types and their relationship with genetic variants. However, comprehensive databases for exploring these mechanisms across different tissues are lacking. We present the Disease-Related Cell Type database (DRCTdb), integrating GWAS and single-cell multiomics data to identify disease-related cell types and elucidate their regulatory mechanisms. DRCTdb contains well-processed data from 16 studies, covering 4 million cells within 28 tissues. Users can browse relationships and regulatory mechanisms between SNPs of 42 genetic diseases and cell types based on GWAS and single-cell data. DRCTdb also offers data downloads and is available at https://singlecellatlas.top/DRCTDB .
Collapse
Affiliation(s)
- Yunhui Kong
- Jiangsu Key Laboratory of Sericultural Biology and Biotechnology, School of Biotechnology, Jiangsu University of Science and Technology, Zhenjiang, China
- Institute of Modern Biology, Nanjing University, Nanjing, China
| | - Junyao Jiang
- School of Life Sciences, Westlake University, Hangzhou, China.
| | - Weikang Kong
- School of Environmental Science and Engineering, University of Science and Technology of Suzhou, Suzhou, Jiangsu, China
| | - Sheng Qin
- Jiangsu Key Laboratory of Sericultural Biology and Biotechnology, School of Biotechnology, Jiangsu University of Science and Technology, Zhenjiang, China.
- Key Laboratory of Silkworm and Mulberry Genetic Improvement, Ministry of Agriculture and Rural Affairs, Sericultural Research Institute, Chinese Academy of Agricultural Science, Zhenjiang, China.
| |
Collapse
|
28
|
Fu C, Zhao Y, Zhou X, Lv J, Jin S, Zhou Y, Liu F, Feng N. Gut microbiota and interstitial cystitis: exploring the gut-bladder axis through mendelian randomization, biological annotation and bulk RNA sequencing. Front Immunol 2024; 15:1395580. [PMID: 39399486 PMCID: PMC11466805 DOI: 10.3389/fimmu.2024.1395580] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2024] [Accepted: 09/10/2024] [Indexed: 10/15/2024] Open
Abstract
Background Several observational studies have indicated an association between interstitial cystitis and the composition of the gut microbiota; however, the causality and underlying mechanisms remain unclear. Understanding the link between gut microbiota and interstitial cystitis could inform strategies for prevention and treatment. Methods A two-sample Mendelian randomization analysis was conducted using published genome-wide association study summary statistics. We employed inverse variance weighted, weighted mode, MR-Egger, weighted median, simple mode, and cML-MA methods to investigate the causal relationship between gut microbiota and interstitial cystitis. Sensitivity analysis was performed to validate the results. Relevant gut microbiota was examined through reverse MR. Single nucleotide polymorphisms were annotated using FUMA to identify genes associated with these genetic variants, thereby revealing potential host gene-microbiota associations in interstitial cystitis patients. Results Eight bacterial taxa were identified in our analysis as associated with interstitial cystitis. Among these, Butyricimonas, Coprococcus, Lactobacillales, Lentisphaerae, and Bilophila wadsworthia were positively correlated with interstitial cystitis risk, while taxa such as Desulfovibrio piger, Oscillibacter unclassified and Ruminococcus lactaris exhibited protective effects against interstitial cystitis. The robustness of these associations was confirmed through sensitivity analyses. Reverse MR analysis did not reveal evidence of reverse causality. Single nucleotide polymorphisms were annotated using FUMA and subjected to biological analysis. Seven hub genes (SPTBN1, PSME4, CHAC2, ERLEC1, ASB3, STAT5A, and STAT3) were identified as differentially expressed between interstitial cystitis patients and healthy individuals, representing potential therapeutic targets. Conclusion Our two-sample Mendelian randomization study established a causal relationship between gut microbiota and interstitial cystitis. Furthermore, our identification of a host gene-microbiota association offers a new avenue for investigating the potential pathogenesis of interstitial cystitis and suggests avenues for the development of personalized treatment strategies.
Collapse
Affiliation(s)
- Chaowei Fu
- Jiangnan University Medical Center, Wuxi School of Medicine, Jiangnan University, Wuxi, Jiangsu, China
| | - Yu Zhao
- Jiangnan University Medical Center, Wuxi School of Medicine, Jiangnan University, Wuxi, Jiangsu, China
| | - Xiang Zhou
- Department of Urology, Jiangnan University Medical Center, Wuxi, Jiangsu, China
| | - Jing Lv
- Jiangnan University Medical Center, Wuxi School of Medicine, Jiangnan University, Wuxi, Jiangsu, China
| | - Shengkai Jin
- Jiangnan University Medical Center, Wuxi School of Medicine, Jiangnan University, Wuxi, Jiangsu, China
| | - Yuhua Zhou
- Jiangnan University Medical Center, Wuxi School of Medicine, Jiangnan University, Wuxi, Jiangsu, China
| | - Fengping Liu
- Jiangnan University Medical Center, Wuxi School of Medicine, Jiangnan University, Wuxi, Jiangsu, China
| | - Ninghan Feng
- Jiangnan University Medical Center, Wuxi School of Medicine, Jiangnan University, Wuxi, Jiangsu, China
- Department of Urology, Jiangnan University Medical Center, Wuxi, Jiangsu, China
| |
Collapse
|
29
|
Du J, Baranova A, Zhang G, Zhang F. The causal relationship between immune cell traits and schizophrenia: a Mendelian randomization analysis. Front Immunol 2024; 15:1452214. [PMID: 39399496 PMCID: PMC11466782 DOI: 10.3389/fimmu.2024.1452214] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2024] [Accepted: 09/09/2024] [Indexed: 10/08/2024] Open
Abstract
INTRODUCTION The complex and unresolved pathogenesis of schizophrenia has posed significant challenges to its diagnosis and treatment. While recent research has established a clear association between immune function and schizophrenia, the causal relationship between the two remains elusive. METHODS We employed a bidirectional two-sample Mendelian randomization approach to investigate the causal relationship between schizophrenia and 731 immune cell traits by utilizing public GWAS data. We further validated the causal relationship between schizophrenia and six types of white cell measures. RESULTS We found the overall causal effects of schizophrenia on immune cell traits were significantly higher than the reverse ones (0.011 ± 0.049 vs 0.001 ± 0.016, p < 0.001), implying that disease may lead to an increase in immune cells by itself. We also identified four immune cell traits that may increase the risk of schizophrenia: CD11c+ monocyte %monocyte (odds ratio (OR): 1.06, 95% confidence interval (CI): 1.03~1.09, FDR = 0.027), CD11c+ CD62L- monocyte %monocyte (OR:1.06, 95% CI: 1.03~1.09, FDR = 0.027), CD25 on IgD+ CD38- naive B cell (OR:1.03, 95% CI:1.01~1.06, FDR = 0.042), and CD86 on monocyte (OR = 1.04, 95% CI:1.01~1.06, FDR = 0.042). However, we did not detect any significant causal effects of schizophrenia on immune cell traits. Using the white blood cell traits data, we identified that schizophrenia increases the lymphocyte counts (OR:1.03, 95%CI: 1.01-1.04, FDR = 0.007), total white blood cell counts (OR:1.02, 95%CI: 1.01-1.04, FDR = 0.021) and monocyte counts (OR:1.02, 95%CI: 1.00-1.03, FDR = 0.034). The lymphocyte counts were nominally associated with the risk of schizophrenia (OR:1.08,95%CI:1.01-1.16, P=0.019). DISCUSSION Our study found that the causal relationship between schizophrenia and the immune system is complex, enhancing our understanding of the role of immune regulation in the development of this disorder. These findings offer new insights for exploring diagnostic and therapeutic options for schizophrenia.
Collapse
Affiliation(s)
- Jianbin Du
- Department of Geriatric Psychiatry, The Affiliated Mental Health Center of Jiangnan University, Wuxi Central Rehabilitation Hospital, Wuxi, Jiangsu, China
| | - Ancha Baranova
- School of Systems Biology, George Mason University, Fairfax, VA, United States
- Research Centre for Medical Genetics, Moscow, Russia
| | - Guofu Zhang
- Department of Geriatric Psychiatry, The Affiliated Mental Health Center of Jiangnan University, Wuxi Central Rehabilitation Hospital, Wuxi, Jiangsu, China
| | - Fuquan Zhang
- Department of Psychiatry, The Affiliated Brain Hospital of Nanjing Medical University, Nanjing, China
- Institute of Neuropsychiatry, The Affiliated Brain Hospital of Nanjing Medical University, Nanjing, China
| |
Collapse
|
30
|
Liu C, Zhang Y, Liang Y, Zhang T, Wang G. DrugReSC: targeting disease-critical cell subpopulations with single-cell transcriptomic data for drug repurposing in cancer. Brief Bioinform 2024; 25:bbae490. [PMID: 39350337 PMCID: PMC11442150 DOI: 10.1093/bib/bbae490] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2024] [Revised: 08/25/2024] [Accepted: 09/17/2024] [Indexed: 10/04/2024] Open
Abstract
The field of computational drug repurposing aims to uncover novel therapeutic applications for existing drugs through high-throughput data analysis. However, there is a scarcity of drug repurposing methods leveraging the cellular-level information provided by single-cell RNA sequencing data. To address this need, we propose DrugReSC, an innovative approach to drug repurposing utilizing single-cell RNA sequencing data, intending to target specific cell subpopulations critical to disease pathology. DrugReSC constructs a drug-by-cell matrix representing the transcriptional relationships between individual cells and drugs and utilizes permutation-based methods to assess drug contributions to cellular phenotypic changes. We demonstrate DrugReSC's superior performance compared to existing drug repurposing methods based on bulk or single-cell RNA sequencing data across multiple cancer case studies. In summary, DrugReSC offers a novel perspective on the utilization of single-cell sequencing data in drug repurposing methods, contributing to the advancement of precision medicine for cancer.
Collapse
Affiliation(s)
- Chonghui Liu
- College of Life Science, Northeast Forestry University, 26 Hexing Road, Xiangfang District, Harbin 150040, China
- College of Computer and Control Engineering, Northeast Forestry University, 26 Hexing Road, Xiangfang District, Harbin 150040, China
| | - Yan Zhang
- Kunming Institute of Zoology, Chinese Academy of Sciences, 17 Longxin Road, Panlong District, Kunming 650201, Yunnan, China
- University of Chinese Academy of Sciences, 1 Yanxi Lake East Road, Huairou District, Beijing 100049, China
| | - Yingjian Liang
- Department of General Surgery, the First Affiliated Hospital of Harbin Medical University, 23 Youzheng Street, Nangang District, Harbin 150007, China
| | - Tianjiao Zhang
- College of Computer and Control Engineering, Northeast Forestry University, 26 Hexing Road, Xiangfang District, Harbin 150040, China
| | - Guohua Wang
- College of Computer and Control Engineering, Northeast Forestry University, 26 Hexing Road, Xiangfang District, Harbin 150040, China
| |
Collapse
|
31
|
Du Y, Wang Q, Zheng Z, Zhou H, Han Y, Qi A, Jiao L, Gong Y. Gut microbiota influence on lung cancer risk through blood metabolite mediation: from a comprehensive Mendelian randomization analysis and genetic analysis. Front Nutr 2024; 11:1425802. [PMID: 39323566 PMCID: PMC11423778 DOI: 10.3389/fnut.2024.1425802] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Accepted: 08/26/2024] [Indexed: 09/27/2024] Open
Abstract
Background Gut microbiota (GM) and metabolic alterations play pivotal roles in lung cancer (LC) development and host genetic variations are known to contribute to LC susceptibility by modulating the GM. However, the causal links among GM, metabolite, host genes, and LC remain to be fully delineated. Method Through bidirectional MR analyses, we examined the causal links between GM and LC, and utilized two-step mediation analysis to identify potential mediating blood metabolite. We employed diverse MR methods, including inverse-variance-weighted (IVW), weighted median, MR-Egger, weighted mode, and simple mode, to ensure a robust examination of the data. MR-Egger intercept test, Radial MR, MR-PRESSO, Cochran Q test and Leave-one-out (LOO) analysis were used for sensitivity analyses. Analyses were adjusted for smoking, alcohol intake frequency and air pollution. Linkage disequilibrium score regression and Steiger test were used to probe genetic causality. The study also explored the association between specific host genes and the abundance of gut microbes in LC patients. Results The presence of Bacteroides clarus was associated with an increased risk of LC (odds ratio [OR] = 1.07, 95% confidence interval [CI]: 1.03-1.11, p = 0.012), whereas the Eubacteriaceae showed a protective effect (OR = 0.82, 95% CI: 0.75-0.89, p = 0.001). These findings remained robust after False Discovery Rate (FDR) correction. Our mediator screening identified 13 blood metabolites that significantly influence LC risk after FDR correction, underscoring cystine and propionylcarnitine in reducing LC risk, while linking specific lipids and hydroxy acids to an increased risk. Our two-step mediation analysis demonstrated that the association between the bacterial pathway of synthesis of guanosine ribonucleotides and LC was mediated by Fructosyllysine, with mediated proportions of 11.38% (p = 0.037). LDSC analysis confirmed the robustness of these associations. Our study unveiled significant host genes ROBO2 may influence the abundance of pathogenic gut microbes in LC patients. Metabolic pathway analysis revealed glutathione metabolism and glutamate metabolism are the pathways most enriched with significant metabolites related to LC. Conclusion These findings underscore the importance of GM in the development of LC, with metabolites partly mediating this effect, and provide dietary and lifestyle recommendations for high-risk lung cancer populations.
Collapse
Affiliation(s)
- Yizhao Du
- Department of Oncology, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Qin Wang
- Department of Oncology, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Zongmei Zheng
- Department of Oncology, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Hailun Zhou
- Department of Oncology, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Yang Han
- Department of Oncology, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Ao Qi
- Department of Oncology, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Lijing Jiao
- Department of Oncology, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Institute of Translational Cancer Research for Integrated Chinese and Western Medicine, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Yabin Gong
- Department of Oncology, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| |
Collapse
|
32
|
Mo X, Wang C, Pu Q, Zhang Z, Wu D. Revealing genetic causality between blood-based biomarkers and major depression in east Asian ancestry. Front Psychiatry 2024; 15:1424958. [PMID: 39323965 PMCID: PMC11423294 DOI: 10.3389/fpsyt.2024.1424958] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/29/2024] [Accepted: 08/26/2024] [Indexed: 09/27/2024] Open
Abstract
Introduction Major Depression (MD) is a common mental disorder. In East Asian ancestry, the association, causality, and shared genetic basis between blood-based biomarkers and MD remain unclear. Methods We investigated the relationships between blood-based biomarkers and MD through a cross-sectional study and Mendelian randomization (MR) analysis. Cross-trait analysis and enrichment analyses were used to highlight the shared genetic determinants and biological pathways. We conducted summary data-based MR to identify shared genes, which were then validated using a transcriptome dataset from drug-naïve patients with MD. Results In the cross-sectional study, C-Reactive Protein showed the significantly positive correlation with depressive symptoms, while hematocrit, hemoglobin, and uric acid exhibited significantly negative correlations. In MR analysis, basophil count (BASO) and low-density lipoprotein cholesterol (LDLc) had a significant causal effect on MD. The enrichment analysis indicated a significant role of inflammatory cytokines and oxidative stress. The shared genes MFN2, FAM55C, GCC2, and SCAPER were validated, with MFN2 identified as a pleiotropic gene involved in MD, BASO, and LDLc. Discussion This study highlighted that BASO and LDLc have a causal effect on MD in East Asian ancestry. The pathological mechanisms of MD are related not only to inflammatory cytokines and oxidative stress but also to down regulation of MFN2 expression and mitochondrial dysfunction.
Collapse
Affiliation(s)
- Xiaoxiao Mo
- Department of Genetic Toxicology, The Key Laboratory of Modern Toxicology of Ministry Education, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, China
- Department of Environmental Genomics, School of Public Health, Nanjing Medical University, Nanjing, China
| | - Chao Wang
- Department of Genetic Toxicology, The Key Laboratory of Modern Toxicology of Ministry Education, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, China
- Department of Environmental Genomics, School of Public Health, Nanjing Medical University, Nanjing, China
| | - Qiuyi Pu
- Department of Genetic Toxicology, The Key Laboratory of Modern Toxicology of Ministry Education, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, China
- Department of Environmental Genomics, School of Public Health, Nanjing Medical University, Nanjing, China
| | - Zhengdong Zhang
- Department of Environmental Genomics, School of Public Health, Nanjing Medical University, Nanjing, China
| | - Dongmei Wu
- Department of Genetic Toxicology, The Key Laboratory of Modern Toxicology of Ministry Education, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, China
- Department of Environmental Genomics, School of Public Health, Nanjing Medical University, Nanjing, China
| |
Collapse
|
33
|
Yuan J, Zhuang YY, Liu X, Zhang Y, Li K, Chen ZJ, Li D, Chen H, Liang J, Yao Y, Yu X, Zhuo R, Zhao F, Zhou X, Yu X, Qu J, Su J. Exome-wide association study identifies KDELR3 mutations in extreme myopia. Nat Commun 2024; 15:6703. [PMID: 39112444 PMCID: PMC11306401 DOI: 10.1038/s41467-024-50580-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2023] [Accepted: 07/15/2024] [Indexed: 08/10/2024] Open
Abstract
Extreme myopia (EM), defined as a spherical equivalent (SE) ≤ -10.00 diopters (D), is one of the leading causes of sight impairment. Known EM-associated variants only explain limited risk and are inadequate for clinical decision-making. To discover risk genes, we performed a whole-exome sequencing (WES) on 449 EM individuals and 9606 controls. We find a significant excess of rare protein-truncating variants (PTVs) in EM cases, enriched in the retrograde vesicle-mediated transport pathway. Employing single-cell RNA-sequencing (scRNA-seq) and a single-cell polygenic burden score (scPBS), we pinpointed PI16 + /SFRP4+ fibroblasts as the most relevant cell type. We observed that KDELR3 is highly expressed in scleral fibroblast and involved in scleral extracellular matrix (ECM) organization. The zebrafish model revealed that kdelr3 downregulation leads to elongated ocular axial length and increased lens diameter. Together, our study provides insight into the genetics of EM in humans and highlights KDELR3's role in EM pathogenesis.
Collapse
Affiliation(s)
- Jian Yuan
- National Engineering Research Center of Ophthalmology and Optometry, Eye Hospital, Wenzhou Medical University, Wenzhou, China
- Oujiang Laboratory, Zhejiang Lab for Regenerative Medicine, Vision and Brain Health, Wenzhou, Zhejiang, China
| | - You-Yuan Zhuang
- National Engineering Research Center of Ophthalmology and Optometry, Eye Hospital, Wenzhou Medical University, Wenzhou, China
| | - Xiaoyu Liu
- National Engineering Research Center of Ophthalmology and Optometry, Eye Hospital, Wenzhou Medical University, Wenzhou, China
| | - Yue Zhang
- National Engineering Research Center of Ophthalmology and Optometry, Eye Hospital, Wenzhou Medical University, Wenzhou, China
| | - Kai Li
- Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, China
| | - Zhen Ji Chen
- National Engineering Research Center of Ophthalmology and Optometry, Eye Hospital, Wenzhou Medical University, Wenzhou, China
| | - Dandan Li
- National Engineering Research Center of Ophthalmology and Optometry, Eye Hospital, Wenzhou Medical University, Wenzhou, China
| | - He Chen
- School of Biomedical Engineering, Hainan University, Haikou, China
| | - Jiacheng Liang
- National Engineering Research Center of Ophthalmology and Optometry, Eye Hospital, Wenzhou Medical University, Wenzhou, China
| | - Yinghao Yao
- Oujiang Laboratory, Zhejiang Lab for Regenerative Medicine, Vision and Brain Health, Wenzhou, Zhejiang, China
| | - Xiangyi Yu
- Institute of PSI Genomics, Wenzhou, China
| | - Ran Zhuo
- National Engineering Research Center of Ophthalmology and Optometry, Eye Hospital, Wenzhou Medical University, Wenzhou, China
| | - Fei Zhao
- National Engineering Research Center of Ophthalmology and Optometry, Eye Hospital, Wenzhou Medical University, Wenzhou, China
| | - Xiangtian Zhou
- National Engineering Research Center of Ophthalmology and Optometry, Eye Hospital, Wenzhou Medical University, Wenzhou, China
- Oujiang Laboratory, Zhejiang Lab for Regenerative Medicine, Vision and Brain Health, Wenzhou, Zhejiang, China
| | | | - Jia Qu
- National Engineering Research Center of Ophthalmology and Optometry, Eye Hospital, Wenzhou Medical University, Wenzhou, China.
- Oujiang Laboratory, Zhejiang Lab for Regenerative Medicine, Vision and Brain Health, Wenzhou, Zhejiang, China.
- School of Biomedical Engineering, Hainan University, Haikou, China.
| | - Jianzhong Su
- National Engineering Research Center of Ophthalmology and Optometry, Eye Hospital, Wenzhou Medical University, Wenzhou, China.
- Oujiang Laboratory, Zhejiang Lab for Regenerative Medicine, Vision and Brain Health, Wenzhou, Zhejiang, China.
- Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, China.
| |
Collapse
|
34
|
Yang F, Song H, Tang W, Liu L, Zhu Z, Ouyang B, Zhang L, He G, Qin W. Causal relationship between the gut microbiota, immune cells, and coronary heart disease: a mediated Mendelian randomization analysis. Front Microbiol 2024; 15:1449935. [PMID: 39161605 PMCID: PMC11332803 DOI: 10.3389/fmicb.2024.1449935] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2024] [Accepted: 07/16/2024] [Indexed: 08/21/2024] Open
Abstract
Background Recent studies have shown that the gut microbiota (GM), immune cells, and coronary heart disease (CHD) are closely related, but the causal nature of these relationships is largely unknown. This study aimed to investigate this causal relationship and reveal the effect of GM and immune cells on the risk of developing CHD using mediated Mendelian randomization (MR) analysis. Methods First, we searched for data related to GM, immune cells, and CHD through published genome-wide association studies (GWAS). We filtered the single nucleotide polymorphisms (SNPs) associated with GM and immune cells and then performed the first MR analysis to identify disease-associated intestinal bacteria and disease-associated immune cells. Subsequently, three MR analyses were conducted: from disease-associated GM to disease-associated immune cells, from disease-associated immune cells to CHD, and from disease-associated GM to CHD. Each MR analysis was conducted using inverse variance weighting (IVW), MR-Egger regression, weighted median, weighted models, and simple models. Results A total of six GM and 25 immune cells were found to be associated with CHD. In the MR analysis using the inverse variance weighting (IVW) method, g__Desulfovibrio.s__Desulfovibrio_piger was associated with EM DN (CD4-CD8-) %T cells (P < 0.05 and OR > 1), EM DN (CD4-CD8-) %T cells was associated with CHD (P < 0.05 and OR < 1), and g__Desulfovibrio.s__Desulfovibrio_piger was associated with CHD (P < 0.05 and OR < 1). Conclusion An increase in the abundance of g__Desulfovibrio.s__Desulfovibrio_piger leads to an increase in the amount of EM DN (CD4-CD8-) %T cells, and an increase in the amount of EM DN (CD4-CD8-) %T cells reduces the risk of developing CHD. Our study provides some references for reducing the incidence of CHD by regulating GM and immune cells.
Collapse
Affiliation(s)
- Feifei Yang
- Graduate School of Guangxi University of Chinese Medicine, Nanning, China
| | - Hui Song
- Graduate School of Guangxi University of Chinese Medicine, Nanning, China
| | - Weizhi Tang
- Graduate School of Guangxi University of Chinese Medicine, Nanning, China
| | - Lingyun Liu
- Graduate School of Guangxi University of Chinese Medicine, Nanning, China
| | - Ziyi Zhu
- Graduate School of Guangxi University of Chinese Medicine, Nanning, China
| | - Bin Ouyang
- Graduate School of Guangxi University of Chinese Medicine, Nanning, China
| | - Liwen Zhang
- Graduate School of Guangxi University of Chinese Medicine, Nanning, China
| | - Guixin He
- Department of Cardiovascular Medicine, The First Affiliated Hospital of Guangxi University of Chinese Medicine, Nanning, China
| | - Weibin Qin
- Department of Cardiovascular Medicine, The First Affiliated Hospital of Guangxi University of Chinese Medicine, Nanning, China
| |
Collapse
|
35
|
Shen S, Sobczyk MK, Paternoster L, Brown SJ. From GWASs toward Mechanistic Understanding with Case Studies in Dermatogenetics. J Invest Dermatol 2024; 144:1189-1199.e8. [PMID: 38782533 DOI: 10.1016/j.jid.2024.03.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2023] [Revised: 02/13/2024] [Accepted: 03/06/2024] [Indexed: 05/25/2024]
Abstract
Many human skin diseases result from the complex interplay of genetic and environmental mechanisms that are largely unknown. GWASs have yielded insight into the genetic aspect of complex disease by highlighting regions of the genome or specific genetic variants associated with disease. Leveraging this information to identify causal genes and cell types will provide insight into fundamental biology, inform diagnostics, and aid drug discovery. However, the etiological mechanisms from genetic variant to disease are still unestablished in most cases. There now exists an unprecedented wealth of data and computational methods for variant interpretation in a functional context. It can be challenging to decide where to start owing to a lack of consensus on the best way to identify causal genetic mechanisms. This article highlights 3 key aspects of genetic variant interpretation: prioritizing causal genes, cell types, and pathways. We provide a practical overview of the main methods and datasets, giving examples from recent atopic dermatitis studies to provide a blueprint for variant interpretation. A collection of resources, including brief description and links to the packages and web tools, is provided for researchers looking to start in silico follow-up genetic analysis of associated genetic variants.
Collapse
Affiliation(s)
- Silvia Shen
- Centre for Genomic & Experimental Medicine, Institute of Genetics and Cancer, The University of Edinburgh, Edinburgh, United Kingdom; Institute for Evolution and Ecology, School of Biological Sciences, The University of Edinburgh, Edinburgh, United Kingdom.
| | - Maria K Sobczyk
- MRC Integrative Epidemiology Unit, Bristol Medical School, University of Bristol, Bristol, United Kingdom
| | - Lavinia Paternoster
- MRC Integrative Epidemiology Unit, Bristol Medical School, University of Bristol, Bristol, United Kingdom
| | - Sara J Brown
- Centre for Genomic & Experimental Medicine, Institute of Genetics and Cancer, The University of Edinburgh, Edinburgh, United Kingdom; Department of Dermatology, NHS Lothian, Edinburgh, United Kingdom
| |
Collapse
|
36
|
Dorans E, Jagadeesh K, Dey K, Price AL. Linking regulatory variants to target genes by integrating single-cell multiome methods and genomic distance. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.05.24.24307813. [PMID: 38826240 PMCID: PMC11142273 DOI: 10.1101/2024.05.24.24307813] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/04/2024]
Abstract
Methods that analyze single-cell paired RNA-seq and ATAC-seq multiome data have shown great promise in linking regulatory elements to genes. However, existing methods differ in their modeling assumptions and approaches to account for biological and technical noise-leading to low concordance in their linking scores-and do not capture the effects of genomic distance. We propose pgBoost, an integrative modeling framework that trains a non-linear combination of existing linking strategies (including genomic distance) on fine-mapped eQTL data to assign a probabilistic score to each candidate SNP-gene link. We applied pgBoost to single-cell multiome data from 85k cells representing 6 major immune/blood cell types. pgBoost attained higher enrichment for fine-mapped eSNP-eGene pairs (e.g. 21x at distance >10kb) than existing methods (1.2-10x; p-value for difference = 5e-13 vs. distance-based method and < 4e-35 for each other method), with larger improvements at larger distances (e.g. 35x vs. 0.89-6.6x at distance >100kb; p-value for difference < 0.002 vs. each other method). pgBoost also outperformed existing methods in enrichment for CRISPR-validated links (e.g. 4.8x vs. 1.6-4.1x at distance >10kb; p-value for difference = 0.25 vs. distance-based method and < 2e-5 for each other method), with larger improvements at larger distances (e.g. 15x vs. 1.6-2.5x at distance >100kb; p-value for difference < 0.009 for each other method). Similar improvements in enrichment were observed for links derived from Activity-By-Contact (ABC) scores and GWAS data. We further determined that restricting pgBoost to features from a focal cell type improved the identification of SNP-gene links relevant to that cell type. We highlight several examples where pgBoost linked fine-mapped GWAS variants to experimentally validated or biologically plausible target genes that were not implicated by other methods. In conclusion, a non-linear combination of linking strategies, including genomic distance, improves power to identify target genes underlying GWAS associations.
Collapse
|
37
|
Zhang S, Shu H, Zhou J, Rubin-Sigler J, Yang X, Liu Y, Cooper-Knock J, Monte E, Zhu C, Tu S, Li H, Tong M, Ecker JR, Ichida JK, Shen Y, Zeng J, Tsao PS, Snyder MP. Deconvolution of polygenic risk score in single cells unravels cellular and molecular heterogeneity of complex human diseases. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.05.14.594252. [PMID: 38798507 PMCID: PMC11118500 DOI: 10.1101/2024.05.14.594252] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/29/2024]
Abstract
Polygenic risk scores (PRSs) are commonly used for predicting an individual's genetic risk of complex diseases. Yet, their implication for disease pathogenesis remains largely limited. Here, we introduce scPRS, a geometric deep learning model that constructs single-cell-resolved PRS leveraging reference single-cell chromatin accessibility profiling data to enhance biological discovery as well as disease prediction. Real-world applications across multiple complex diseases, including type 2 diabetes (T2D), hypertrophic cardiomyopathy (HCM), and Alzheimer's disease (AD), showcase the superior prediction power of scPRS compared to traditional PRS methods. Importantly, scPRS not only predicts disease risk but also uncovers disease-relevant cells, such as hormone-high alpha and beta cells for T2D, cardiomyocytes and pericytes for HCM, and astrocytes, microglia and oligodendrocyte progenitor cells for AD. Facilitated by a layered multi-omic analysis, scPRS further identifies cell-type-specific genetic underpinnings, linking disease-associated genetic variants to gene regulation within corresponding cell types. We substantiate the disease relevance of scPRS-prioritized HCM genes and demonstrate that the suppression of these genes in HCM cardiomyocytes is rescued by Mavacamten treatment. Additionally, we establish a novel microglia-specific regulatory relationship between the AD risk variant rs7922621 and its target genes ANXA11 and TSPAN14. We further illustrate the detrimental effects of suppressing these two genes on microglia phagocytosis. Our work provides a multi-tasking, interpretable framework for precise disease prediction and systematic investigation of the genetic, cellular, and molecular basis of complex diseases, laying the methodological foundation for single-cell genetics.
Collapse
Affiliation(s)
- Sai Zhang
- Department of Epidemiology, University of Florida, Gainesville, FL, USA
- Departments of Biostatistics & Biomedical Engineering, Genetics Institute, McKnight Brain Institute, University of Florida, Gainesville, FL, USA
- Department of Genetics, Center for Genomics and Personalized Medicine, Stanford University School of Medicine, Stanford, CA, USA
- These authors contributed equally: Sai Zhang, Hantao Shu, and Jingtian Zhou
| | - Hantao Shu
- Institute for Interdisciplinary Information Sciences, Tsinghua University, Beijing, China
- These authors contributed equally: Sai Zhang, Hantao Shu, and Jingtian Zhou
| | - Jingtian Zhou
- Arc Institute, Palo Alto, CA, USA
- Genomic Analysis Laboratory, The Salk Institute for Biological Studies, La Jolla, CA, USA
- Bioinformatics and Systems Biology Program, University of California San Diego, La Jolla, CA, USA
- These authors contributed equally: Sai Zhang, Hantao Shu, and Jingtian Zhou
| | - Jasper Rubin-Sigler
- Department of Stem Cell Biology and Regenerative Medicine, Eli and Edythe Broad Center for Regenerative Medicine and Stem Cell Research, University of Southern California, Los Angeles, CA, USA
| | - Xiaoyu Yang
- Institute for Human Genetics, University of California San Francisco, San Francisco, CA, USA
| | - Yuxi Liu
- Institute for Human Genetics, University of California San Francisco, San Francisco, CA, USA
| | - Johnathan Cooper-Knock
- Sheffield Institute for Translational Neuroscience, University of Sheffield, Sheffield, UK
| | - Emma Monte
- Department of Genetics, Center for Genomics and Personalized Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Chenchen Zhu
- Department of Genetics, Center for Genomics and Personalized Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Sharon Tu
- Department of Stem Cell Biology and Regenerative Medicine, Eli and Edythe Broad Center for Regenerative Medicine and Stem Cell Research, University of Southern California, Los Angeles, CA, USA
| | - Han Li
- Institute for Interdisciplinary Information Sciences, Tsinghua University, Beijing, China
| | - Mingming Tong
- Department of Genetics, Center for Genomics and Personalized Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Joseph R. Ecker
- Genomic Analysis Laboratory, The Salk Institute for Biological Studies, La Jolla, CA, USA
- Howard Hughes Medical Institute, The Salk Institute for Biological Studies, La Jolla, CA, USA
| | - Justin K. Ichida
- Department of Stem Cell Biology and Regenerative Medicine, Eli and Edythe Broad Center for Regenerative Medicine and Stem Cell Research, University of Southern California, Los Angeles, CA, USA
| | - Yin Shen
- Institute for Human Genetics, University of California San Francisco, San Francisco, CA, USA
- Department of Neurology, Weill Institute for Neurosciences, University of California San Francisco, San Francisco, CA, USA
| | - Jianyang Zeng
- School of Engineering, Research Center for Industries of the Future, Westlake University, Hangzhou, Zhejiang, China
| | - Philip S. Tsao
- VA Palo Alto Healthcare System, Palo Alto, CA, USA
- Department of Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Michael P. Snyder
- Department of Genetics, Center for Genomics and Personalized Medicine, Stanford University School of Medicine, Stanford, CA, USA
| |
Collapse
|
38
|
Ma Y, Zhou Y, Jiang D, Dai W, Li J, Deng C, Chen C, Zheng G, Zhang Y, Qiu F, Sun H, Xing S, Han H, Qu J, Wu N, Yao Y, Su J. Integration of human organoids single-cell transcriptomic profiles and human genetics repurposes critical cell type-specific drug targets for severe COVID-19. Cell Prolif 2024; 57:e13558. [PMID: 37807299 PMCID: PMC10905359 DOI: 10.1111/cpr.13558] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2023] [Revised: 08/31/2023] [Accepted: 09/18/2023] [Indexed: 10/10/2023] Open
Abstract
Human organoids recapitulate the cell type diversity and function of their primary organs holding tremendous potentials for basic and translational research. Advances in single-cell RNA sequencing (scRNA-seq) technology and genome-wide association study (GWAS) have accelerated the biological and therapeutic interpretation of trait-relevant cell types or states. Here, we constructed a computational framework to integrate atlas-level organoid scRNA-seq data, GWAS summary statistics, expression quantitative trait loci, and gene-drug interaction data for distinguishing critical cell populations and drug targets relevant to coronavirus disease 2019 (COVID-19) severity. We found that 39 cell types across eight kinds of organoids were significantly associated with COVID-19 outcomes. Notably, subset of lung mesenchymal stem cells increased proximity with fibroblasts predisposed to repair COVID-19-damaged lung tissue. Brain endothelial cell subset exhibited significant associations with severe COVID-19, and this cell subset showed a notable increase in cell-to-cell interactions with other brain cell types, including microglia. We repurposed 33 druggable genes, including IFNAR2, TYK2, and VIPR2, and their interacting drugs for COVID-19 in a cell-type-specific manner. Overall, our results showcase that host genetic determinants have cellular-specific contribution to COVID-19 severity, and identification of cell type-specific drug targets may facilitate to develop effective therapeutics for treating severe COVID-19 and its complications.
Collapse
Affiliation(s)
- Yunlong Ma
- National Engineering Research Center of Ophthalmology and Optometry, Eye Hospital, Wenzhou Medical University, Wenzhou, China
- Department of Biomedical Informatics, Institute of Biomedical Big Data, Wenzhou Medical University, Wenzhou, China
- Oujiang Laboratory, Zhejiang Lab for Regenerative Medicine, Vision and Brain Health, Zhejiang, China
| | - Yijun Zhou
- National Engineering Research Center of Ophthalmology and Optometry, Eye Hospital, Wenzhou Medical University, Wenzhou, China
- Department of Biomedical Informatics, Institute of Biomedical Big Data, Wenzhou Medical University, Wenzhou, China
| | - Dingping Jiang
- National Engineering Research Center of Ophthalmology and Optometry, Eye Hospital, Wenzhou Medical University, Wenzhou, China
- Oujiang Laboratory, Zhejiang Lab for Regenerative Medicine, Vision and Brain Health, Zhejiang, China
| | - Wei Dai
- Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, China
| | - Jingjing Li
- National Engineering Research Center of Ophthalmology and Optometry, Eye Hospital, Wenzhou Medical University, Wenzhou, China
- Department of Biomedical Informatics, Institute of Biomedical Big Data, Wenzhou Medical University, Wenzhou, China
| | - Chunyu Deng
- School of Life Science and Technology, Harbin Institute of Technology, Harbin, China
| | - Cheng Chen
- National Engineering Research Center of Ophthalmology and Optometry, Eye Hospital, Wenzhou Medical University, Wenzhou, China
- Department of Biomedical Informatics, Institute of Biomedical Big Data, Wenzhou Medical University, Wenzhou, China
| | - Gongwei Zheng
- National Engineering Research Center of Ophthalmology and Optometry, Eye Hospital, Wenzhou Medical University, Wenzhou, China
- Department of Biomedical Informatics, Institute of Biomedical Big Data, Wenzhou Medical University, Wenzhou, China
| | - Yaru Zhang
- National Engineering Research Center of Ophthalmology and Optometry, Eye Hospital, Wenzhou Medical University, Wenzhou, China
- Department of Biomedical Informatics, Institute of Biomedical Big Data, Wenzhou Medical University, Wenzhou, China
- Oujiang Laboratory, Zhejiang Lab for Regenerative Medicine, Vision and Brain Health, Zhejiang, China
| | - Fei Qiu
- National Engineering Research Center of Ophthalmology and Optometry, Eye Hospital, Wenzhou Medical University, Wenzhou, China
- Department of Biomedical Informatics, Institute of Biomedical Big Data, Wenzhou Medical University, Wenzhou, China
| | - Haojun Sun
- National Engineering Research Center of Ophthalmology and Optometry, Eye Hospital, Wenzhou Medical University, Wenzhou, China
- Department of Biomedical Informatics, Institute of Biomedical Big Data, Wenzhou Medical University, Wenzhou, China
| | - Shilai Xing
- National Engineering Research Center of Ophthalmology and Optometry, Eye Hospital, Wenzhou Medical University, Wenzhou, China
- Department of Biomedical Informatics, Institute of Biomedical Big Data, Wenzhou Medical University, Wenzhou, China
| | - Haijun Han
- School of Medicine, Hangzhou City University, Hangzhou, China
| | - Jia Qu
- National Engineering Research Center of Ophthalmology and Optometry, Eye Hospital, Wenzhou Medical University, Wenzhou, China
| | - Nan Wu
- Beijing Key Laboratory for Genetic Research of Skeletal Deformity, Key Laboratory of Big Data for Spinal Deformities, Department of Orthopedic Surgery, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
| | - Yinghao Yao
- Oujiang Laboratory, Zhejiang Lab for Regenerative Medicine, Vision and Brain Health, Zhejiang, China
| | - Jianzhong Su
- National Engineering Research Center of Ophthalmology and Optometry, Eye Hospital, Wenzhou Medical University, Wenzhou, China
- Department of Biomedical Informatics, Institute of Biomedical Big Data, Wenzhou Medical University, Wenzhou, China
- Oujiang Laboratory, Zhejiang Lab for Regenerative Medicine, Vision and Brain Health, Zhejiang, China
| |
Collapse
|
39
|
Hamel AR, Yan W, Rouhana JM, Monovarfeshani A, Jiang X, Mehta PA, Advani J, Luo Y, Liang Q, Rajasundaram S, Shrivastava A, Duchinski K, Mantena S, Wang J, van Zyl T, Pasquale LR, Swaroop A, Gharahkhani P, Khawaja AP, MacGregor S, Chen R, Vitart V, Sanes JR, Wiggs JL, Segrè AV. Integrating genetic regulation and single-cell expression with GWAS prioritizes causal genes and cell types for glaucoma. Nat Commun 2024; 15:396. [PMID: 38195602 PMCID: PMC10776627 DOI: 10.1038/s41467-023-44380-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2022] [Accepted: 12/12/2023] [Indexed: 01/11/2024] Open
Abstract
Primary open-angle glaucoma (POAG), characterized by retinal ganglion cell death, is a leading cause of irreversible blindness worldwide. However, its molecular and cellular causes are not well understood. Elevated intraocular pressure (IOP) is a major risk factor, but many patients have normal IOP. Colocalization and Mendelian randomization analysis of >240 POAG and IOP genome-wide association study (GWAS) loci and overlapping expression and splicing quantitative trait loci (e/sQTLs) in 49 GTEx tissues and retina prioritizes causal genes for 60% of loci. These genes are enriched in pathways implicated in extracellular matrix organization, cell adhesion, and vascular development. Analysis of single-nucleus RNA-seq of glaucoma-relevant eye tissues reveals that the POAG and IOP colocalizing genes and genome-wide associations are enriched in specific cell types in the aqueous outflow pathways, retina, optic nerve head, peripapillary sclera, and choroid. This study nominates IOP-dependent and independent regulatory mechanisms, genes, and cell types that may contribute to POAG pathogenesis.
Collapse
Affiliation(s)
- Andrew R Hamel
- Ocular Genomics Institute, Department of Ophthalmology, Massachusetts Eye and Ear, Boston, MA, USA
- Department of Ophthalmology, Harvard Medical School, Boston, MA, USA
- Broad Institute of Harvard and MIT, Cambridge, MA, USA
| | - Wenjun Yan
- Department of Molecular and Cellular Biology and Center for Brain Science, Harvard University, Cambridge, MA, USA
| | - John M Rouhana
- Ocular Genomics Institute, Department of Ophthalmology, Massachusetts Eye and Ear, Boston, MA, USA
- Department of Ophthalmology, Harvard Medical School, Boston, MA, USA
- Broad Institute of Harvard and MIT, Cambridge, MA, USA
| | - Aboozar Monovarfeshani
- Department of Molecular and Cellular Biology and Center for Brain Science, Harvard University, Cambridge, MA, USA
| | - Xinyi Jiang
- MRC Human Genetics Unit, Institute of Genetics and Cancer, The University of Edinburgh, Edinburgh, UK
- Centre for Genomic and Experimental Medicine, Institute of Genetics and Molecular Medicine, The University of Edinburgh, Edinburgh, UK
| | - Puja A Mehta
- Ocular Genomics Institute, Department of Ophthalmology, Massachusetts Eye and Ear, Boston, MA, USA
- Department of Ophthalmology, Harvard Medical School, Boston, MA, USA
- Broad Institute of Harvard and MIT, Cambridge, MA, USA
| | - Jayshree Advani
- Neurobiology, Neurodegeneration and Repair Laboratory, National Eye Institute, National Institutes of Health, Bethesda, MA, USA
| | - Yuyang Luo
- Ocular Genomics Institute, Department of Ophthalmology, Massachusetts Eye and Ear, Boston, MA, USA
- Department of Ophthalmology, Harvard Medical School, Boston, MA, USA
- Broad Institute of Harvard and MIT, Cambridge, MA, USA
| | - Qingnan Liang
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA
| | - Skanda Rajasundaram
- Department of Ophthalmology, Harvard Medical School, Boston, MA, USA
- Centre for Evidence-Based Medicine, University of Oxford, Oxford, UK
- Faculty of Medicine, Imperial College London, London, UK
| | - Arushi Shrivastava
- Ocular Genomics Institute, Department of Ophthalmology, Massachusetts Eye and Ear, Boston, MA, USA
- Department of Ophthalmology, Harvard Medical School, Boston, MA, USA
- Broad Institute of Harvard and MIT, Cambridge, MA, USA
| | - Katherine Duchinski
- Ocular Genomics Institute, Department of Ophthalmology, Massachusetts Eye and Ear, Boston, MA, USA
- Department of Ophthalmology, Harvard Medical School, Boston, MA, USA
- Broad Institute of Harvard and MIT, Cambridge, MA, USA
- Bioinformatics and Integrative Genomics (BIG) PhD Program, Harvard Medical School, Boston, MA, USA
| | - Sreekar Mantena
- Ocular Genomics Institute, Department of Ophthalmology, Massachusetts Eye and Ear, Boston, MA, USA
- Harvard/MIT MD-PhD Program, Harvard Medical School, Boston, MA, USA
| | - Jiali Wang
- Ocular Genomics Institute, Department of Ophthalmology, Massachusetts Eye and Ear, Boston, MA, USA
- Department of Ophthalmology, Harvard Medical School, Boston, MA, USA
- Broad Institute of Harvard and MIT, Cambridge, MA, USA
| | - Tavé van Zyl
- Department of Molecular and Cellular Biology and Center for Brain Science, Harvard University, Cambridge, MA, USA
- Department of Ophthalmology and Visual Sciences, Yale School of Medicine, New Haven, CT, USA
| | - Louis R Pasquale
- Department of Ophthalmology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Anand Swaroop
- Neurobiology, Neurodegeneration and Repair Laboratory, National Eye Institute, National Institutes of Health, Bethesda, MA, USA
| | - Puya Gharahkhani
- QIMR Berghofer Medical Research Institute, Brisbane, QLD, 4029, Australia
| | - Anthony P Khawaja
- NIHR Biomedical Research Centre, Moorfields Eye Hospital NHS Foundation Trust and UCL Institute of Ophthalmology, London, UK
| | - Stuart MacGregor
- QIMR Berghofer Medical Research Institute, Brisbane, QLD, 4029, Australia
| | - Rui Chen
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA
| | - Veronique Vitart
- MRC Human Genetics Unit, Institute of Genetics and Cancer, The University of Edinburgh, Edinburgh, UK
| | - Joshua R Sanes
- Department of Molecular and Cellular Biology and Center for Brain Science, Harvard University, Cambridge, MA, USA
| | - Janey L Wiggs
- Ocular Genomics Institute, Department of Ophthalmology, Massachusetts Eye and Ear, Boston, MA, USA
- Department of Ophthalmology, Harvard Medical School, Boston, MA, USA
- Broad Institute of Harvard and MIT, Cambridge, MA, USA
| | - Ayellet V Segrè
- Ocular Genomics Institute, Department of Ophthalmology, Massachusetts Eye and Ear, Boston, MA, USA.
- Department of Ophthalmology, Harvard Medical School, Boston, MA, USA.
- Broad Institute of Harvard and MIT, Cambridge, MA, USA.
| |
Collapse
|
40
|
Chen L, Mou X, Li J, Li M, Ye C, Gao X, Liu X, Ma Y, Xu Y, Zhong Y. Alterations in gut microbiota and host transcriptome of patients with coronary artery disease. BMC Microbiol 2023; 23:320. [PMID: 37924005 PMCID: PMC10623719 DOI: 10.1186/s12866-023-03071-w] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2023] [Accepted: 10/16/2023] [Indexed: 11/06/2023] Open
Abstract
BACKGROUND Coronary artery disease (CAD) is a widespread heart condition caused by atherosclerosis and influences millions of people worldwide. Early detection of CAD is challenging due to the lack of specific biomarkers. The gut microbiota and host-microbiota interactions have been well documented to affect human health. However, investigation that reveals the role of gut microbes in CAD is still limited. This study aims to uncover the synergistic effects of host genes and gut microbes associated with CAD through integrative genomic analyses. RESULTS Herein, we collected 52 fecal and 50 blood samples from CAD patients and matched controls, and performed amplicon and transcriptomic sequencing on these samples, respectively. By comparing CAD patients with health controls, we found that dysregulated gut microbes were significantly associated with CAD. By leveraging the Random Forest method, we found that combining 20 bacteria and 30 gene biomarkers could distinguish CAD patients from health controls with a high performance (AUC = 0.92). We observed that there existed prominent associations of gut microbes with several clinical indices relevant to heart functions. Integration analysis revealed that CAD-relevant gut microbe genus Fusicatenibacter was associated with expression of CAD-risk genes, such as GBP2, MLKL, and CPR65, which is in line with previous evidence (Tang et al., Nat Rev Cardiol 16:137-154, 2019; Kummen et al., J Am Coll Cardiol 71:1184-1186, 2018). In addition, the upregulation of immune-related pathways in CAD patients were identified to be primarily associated with higher abundance of genus Blautia, Eubacterium, Fusicatenibacter, and Monoglobus. CONCLUSIONS Our results highlight that dysregulated gut microbes contribute risk to CAD by interacting with host genes. These identified microbes and interacted risk genes may have high potentials as biomarkers for CAD.
Collapse
Affiliation(s)
- Liuying Chen
- Department of Cardiology, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Xuanting Mou
- Department of Cardiology, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Jingjing Li
- National Engineering Research Center of Ophthalmology and Optometry, Eye Hospital, Wenzhou Medical University, Wenzhou, 325027, China
| | - Miaofu Li
- Department of Cardiology, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Caijie Ye
- Zhejiang Chinese Medical University, Hangzhou, China
| | - Xiaofei Gao
- Department of Cardiology, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Xiaohua Liu
- Department of Cardiology, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Yunlong Ma
- National Engineering Research Center of Ophthalmology and Optometry, Eye Hospital, Wenzhou Medical University, Wenzhou, 325027, China.
- Department of Biomedical Informatics, Institute of Biomedical Big Data, Wenzhou Medical University, Wenzhou, 325027, China.
- Oujiang Laboratory, Zhejiang Lab for Regenerative Medicine, Vision and Brain Health, Wenzhou, 325101, Zhejiang, China.
| | - Yizhou Xu
- Department of Cardiology, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou, China.
| | - Yigang Zhong
- Department of Cardiology, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou, China.
| |
Collapse
|